1
|
Hobsteter AW, Irazoqui AP, Gonzalez A, Picco AS, Rubert AA, Buitrago CG, Lo Fiego MJ, Silbestri GF. Acetylated galactopyranosyl N-heterocyclic monocarbene complexes of Silver(I) as novel anti-proliferative agents in a rhabdomyosarcoma cell line. Bioorg Med Chem 2024; 107:117756. [PMID: 38759255 DOI: 10.1016/j.bmc.2024.117756] [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: 04/15/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Herein, four silver(I) complexes bearing acetylated d-galactopyranoside-based N-heterocyclic carbene ligands were synthesized and fully characterized by elemental analysis, NMR, and X-ray photoelectron spectroscopy. All complexes were obtained with an anomeric β-configuration and as monocarbene species. In this study, we investigated the biological effects of the silver(I) complexes 2a-d on the human rhabdomyosarcoma cell line, RD. Our results show concentration-dependent effects on cell density, growth inhibition, and activation of key signaling pathways such as Akt 1/2, ERK 1/2, and p38-MAPK, indicating their potential as anticancer agents. Notably, at 35.5 µM, the complexes induced mitochondrial network disruption, as observed with 2b and 2c, whereas with 2a, this disruption was accompanied by nuclear content release. These results provide insight into the utility of carbohydrate incorporated NHC complexes of silver(I) as new agents in cancer therapy.
Collapse
Affiliation(s)
- Ariana W Hobsteter
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina
| | - Ana P Irazoqui
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina; Comisión de Investigaciones Científicas de la provincia de Buenos Aires (CIC PBA), Argentina.
| | - Agustina Gonzalez
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - Agustín S Picco
- INIFTA, Fac. de Cs. Exactas, Universidad Nacional de La Plata-CONICET, 1900 La Plata, Argentina
| | - Aldo A Rubert
- INIFTA, Fac. de Cs. Exactas, Universidad Nacional de La Plata-CONICET, 1900 La Plata, Argentina
| | - Claudia G Buitrago
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - Marcos J Lo Fiego
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina.
| | - Gustavo F Silbestri
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina
| |
Collapse
|
2
|
Naeem A, Knoer G, Avantaggiati ML, Rodriguez O, Albanese C. Provocative non-canonical roles of p53 and AKT signaling: A role for Thymosin β4 in medulloblastoma. Int Immunopharmacol 2023; 116:109785. [PMID: 36720193 DOI: 10.1016/j.intimp.2023.109785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/30/2023]
Abstract
The PI3K/AKT and p53 pathways are key regulators of cancer cell survival and death, respectively. Contrary to their generally accepted roles, several lines of evidence, including ours in medulloblastoma, the most common childhood brain cancer, highlight non-canonical functions for both proteins and show a complex context-dependent dynamic behavior in determining cell fate. Interestingly, p53-mediated cell survival and AKT-mediated cell death can dominate in certain conditions, and these interchangeable physiological functions may potentially be manipulated for better clinical outcomes. This review article presents studies in which p53 and AKT behave contrary to their well-established functions. We discuss the factors and circumstances that may be involved in mediating these changes and the implications of these unique roles of p53 and AKT in devising therapeutic strategies. Lastly, based on our recent finding of Thymosin beta 4-mediated chemosensitivity via an AKT-p53 interaction in medulloblastoma cells, we also discuss the possible implications of Thymosin beta-4 in enhancing drug sensitivity in this deadly childhood disease.
Collapse
Affiliation(s)
- Aisha Naeem
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Health Research Governance Department, Ministry of Public Health, Qatar.
| | - Grace Knoer
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Maria Laura Avantaggiati
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Olga Rodriguez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Center for Translational Imaging, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Chris Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Radiology, Georgetown University Medical Center, Washington, DC 20057, USA; Center for Translational Imaging, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
3
|
Zhou Y, Ji M, Xia Y, Han X, Li M, Li W, Sun T, Zhang J, Lu F, Sun Y, Liu N, Li J, Ma D, Ye J, Ji C. Silencing of IRF8 Mediated by m6A Modification Promotes the Progression of T-Cell Acute Lymphoblastic Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2201724. [PMID: 36478193 PMCID: PMC9839875 DOI: 10.1002/advs.202201724] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/11/2022] [Indexed: 06/17/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy with a poor prognosis, urging for novel therapeutic targets and treatment strategies. N6-methyladenosine (m6A) is a crucial methylation modification that affects the pathogenesis of leukemia by regulating the mRNA of key genes. Interferon regulatory factor 8 (IRF8) is a crucial transcription factor for hematological lineage commitment, but its role in T-ALL is unclear. Here, IRF8 is shown to suppress T-ALL. The expression of IRF8 is abnormally silenced in patients with T-ALL. Knockout of Irf8 significantly hastens the progression of Notch1-induced T-ALL in vivo. Overexpression of IRF8 suppresses the proliferation and invasion of T-ALL cells by inhibiting the phosphatidylinositol 3-kinase/AKT signaling pathway. The fat mass- and obesity-associated protein (FTO), an m6A demethylase, is responsible for directly binding to m6A sites in 3' untranslated region of IRF8 messenger RNA (mRNA) and inducing mRNA degradation via m6A modification. Targeting the FTO-IRF8 axis is used as a proof of concept therapy; inhibition of FTO's demethylase activity drastically alleviates the proliferation of leukemic cells and prolongs the survival of T-ALL mice by restoring IRF8 expression. This study elucidates the pathogenesis of T-ALL from the perspective of epitranscriptomics and provides new insight into the genetic mechanisms and targeted therapy of T-ALL.
Collapse
Affiliation(s)
- Ying Zhou
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Min Ji
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Yuan Xia
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Xiaoyu Han
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Mingying Li
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Wei Li
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Tao Sun
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
- Key Laboratory of ImmunohematologyQilu HospitalShandong UniversityJinan250012P. R. China
| | - Jingru Zhang
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Fei Lu
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Yanping Sun
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Na Liu
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Jingxin Li
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Daoxin Ma
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
- Key Laboratory of ImmunohematologyQilu HospitalShandong UniversityJinan250012P. R. China
| | - Jingjing Ye
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| | - Chunyan Ji
- Department of HematologyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012P. R. China
| |
Collapse
|
4
|
Diwan D, Cheng L, Usmani Z, Sharma M, Holden N, Willoughby N, Sangwan N, Baadhe RR, Liu C, Gupta VK. Microbial cancer therapeutics: A promising approach. Semin Cancer Biol 2022; 86:931-950. [PMID: 33979677 DOI: 10.1016/j.semcancer.2021.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/24/2021] [Accepted: 05/04/2021] [Indexed: 01/27/2023]
Abstract
The success of conventional cancer therapeutics is hindered by associated dreadful side-effects of antibiotic resistance and the dearth of antitumor drugs' selectivity and specificity. Hence, the conceptual evolution of anti-cancerous therapeutic agents that selectively target cancer cells without impacting the healthy cells or tissues, has led to a new wave of scientific interest in microbial-derived bioactive molecules. Such strategic solutions may pave the way to surmount the shortcomings of conventional therapies and raise the potential and hope for the cure of wide range of cancer in a selective manner. This review aims to provide a comprehensive summary of anti-carcinogenic properties and underlying mechanisms of bioactive molecules of microbial origin, and discuss the current challenges and effective therapeutic application of combinatorial strategies to attain minimal systemic side-effects.
