1
|
Corridore S, Verreault M, Martin H, Delobel T, Carrère C, Idbaih A, Ballesta A. Circumventing glioblastoma resistance to temozolomide through optimal drug combinations designed by systems pharmacology and machine learning. Br J Pharmacol 2025. [PMID: 40229949 DOI: 10.1111/bph.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND AND PURPOSE Glioblastoma (GBM), the most frequent and aggressive brain tumour in adults, is associated with a dismal prognostic despite intensive treatment involving surgery, radiotherapy and temozolomide (TMZ)-based chemotherapy. The initial or acquired resistance of GBM to TMZ appeals for precision medicine approaches to the design of novel efficient combination pharmacotherapies. Such investigation needs to account for the overexpression of the O6-methylguanine-DNA methyl-transferase (MGMT) repair enzyme which is responsible for TMZ resistance in patients. EXPERIMENTAL APPROACH A comprehensive approach combining quantitative systems pharmacology (QSP) models and machine learning (ML) was undertaken to design TMZ-based drug combinations circumventing the initial resistance to the alkylating agent. KEY RESULTS A QSP model representing TMZ cellular pharmacokinetics-pharmacodynamics and dysregulated pathways in GBM was developed and validated using multi-type time- and dose-resolved datasets, available in control or MGMT-overexpressing cells. In silico drug screening and subsequent experimental validation identified a strategy to re-sensitise TMZ-resistant cells consisting in combining TMZ with inhibitors of the base excision repair and of homologous recombination. Using ML, functional signatures of response to such optimal multi-agent therapy were derived to assist decision-making in patients. CONCLUSION AND IMPLICATIONS We successfully demonstrated the relevance of combined QSP and ML to design efficient drug combinations re-sensitising glioblastoma cells initially resistant to TMZ. The developed framework may further serve to identify personalised therapies and administration schedules by extending it to account for additional patient-specific altered pathways and whole-body features.
Collapse
Affiliation(s)
- Sergio Corridore
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
| | - Maïté Verreault
- AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, Paris, France
| | - Hugo Martin
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
- University of Rennes, EHESP, CNRS, Inserm, Arènes - UMR 6051, RSMS - U 1309, Rennes, France
| | - Thibault Delobel
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
| | - Cécile Carrère
- Institut Denis Poisson, Université d'Orléans, CNRS, Orléans, France
| | - Ahmed Idbaih
- AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, Paris, France
| | - Annabelle Ballesta
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
| |
Collapse
|
2
|
Abstract
Background: Cell cycle is critical for a wide range of cellular processes such as proliferation, differentiation and apoptosis in dividing cells. Neurons are postmitotic cells which have withdrawn from the cell division cycle. Recent data show us that inappropriate activation of cell cycle regulators including cyclins, cyclin dependent kinases (CDKs) and endogenous cyclin dependent kinase inhibitors (CDKIs) may take part in the aetiology of neurodegenerative diseases. However, the mechanisms for cell cycle reentry in neurodegenerative disease remain unclear.Methods: Electronic databases such as Pubmed, Science Direct, Directory of Open Access Journals, PLOS were searched for relevant articles.Conclusion: The present work reviews basic aspects of cell cycle mechanism, as well as the evidence showing the expression of cell cycle proteins in neurodegenerative disease. We provide a brief summary of these findings and hope to highlight the interaction between the cell cycle reentry and neurodegenerative diseases. Moreover, we outline the possible signaling pathways. However more understanding of the mechanism of cell cycle is of great importance. Because these represents an alternative target for therapeutic interventions, leading to novel treatments of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuxin Song
- School of Integrated Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenpeng Peng
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Radwan RR, Mohamed HA. Mechanistic approach of the therapeutic potential of mesenchymal stem cells on brain damage in irradiated mice: emphasis on anti-inflammatory and anti-apoptotic effects. Int J Radiat Biol 2023; 99:1463-1472. [PMID: 35647928 DOI: 10.1080/09553002.2022.2084170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/04/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Brain damage which has been induced by radiation generally occurs in radiotherapeutics patients. Stem cell transplantation represents a vital applicant for alleviating neurodegenerative disorders. This work aims at exploring the potential of bone marrow-derived mesenchymal stem cells (BM-MSCs) on brain injury induced by γ radiation in mice and the possible underlying mechanisms were elucidated. MATERIALS AND METHODS Mice were allocated into three groups; Group I (Control), Group II (Irradiated control) where mice submitted to 5 Gy of whole-body γ radiation, Group III (Irradiated + BM-MSCs) where mice were intravenously injected of BM-MSCs at a dose of 106 cells/mice 24 h following irradiation. Animals were sacrificed 28 d following exposure to γ radiation. RESULTS It was observed that BM-MSCs therapy provided a valuable tissue repair as evidenced by a reduction in inflammatory mediators including tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), nuclear factor kappa (NF-κβ), phosphorylated NF-κβ-p65 (P-NF-κβ-p65), interferon-gamma (IFNγ) and monocyte chemoattractant protein-1 (MCP-1) associated with decreased levels of transforming growth factor-β (TGF-β) and vascular endothelial growth factor (VEGF) in brain tissues of irradiated mice. Furthermore, neuronal apoptosis was declined in brain tissues of the BM-MSCs group as remarkable inhibition of caspase-3 and Bax accompanied by elevation of Bcl-2 proteins expression. These results were supported by histopathological investigation. CONCLUSIONS In conclusion, BM-MSCs could display a vital rule in alleviating brain injury in radio-therapeutic patients.
Collapse
Affiliation(s)
- Rasha R Radwan
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Egypt
| | - Heba A Mohamed
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Egypt
| |
Collapse
|
4
|
Mathematical Modelling of p53 Signalling during DNA Damage Response: A Survey. Int J Mol Sci 2021; 22:ijms221910590. [PMID: 34638930 PMCID: PMC8508851 DOI: 10.3390/ijms221910590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/14/2021] [Accepted: 09/26/2021] [Indexed: 02/05/2023] Open
Abstract
No gene has garnered more interest than p53 since its discovery over 40 years ago. In the last two decades, thanks to seminal work from Uri Alon and Ghalit Lahav, p53 has defined a truly synergistic topic in the field of mathematical biology, with a rich body of research connecting mathematic endeavour with experimental design and data. In this review we survey and distill the extensive literature of mathematical models of p53. Specifically, we focus on models which seek to reproduce the oscillatory dynamics of p53 in response to DNA damage. We review the standard modelling approaches used in the field categorising them into three types: time delay models, spatial models and coupled negative-positive feedback models, providing sample model equations and simulation results which show clear oscillatory dynamics. We discuss the interplay between mathematics and biology and show how one informs the other; the deep connections between the two disciplines has helped to develop our understanding of this complex gene and paint a picture of its dynamical response. Although yet more is to be elucidated, we offer the current state-of-the-art understanding of p53 response to DNA damage.
Collapse
|
5
|
Huang Y, Mei X, Jiang W, Zhao H, Yan Z, Zhang H, Liu Y, Hu X, Zhang J, Peng W, Zhang J, Qi Q, Chen N. Mesenchymal Stem Cell-Conditioned Medium Protects Hippocampal Neurons From Radiation Damage by Suppressing Oxidative Stress and Apoptosis. Dose Response 2021; 19:1559325820984944. [PMID: 33716588 PMCID: PMC7923989 DOI: 10.1177/1559325820984944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 11/15/2022] Open
Abstract
Objective To investigate the effects of mesenchymal stem cell-conditioned medium (MSC-CM) on radiation-induced oxidative stress, survival and apoptosis in hippocampal neurons. Methods The following groups were defined: Control, radiation treatment (RT), RT+MSC-CM, MSC-CM, RT + N-Acetylcysteine (RT+NAC), and RT + MSC-CM + PI3 K inhibitor (LY294002). A cell Counting Kit-8 (CCK-8) was used to measure cell proliferation. Apoptosis was examined by AnnexinV/PI flow cytometric analyses. Intracellular reactive oxygen species (ROS) were detected by DCFH-DA. Intracellular glutathione (GSH), malondialdehyde (MDA) content, and superoxide dismutase (SOD) activity were detected by colorimetric assays. Protein levels of γ-H2AX, PI3K-AKT, P53, cleaved caspase-3, Bax, and BCl-2 were analyzed by Western blotting. Results The proliferation of HT22 cells was significantly inhibited in the RT group, but was significantly preserved in the RT + MSC-CM group (P < 0.01). Apoptosis was significantly higher in the RT group than in the RT+ MSC-CM group (P < 0.01). MSC-CM decreased intracellular ROS and MDA content after irradiation (P < 0.01). GSH level and SOD activity were higher in the RT + MSC-CM group than in the RT group, as was MMP (P < 0.01). MSC-CM decreased expression of γ-H2AX, P53, Bax, and cleaved-caspase-3, but increased Bcl-2 expression (P < 0.01). Conclusion MSC-CM attenuated radiation-induced hippocampal neuron cell line damage by alleviating oxidative stress and suppressing apoptosis.