Collapse
Affiliation(s)
- Deepti Diwan
- Washington University, School of Medicine, Saint Louis, MO, USA
| | - Lei Cheng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 230032, China
| | - Zeba Usmani
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Minaxi Sharma
- Department of Food Technology, Akal College of Agriculture, Eternal University, Baru Sahib, Himachal Pradesh, 173101, India
| | - Nicola Holden
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK
| | - Nicholas Willoughby
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Neelam Sangwan
- Department of Biochemistry, Central University of Haryana, Mahendergarh, Haryana, 123031, India
| | - Rama Raju Baadhe
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Vijai Kumar Gupta
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK; Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK.
| |
Collapse
|
5
|
Shati AA, El-Kott AF, Alkhateeb MA. Resolvin D1 prevents cadmium chloride-induced memory loss and hippocampal damage in rats by activation/upregulation of PTEN-induced suppression of PI3K/Akt/mTOR signaling pathway. Clin Exp Pharmacol Physiol 2021; 49:275-290. [PMID: 34570918 DOI: 10.1111/1440-1681.13596] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/16/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022]
Abstract
This study evaluated the protective effect of resolvin D1 (RVD1) against cadmium chloride (CdCl2 )-induced hippocampal damage and memory loss in rats and investigated if such protection is mediated by modulating the PTEN/PI3K/Akt/mTOR pathway. Adult male Wistar rats (n = 18/group) were divided as control, control + RVD1, CdCl2 , CdCl2 + RVD1 and CdCl2 + RVD1 + bpV(pic), a PTEN inhibitor. All treatments were conducted for 4 weeks. Resolvin D1 improved the memory function as measured by Morris water maze (MWM), preserved the structure of CA1 area of the hippocampus, and increased hippocampal levels of RVD1 in the CdCl2 -treated rats. Resolvin D1 also suppressed the generation of reactive oxygen species (ROS), tumour necrosis factor-α and interleukine-6 (IL-6), inhibited nuclear factor κB (NF-κB) p65, stimulated levels of glutathione (GSH), manganese superoxide dismutase (MnSOD), and Bcl2 but reduced the expression of Bax and cleaved caspase 3 in hippocampi of CdCl2 -treated rats. Concomitantly, it stimulated levels and activity of PTEN and reduced the phosphorylation (activation) of PI3K, Akt and mTOR in hippocampi of CdCl2 -treated rats. In conclusion, RVD1 attenuates CdCl2 -induced memory loss and hippocampal damage in rats mainly by activating PTEN-induced suppression of PI3K/Akt/mTOR, an effect that seems secondary to its' anti-oxidant and anti-inflammatory potential.
Collapse
Affiliation(s)
- Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Mahmoud A Alkhateeb
- Department of Basic Medical Sciences, College of Medicine, King Saud University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Zhang Y, Zhang X, Cheng A, Wang M, Yin Z, Huang J, Jia R. Apoptosis Triggered by ORF3 Proteins of the Circoviridae Family. Front Cell Infect Microbiol 2021; 10:609071. [PMID: 33604306 PMCID: PMC7884757 DOI: 10.3389/fcimb.2020.609071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Apoptosis, a form of the programmed cell death, is an indispensable defense mechanism regulating cellular homeostasis and is triggered by multiple stimuli. Because of the regulation of apoptosis in cellular homeostasis, viral proteins with apoptotic activity are particular foci of on antitumor therapy. One representative viral protein is the open reading frame 3 (ORF3) protein, also named as apoptin in the Circoviridae chicken anemia virus (CAV), and has the ability to induce tumor-specific apoptosis. Proteins encoded by ORF3 in other circovirus species, such as porcine circovirus (PCV) and duck circovirus (DuCV), have also been reported to induce apoptosis, with subtle differences in apoptotic activity based on cell types. This article is aimed at reviewing the latest research advancements in understanding ORF3 protein-mediated apoptosis mechanisms of Circoviridae from three perspectives: subcellular localization, interactions with host proteins, and participation in multiple apoptotic signaling pathways, providing a scientific basis for circovirus pathogenesis and a reference on its potential anticancer function.
Collapse
Affiliation(s)
- Yanting Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xingcui Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
7
|
Niesler N, Arndt J, Silberreis K, Fuchs H. Generation of a soluble and stable apoptin-EGF fusion protein, a targeted viral protein applicable for tumor therapy. Protein Expr Purif 2020; 175:105687. [PMID: 32681952 DOI: 10.1016/j.pep.2020.105687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 11/18/2022]
Abstract
A promising candidate for tumor targeted toxins is the chicken anemia-derived protein apoptin that induces tumor-specific apoptosis. It was aimed to design a novel apoptin-based targeted toxin by genetic fusion of apoptin with the tumor-directed ligand epidermal growth factor (EGF) using Escherichia coli as expression host. However, apoptin is highly hydrophobic and tends to form insoluble aggregates. Therefore, three different apoptin-EGF variants were generated. The fusion protein hexa-histidine (His)-apoptin-EGF (HAE) was expressed in E. coli and purified under denaturing conditions due to inclusion bodies. The protein solubility was improved by maltose-binding protein (MBP) or glutathione S-transferase. The protein MBP-apoptin-EGFHis (MAEH) was found favorable as a targeted toxin regarding final yield (4-6 mg/L) and stability. MBP was enzymatically removed using clotting factor Xa, which resulted in low yield and poor separation. MAEH was tested on target and non-target cell lines. The targeted tumor cell line A431 showed significant toxicity with an IC50 of 69.55 nM upon incubation with MAEH while fibroblasts and target receptor-free cells remained unaffected. Here we designed a novel EGF receptor targeting drug with high yield, purity and stability.
Collapse
Affiliation(s)
- Nicole Niesler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany
| | - Janine Arndt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Kim Silberreis
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Hendrik Fuchs
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
8
|
Malla WA, Arora R, Khan RIN, Mahajan S, Tiwari AK. Apoptin as a Tumor-Specific Therapeutic Agent: Current Perspective on Mechanism of Action and Delivery Systems. Front Cell Dev Biol 2020; 8:524. [PMID: 32671070 PMCID: PMC7330108 DOI: 10.3389/fcell.2020.00524] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer remains one of the leading causes of death worldwide in humans and animals. Conventional treatment regimens often fail to produce the desired outcome due to disturbances in cell physiology that arise during the process of transformation. Additionally, development of treatment regimens with no or minimum side-effects is one of the thrust areas of modern cancer research. Oncolytic viral gene therapy employs certain viral genes which on ectopic expression find and selectively destroy malignant cells, thereby achieving tumor cell death without harming the normal cells in the neighborhood. Apoptin, encoded by Chicken Infectious Anemia Virus' VP3 gene, is a proline-rich protein capable of inducing apoptosis in cancer cells in a selective manner. In normal cells, the filamentous Apoptin becomes aggregated toward the cell margins, but is eventually degraded by proteasomes without harming the cells. In malignant cells, after activation by phosphorylation by a cancer cell-specific kinase whose identity is disputed, Apoptin accumulates in the nucleus, undergoes aggregation to form multimers, and prevents the dividing cancer cells from repairing their DNA lesions, thereby forcing them to undergo apoptosis. In this review, we discuss the present knowledge about the structure of Apoptin protein, elaborate on its mechanism of action, and summarize various strategies that have been used to deliver it as an anticancer drug in various cancer models.