Collapse
Affiliation(s)
- Yue Huang
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Xiaolong Mei
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Weishi Jiang
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Hui Zhao
- Tianjin Key Laboratory of Food and Biotechnology, State Experimental and Training Centre of Food and Drug, School of Biotechnology and Food Science, Tianjin University of Commerce, Beichen, Tianjin, China
| | - Zhenyu Yan
- Department of Hematology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Haixia Zhang
- Department of Hematology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Ying Liu
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Xia Hu
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Jingyi Zhang
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Wenshuo Peng
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Jing Zhang
- The Third Central Hospital of Tianjin, Hedong District, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Qingling Qi
- Department of Anesthesiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Naiyao Chen
- Department of Hematology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| |
Collapse
|
6
|
Moiseeva O, Guillon J, Ferbeyre G. Senescence: A program in the road to cell elimination and cancer. Semin Cancer Biol 2020; 81:48-53. [DOI: 10.1016/j.semcancer.2020.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/13/2020] [Accepted: 12/20/2020] [Indexed: 02/08/2023]
|
7
|
Zhang Y, Liu H, Li Z, Miao Z, Zhou J. Oscillatory Dynamics of p53-Mdm2 Circuit in Response to DNA Damage Caused by Ionizing Radiation. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1703-1713. [PMID: 30762566 DOI: 10.1109/tcbb.2019.2899574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Although the dynamical behavior of the p53-Mdm2 loop has been extensively studied, the understanding of the mechanism underlying the regulation of this pathway still remains limited. Herein, we developed an integrated model with five basic components and three ubiquitous time delays for the p53-Mdm2 interaction in response to DNA damage following ionizing radiation (IR). We showed that a sufficient amount of activated ATM level can initiate the p53 oscillations with nearly the same amplitude over a wide range of the ATM level; a proper range of p53 level is also required for generating the oscillations, for too high or too low levels it would fail to generate the oscillations; and increased Mdm2 level leads to decreased amplitude of the p53 oscillation and reduced expression of the p53 activity. Moreover, we found that the negative feedback loop formed between p53 and nuclear Mdm2 plays a dominant role in determining the p53 dynamics, whereas when interaction strength of the negative feedback loop becomes weaker, the positive feedback loop formed between p53 and cytoplasmatic Mdm2 can induce different types of dynamics. Furthermore, we demonstrated that the total time delay required for protein production and nuclear translocation of Mdm2 can induce p53 oscillations even when the p53 level is at a certain stable high steady state or at a certain stable low steady state. In addition, the two important features of the oscillatory dynamics-amplitude and period-can be controlled by such time delay. These results are in agreement with multiple experimental observations and may enrich our understanding of the dynamics of the p53 network.
Collapse
|
8
|
Tojyo I, Shintani Y, Nakanishi T, Okamoto K, Hiraishi Y, Fujita S, Enaka M, Sato F, Muragaki Y. PD-L1 expression correlated with p53 expression in oral squamous cell carcinoma. Maxillofac Plast Reconstr Surg 2019; 41:56. [PMID: 31857991 PMCID: PMC6892985 DOI: 10.1186/s40902-019-0239-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
Background Programmed cell death ligand 1 (PD-L1) is an immune checkpoint molecule that attenuates the immune response. PD-L1 contributes to failed antitumor immunity; thereby, blockade of PD-L1 with monoclonal antibody enhances the immune response. Recently, it was reported that PD-L1 was regulated by protein 53 (p53). Besides, cytokeratin 17 (CK17) is thought to be a diagnostic marker of oral squamous cell carcinoma (OSCC). Our aim was to evaluate the correlation between the immunohistochemical expression of PD-L1, p53 and CK17 with clinicopathological characteristics and disease-specific survival in patients with OSCC. Methods A total of 48 patients with OSCC were included in this study. Immunohistochemical staining was performed to evaluate the correlation among the expressions of PD-L1, p53 and CK17, and furthermore the correlation among various clinicopathological factors, PD-L1, p53 and CK17. Results The positive rate of p53, CK17, PD-L1 (tumor cells) and PD-L1 (tumor-infiltrating lymphocytes) was 63.2%, 91.7%, 48.9% and 57.1%. A statistically significant correlation between p53 expression and T stage and TNM stage (p = 0.049, p = 0.03, respectively) was observed. Also, a statistically significant correlation between p53 and PD-L1 (TCs) expression (p = 0.0009) was observed. Five-year disease-specific survival rate was not significantly correlated with gender, TNM stage, p53 expression, PD-L1 expression and CK17 expression. Conclusion The expression of p53 and PD-L1 shows significantly positive correlation in oral squamous cell carcinoma in tumor cells. Also, a significant correlation between p53 expression and T stage and TNM stage was observed. No other significant correlation between PD-L1 staining or CK17 and clinical or pathologic characteristics was identified.
Collapse
Affiliation(s)
- Itaru Tojyo
- 1Department of Oral and Maxillofacial Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| | - Yukari Shintani
- 1Department of Oral and Maxillofacial Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| | - Takashi Nakanishi
- 1Department of Oral and Maxillofacial Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| | - Kenjiro Okamoto
- 1Department of Oral and Maxillofacial Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| | - Yukihiro Hiraishi
- 2Department of Dentistry and Oral Surgery, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubara-dori, Wakayama, Wakayama 640-8558 Japan
| | - Shigeyuki Fujita
- 1Department of Oral and Maxillofacial Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| | - Mayu Enaka
- 3Department of Pathology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| | - Fuyuki Sato
- 3Department of Pathology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| | - Yasuteru Muragaki
- 3Department of Pathology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-8509 Japan
| |
Collapse
|
9
|
Kim E, Kim JY, Lee JY. Mathematical Modeling of p53 Pathways. Int J Mol Sci 2019; 20:ijms20205179. [PMID: 31635420 PMCID: PMC6834204 DOI: 10.3390/ijms20205179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cells have evolved balanced systems that ensure an appropriate response to stress. The systems elicit repair responses in temporary or moderate stress but eliminate irreparable cells via apoptosis in detrimental conditions of prolonged or severe stress. The tumor suppressor p53 is a central player in these stress response systems. When activated under DNA damage stress, p53 regulates hundreds of genes that are involved in DNA repair, cell cycle, and apoptosis. Recently, increasing studies have demonstrated additional regulatory roles of p53 in metabolism and mitochondrial physiology. Due to the inherent complexity of feedback loops between p53 and its target genes, the application of mathematical modeling has emerged as a novel approach to better understand the multifaceted functions and dynamics of p53. In this review, we discuss several mathematical modeling approaches in exploring the p53 pathways.
Collapse
Affiliation(s)
- Eunjung Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
- Korea Basic Science Institute, Daejeon 34133, Korea.
| | - Joo-Yong Lee
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
- Korea Basic Science Institute, Daejeon 34133, Korea.
| |
Collapse
|
10
|
Deist TM, Patti A, Wang Z, Krane D, Sorenson T, Craft D. Simulation-assisted machine learning. Bioinformatics 2019; 35:4072-4080. [PMID: 30903692 PMCID: PMC6792064 DOI: 10.1093/bioinformatics/btz199] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 11/15/2018] [Accepted: 03/21/2019] [Indexed: 01/02/2023] Open
Abstract
MOTIVATION In a predictive modeling setting, if sufficient details of the system behavior are known, one can build and use a simulation for making predictions. When sufficient system details are not known, one typically turns to machine learning, which builds a black-box model of the system using a large dataset of input sample features and outputs. We consider a setting which is between these two extremes: some details of the system mechanics are known but not enough for creating simulations that can be used to make high quality predictions. In this context we propose using approximate simulations to build a kernel for use in kernelized machine learning methods, such as support vector machines. The results of multiple simulations (under various uncertainty scenarios) are used to compute similarity measures between every pair of samples: sample pairs are given a high similarity score if they behave similarly under a wide range of simulation parameters. These similarity values, rather than the original high dimensional feature data, are used to build the kernel. RESULTS We demonstrate and explore the simulation-based kernel (SimKern) concept using four synthetic complex systems-three biologically inspired models and one network flow optimization model. We show that, when the number of training samples is small compared to the number of features, the SimKern approach dominates over no-prior-knowledge methods. This approach should be applicable in all disciplines where predictive models are sought and informative yet approximate simulations are available. AVAILABILITY AND IMPLEMENTATION The Python SimKern software, the demonstration models (in MATLAB, R), and the datasets are available at https://github.com/davidcraft/SimKern. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Timo M Deist
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- The D-Lab: Decision Support for Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht ER, The Netherlands
| | - Andrew Patti
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhaoqi Wang
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David Krane
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Taylor Sorenson
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David Craft
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
El Majzoub R, Fayyad-kazan M, Nasr El Dine A, Makki R, Hamade E, Grée R, Hachem A, Talhouk R, Fayyad-Kazan H, Badran B. A thiosemicarbazone derivative induces triple negative breast cancer cell apoptosis: possible role of miRNA-125a-5p and miRNA-181a-5p. Genes Genomics 2019; 41:1431-1443. [DOI: 10.1007/s13258-019-00866-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022]
|
12
|
Macnamara CK, Mitchell EI, Chaplain MA. Spatial-Stochastic modelling of synthetic gene regulatory networks. J Theor Biol 2019; 468:27-44. [DOI: 10.1016/j.jtbi.2019.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/18/2019] [Accepted: 02/05/2019] [Indexed: 02/06/2023]
|
13
|
Wang X, Lu S, Xiao Y, Xu L, Zhou L, Hu J, Li B, Zeng C, Li Y. Alteration of gene expression profile in CD3 + T-cells after downregulating MALT1. Immunotargets Ther 2018; 7:77-81. [PMID: 30538965 PMCID: PMC6251356 DOI: 10.2147/itt.s179656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background T cell immunodeficiency is a common feature in patients with different kinds of hematological disease such as T cell non-Hodgkin lymphoma (T-NHL), B cells NHL (B-NHL), NK/T cell NHL (NK/T-CL) and acute myeloid leukemia (AML). In our recent research, we found that significantly lower expression levels in MALT1 and NF-κB were related to suppression of T cell activation. Therefore, this study was conducted to further investigate the role of downregulating MALT1 in the development of immunodeficiency in T cells. Methods We induced activation inhibition in CD3+ T cells by MALT1 knockdown. Then we characterized the gene expression profile after MALT1 suppression by microarray analysis. Result The differentially expressed genes were ZAP-70, p65, MDM2, ATM, NFATC2 which participate in the NF-κB, p53, and NFAT pathways in CD3+ T cells after MALT1 downregulation. Conclusion MALT1 suppression may contribute to immunodeficiency in T cells via suppression of T cell activation and proliferation pathways. These data may help to explain some of the characteristics of immunodeficiency of T cells.