Collapse
Affiliation(s)
- Waseem Akram Malla
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Richa Arora
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Sonalika Mahajan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Ashok Kumar Tiwari
- Division of Biological Standardisation, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| |
Collapse
|
9
|
Feng C, Liang Y, Teodoro JG. The Role of Apoptin in Chicken Anemia Virus Replication. Pathogens 2020; 9:pathogens9040294. [PMID: 32316372 PMCID: PMC7238243 DOI: 10.3390/pathogens9040294] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 12/16/2022] Open
Abstract
Apoptin is the Vp3 protein of chicken anemia virus (CAV), which infects the thymocytes and erythroblasts in young chickens, causing chicken infectious anemia and immunosuppression. Apoptin is highly studied for its ability to selectively induce apoptosis in human tumor cells and, thus, is a protein of interest in anti-tumor therapy. CAV apoptin is known to localize to different subcellular compartments in transformed and non-transformed cells, depending on the DNA damage response, and the phosphorylation of several identified threonine residues. In addition, apoptin interacts with molecular machinery such as the anaphase promoting complex/cyclosome (APC/C) to inhibit the cell cycle and induce arrest in G2/M phase. While these functions of apoptin contribute to the tumor-selective effect of the protein, they also provide an important fundamental framework to apoptin’s role in viral infection, pathogenesis, and propagation. Here, we reviewed how the regulation, localization, and functions of apoptin contribute to the viral life cycle and postulated its importance in efficient replication of CAV. A model of the molecular biology of infection is critical to informing our understanding of CAV and other related animal viruses that threaten the agricultural industry.
Collapse
Affiliation(s)
- Cynthia Feng
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Yingke Liang
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Jose G. Teodoro
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
- Goodman Cancer Research Centre, Montreal, QC H3G 1A1, Canada
- Correspondence:
| |
Collapse
|
10
|
Cancer Treatment Goes Viral: Using Viral Proteins to Induce Tumour-Specific Cell Death. Cancers (Basel) 2019; 11:cancers11121975. [PMID: 31817939 PMCID: PMC6966515 DOI: 10.3390/cancers11121975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/24/2022] Open
Abstract
Cell death is a tightly regulated process which can be exploited in cancer treatment to drive the killing of the tumour. Several conventional cancer therapies including chemotherapeutic agents target pathways involved in cell death, yet they often fail due to the lack of selectivity they have for tumour cells over healthy cells. Over the past decade, research has demonstrated the existence of numerous proteins which have an intrinsic tumour-specific toxicity, several of which originate from viruses. These tumour-selective viral proteins, although from distinct backgrounds, have several similar and interesting properties. Though the mechanism(s) of action of these proteins are not fully understood, it is possible that they can manipulate several cell death modes in cancer exemplifying the intricate interplay between these pathways. This review will discuss our current knowledge on the topic and outstanding questions, as well as deliberate the potential for viral proteins to progress into the clinic as successful cancer therapeutics.
Collapse
|
11
|
Liang J, Lu F, Li B, Liu L, Zeng G, Zhou Q, Chen L. IRF8 induces senescence of lung cancer cells to exert its tumor suppressive function. Cell Cycle 2019; 18:3300-3312. [PMID: 31594449 DOI: 10.1080/15384101.2019.1674053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. However, tumor suppressor genes remain to be systemically determined for lung cancer. Here we report interferon regulatory factor 8 (IRF8), a member of the IRF family of transcription factors, as a potent lung tumor suppressor gene. Expression of IRF8 is frequently diminished in lung tumoral tissues and is associated with prognosis of non-small cell lung cancer (NSCLC) patients. Ectopic expression of IRF8 suppresses the NSCLC cells proliferation in vitro and tumorigenic potential in vivo. More importantly, forced expression of IRF8 through infection of recombinant virus inhibits lung tumorigenesis in genetically engineered mouse model (GEMM). Mechanistically, IRF8 inhibits AKT signaling and promotes accumulation of P27 protein, which results in senescence of lung cancer cells. Ectopic expression of IRF8 in tumor cells leads to regression of lung cancer tumor nodules in a xenograft tumor model. Our data, therefore, solidly shows IRF8 to be a lung cancer suppressor gene and may denote an opportunity for therapeutic intervention of NSCLC.
Collapse
Affiliation(s)
- Jinxia Liang
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Feng Lu
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Bo Li
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Lu Liu
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Guandi Zeng
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Qian Zhou
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Liang Chen
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| |
Collapse
|
12
|
Akbari A, Arabsolghar R, Behzad Behbahani A, Rafiei Dehbidi G, Zare F, Hadi M. Human Gyrovirus Apoptin as a Potential Selective Anticancer Agent: An In Vitro Study. PHARMACEUTICAL SCIENCES 2019. [DOI: 10.15171/ps.2019.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Selective therapy has always been the main challenge in cancer treatments. Recently, it has been shown that Human Gyrovirus-derived protein apoptin (HGV-Apoptin) has selective cytotoxic effects on cancer cells similar to its homologue, Chicken Anemia Virus-derived Apoptin (CAV-Apoptin). However, apoptotic effects of Human Gyrovirus apoptin have been only evaluated on a few cancerous cell lines and need to be further investigated. In this study, we have evaluated the apoptotic effects of HGV-Apoptin and CAV-Apoptin expression on lung cancer (A549) and normal (HEK-293) cell lines, in order to provide more information about the specificity of these proteins on cancerous cells. Methods: Target cells were transfected by the calcium-phosphate precipitation method with constructed plasmids expressing HGV-Apoptin and CAV-Apoptin proteins as well as the control plasmid. Transfection efficiency was followed and imaged by fluorescence microscopy. Quantification of apoptosis was performed by flow cytometry. Measurements were compared by paired Student t-test. Results: Cells were successfully transfected with control and constructed plasmids. Flowcytometry analysis showed that A549 cells transfected with HGV-Apoptin and CAV-Apoptin expressing plasmids, undergone the apoptosis compared to A549 cells transfected with control plasmid (P<0.001). None of the plasmids could induce apoptosis in HEK-293 cells. Conclusion: Human Gyrovirus-derived apoptin (HGV-Apoptin) similar to its homologue, chicken anemia virus derived Apoptin (CAV-Apoptin) can induce apoptosis in Non-small-cell lung carcinoma cell line A549, but not in normal human embryonic kidney cell line HEK-293, which can be introduced as a promising novel specific antitumor agent.
Collapse
Affiliation(s)
- Amir Akbari
- Department of Medical Laboratory Science, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rita Arabsolghar
- Department of Medical Laboratory Science, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Behzad Behbahani
- Department of Medical Laboratory Science, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Rafiei Dehbidi
- Department of Medical Laboratory Science, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farahnaz Zare
- Department of Medical Laboratory Science, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahdieh Hadi
- Department of Medical Laboratory Science, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Kawahara R, Rosa-Fernandes L, Dos Santos AF, Bandeira CL, Dombrowski JG, Souza RM, Da Fonseca MP, Festuccia WT, Labriola L, Larsen MR, Marinho CRF, Palmisano G. Integrated Proteomics Reveals Apoptosis-related Mechanisms Associated with Placental Malaria. Mol Cell Proteomics 2019; 18:182-199. [PMID: 30242111 PMCID: PMC6356084 DOI: 10.1074/mcp.ra118.000907] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/15/2018] [Indexed: 12/27/2022] Open
Abstract
Malaria in pregnancy is a public health concern in malaria-endemic areas. Accumulation of maternal immune cells in the placenta and increased levels of inflammatory cytokines caused by sequestration of Plasmodium falciparum-infected erythrocytes have been associated to poor neonatal outcomes, including low birth weight because of fetal growth restriction. Little is known about the molecular changes occurring in a P. falciparum-infected placenta that has developed placental malaria during pregnancy but had the parasites cleared by pharmacological treatment (past infection). We conducted an integrated proteome, phosphoproteome and glycoproteome analysis in past P. falciparum-infected placentas aiming to find molecular changes associated with placental malaria. A total of 2946 proteins, 1733 N-linked glycosites and 4100 phosphosites were identified and quantified in this study, disclosing overrepresented processes related to oxidative stress, protein folding and regulation of apoptosis in past-infected placentas Moreover, AKT and ERK signaling pathways activation, together with clinical data, were further correlated to an increased apoptosis in past-infected placentas. This study showed apoptosis-related mechanisms associated with placental malaria that can be further explored as therapeutic target against adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Rebeca Kawahara
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Livia Rosa-Fernandes
- Department of Biochemistry and Molecular biology, University of Southern Denmark, Odense, Denmark
| | | | - Carla Letícia Bandeira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Jamille G Dombrowski
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Rodrigo M Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | | | - William T Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | - Leticia Labriola
- Department of Biochemistry, Chemistry Institute, University of São Paulo, Brazil
| | - Martin R Larsen
- Department of Biochemistry and Molecular biology, University of Southern Denmark, Odense, Denmark
| | - Claudio R F Marinho
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil;.
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil;.
| |
Collapse
|
14
|
Brown AC, Reddy VRAP, Lee J, Nair V. Marek's disease virus oncoprotein Meq physically interacts with the chicken infectious anemia virus-encoded apoptotic protein apoptin. Oncotarget 2018; 9:28910-28920. [PMID: 29988968 PMCID: PMC6034753 DOI: 10.18632/oncotarget.25628] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/31/2018] [Indexed: 12/17/2022] Open
Abstract
Marek's disease (MD) is a neoplastic disease of poultry caused by Marek's disease virus (MDV), a highly contagious alphaherpesvirus. Meq, the major MDV oncoprotein, induces neoplastic transformation of T-cells through several mechanisms, including inhibition of apoptosis. In contrast, the chicken anemia virus (CAV)-encoded protein apoptin (VP3) is a powerful inducer of apoptosis of tumor cells, a property that is exploited for anticancer therapeutics. Although the molecular mechanisms of selective induction of tumor cell apoptosis by apoptin are not fully understood, its tumor cell–restricted nuclear translocation is thought to be important. Co-infection with MDV and CAV is common in many countries, CAV antigens are readily detectable in MD lymphomas, and the MDV-transformed T-lymphoblastoid cell lines such as MSB-1 is widely used for propagating CAV for vaccine production. As MDV-transformed cell lines express high levels of Meq, we examined here whether CAV-encoded apoptin interacts with Meq in these cells. Using immunofluorescence microscopy, we found that apoptin and Meq co-localize to the nucleus, and biochemical analysis indicated that the two proteins do physically interact. Using a combination of Meq mutagenesis and co-immunoprecipitation, we demonstrate that apoptin interacts with Meq within a region between amino acids 130 and 140. Results from the IncuCyte assay suggested that Meq inhibits apoptin-induced apoptosis activity. In summary, our findings indicate that Meq interacts with and inhibits apoptin. Insights into this novel interaction between Meq and apoptin will relevance for pathogenesis of coinfections of the two viruses and in CAV vaccine production using MDV-transformed cell lines.
Collapse
Affiliation(s)
- Andrew C Brown
- Wellcome Trust Centre for Human Genetics, Oxford, OX3 7BN, UK
| | | | - Joshua Lee
- Bristol University, Bristol, BS8 1TH, UK
| | - Venugopal Nair
- Viral Oncogenesis Group, The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
| |
Collapse
|
15
|
Human Gyrovirus-Apoptin Interferes with the Cell Cycle and Induces G2/M Arrest Prior to Apoptosis. Arch Immunol Ther Exp (Warsz) 2017; 65:545-552. [DOI: 10.1007/s00005-017-0464-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/20/2017] [Indexed: 01/06/2023]
|
16
|
Insights into the mechanism of Apoptin's exquisitely selective anti-tumor action from atomic level characterization of its conformation and dynamics. Arch Biochem Biophys 2016; 614:53-64. [PMID: 28034642 DOI: 10.1016/j.abb.2016.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 01/08/2023]
Abstract
Apoptin is a 121 residue protein which forms large, soluble aggregates and possesses an exceptionally selectively cytotoxic action on cancer cells. In the accompanying paper, we described the design, production and initial characterization of an Apoptin truncated variant called H6-ApopΔProΔLeu. Whereas both the variant and wild type protein possess similar selective cytotoxicity against cancer cells following transfection, only the variant is cytotoxic when added externally. Remarkably, as observed by gel filtration chromatography and dynamic light scattering, H6-ApopΔProΔLeu lacks the tendency of wild type Apoptin to form large aggregates, which greatly facilitated the study of its biological properties. Here, we characterize the conformation and dynamics of H6-ApopΔProΔLeu. Using a battery of 2D, 3D and (4,2)D NMR spectra, the essentially complete 1H, 13C and 15N resonance assignments of H6-ApopΔProΔLeu were obtained. The analysis of these data shows that the variant is an intrinsically disordered protein, which lacks a preferred conformation. This conclusion is corroborated by a lack of protection against proteolytic cleavage and hydrogen/deuterium exchange. Moreover, the CD spectra are dominated by random coil contributions. Finally, 1H-15N NOE ratios are low, which indicates flexibility on the ps-ns time scale. Interestingly, H6-ApopΔProΔLeu's intrinsically disordered ensemble is not significantly altered by the redox state of its Cys residues or by Thr phosphorylation, which has been proposed to play a key role in Apoptin's selective cytotoxicity. These results serve to better comprehend Apoptin's remarkably selective anticancer action and provide a framework for the future design of improved Apoptin variants.
Collapse
|
17
|
Activation of the Chicken Anemia Virus Apoptin Protein by Chk1/2 Phosphorylation Is Required for Apoptotic Activity and Efficient Viral Replication. J Virol 2016; 90:9433-45. [PMID: 27512067 DOI: 10.1128/jvi.00936-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/03/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Chicken anemia virus (CAV) is a single-stranded circular DNA virus that carries 3 genes, the most studied of which is the gene encoding VP3, also known as apoptin. This protein has been demonstrated to specifically kill transformed cells while leaving normal cells unharmed in a manner that is independent of p53 status. Although the mechanistic basis for this differential activity is unclear, it is evident that the subcellular localization of the protein is important for the difference. In normal cells, apoptin exists in filamentous networks in the cytoplasm, whereas in transformed cells, apoptin is present in the nucleus and appears as distinct foci. We have previously demonstrated that DNA damage signaling through the ataxia telangiectasia mutated (ATM) pathway induces the translocation of apoptin from the cytoplasm to the nucleus, where it induces apoptosis. We found that apoptin contains four checkpoint kinase consensus sites and that mutation of either threonine 56 or 61 to alanine restricts apoptin to the cytoplasm. Furthermore, treatment of tumor cells expressing apoptin with inhibitors of checkpoint kinase 1 (Chk1) and Chk2 causes apoptin to localize to the cytoplasm. Importantly, silencing of Chk2 rescues cancer cells from the cytotoxic effects of apoptin. Finally, treatment of virus-producing cells with Chk inhibitor protects them from virus-mediated toxicity and reduces the titer of progeny virus. Taken together, our results indicate that apoptin is a sensor of DNA damage signaling through the ATM-Chk2 pathway, which induces it to migrate to the nucleus during viral replication. IMPORTANCE The chicken anemia virus (CAV) protein apoptin is known to induce tumor cell-specific death when expressed. Therefore, understanding its regulation and mechanism of action could provide new insights into tumor cell biology. We have determined that checkpoint kinase 1 and 2 signaling is important for apoptin regulation and is a likely feature of both tumor cells and host cells producing virus progeny. Inhibition of checkpoint signaling prevents apoptin toxicity in tumor cells and attenuates CAV replication, suggesting it may be a future target for antiviral therapy.