Collapse
Affiliation(s)
- Xu Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, ,
| | - Shuai Lu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, ,
| | - Yankai Xiao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, ,
| | - Ling Xu
- Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People's Republic of China,
| | - Lingling Zhou
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, ,
| | - Junyan Hu
- Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, ,
| | - Bo Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, ,
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, ,
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Institute of Hematology, Jinan University, Guangzhou, 510632, People's Republic of China, , .,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People's Republic of China,
| |
Collapse
|
14
|
Liu N, Wang KS, Qi M, Zhou YJ, Zeng GY, Tao J, Zhou JD, Zhang JL, Chen X, Peng C. Vitexin compound 1, a novel extraction from a Chinese herb, suppresses melanoma cell growth through DNA damage by increasing ROS levels. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:269. [PMID: 30400954 PMCID: PMC6219156 DOI: 10.1186/s13046-018-0897-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/31/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Vitex negundo L (Verbenaceae) is an aromatic shrub that is abundant in Asian countries. A series of compounds from Vitex negundo have been used in traditional Chinese medicine for the treatment of various diseases. Cutaneous melanoma is one of the most aggressive malignancies. A significant feature of melanoma is its resistance to traditional chemotherapy and radiotherapy; therefore, there is an urgent need to develop novel treatments for melanoma. METHODS We first examined the effects of VB1 (vitexin compound 1) on cell viability by CCK-8 (cell counting kit) and Colony Formation Assay; And then, we analyzed the apoptosis and cell cycle by flow cytometry, verified apoptosis by Immunoblotting. The in vivo effect of VB1 was evaluated in xenograft mouse model. Potential mechanisms of VB1's antitumor effects were explored by RNA sequencing and the key differential expression genes were validated by real-time quantitative PCR. Finally, the intracellular reactive oxygen species (ROS) level was detected by flow cytometry, and the DNA damage was revealed by Immunofluorescence and Immunoblotting. RESULTS In this study, we show that VB1, which is a compound purified from the seed of the Chinese herb Vitex negundo, blocks melanoma cells growth in vitro and in vivo, arrests the cell cycle in G2/M phase and induces apoptosis in melanoma cell lines, whereas the effects are not significantly observed in normal cells. To study the details of VB1, we analyzed the alteration of gene expression profiles after treatment with VB1 in melanoma cells. The findings showed that VB1 can affect various pathways, including p53, apoptosis and the cell cycle pathway, in a variety of melanoma cell lines. Furthermore, we confirmed that VB1 restored the P53 pathway protein level, and then we demonstrated that VB1 significantly induced the accumulation of ROS, which resulted in DNA damage in melanoma cell lines. Interestingly, our results showed that VB1 also increased the ROS levels in BRAFi (BRAF inhibitor)-resistant melanoma cells, leading to DNA cytotoxicity, which caused G2/M phase arrest and apoptosis. CONCLUSIONS Taken together, our findings indicate that vitexin compound 1 might be a promising therapeutic Chinese medicine for melanoma treatment regardless of BRAFi resistance.
Collapse
Affiliation(s)
- Nian Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
| | - Kuan Song Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Min Qi
- Department of Plastic and Cosmetic Surgery, XiangYa Hospital, Central South University, Changsha, Hunan, China
| | - Ying Jun Zhou
- School of Pharmaceutical Science,Central, South University, Changsha, Hunan, China
| | - Guang Yao Zeng
- School of Pharmaceutical Science,Central, South University, Changsha, Hunan, China
| | - Juan Tao
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Da Zhou
- Department of Plastic Surgery of Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiang Lin Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
| |
Collapse
|
15
|
Sasaki S, Baba T, Muranaka H, Tanabe Y, Takahashi C, Matsugo S, Mukaida N. Involvement of Prokineticin 2-expressing Neutrophil Infiltration in 5-Fluorouracil-induced Aggravation of Breast Cancer Metastasis to Lung. Mol Cancer Ther 2018; 17:1515-1525. [PMID: 29643149 DOI: 10.1158/1535-7163.mct-17-0845] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/23/2018] [Accepted: 04/05/2018] [Indexed: 11/16/2022]
Abstract
Adjuvant chemotherapy is used for human breast cancer patients, even after curative surgery of primary tumor, to prevent tumor recurrence primarily as a form of metastasis. However, anticancer drugs can accelerate metastasis in several mouse metastasis models. Hence, we examined the effects of postsurgical administration with 5-fluorouracil (5-FU), doxorubicin, and cyclophosphamide, on lung metastasis process, which developed after the resection of the primary tumor arising from the orthotopic injection of a mouse triple-negative breast cancer cell line, 4T1. Only 5-FU markedly increased the numbers and sizes of lung metastasis foci, with enhanced tumor cell proliferation and angiogenesis as evidenced by increases in Ki67-positive cell numbers and CD31-positive areas, respectively. 5-FU-mediated augmented lung metastasis was associated with increases in intrapulmonary neutrophil numbers and expression of neutrophilic chemokines, Cxcl1 and Cxcl2 in tumor cells, with few effects on intrapulmonary T-cell or macrophage numbers. 5-FU enhanced Cxcl1 and Cxcl2 expression in 4T1 cells in a NFκB-dependent manner. Moreover, the administration of a neutrophil-depleting antibody or a Cxcr2 antagonist, SB225002, significantly attenuated 5-FU-mediated enhanced lung metastasis with depressed neutrophil infiltration. Furthermore, infiltrating neutrophils and 4T1 cells abundantly expressed prokineticin-2 (Prok2) and its receptor, Prokr1, respectively. Finally, the administration of 5-FU after the resection of the primary tumor failed to augment lung metastasis in the mice receiving Prokr1-deleted 4T1 cells. Collectively, 5-FU can enhance lung metastasis by inducing tumor cells to produce Cxcl1 and Cxcl2, which induced the migration of neutrophils expressing Prok2 with a capacity to enhance 4T1 cell proliferation. Mol Cancer Ther; 17(7); 1515-25. ©2018 AACR.
Collapse
Affiliation(s)
- Soichiro Sasaki
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hayato Muranaka
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Chiaki Takahashi
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Seiichi Matsugo
- School of Natural System, College of Science and Engineering, Kanazawa University, Kanazawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
16
|
Sun Y, Heidary DK, Zhang Z, Richards CI, Glazer EC. Bacterial Cytological Profiling Reveals the Mechanism of Action of Anticancer Metal Complexes. Mol Pharm 2018; 15:3404-3416. [PMID: 29865789 PMCID: PMC6083414 DOI: 10.1021/acs.molpharmaceut.8b00407] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
![]()
Target
identification and mechanistic studies of cytotoxic agents
are challenging processes that are both time-consuming and costly.
Here we describe an approach to mechanism of action studies for potential
anticancer compounds by utilizing the simple prokaryotic system, E. coli, and we demonstrate its utility with the characterization
of a ruthenium polypyridyl complex [Ru(bpy)2dmbpy2+]. Expression of the photoconvertible fluorescent protein Dendra2
facilitated both high throughput studies and single-cell imaging.
This allowed for simultaneous ratiometric analysis of inhibition of
protein production and phenotypic investigations. The profile of protein
production, filament size and population, and nucleoid morphology
revealed important differences between inorganic agents that damage
DNA vs more selective inhibitors of transcription and translation.
Trace metal analysis demonstrated that DNA is the preferred nucleic
acid target of the ruthenium complex, but further studies in human
cancer cells revealed altered cell signaling pathways compared to
the commonly administrated anticancer agent cisplatin. This study
demonstrates E. coli can be used to rapidly distinguish
between compounds with disparate mechanisms of action and also for
more subtle distinctions within in studies in mammalian cells.
Collapse
Affiliation(s)
- Yang Sun
- Department of Chemistry , University of Kentucky , Lexington , Kentucky 40506 , United States
| | - David K Heidary
- Department of Chemistry , University of Kentucky , Lexington , Kentucky 40506 , United States
| | - Zhihui Zhang
- Department of Chemistry , University of Kentucky , Lexington , Kentucky 40506 , United States
| | - Christopher I Richards
- Department of Chemistry , University of Kentucky , Lexington , Kentucky 40506 , United States
| | - Edith C Glazer
- Department of Chemistry , University of Kentucky , Lexington , Kentucky 40506 , United States
| |
Collapse
|
17
|
Azam MR, Fazal S, Ullah M, Bhatti AI. System-based strategies for p53 recovery. IET Syst Biol 2018; 12:101-107. [PMID: 29745903 PMCID: PMC8687347 DOI: 10.1049/iet-syb.2017.0025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/25/2017] [Accepted: 11/19/2017] [Indexed: 11/19/2022] Open
Abstract
The authors have proposed a systems theory-based novel drug design approach for the p53 pathway. The pathway is taken as a dynamic system represented by ordinary differential equations-based mathematical model. Using control engineering practices, the system analysis and subsequent controller design is performed for the re-activation of wild-type p53. p53 revival is discussed for both modes of operation, i.e. the sustained and oscillatory. To define the problem in control system paradigm, modification in the existing mathematical model is performed to incorporate the effect of Nutlin. Attractor point analysis is carried out to select the suitable domain of attraction. A two-loop negative feedback control strategy is devised to drag the system trajectories to the attractor point and to regulate cellular concentration of Nutlin, respectively. An integrated framework is constituted to incorporate the pharmacokinetic effects of Nutlin in the cancerous cells. Bifurcation analysis is also performed on the p53 model to see the conditions for p53 oscillation.
Collapse
Affiliation(s)
- Muhammad Rizwan Azam
- CASPR, Department of Electronics Engineering, Capital University of Science and Technology, Islamabad, Pakistan
| | - Sahar Fazal
- Department of Bioinformatics and Biosciences, Capital University of Science and Technology, Islamabad, Pakistan
| | - Mukhtar Ullah
- Department of Electrical Engineering, National University of Computer and Emerging Sciences, Islamabad, Pakistan
| | - Aamer I Bhatti
- CASPR, Department of Electronics Engineering, Capital University of Science and Technology, Islamabad, Pakistan.
| |
Collapse
|
18
|
Circulating exosomal microRNAs reveal the mechanism of Fructus Meliae Toosendan-induced liver injury in mice. Sci Rep 2018; 8:2832. [PMID: 29434260 PMCID: PMC5809479 DOI: 10.1038/s41598-018-21113-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/29/2018] [Indexed: 02/07/2023] Open
Abstract
The toxicological mechanisms of liver injury caused by most traditional Chinese medicine (TCM) remain largely unknown. Due to the unique features, exosomal microRNAs (miRNAs) are currently attracting major interests to provide further insights into toxicological mechanisms. Thus, taking Fructus Meliae Toosendan as an example of hepatoxic TCM, this study aimed to elucidate its hepatotoxicity mechanisms through profiling miRNAs in circulating exosomes of Fructus Meliae Toosendan water extract (FMT)-exposed mice. Biological pathway analysis of the 64 differentially expressed exosomal miRNAs (DEMs) showed that hepatic dysfunction induced by FMT likely related to apoptosis, mitochondrial dysfunction, and cell cycle dysregulation. Integrated analysis of serum exosomal DEMs and hepatic differentially expressed mRNAs further enriched oxidative stress and apoptosis related pathways. In vitro validation studies for omics results suggested that FMT-induced DNA damage was mediated by generating intracellular reactive oxygen species, leading to cell apoptosis through p53-dependent mitochondrial damage and S-phase arrest. Nrf2-mediated antioxidant response was activated to protect liver cells. Moreover, serum exosomal miR-370-3p, the most down-regulated miRNA involving in these pathways, might be the momentous event in aggravating cytotoxic effect of FMT by elevating p21 and Cyclin E. In conclusion, circulating exosomal miRNAs profiling could contribute to deepen the understanding of TCM-induced hepatotoxicity.
Collapse
|
19
|
Vale N, Gouveia MJ, Rinaldi G, Santos J, Santos LL, Brindley PJ, da Costa JMC. The role of estradiol metabolism in urogenital schistosomiasis-induced bladder cancer. Tumour Biol 2017; 39:1010428317692247. [PMID: 28345469 DOI: 10.1177/1010428317692247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Urogenital schistosomiasis is a neglected tropical disease that can lead to bladder cancer. How urogenital schistosomiasis induces carcinogenesis remains unclear, although there is evidence that the human blood fluke Schistosoma haematobium, the infectious agent of urogenital schistosomiasis, releases estradiol-like metabolites. These kind of compounds have been implicated in other cancers. Aiming for enhanced understanding of the pathogenesis of the urogenital schistosomiasis-induced bladder cancer, here we review, interpret, and discuss findings of estradiol-like metabolites detected in both the parasite and in the human urine during urogenital schistosomiasis. Moreover, we predict pathways and enzymes that are involved in the production of these metabolites emphasizing their potential effects on the dysregulation of the tumor suppressor gene p53 expression during urogenital schistosomiasis. Enhanced understanding of these potential carcinogens may not only shed light on urogenital schistosomiasis-induced neoplasia of the bladder, but would also facilitate development of interventions and biomarkers for this and other infection-associated cancers at large.
Collapse
Affiliation(s)
- Nuno Vale
- 1 UCIBIO/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Maria J Gouveia
- 1 UCIBIO/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.,2 Center for the Study of Animal Science, ICETA, University of Porto, Porto, Portugal
| | - Gabriel Rinaldi
- 3 Department of Microbiology, Immunology, & Tropical Medicine and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA.,4 The Wellcome Trust Sanger Institute, Cambridge, UK
| | - Júlio Santos
- 5 Clínica da Sagrada Esperança, Luanda, Angola.,6 Experimental Pathology and Therapeutics Group, Research Center of Instituto Português de Oncologia, Porto, Portugal
| | - Lúcio Lara Santos
- 6 Experimental Pathology and Therapeutics Group, Research Center of Instituto Português de Oncologia, Porto, Portugal
| | - Paul J Brindley
- 3 Department of Microbiology, Immunology, & Tropical Medicine and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| | - José M Correia da Costa
- 2 Center for the Study of Animal Science, ICETA, University of Porto, Porto, Portugal.,7 Department of Infectious Diseases, R&D Unit, National Health Institute Doutor Ricardo Jorge (INSA), Porto, Portugal
| |
Collapse
|
20
|
Eliaš J. Positive effect of Mdm2 on p53 expression explains excitability of p53 in response to DNA damage. J Theor Biol 2017; 418:94-104. [DOI: 10.1016/j.jtbi.2017.01.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 09/07/2016] [Accepted: 01/21/2017] [Indexed: 11/28/2022]
|
21
|
Oghabi Bakhshaiesh T, Majidzadeh-A K, Esmaeili R. Wip1: A candidate phosphatase for cancer diagnosis and treatment. DNA Repair (Amst) 2017; 54:63-66. [PMID: 28385459 DOI: 10.1016/j.dnarep.2017.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 12/28/2022]
Abstract
The critical regulatory mechanisms in numerous cellular pathways including cell survival and DNA damage response mostly depend on phosphorylation and dephosphorylation of proteins. The serine/threonine phosphatase wild-type p53-induced phosphatase 1 (Wip1) is a growth-promoting phosphatase and its numerous downstream targets are important tumor suppressors. Here, we review the Wip1 activity and its relevance to cancer as an oncoprotein. Consecutive investigations about Wip1 and its relation to cancer is critical, as these studies ultimately contribute to the etiology of cancer. A number of innovative studies have recently investigated the importance of Wip1 as a new candidate for cancer diagnosis and prognosis. Accordingly, we discuss the present challenges of using Wip1 as a target for cancer treatment.
Collapse
Affiliation(s)
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Rezvan Esmaeili
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
22
|
Macnamara CK, Chaplain MAJ. Spatio-temporal models of synthetic genetic oscillators. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2017; 14:249-262. [PMID: 27879131 DOI: 10.3934/mbe.2017016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Signal transduction pathways play a major role in many important aspects of cellular function e.g. cell division, apoptosis. One important class of signal transduction pathways is gene regulatory networks (GRNs). In many GRNs, proteins bind to gene sites in the nucleus thereby altering the transcription rate. Such proteins are known as transcription factors. If the binding reduces the transcription rate there is a negative feedback leading to oscillatory behaviour in mRNA and protein levels, both spatially (e.g. by observing fluorescently labelled molecules in single cells) and temporally (e.g. by observing protein/mRNA levels over time). Recent computational modelling has demonstrated that spatial movement of the molecules is a vital component of GRNs and may cause the oscillations. These numerical findings have subsequently been proved rigorously i.e. the diffusion coefficient of the protein/mRNA acts as a bifurcation parameter and gives rise to a Hopf bifurcation. In this paper we first present a model of the canonical GRN (the Hes1 protein) and show the effect of varying the spatial location of gene and protein production sites on the oscillations. We then extend the approach to examine spatio-temporal models of synthetic gene regulatory networks e.g. n-gene repressilators and activator-repressor systems.
Collapse
Affiliation(s)
- Cicely K Macnamara
- School of Mathematics and Statistics, Mathematical Institute, North Haugh, University of St Andrews, St Andrews KY16 9SS, Scotland.
| | | |
Collapse
|
23
|
Macnamara CK, Chaplain MAJ. Diffusion driven oscillations in gene regulatory networks. J Theor Biol 2016; 407:51-70. [DOI: 10.1016/j.jtbi.2016.07.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 06/24/2016] [Accepted: 07/16/2016] [Indexed: 10/21/2022]
|
24
|
Kuijper IA, Yang H, Van De Water B, Beltman JB. Unraveling cellular pathways contributing to drug-induced liver injury by dynamical modeling. Expert Opin Drug Metab Toxicol 2016; 13:5-17. [PMID: 27609146 DOI: 10.1080/17425255.2017.1234607] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Drug-induced liver injury (DILI) is a significant threat to human health and a major problem in drug development. It is hard to predict due to its idiosyncratic nature and which does not show up in animal trials. Hepatic adaptive stress response pathway activation is generally observed in drug-induced liver injury. Dynamical pathway modeling has the potential to foresee adverse effects of drugs before they go in trial. Ordinary differential equation modeling can offer mechanistic insight, and allows us to study the dynamical behavior of stress pathways involved in DILI. Areas covered: This review provides an overview on the progress of the dynamical modeling of stress and death pathways pertinent to DILI, i.e. pathways relevant for oxidative stress, inflammatory stress, DNA damage, unfolded proteins, heat shock and apoptosis. We also discuss the required steps for applying such modeling to the liver. Expert opinion: Despite the strong progress made since the turn of the century, models of stress pathways have only rarely been specifically applied to describe pathway dynamics for DILI. We argue that with minor changes, in some cases only to parameter values, many of these models can be repurposed for application in DILI research. Combining both dynamical models with in vitro testing might offer novel screening methods for the harmful side-effects of drugs.