Collapse
|
18
|
Rewiring of the apoptotic TGF-β-SMAD/NFκB pathway through an oncogenic function of p27 in human papillary thyroid cancer. Oncogene 2016; 36:652-666. [PMID: 27452523 DOI: 10.1038/onc.2016.233] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/29/2016] [Accepted: 05/24/2016] [Indexed: 12/25/2022]
Abstract
Papillary thyroid carcinoma (PTC), the most frequent thyroid cancer, is characterized by low proliferation but no apoptosis, presenting frequent lymph-node metastasis. Papillary thyroid carcinoma overexpress transforming growth factor-beta (TGF-β). In human cells, TGF-β has two opposing actions: antitumoral through pro-apoptotic and cytostatic activities, and pro-tumoral promoting growth and metastasis. The switch converting TGF-β from a tumor-suppressor to tumor-promoter has not been identified. In the current study, we have quantified a parallel upregulation of TGF-β and nuclear p27, a CDK2 inhibitor, in samples from PTC. We established primary cultures from follicular epithelium in human homeostatic conditions (h7H medium). TGF-β-dependent cytostasis occurred in normal and cancer cells through p15/CDKN2B induction. However, TGF-β induced apoptosis in normal and benign but not in carcinoma cultures. In normal thyroid cells, TGF-β/SMAD repressed the p27/CDKN1B gene, activating CDK2-dependent SMAD3 phosphorylation to induce p50 NFκB-dependent BAX upregulation and apoptosis. In thyroid cancer cells, oncogene activation prevented TGF-β/SMAD-dependent p27 repression, and CDK2/SMAD3 phosphorylation, leading to p65 NFκB upregulation which repressed BAX, induced cyclin D1 and promoted TGF-β-dependent growth. In PTC samples from patients, upregulation of TGF-β, p27, p65 and cyclin D1 mRNA were significantly correlated, while the expression of the isoform BAX-β, exclusively transcribed in apoptotic cells, was negatively correlated. Additionally, combined ERK and p65 NFκB inhibitors reduced p27 expression and potentiated apoptosis in thyroid cancer cells while not affecting survival in normal thyroid cells. Our results therefore suggest that the oncoprotein p27 reorganizes the effects of TGF-β in thyroid cancer, explaining the slow proliferation but lack of apoptosis and metastatic behavior of PTC.
Collapse
|
19
|
Could drugs inhibiting the mevalonate pathway also target cancer stem cells? Drug Resist Updat 2016; 25:13-25. [PMID: 27155373 DOI: 10.1016/j.drup.2016.02.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 12/12/2015] [Accepted: 01/28/2016] [Indexed: 02/07/2023]
Abstract
Understanding the connection between metabolic pathways and cancer is very important for the development of new therapeutic approaches based on regulatory enzymes in pathways associated with tumorigenesis. The mevalonate cascade and its rate-liming enzyme HMG CoA-reductase has recently drawn the attention of cancer researchers because strong evidences arising mostly from epidemiologic studies, show that it could promote transformation. Hence, these studies pinpoint HMG CoA-reductase as a candidate proto-oncogene. Several recent epidemiological studies, in different populations, have proven that statins are beneficial for the treatment-outcome of various cancers, and may improve common cancer therapy strategies involving alkylating agents, and antimetabolites. Cancer stem cells/cancer initiating cells (CSC) are key to cancer progression and metastasis. Therefore, in the current review we address the different effects of statins on cancer stem cells. The mevalonate cascade is among the most pleiotropic, and highly interconnected signaling pathways. Through G-protein-coupled receptors (GRCP), it integrates extra-, and intracellular signals. The mevalonate pathway is implicated in cell stemness, cell proliferation, and organ size regulation through the Hippo pathway (e.g. Yap/Taz signaling axis). This pathway is a prime preventive target through the administration of statins for the prophylaxis of obesity-related cardiovascular diseases. Its prominent role in regulation of cell growth and stemness also invokes its role in cancer development and progression. The mevalonate pathway affects cancer metastasis in several ways by: (i) affecting epithelial-to-mesenchymal transition (EMT), (ii) affecting remodeling of the cytoskeleton as well as cell motility, (iii) affecting cell polarity (non-canonical Wnt/planar pathway), and (iv) modulation of mesenchymal-to-epithelial transition (MET). Herein we provide an overview of the mevalonate signaling network. We then briefly highlight diverse functions of various elements of this mevalonate pathway. We further discuss in detail the role of elements of the mevalonate cascade in stemness, carcinogenesis, cancer progression, metastasis and maintenance of cancer stem cells.
Collapse
|
20
|
Bullenkamp J, Gäken J, Festy F, Chong EZ, Ng T, Tavassoli M. Apoptin interacts with and regulates the activity of protein kinase C beta in cancer cells. Apoptosis 2016; 20:831-42. [PMID: 25828882 DOI: 10.1007/s10495-015-1120-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Apoptin, the VP3 protein from chicken anaemia virus (CAV), induces tumour cell-specific cell death and represents a potential future anti-cancer therapeutic. In tumour but not in normal cells, Apoptin is phosphorylated and translocates to the nucleus, enabling its cytotoxic activity. Recently, the β isozyme of protein kinase C (PKCβ) was shown to phosphorylate Apoptin in multiple myeloma cell lines. However, the exact mechanism and nature of interaction between PKCβ and Apoptin remain unclear. Here we investigated the physical and functional link between PKCβ and CAV-Apoptin as well as with the recently identified Apoptin homologue derived from human Gyrovirus (HGyV). In contrast to HCT116 colorectal cancer cells the normal colon mucosa cell lines expressed low levels of PKCβI and showed reduced Apoptin activation, as evident by cytoplasmic localisation, decreased phosphorylation and lack of cytotoxic activity. Co-immunoprecipitation and proximity ligation assay studies identified binding of both CAV- and HGyV-Apoptin to PKCβI in HCT116 cells. Using Apoptin deletion constructs the N-terminal domain of Apoptin was found to be required for interacting with PKCβI. FRET-based PKC activity reporter assays by fluorescence lifetime imaging microscopy showed that expression of Apoptin in cancer cells but not in normal cells triggers a significant increase in PKC activity. Collectively, the results demonstrate a novel cancer specific interplay between Apoptin and PKCβI. Direct interaction between the two proteins leads to Apoptin-induced activation of PKC and consequently activated PKCβI mediates phosphorylation of Apoptin to promote its tumour-specific nuclear translocation and cytotoxic function.