Collapse
Affiliation(s)
- Isoude A Kuijper
- a Division of Toxicology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Huan Yang
- a Division of Toxicology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Bob Van De Water
- a Division of Toxicology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Joost B Beltman
- a Division of Toxicology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| |
Collapse
|
25
|
Jonak K, Kurpas M, Szoltysek K, Janus P, Abramowicz A, Puszynski K. A novel mathematical model of ATM/p53/NF- κB pathways points to the importance of the DDR switch-off mechanisms. BMC SYSTEMS BIOLOGY 2016; 10:75. [PMID: 27526774 PMCID: PMC4986247 DOI: 10.1186/s12918-016-0293-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 06/27/2016] [Indexed: 12/20/2022]
Abstract
Background Ataxia telangiectasia mutated (ATM) is a detector of double-strand breaks (DSBs) and a crucial component of the DNA damage response (DDR) along with p53 and NF- κB transcription factors and Wip1 phosphatase. Despite the recent advances in studying the DDR, the mechanisms of cell fate determination after DNA damage induction is still poorly understood. Results To investigate the importance of various DDR elements with particular emphasis on Wip1, we developed a novel mathematical model of ATM/p53/NF- κB pathways. Our results from in silico and in vitro experiments performed on U2-OS cells with Wip1 silenced to 25 % (Wip1-RNAi) revealed a strong dependence of cellular response to DNA damages on this phosphatase. Notably, Wip1-RNAi cells exhibited lower resistance to ionizing radiation (IR) resulting in smaller clonogenicity and higher apoptotic fraction. Conclusions In this article, we demonstrated that Wip1 plays a role as a gatekeeper of apoptosis and influences the pro-survival behaviour of cells – the level of Wip1 increases to block the apoptotic decision when DNA repair is successful. Moreover, we were able to verify the dynamics of proteins and transcripts, apoptotic fractions and cells viability obtained from stochastic simulations using in vitro approaches. Taken together, we demonstrated that the model can be successfully used in prediction of cellular behaviour after exposure to IR. Thus, our studies may provide further insights into key elements involved in the underlying mechanisms of the DDR. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0293-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katarzyna Jonak
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka, Gliwice, 16, 44-100, Poland
| | - Monika Kurpas
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka, Gliwice, 16, 44-100, Poland
| | - Katarzyna Szoltysek
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Wybrzeze Armii Krajowej, Gliwice, 15, 44-400, Poland
| | - Patryk Janus
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Wybrzeze Armii Krajowej, Gliwice, 15, 44-400, Poland
| | - Agata Abramowicz
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Wybrzeze Armii Krajowej, Gliwice, 15, 44-400, Poland
| | - Krzysztof Puszynski
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka, Gliwice, 16, 44-100, Poland.
| |
Collapse
|
26
|
Murphy H, Jaafari H, Dobrovolny HM. Differences in predictions of ODE models of tumor growth: a cautionary example. BMC Cancer 2016; 16:163. [PMID: 26921070 PMCID: PMC4768423 DOI: 10.1186/s12885-016-2164-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 02/14/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND While mathematical models are often used to predict progression of cancer and treatment outcomes, there is still uncertainty over how to best model tumor growth. Seven ordinary differential equation (ODE) models of tumor growth (exponential, Mendelsohn, logistic, linear, surface, Gompertz, and Bertalanffy) have been proposed, but there is no clear guidance on how to choose the most appropriate model for a particular cancer. METHODS We examined all seven of the previously proposed ODE models in the presence and absence of chemotherapy. We derived equations for the maximum tumor size, doubling time, and the minimum amount of chemotherapy needed to suppress the tumor and used a sample data set to compare how these quantities differ based on choice of growth model. RESULTS We find that there is a 12-fold difference in predicting doubling times and a 6-fold difference in the predicted amount of chemotherapy needed for suppression depending on which growth model was used. CONCLUSION Our results highlight the need for careful consideration of model assumptions when developing mathematical models for use in cancer treatment planning.
Collapse
Affiliation(s)
- Hope Murphy
- Department of Physics, Utica College, Utica, NY, USA.
| | - Hana Jaafari
- Department of Physics & Astronomy, Texas Christian University, 2800 S. University Drive, TX, 76129, Fort Worth, USA.
| | - Hana M Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, 2800 S. University Drive, TX, 76129, Fort Worth, USA.
| |
Collapse
|
27
|
Zong J, Wang D, Jiao W, Zhang L, Bao G, Ho CT, Hou R, Wan X. Oleiferasaponin C6 from the seeds of Camellia oleifera Abel.: a novel compound inhibits proliferation through inducing cell-cycle arrest and apoptosis on human cancer cell lines in vitro. RSC Adv 2016. [DOI: 10.1039/c6ra14467e] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Oleiferasaponin C6 was isolated from Camellia oleifera Abel. and inhibits proliferation through inducing cell-cycle arrest and apoptosis on cancer cell lines in vitro.
Collapse
Affiliation(s)
- Jianfa Zong
- State Key Laboratory of Tea Plant Biology and Utilization
- Anhui Agricultural University
- Hefei 230036
- China
| | - Dongxu Wang
- State Key Laboratory of Tea Plant Biology and Utilization
- Anhui Agricultural University
- Hefei 230036
- China
| | - Weiting Jiao
- State Key Laboratory of Tea Plant Biology and Utilization
- Anhui Agricultural University
- Hefei 230036
- China
| | - Liang Zhang
- State Key Laboratory of Tea Plant Biology and Utilization
- Anhui Agricultural University
- Hefei 230036
- China
| | - Guanhu Bao
- State Key Laboratory of Tea Plant Biology and Utilization
- Anhui Agricultural University
- Hefei 230036
- China
| | - Chi-Tang Ho
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
| | - Ruyan Hou
- State Key Laboratory of Tea Plant Biology and Utilization
- Anhui Agricultural University
- Hefei 230036
- China
| | - Xiaochun Wan
- State Key Laboratory of Tea Plant Biology and Utilization
- Anhui Agricultural University
- Hefei 230036
- China
| |
Collapse
|
28
|
Zhang G, An Y, Lu X, Zhong H, Zhu Y, Wu Y, Ma F, Yang J, Liu Y, Zhou Z, Peng Y, Chen Z. A Novel Naphthalimide Compound Restores p53 Function in Non-small Cell Lung Cancer by Reorganizing the Bak·Bcl-xl Complex and Triggering Transcriptional Regulation. J Biol Chem 2015; 291:4211-25. [PMID: 26668309 DOI: 10.1074/jbc.m115.669978] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Indexed: 12/16/2022] Open
Abstract
p53 inactivation is a hallmark in non-small-cell lung cancer (NSCLC). It is therefore highly desirable to develop tumor-specific treatment for NSCLC therapy by restoring p53 function. Herein, a novel naphthalimide compound, NA-17, was identified as a promising drug candidate in view of both its anticancer activity and mechanism of action. NA-17 exhibited strong anticancer activity on a broad range of cancer cell lines but showed low toxicity to normal cell lines, such as HL-7702 and WI-38. Moreover, NA-17 showed p53-dependent inhibition selectivity in different NSCLC cell lines due to the activation state of endogenous p53 in the background level. Further studies revealed that NA-17 caused cell cycle arrest at the G1 phase, changed cell size, and induced apoptosis and cell death by increasing the proportion of sub-G1 cells. Molecular mechanism studies suggested that targeted accumulation of phospho-p53 in mitochondria and nuclei induced by NA-17 resulted in activation of Bak and direct binding of phospho-p53 to the target DNA sequences, thereby evoking cell apoptosis and cell cycle arrest and eventually leading to irreversible cancer cell inhibition. This work provided new insights into the molecular interactions and anticancer mechanisms of phospho-p53-dependent naphthalimide compounds.
Collapse
Affiliation(s)
- Guohai Zhang
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Yunfeng An
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Xing Lu
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Hui Zhong
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Yanhong Zhu
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Yiming Wu
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Feng'e Ma
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Jingmei Yang
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Yancheng Liu
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Zuping Zhou
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Yan Peng
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Zhenfeng Chen
- From the State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
29
|
Cortez MA, Ivan C, Valdecanas D, Wang X, Peltier HJ, Ye Y, Araujo L, Carbone DP, Shilo K, Giri DK, Kelnar K, Martin D, Komaki R, Gomez DR, Krishnan S, Calin GA, Bader AG, Welsh JW. PDL1 Regulation by p53 via miR-34. J Natl Cancer Inst 2015; 108:djv303. [PMID: 26577528 PMCID: PMC4862407 DOI: 10.1093/jnci/djv303] [Citation(s) in RCA: 494] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 09/25/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Although clinical studies have shown promise for targeting PD1/PDL1 signaling in non-small cell lung cancer (NSCLC), the regulation of PDL1 expression is poorly understood. Here, we show that PDL1 is regulated by p53 via miR-34. METHODS p53 wild-type and p53-deficient cell lines (p53(-/-) and p53(+/+) HCT116, p53-inducible H1299, and p53-knockdown H460) were used to determine if p53 regulates PDL1 via miR-34. PDL1 and miR-34a expression were analyzed in samples from patients with NSCLC and mutated p53 vs wild-type p53 tumors from The Cancer Genome Atlas for Lung Adenocarcinoma (TCGA LUAD). We confirmed that PDL1 is a direct target of miR-34 with western blotting and luciferase assays and used a p53(R172HΔ)g/+K-ras(LA1/+) syngeneic mouse model (n = 12) to deliver miR-34a-loaded liposomes (MRX34) plus radiotherapy (XRT) and assessed PDL1 expression and tumor-infiltrating lymphocytes (TILs). A two-sided t test was applied to compare the mean between different treatments. RESULTS We found that p53 regulates PDL1 via miR-34, which directly binds to the PDL1 3' untranslated region in models of NSCLC (fold-change luciferase activity to control group, mean for miR-34a = 0.50, SD = 0.2, P < .001; mean for miR-34b = 0.52, SD = 0.2, P = .006; and mean for miR-34c = 0.59, SD = 0.14, and P = .006). Therapeutic delivery of MRX34, currently the subject of a phase I clinical trial, promoted TILs (mean of CD8 expression percentage of control group = 22.5%, SD = 1.9%; mean of CD8 expression percentage of MRX34 = 30.1%, SD = 3.7%, P = .016, n = 4) and reduced CD8(+)PD1(+) cells in vivo (mean of CD8/PD1 expression percentage of control group = 40.2%, SD = 6.2%; mean of CD8/PD1 expression percentage of MRX34 = 20.3%, SD = 5.1%, P = .001, n = 4). Further, MRX34 plus XRT increased CD8(+) cell numbers more than either therapy alone (mean of CD8 expression percentage of MRX34 plus XRT to control group = 44.2%, SD = 8.7%, P = .004, n = 4). Finally, miR-34a delivery reduced the numbers of radiation-induced macrophages (mean of F4-80 expression percentage of control group = 52.4%, SD = 1.7%; mean of F4-80 expression percentage of MRX34 = 40.1%, SD = 3.5%, P = .008, n = 4) and T-regulatory cells. CONCLUSIONS We identified a novel mechanism by which tumor immune evasion is regulated by p53/miR-34/PDL1 axis. Our results suggest that delivery of miRNAs with standard therapies, such as XRT, may represent a novel therapeutic approach for lung cancer.
Collapse
Affiliation(s)
- Maria Angelica Cortez
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Cristina Ivan
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - David Valdecanas
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Xiaohong Wang
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Heidi J Peltier
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Yuping Ye
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Luiz Araujo
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - David P Carbone
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Konstantin Shilo
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Dipak K Giri
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Kevin Kelnar
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Desiree Martin
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Ritsuko Komaki
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Daniel R Gomez
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Sunil Krishnan
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - George A Calin
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Andreas G Bader
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - James W Welsh
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG).
| |
Collapse
|
30
|
Thorsteinsdottir UA, Thorsteinsdottir M, Lambert IH. Protolichesterinic Acid, Isolated from the Lichen Cetraria islandica
, Reduces LRRC8A Expression and Volume-Sensitive Release of Organic Osmolytes in Human Lung Epithelial Cancer Cells. Phytother Res 2015; 30:97-104. [DOI: 10.1002/ptr.5507] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Unnur Arna Thorsteinsdottir
- Department of Biology, Section of Cell biology and Physiology; University of Copenhagen; 13 Universitetsparken Copenhagen DK-2100 Denmark
- School of Health Sciences, Faculty of Pharmaceutical Sciences; University of Iceland; 101 Reykjavik Iceland
| | - Margret Thorsteinsdottir
- School of Health Sciences, Faculty of Pharmaceutical Sciences; University of Iceland; 101 Reykjavik Iceland
| | - Ian Henry Lambert
- Department of Biology, Section of Cell biology and Physiology; University of Copenhagen; 13 Universitetsparken Copenhagen DK-2100 Denmark
| |
Collapse
|
31
|
Zhang GN, Ashby CR, Zhang YK, Chen ZS, Guo H. The reversal of antineoplastic drug resistance in cancer cells by β-elemene. CHINESE JOURNAL OF CANCER 2015; 34:488-95. [PMID: 26370907 PMCID: PMC4593349 DOI: 10.1186/s40880-015-0048-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/26/2015] [Indexed: 12/30/2022]
Abstract
Multidrug resistance (MDR), defined as the resistance of cancer cells to compounds with diverse structures and mechanisms of actions, significantly limits the efficacy of antitumor drugs. A major mechanism that mediates MDR in cancer is the overexpression of adenosine triphosphate (ATP)-binding cassette transporters. These transporters bind to their respective substrates and catalyze their efflux from cancer cells, thereby lowering the intracellular concentrations of the substrates and thus attenuating or even abolishing their efficacy. In addition, cancer cells can become resistant to drugs via mechanisms that attenuate apoptosis and cell cycle arrest such as alterations in the p53, check point kinase, nuclear factor kappa B, and the p38 mitogen-activated protein kinase pathway. In this review, we discuss the mechanisms by which β-elemene, a compound extracted from Rhizoma zedoariae that has clinical antitumor efficacy, overcomes drug resistance in cancer.
Collapse
Affiliation(s)
- Guan-Nan Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Yun-Kai Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Huiqin Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing, 100730, P. R. China.
| |
Collapse
|
32
|
Wang C, Jiang L, Wang S, Shi H, Wang J, Wang R, Li Y, Dou Y, Liu Y, Hou G, Ke Y, Liu H. The Antitumor Activity of the Novel Compound Jesridonin on Human Esophageal Carcinoma Cells. PLoS One 2015; 10:e0130284. [PMID: 26103161 PMCID: PMC4477902 DOI: 10.1371/journal.pone.0130284] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 05/18/2015] [Indexed: 11/18/2022] Open
Abstract
Jesridonin, a small molecule obtained through the structural modification of Oridonin, has extensive antitumor activity. In this study, we evaluated both its in vitro activity in the cancer cell line EC109 and its in vivo effect on tumor xenografts in nude mice. Apoptosis induced by Jesridonin was determined using an MTT assay, Annexin-V FITC assay and Hoechest 33258 staining. Apoptosis via mitochondrial and death receptor pathways were confirmed by detecting the regulation of MDM2, p53, and Bcl-2 family members and by activation of caspase-3/-8/-9. In addition, vena caudalis injection of Jesridonin showed significant inhibition of tumor growth in the xenograft model, and Jesridonin-induced cell apoptosis in tumor tissues was determined using TUNEL. Biochemical serum analysis of alkaline phosphatase (ALP), alanine transaminase (ALT), aspartate transaminase (AST), gamma-glutamyl transferase (GGT), total protein (TP) and albumin (ALB) indicated no obvious effects on liver function. Histopathological examination of the liver, kidney, lung, heart and spleen revealed no signs of JD-induced toxicity. Taken together, these results demonstrated that Jesridonin exhibits antitumor activity in human esophageal carcinomas EC109 cells both in vitro and in vivo and demonstrated no adverse effects on major organs in nude mice. These studies provide support for new drug development.
Collapse
Affiliation(s)
- Cong Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Liping Jiang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Saiqi Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Hongge Shi
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Junwei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Ran Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yongmei Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yinhui Dou
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Ying Liu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Guiqin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yu Ke
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- * E-mail: (HML); (YK)
| | - Hongmin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- * E-mail: (HML); (YK)
| |
Collapse
|
33
|
Lambert IH, Enghoff MS, Brandi ML, Hoffmann EK. Regulation of p53 in NIH3T3 mouse fibroblasts following hyperosmotic stress. Physiol Rep 2015; 3:3/6/e12412. [PMID: 26056062 PMCID: PMC4510620 DOI: 10.14814/phy2.12412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The aim of this project was to analyze the regulation of p53 expression in NIH3T3 fibroblasts under the influence of increasing hyperosmotic stress. Expression of p53 showed a biphasic response pattern in NIH3T3 cells under increasing osmotic stress (337 mOsm to 737 mOsm) with a maximum at 587 mOsm. Under isotonic conditions p53 expression increased after addition of the proteasome inhibitor MG132 indicating that cellular p53 levels in unperturbed cells is kept low by proteasomal degradation. However, under hypertonic conditions p53 synthesis as well as p53 degradation were significantly reduced and it is demonstrated that the increase in p53 expression observed when tonicity is increased from 337 to 587 mOsm reflects that degradation is more inhibited than synthesis, whereas the decrease in p53 expression at higher tonicities reflects that synthesis is more inhibited than degradation. The activity of the p53 regulating proteins p38 MAP kinase and the ubiquitin ligase MDM2 were studied as a function of increasing osmolarity. MDM2 protein expression was unchanged at all osmolarities, whereas MDM2 phosphorylation (Ser166) increased at osmolarities up to 537 mOsm and remained constant at higher osmolarities. Phosphorylation of p38 increased at osmolarities up to 687 mOsm which correlated with an increased phosphorylation of p53 (Ser15) and the decreased p53 degradation. Caspase-3 activity increased gradually with hypertonicity and at 737 mOsm both Caspase-3 activity and annexin V binding are high even though p53 expression and activity are low, indicating that initiation of apoptosis under severe hypertonic conditions is not strictly controlled by p53.