Collapse
Affiliation(s)
- Jessica Bullenkamp
- Department of Molecular Oncology, King's College London, Guy's Campus, Hodgkin Building, London, SE1 1UL, UK
| | | | | | | | | | | |
Collapse
|
21
|
Gierut JJ, Wood LB, Lau KS, Lin YJ, Genetti C, Samatar AA, Lauffenburger DA, Haigis KM. Network-level effects of kinase inhibitors modulate TNF-α-induced apoptosis in the intestinal epithelium. Sci Signal 2015; 8:ra129. [PMID: 26671150 DOI: 10.1126/scisignal.aac7235] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Individual signaling pathways operate in the context of the broader signaling network. Thus, the response of a cell to signals from the environment is affected by the state of the signaling network, such as the clinically relevant example of whether some components in the network are inhibited. The cytokine tumor necrosis factor-α (TNF-α) promotes opposing cellular behaviors under different conditions; the outcome is influenced by the state of the network. For example, in the mouse intestinal epithelium, inhibition of the mitogen-activated protein kinase (MAPK) kinase MEK alters the timing of TNF-α-induced apoptosis. We investigated whether MAPK signaling directly influences TNF-α-induced apoptosis or whether network-level effects secondary to inhibition of the MAPK pathway alter the cellular response. We found that inhibitors of the MAPK kinase kinase Raf, MEK, or extracellular signal-regulated kinase (ERK) exerted distinct effects on the timing and magnitude of TNF-α-induced apoptosis in the mouse intestine. Furthermore, even different MEK inhibitors exerted distinct effects; one, CH5126766, potentiated TNF-α-induced apoptosis, and the others reduced cell death. Computational modeling and experimental perturbation identified the kinase Akt as the primary signaling node that enhanced apoptosis in the context of TNF-α signaling in the presence of CH5126766. Our work emphasizes the importance of integrated network signaling in specifying cellular behavior in response to experimental or therapeutic manipulation. More broadly, this study highlighted the importance of considering the network-level effects of pathway inhibitors and showed the distinct effects of inhibitors that share the same target.
Collapse
Affiliation(s)
- Jessica J Gierut
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Levi B Wood
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ken S Lau
- Department of Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Yi-Jang Lin
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Casie Genetti
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | | | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kevin M Haigis
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
22
|
Sabbineni H, Alwhaibi A, Goc A, Gao F, Pruitt A, Somanath PR. Genetic deletion and pharmacological inhibition of Akt1 isoform attenuates bladder cancer cell proliferation, motility and invasion. Eur J Pharmacol 2015; 764:208-214. [PMID: 26148825 DOI: 10.1016/j.ejphar.2015.06.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/09/2015] [Accepted: 06/29/2015] [Indexed: 01/17/2023]
Abstract
Isoform specific expression, intracellular localization and function of Akt in bladder cancer are not known. In the current study, we identified Akt1, followed by Akt2 and Akt3 as the predominant Akt isoform in human T24 and UM-UC-3 metastatic bladder cancer cells. Whereas Akt1 is localized at the membrane, cytoplasm and nucleus, Akt2 is solely cytoplasmic and Akt3 is mostly localized in the nucleus in T24 cells. ShRNA-mediated Akt1 knockdown resulted in impaired T24 cell survival, proliferation, colony formation, migration and microinvasion. Whereas pharmacological inhibition of Akt1 resulted in impaired T24 and UM-UC-3 cell motility, viability and proliferation, effect of pharmacological inhibition by Akt2 inhibitor was limited to proliferation in T24, but not UM-UC-3 cells. Our data provide important clues on the therapeutic benefits of targeting Akt1 for bladder cancer therapy.
Collapse
Affiliation(s)
- Harika Sabbineni
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Abdulrahman Alwhaibi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Anna Goc
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Fei Gao
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Alanna Pruitt
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States; Department of Medicine, Vascular Biology Center and Cancer Center, Georgia Regents University, Augusta, GA, United States.
| |
Collapse
|
23
|
Jain MV, Jangamreddy JR, Grabarek J, Schweizer F, Klonisch T, Cieślar-Pobuda A, Łos MJ. Nuclear localized Akt enhances breast cancer stem-like cells through counter-regulation of p21(Waf1/Cip1) and p27(kip1). Cell Cycle 2015; 14:2109-20. [PMID: 26030190 DOI: 10.1080/15384101.2015.1041692] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Cancer stem-like cells (CSCs) are a rare subpopulation of cancer cells capable of propagating the disease and causing cancer recurrence. In this study, we found that the cellular localization of PKB/Akt kinase affects the maintenance of CSCs. When Akt tagged with nuclear localization signal (Akt-NLS) was overexpressed in SKBR3 and MDA-MB468 cells, these cells showed a 10-15% increase in the number of cells with CSCs enhanced ALDH activity and demonstrated a CD44(+High)/CD24(-Low) phenotype. This effect was completely reversed in the presence of Akt-specific inhibitor, triciribine. Furthermore, cells overexpressing Akt or Akt-NLS were less likely to be in G0/G1 phase of the cell cycle by inactivating p21(Waf1/Cip1) and exhibited increased clonogenicity and proliferation as assayed by colony-forming assay (mammosphere formation). Thus, our data emphasize the importance the intracellular localization of Akt has on stemness in human breast cancer cells. It also indicates a new robust way for improving the enrichment and culture of CSCs for experimental purposes. Hence, it allows for the development of simpler protocols to study stemness, clonogenic potency, and screening of new chemotherapeutic agents that preferentially target cancer stem cells. SUMMARY The presented data, (i) shows new, stemness-promoting role of nuclear Akt/PKB kinase, (ii) it underlines the effects of nuclear Akt on cell cycle regulation, and finally (iii) it suggests new ways to study cancer stem-like cells.