Collapse
Affiliation(s)
- Ian Henry Lambert
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Stine Enghoff
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Luise Brandi
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Else Kay Hoffmann
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Ma X, Han J, Wu Q, Liu H, Shi S, Wang C, Wang Y, Xiao J, Zhao J, Jiang J, Wan C. Involvement of dysregulated Wip1 in manganese-induced p53 signaling and neuronal apoptosis. Toxicol Lett 2015; 235:17-27. [PMID: 25791630 DOI: 10.1016/j.toxlet.2014.12.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 12/22/2014] [Accepted: 12/29/2014] [Indexed: 02/07/2023]
Abstract
Overexposure to manganese (Mn) has been known to induce neuronal death and neurodegenerative symptoms. However, the precise mechanisms underlying Mn neurotoxicity remain incompletely understood. In the present study, we established a Mn-exposed rat model and found that downregulation of wild type p53-induced phosphatase 1 (Wip1) might contribute to p53 activation and resultant neuronal apoptosis following Mn exposure. Western blot and immunohistochemical analyses revealed that the expression of Wip1 was markedly decreased following Mn exposure. In addition, immunofluorescence assay demonstrated that Mn exposure led to significant reduction in the number of Wip1-positive neurons. Accordingly, the expression of Mdm2 was progressively decreased, which was accompanied with markedly increased expression of p53, as well as the ratio of Bax/Bcl-xl. Furthermore, we showed that Mn exposure decreased the viability and induced apparent apoptosis in NFG-differentiated neuron-like PC12 cells. Importantly, the expression of Wip1 decreased progressively, whereas the level of cellular p53 and the ratio of Bax/Bcl-xl were elevated, which resembled the expression of the proteins in animal model studies. Depletion of p53 significantly ameliorated Mn-mediated cytotoxic effect in PC12 cells. In addition, ectopic expression of Wip1 attenuated Mn-induced p53 signaling as well as apoptosis in PC12 cells. Finally, we observed that depletion of Wip1 augmented Mn-induced apoptosis in PC12 cells. Collectively, these findings suggest that downregulated Wip1 expression plays an important role in Mn-induced neuronal death in the brain striatum via the modulation of p53 signaling.
Collapse
Affiliation(s)
- Xia Ma
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Jingling Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Qiyun Wu
- The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| | - Hanzhang Liu
- The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| | - Shangshi Shi
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Cheng Wang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Yueran Wang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Jing Xiao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Jianya Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, People's Republic of China; The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China.
| | - Chunhua Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, People's Republic of China; The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China.
| |
Collapse
|
35
|
|
36
|
Lambert IH, Kristensen DM, Holm JB, Mortensen OH. Physiological role of taurine--from organism to organelle. Acta Physiol (Oxf) 2015; 213:191-212. [PMID: 25142161 DOI: 10.1111/apha.12365] [Citation(s) in RCA: 241] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 08/01/2014] [Accepted: 08/13/2014] [Indexed: 12/21/2022]
Abstract
Taurine is often referred to as a semi-essential amino acid as newborn mammals have a limited ability to synthesize taurine and have to rely on dietary supply. Taurine is not thought to be incorporated into proteins as no aminoacyl tRNA synthetase has yet been identified and is not oxidized in mammalian cells. However, taurine contributes significantly to the cellular pool of organic osmolytes and has accordingly been acknowledged for its role in cell volume restoration following osmotic perturbation. This review describes taurine homeostasis in cells and organelles with emphasis on taurine biophysics/membrane dynamics, regulation of transport proteins involved in active taurine uptake and passive taurine release as well as physiological processes, for example, development, lung function, mitochondrial function, antioxidative defence and apoptosis which seem to be affected by a shift in the expression of the taurine transporters and/or the cellular taurine content.
Collapse
Affiliation(s)
- I. H. Lambert
- Section of Cellular and Developmental Biology; Department of Biology; University of Copenhagen; Copenhagen Ø Denmark
| | - D. M. Kristensen
- Section of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
- Cellular and Metabolic Research Section; Department of Biomedical Sciences; Panum Institute; University of Copenhagen; Copenhagen N Denmark
| | - J. B. Holm
- Section of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - O. H. Mortensen
- Cellular and Metabolic Research Section; Department of Biomedical Sciences; Panum Institute; University of Copenhagen; Copenhagen N Denmark
| |
Collapse
|
37
|
Zhao P, Chen L, Li LH, Wei ZF, Tong B, Jia YG, Kong LY, Xia YF, Dai Y. SC-III3, a novel scopoletin derivative, induces cytotoxicity in hepatocellular cancer cells through oxidative DNA damage and ataxia telangiectasia-mutated nuclear protein kinase activation. BMC Cancer 2014; 14:987. [PMID: 25527123 PMCID: PMC4320555 DOI: 10.1186/1471-2407-14-987] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 12/11/2014] [Indexed: 11/16/2022] Open
Abstract
Background Natural products from plants have been proven to be important resources of antitumor agents. In this study, we exploited the antitumor activity of (E)-3-(4-chlorophenyl)-N-(7-hydroxy-6-methoxy-2-oxo-2H-chromen-3-yl) acrylamide (SC-III3), a newly synthesized derivative of scopoletin, by in vitro and in vivo experiments. Methods Human hepatocellular carcinoma cell line HepG2 cells and xenograft of HepG2 cells in BALB/c nude mice were used to investigate the effects of SC-III3 on hepatocellular cancers. Cell cycle arrest and apoptosis were analyzed by flow cytometry. Cell cycle arrest, apoptosis and ATM-Chk pathway-related proteins were characterized by western blot. Results SC-III3 selectively inhibited the viability of HepG2 cells without significant cytotoxicity against human normal liver cells LO2. In mouse xenograft model of HepG2 cells, SC-III3 showed a marked inhibition of tumor growth in a dose-dependent manner. Cell cycle analysis revealed that SC-III3 induced cells to accumulate in S phase, which was accompanied by a marked decrease of the expressions of cyclin A, cyclin B, cyclin E and Cdk2 proteins, the crucial regulators of S phase cell cycle. SC-III3 treatment resulted in DNA breaks in HepG2 cells, which might contribute to its S phase arrest. The S arrest and the activation of ATM-Chk1/Chk2-Cdc25A-Cdk2 pathways induced by SC-III3 in HepG2 cells could be efficiently abrogated by pretreatments of either Ku55933 (an inhibitor of ATM) or UCN-01 (an inhibitor of Chk1/Chk2). The activation of p53-p21 pathway by SC-III3 was also reversed by Ku55933 treatment. SC-III3 led to significant accumulation of intracellular reactive oxygen species (ROS), a breaker of DNA strand, in HepG2 cells but not LO2 cells. Pretreatment with N-acetyl-l-cysteine (NAC), a ROS scavenger, could reverse SC-III3-caused ROS accumulation, DNA damage, activation of signal pathways relevant to DNA damage, S phase arrest and cell viability decrease in HepG2 cells. Conclusion SC-III3 is able to efficiently inhibit the growth of hepatocellular carcinoma through inducing the generation of intracellular ROS, DNA damage and consequent S phase arrest, but lack of significant cytotoxicity against normal liver cells. This compound deserves further studies as a candidate of anticancer drugs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yu-Feng Xia
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| | | |
Collapse
|
38
|
Knockdown of PSF1 expression inhibits cell proliferation in lung cancer cells in vitro. Tumour Biol 2014; 36:2163-8. [PMID: 25398693 DOI: 10.1007/s13277-014-2826-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/06/2014] [Indexed: 02/06/2023] Open
Abstract
Partner of sld five 1 (PSF1) is a member of the heterotetrameric complex termed GINS. Previous studies have shown that PSF1 is unregulated in several cancer and associated with tumor malignant characters. However, the effects of PSF1 in lung cancer are still unclear. The goal of this study was to investigate the effects of PSF1 on the proliferation capacities of lung cancer. To start with, expression of PSF1 in 22 human lung cancer samples and adjacent non-tumor samples were detected by real-time RT-PCR and Western blotting. Our results showed that PSF1 was overexpressed in lung cancer samples compared to adjacent non-tumor samples. To achieve better insights of PSF1 functions in lung cancer cells, we used PSF1-specific small interfering RNA (siRNA) successfully inhibit the expression of PSF1 in messenger RNA (mRNA) and protein levels. In addition, we used lung cancer cell lines with different p53 gene background (p53 null and p53 wild-type). The results showed that knockdown of PSF1 inhibited cell proliferation and caused cell cycle arrest of lung cancer cells in a p53-independent manner. Our data indicated that PSF1 is functionally involved in lung cancer cell proliferation and is a potential target for lung cancer therapy.
Collapse
|
39
|
Nussinov R, Jang H. Dynamic multiprotein assemblies shape the spatial structure of cell signaling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 116:158-64. [PMID: 25046855 PMCID: PMC4250281 DOI: 10.1016/j.pbiomolbio.2014.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/07/2014] [Indexed: 11/25/2022]
Abstract
Cell signaling underlies critical cellular decisions. Coordination, efficiency as well as fail-safe mechanisms are key elements. How the cell ensures that these hallmarks are at play are important questions. Cell signaling is often viewed as taking place through discrete and cross-talking pathways; oftentimes these are modularized to emphasize distinct functions. While simple, convenient and clear, such models largely neglect the spatial structure of cell signaling; they also convey inter-modular (or inter-protein) spatial separation that may not exist. Here our thesis is that cell signaling is shaped by a network of multiprotein assemblies. While pre-organized, the assemblies and network are loose and dynamic. They contain transiently-associated multiprotein complexes which are often mediated by scaffolding proteins. They are also typically anchored in the membrane, and their continuum may span the cell. IQGAP1 scaffolding protein which binds proteins including Raf, calmodulin, Mek, Erk, actin, and tens more, with actin shaping B-cell (and likely other) membrane-anchored nanoclusters and allosterically polymerizing in dynamic cytoskeleton formation, and Raf anchoring in the membrane along with Ras, provides a striking example. The multivalent network of dynamic proteins and lipids, with specific interactions forming and breaking, can be viewed as endowing gel-like properties. Collectively, this reasons that efficient, productive and reliable cell signaling takes place primarily through transient, preorganized and cooperative protein-protein interactions spanning the cell rather than stochastic, diffusion-controlled processes.