Collapse
Key Words
- 7-AAD, 7-aminoactinomycin D
- ALDH, aldehyde dehydrogenase
- Akt-NLS
- BPE, bovine pituitary epithelial
- Bcl2, B cell lymphoma 2
- CDK, cyclin-dependent kinase
- CSCs, cancer stem-like cells
- DEAB, diethylaminobenzaldehyde
- FBS, fetal bovine serum
- GAPDH, glucose-6-phosphate dehydrogenase
- GPCR, G-protein-coupled receptor
- GSK3, glycogen synthase kinase-3
- IGF1, insulin like growth factor 1
- JAK, Janus kinase
- NLS, nuclear localization signal
- PDK, phosphoinositide dependent kinase
- PH, pleckstrin-homology
- PI3K
- PI3K, phoshatidylinositol-3-kinase
- PKB, protein kinase B
- PTEN, phosphatase and tensin homolog
- PVDF, polyvinylidene fluoride
- RIPA, radioimmunoprecipitation
- RPMI, Roswell Park Memorial Institute
- RT, room temperature
- RTK, receptor tyrosine kinase
- STAT, signal transducer and activator of transcription
- T-ALL, T-cell acute lymphoblastic leukemia
- WT, wild type
- cancer stem-like cells
- hEGF, human epidermal growth factor
- mTOR
- mTOR, mammalian target of rapamycin
- poly-HEMA, poly-2-hydroxyethyl methacrylate
- stemness
Collapse
Affiliation(s)
- Mayur Vilas Jain
- a Department of Clinical & Experimental Medicine; Division of Cell Biology Integrative Regenerative Med. Center (IGEN); Linköping Univ. ; Linköping , Sweden
| | | | | | | | | | | | | |
Collapse
|
24
|
Affiliation(s)
- Britta Merz
- a Department of Molecular Medicine; Interfaculty Institute for Biochemistry; University of Tübingen ; Tübingen , Germany
| | | | | |
Collapse
|
25
|
Langner E, Walczak K, Jeleniewicz W, Turski WA, Rajtar G. Quinaldic acid inhibits proliferation of colon cancer ht-29 cells in vitro: effects on signaling pathways. Eur J Pharmacol 2015; 757:21-7. [PMID: 25797283 DOI: 10.1016/j.ejphar.2015.02.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 10/23/2022]
Abstract
Quinaldic acid is presumed to be a derivative of kynurenic acid, a tryptophan metabolite with proven antiproliferative activity towards cancer cells in vitro. The aim of present study was to evaluate the activity of quinaldic acid in colon cancer cells. The antiproliferative potential of quinaldic acid was assessed in HT-29, LS180 and Caco-2 cells. Suppression of metabolic activity (IC50 of 0.5mM for HT-29 and LS180 cells, 0.9mM for Caco-2 cells) and DNA synthesis (IC50 of 2.7, 4.3, 2mM for HT-29, LS180 and Caco-2 cells, respectively) were observed in all tested cell lines. It is noteworthy that quinaldic acid in antiproliferative concentrations was non-toxic to normal colon epithelium CCD 841 CoTr cells. Concomitantly, alterations in several signaling pathways in HT-29 cells were observed. Quinaldic acid led to changes in the phosphorylation level of extracellular-signal-regulated kinase (ERK) 1/2, p38, cAMP response element-binding protein (CREB) and Akt (protein kinase B) kinases. Moreover, changes in the CREB transcription factor were also found at the gene expression level. Antiproliferative activity and signaling pathways modulatory potential of quinaldic acid in colon cancer cells in vitro has been stated.
Collapse
Affiliation(s)
- Ewa Langner
- Department of Pharmacology, Medical University in Lublin, Chodźki 4a, 20-093 Lublin, Poland; Department of Medical Biology, Institute of Agricultural Medicine, Jaczewskiego 2, 20-090 Lublin, Poland.
| | - Katarzyna Walczak
- Department of Pharmacology, Medical University in Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Witold Jeleniewicz
- Department of Biochemistry and Molecular Biology, Medical University in Lublin, Chodźki 1, 20-093 Lublin, Poland
| | - Waldemar A Turski
- Department of Experimental and Clinical Pharmacology, Medical University in Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Grażyna Rajtar
- Department of Pharmacology, Medical University in Lublin, Chodźki 4a, 20-093 Lublin, Poland
| |
Collapse
|
26
|
Gupta SK, Gandham RK, Sahoo AP, Tiwari AK. Viral genes as oncolytic agents for cancer therapy. Cell Mol Life Sci 2015; 72:1073-94. [PMID: 25408521 PMCID: PMC11113997 DOI: 10.1007/s00018-014-1782-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 10/29/2014] [Accepted: 11/13/2014] [Indexed: 12/20/2022]
Abstract
Many viruses have the ability to modulate the apoptosis, and to accomplish it; viruses encode proteins which specifically interact with the cellular signaling pathways. While some viruses encode proteins, which inhibit the apoptosis or death of the infected cells, there are viruses whose encoded proteins can kill the infected cells by multiple mechanisms, including apoptosis. A particular class of these viruses has specific gene(s) in their genomes which, upon ectopic expression, can kill the tumor cells selectively without affecting the normal cells. These genes and their encoded products have demonstrated great potential to be developed as novel anticancer therapeutic agents which can specifically target and kill the cancer cells leaving the normal cells unharmed. In this review, we will discuss about the viral genes having specific cancer cell killing properties, what is known about their functioning, signaling pathways and their therapeutic applications as anticancer agents.
Collapse
Affiliation(s)
- Shishir Kumar Gupta
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - Ravi Kumar Gandham
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. P. Sahoo
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. K. Tiwari
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| |
Collapse
|
27
|
Nastasie MS, Thissen H, Jans DA, Wagstaff KM. Enhanced tumour cell nuclear targeting in a tumour progression model. BMC Cancer 2015; 15:76. [PMID: 25885577 PMCID: PMC4342815 DOI: 10.1186/s12885-015-1045-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 01/27/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND There is an urgent need for new approaches to deliver bioactive molecules to cancer cells efficiently and specifically. METHODS Here we fuse the cancer cell nuclear targeting module of the Chicken Anaemia Virus Apoptin protein to the core histones H2B and H3 and utilise them in transfection, protein transduction and DNA binding assays. RESULTS We found subsequent nuclear accumulation of these proteins to be 2-3 fold higher in tumour compared to normal cells in transfected isogenic human osteosarcoma and breast tumour progression models. This represents the first demonstration of enhanced nuclear targeting by Apoptin in a tumour progression model, and its functionality in a heterologous protein context. Excitingly, we found that the innate transduction ability of histones could be exploited in combination with the Apoptin nuclear targeting module to effect an overall 13-fold higher delivery of protein to osteosarcoma cancer cell nuclei compared to their isogenic normal counterparts. CONCLUSIONS This is the first report of cancer-cell specificity by a cell penetrating protein, with important implications for the use of protein transduction as a vehicle for gene/drug delivery in the future, and in particular in the development of highly specific and effective anti-cancer agents.
Collapse
Affiliation(s)
- Michael S Nastasie
- Nuclear Signalling Laboratory, Department Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| | - Helmut Thissen
- CSIRO Molecular and Health Technologies, Bayview Avenue, Clayton, Victoria, 3168, Australia.
| | - David A Jans
- Nuclear Signalling Laboratory, Department Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| | - Kylie M Wagstaff
- Nuclear Signalling Laboratory, Department Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
28
|
Preclinical pharmacology and toxicology study of Ad-hTERT-E1a-Apoptin, a novel dual cancer-specific oncolytic adenovirus. Toxicol Appl Pharmacol 2014; 280:362-9. [DOI: 10.1016/j.taap.2014.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 08/05/2014] [Accepted: 08/12/2014] [Indexed: 11/17/2022]
|
29
|
Rollano Peñaloza OM, Lewandowska M, Stetefeld J, Ossysek K, Madej M, Bereta J, Sobczak M, Shojaei S, Ghavami S, Łos MJ. Apoptins: selective anticancer agents. Trends Mol Med 2014; 20:519-28. [PMID: 25164066 DOI: 10.1016/j.molmed.2014.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 12/20/2022]
Abstract
Therapies that selectively target cancer cells for death have been the center of intense research recently. One potential therapy may involve apoptin proteins, which are able to induce apoptosis in cancer cells leaving normal cells unharmed. Apoptin was originally discovered in the Chicken anemia virus (CAV); however, human gyroviruses (HGyV) have recently been found that also harbor apoptin-like proteins. Although the cancer cell specific activity of these apoptins appears to be well conserved, the precise functions and mechanisms of action are yet to be fully elucidated. Strategies for both delivering apoptin to treat tumors and disseminating the protein inside the tumor body are now being developed, and have shown promise in preclinical animal studies.