Collapse
Affiliation(s)
- Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Sackler Inst. of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Hyunbum Jang
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
40
|
Le Cigne A, Menil-Philippot V, Fleury F, Takahashi M, Thiriet C. Transient expression of RAD51 in the late G2-phase is required for cell cycle progression in synchronous Physarum cells. Genes Cells 2014; 19:755-65. [PMID: 25200281 DOI: 10.1111/gtc.12174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 07/30/2014] [Indexed: 11/27/2022]
Abstract
The homologous recombination factor RAD51 is highly conserved. This criterion enabled us to identify a RAD51 ortholog in Physarum polycephalum. We found that the Physarum protein presents a high homology to the human protein and cross-reacted with antibodies directed against the human RAD51. Taking advantage of the natural synchrony of millions of nuclei within a single cell of Physarum, we investigated the fluctuation of the amount of the PpRAD51 throughout the cell cycle. Our results showed that in the late G2-phase, RAD51 was transiently expressed in a large quantity. Furthermore, knocking-down RAD51 in the G2-phase abolished this transient expression before mitosis and affected cell cycle progression. These results support the idea that RAD51 plays a role in the progression of the cell cycle in the late G2-phase.
Collapse
Affiliation(s)
- Anthony Le Cigne
- Faculté des Sciences et des Techniques, UFIP UMR CNRS 6286 & Université de Nantes, 44322, Nantes Cedex 3, France; Division of Mechanism and Regulation of DNA Repair, Faculté des Sciences et des Techniques, UFIP UMR CNRS 6286 & Université de Nantes, 44322, Nantes Cedex 3, France; Division of Epigenetics: Proliferation and Differentiation, Faculté des Sciences et des Techniques, UFIP UMR CNRS 6286 & Université de Nantes, 44322, Nantes Cedex 3, France
| | | | | | | | | |
Collapse
|
41
|
Eliaš J, Dimitrio L, Clairambault J, Natalini R. The dynamics of p53 in single cells: physiologically based ODE and reaction-diffusion PDE models. Phys Biol 2014; 11:045001. [PMID: 25075792 DOI: 10.1088/1478-3975/11/4/045001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The intracellular signalling network of the p53 protein plays important roles in genome protection and the control of cell cycle phase transitions. Recently observed oscillatory behaviour in single cells under stress conditions has inspired several research groups to simulate and study the dynamics of the protein with the aim of gaining a proper understanding of the physiological meanings of the oscillations. We propose compartmental ODE and PDE models of p53 activation and regulation in single cells following DNA damage and we show that the p53 oscillations can be retrieved by plainly involving p53-Mdm2 and ATM-p53-Wip1 negative feedbacks, which are sufficient for oscillations experimentally, with no further need to introduce any delays into the protein responses and without considering additional positive feedback.
Collapse
Affiliation(s)
- Ján Eliaš
- UPMC, Laboratoire Jacques-Louis Lions, 4 Place Jussieu, F-75005 Paris, France & INRIA Paris-Rocquencourt, MAMBA project-team, Paris and Rocquencourt, France
| | | | | | | |
Collapse
|
42
|
Li KC, Hua KT, Lin YS, Su CY, Ko JY, Hsiao M, Kuo ML, Tan CT. Inhibition of G9a induces DUSP4-dependent autophagic cell death in head and neck squamous cell carcinoma. Mol Cancer 2014; 13:172. [PMID: 25027955 PMCID: PMC4107555 DOI: 10.1186/1476-4598-13-172] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 07/07/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a common cancer worldwide. Emerging evidence indicates that alteration of epigenetics might be a key event in HNSCC progression. Abnormal expression of histone methyltransferase G9a, which contributes to transcriptional repression of tumor suppressors, has been implicated in promoting cancerous malignancies. However, its role in HNSCC has not been previously characterized. In this study, we elucidate the function of G9a and its downstream mechanism in HNSCC. METHODS We investigated the clinical relevance of G9a in HNSCC using immunohistochemistry (IHC) staining. In vitro cell proliferation and tumorigenesis ability of G9a-manipulated HNSCC cells were examined with MTT assays, clonogenic assays, and soft agar assays. We examined different routes of cell death in HNSCC cells induced by G9a-depletion or enzymatic inhibition by immunoblot, flow cytometry, fluorescent and transmission electron microscopy analysis. Specific targets of G9a were identified by affymetrix microarray and quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Lastly, functions of G9a in vivo were confirmed with a xenograft tumor model. RESULTS G9a expression is positively correlated to proliferation marker Ki-67 and to poor prognosis in HNSCC patients. Genetic or pharmacological inhibition of G9a reduced cell proliferation without inducing necrosis or apoptosis. Instead, autophagic cell death was the major consequence, and our investigation of mechanisms suggested it is mediated via the dual specificity phosphatase-4 (DUSP4) dependent ERK inactivation pathway. An orthotopic tumor model further confirmed the growth inhibiting effect and induction of autophagy that followed suppression of G9a. CONCLUSIONS In this study, we provide evidence that G9a confers the survival advantage of HNSCC. Genetic or pharmacological inhibition of G9a induces autophagic cell death; this finding provides a basis for new therapeutic targets for treating HNSCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
43
|
Eliaš J, Clairambault J. Reaction-diffusion systems for spatio-temporal intracellular protein networks: A beginner's guide with two examples. Comput Struct Biotechnol J 2014; 10:12-22. [PMID: 25210594 PMCID: PMC4151873 DOI: 10.1016/j.csbj.2014.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Spatio-temporal dynamics of a variety of proteins is, among other things, regulated by post-translational modifications of these proteins. Such modifications can thus influence stability and biochemical activities of the proteins, activity and stability of their upstream targets within specific signalling pathways. Commonly used mathematical tools for such protein–protein (and/or protein-mRNA) interactions in single cells, namely, Michaelis–Menten and Hill kinetics, yielding a system of ordinary differential equations, are extended here into (non-linear) partial differential equations by taking into account a more realistic spatial representation of the environment where these reactions occur. In the modelling framework under consideration, all interactions occur in a cell divided into two compartments, the nucleus and the cytoplasm, connected by the semipermeable nuclear membrane and bounded by the impermeable cell membrane. Passive transport mechanism, modelled by the so-called Kedem–Katchalsky boundary conditions, is used here to represent migration of species throughout the nuclear membrane. Nonlinear systems of partial differential equations are solved by the semi-implicit Rothe method. Examples of two spatial oscillators are shown. Namely, these are the circadian rhythm for concentration of the FRQ protein in Neurospora crassa and oscillatory dynamics observed in the activation and regulation of the p53 protein following DNA damage in mammalian cells.
Collapse
Affiliation(s)
- Ján Eliaš
- Université Pierre et Marie Curie Paris 06, Sorbonne Universités, Laboratoire Jacques-Louis Lions, boîte courrier 187, F75253 Cedex 05, Paris, France
| | - Jean Clairambault
- Université Pierre et Marie Curie Paris 06, Sorbonne Universités, Laboratoire Jacques-Louis Lions, boîte courrier 187, F75253 Cedex 05, Paris, France
| |
Collapse
|
44
|
Zhang W, Liu N, Wang X, Jin X, Du H, Peng G, Xue J. Benzo(a)pyrene-7,8-diol-9,10-epoxide induced p53-independent necrosis via the mitochondria-associated pathway involving Bax and Bak activation. Hum Exp Toxicol 2014; 34:179-90. [PMID: 24837741 DOI: 10.1177/0960327114533358] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE) is a highly reactive DNA damage agent and can induce cell death through both p53-independent and -dependent pathways. However, little is known about the molecular mechanisms of p53-independent pathways in BPDE-induced cell death. To understand the p53-independent mechanisms, we have now examined BPDE-induced cytotoxicity in p53-deficient baby mouse kidney (BMK) cells. The results showed that BPDE could induce Bax and Bak activation, cytochrome c release, caspases activation, and necrotic cell death in the BMK cells. Bax and Bak, two key molecules of mitochondrial permeability transition pore, were interdependently activated by BPDE, with Bax and Bak translocation to and Bax/Bak homo-oligomerization in mitochondria, release of cytochrome c was induced. Importantly, cytochrome c release and necrotic cell death were diminished in BMK cells (Bax−/−), BMK cells (Bak−/−), and BMK cells (Bax−/−/Bak−/−). Furthermore, overexpression of Bcl-2 could ameliorate BPDE-induced cytochrome c release and necrosis. Together the findings suggested that BPDE-induced necrosis was modulated by the p53-independent pathway, which was related to the translocation of Bax and Bak to mitochondria, release of cytochrome c, and activation of caspases.
Collapse
Affiliation(s)
- W Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - N Liu
- Department of General Surgery, Hainan Provincial People Hospital, Haikou, China
| | - X Wang
- Department of Vascular Surgery, Xuzhou Central Hospital, Xuzhou, China
| | - X Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - H Du
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - G Peng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Xue
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Dynamics of posttranslational modifications of p53. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2014; 2014:245610. [PMID: 24899917 PMCID: PMC4037116 DOI: 10.1155/2014/245610] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 04/06/2014] [Indexed: 11/25/2022]
Abstract
The latest experimental evidence indicates that acetylation of p53 at K164 (lysine 164) and K120 may induce directly cell apoptosis under severe DNA damage. However, previous cell apoptosis models only studied the effects of active and/or inactive p53, that is, phosphorylation/dephosphorylation of p53. In the present paper, based partly on Geva-Zatorsky et al. (2006) and Batchelor et al. (2008), we propose a new cell apoptosis network, in which p53 has three statuses, that is, unphosphorylated p53, phosphorylated p53, and acetylated p53. The time delay differential equations (DDEs) are formulated based on our network to investigate the dynamical insights of p53-induced cell apoptosis. In agreement with experiments (Loewer et al. (2010)), our simulations indicate that acetylated p53 accumulates gradually and then induces the proapoptotic protein Bax under enough DNA damage. Moreover, phosphorylated p53 oscillates and initiates cell repair during DNA damage.
Collapse
|