Collapse
Affiliation(s)
- Oscar M Rollano Peñaloza
- Department Clinical & Experimental Medicine, Division of Cell Biology, and Integrative Regenerative Medical Center, Linköping University, Linköping, Sweden; Instituto de Biologia Molecular y Biotecnologia, La Paz, Bolivia
| | | | - Joerg Stetefeld
- Department of Chemistry, University of Manitoba, Winnipeg, Canada
| | - Karolina Ossysek
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mariusz Madej
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Joanna Bereta
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mateusz Sobczak
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Shahla Shojaei
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Ghavami
- Department of Human Anatomy & Cell Science, College of Medicine, Faculty of Health Sciences, and Manitoba Institute of Child Health, University of Manitoba, Winnipeg, Canada; Health Policy Research Centre, Shiraz University of Medical Science, Shiraz, Iran
| | - Marek J Łos
- Department Clinical & Experimental Medicine, Division of Cell Biology, and Integrative Regenerative Medical Center, Linköping University, Linköping, Sweden; Department of Pathology, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
30
|
Chen KC, Yang TY, Wu CC, Cheng CC, Hsu SL, Hung HW, Chen JW, Chang GC. Pemetrexed induces S-phase arrest and apoptosis via a deregulated activation of Akt signaling pathway. PLoS One 2014; 9:e97888. [PMID: 24847863 PMCID: PMC4029963 DOI: 10.1371/journal.pone.0097888] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/25/2014] [Indexed: 12/16/2022] Open
Abstract
Pemetrexed is approved for first-line and maintenance treatment of patients with advanced or metastatic non-small-cell lung cancer (NSCLC). The protein kinase Akt/protein kinase B is a well-known regulator of cell survival which is activated by pemetrexed, but its role in pemetrexed-mediated cell death and its molecular mechanisms are unclear. This study showed that stimulation with pemetrexed induced S-phase arrest and cell apoptosis and a parallel increase in sustained Akt phosphorylation and nuclear accumulation in the NSCLC A549 cell line. Inhibition of Akt expression by Akt specific siRNA blocked S-phase arrest and protected cells from apoptosis, indicating an unexpected proapoptotic role of Akt in the pemetrexed-mediated toxicity. Treatment of A549 cells with pharmacological inhibitors of phosphatidylinositol 3-kinase (PI3K), wortmannin and Ly294002, similarly inhibited pemetrexed-induced S-phase arrest and apoptosis and Akt phosphorylation, indicating that PI3K is an upstream mediator of Akt and is involved in pemetrexed-mediated cell death. Previously, we identified cyclin A-associated cyclin-dependent kinase 2 (Cdk2) as the principal kinase that was required for pemetrexed-induced S-phase arrest and apoptosis. The current study showed that inhibition of Akt function and expression by pharmacological inhibitors as well as Akt siRNA drastically inhibited cyclin A/Cdk2 activation. These pemetrexed-mediated biological and molecular events were also observed in a H1299 cell line. Overall, our results indicate that, in contrast to its normal prosurvival role, the activated Akt plays a proapoptotic role in pemetrexed-mediated S-phase arrest and cell death through a mechanism that involves Cdk2/cyclin A activation.
Collapse
Affiliation(s)
- Kun-Chieh Chen
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Tsung-Ying Yang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Chun-Chi Wu
- Institute of Medicine, Chung Shang Medical University, Taichung, Taiwan, Republic of China
- Department of Medical Research, Chung-Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Chi-Chih Cheng
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Shih-Lan Hsu
- Department of Medical Research, Chung-Shan Medical University Hospital, Taichung, Taiwan, Republic of China
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Hsiao-Wen Hung
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Jian-Wei Chen
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Gee-Chen Chang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| |
Collapse
|
31
|
Abstract
The virus-derived protein Apoptin has the ability to induce p53-independent apoptosis in a variety of human cancer cells while leaving normal cells unharmed. It thus represents a potential anti-cancer therapeutic agent of the future but a proper understanding of Apoptin-induced signalling events is necessary prior to clinical application. The tumor-specific nuclear translocation and phosphorylation of Apoptin by a cellular kinase such as protein kinase C seem to be required for its function but otherwise the mode of tumor selectivity remains unknown. Apoptin has been shown to interact with several cellular proteins including Akt and the anaphase-promoting complex that regulate its activity and promote caspase-dependent apoptosis. This chapter summarizes the available data on tumor-specific pathways sensed by Apoptin and the mechanism of Apoptin-induced cell death.
Collapse
Affiliation(s)
- Jessica Bullenkamp
- Kings College London, Guy's Hospital, Floor 2 Room 2.66S, Hodgkin Building, London, UK
| | | |
Collapse
|
32
|
Backendorf C, Noteborn MHM. Apoptin Towards Safe and Efficient Anticancer Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 818:39-59. [DOI: 10.1007/978-1-4471-6458-6_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Kochneva GV, Babkina IN, Lupan TA, Grazhdantseva AA, Yudin PV, Sivolobova GF, Shvalov AN, Popov EG, Babkin IV, Netesov SV, Chumakov PM. Apoptin enhances the oncolytic activity of vaccinia virus in vitro. Mol Biol 2013. [DOI: 10.1134/s0026893313050075] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
Zhao J, Han SX, Ma JL, Ying X, Liu P, Li J, Wang L, Zhang Y, Ma J, Zhang L, Zhu Q. The role of CDK1 in apoptin-induced apoptosis in hepatocellular carcinoma cells. Oncol Rep 2013; 30:253-9. [PMID: 23619525 DOI: 10.3892/or.2013.2426] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 03/05/2013] [Indexed: 11/05/2022] Open
Abstract
Apoptin, a small protein derived from the chicken anemia virus, specifically induces apoptosis in transformed cells or tumor cells but not in normal cells. Thus, apoptin is involved in a general, tumor-specific pathway. Apoptin-induced apoptosis presumably requires additional interaction partners that activate specific signaling pathways in cancer cells. A number of molecules interact with apoptin and play an important role in the nuclear localization of apoptin or its tumor-selective cytotoxicity. Our data indicated that apoptin selectively kills HepG2 hepatocellular carcinoma (HCC) cells but has no effect on the normal liver cell line HL-7702. Analyses of human HCC tissue samples confirmed that CDK1 (cyclin-dependent kinase 1) activity was detected in primary malignancies but not in healthy paraneoplastic tissues. shRNA knockdown of CDK1 significantly reduced the tumor-specific killing effects of apoptin, suggesting that CDK1 plays an important role in the regulation of apoptin-induced apoptosis. Furthermore, the majority of apoptin translocated to the cytoplasm from the nucleus after knockdown of CDK1. Collectively, our results revealed for the first time that apoptin interacts with CDK1 in the complex process of tumorigenesis. The link between CDK1 and apoptin may be a novel cellular signaling pathway to modulate apoptosis in cancer; therefore, apoptin may have pharmacological potential to be directly employed for cancer therapy.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical college, Xi'an, Shaanxi 710061, P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Peng C, Yang P, Cui Y, He M, Liang L, Di Y. HSPA9 overexpression inhibits apoptin-induced apoptosis in the HepG2 cell line. Oncol Rep 2013; 29:2431-7. [PMID: 23589050 DOI: 10.3892/or.2013.2399] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/15/2013] [Indexed: 11/06/2022] Open
Abstract
Apoptin, a small protein derived from chicken anemia virus, possesses the capacity to specifically kill tumor cells while leaving normal cells intact. Previous studies have indicated that the subcellular localization of apoptin appears to be crucial for this tumor-selective activity. Apoptin resides in the cytoplasm of normal cells; however, in cancer cells it translocates into the nucleus. In the present study, purified prokaryotic native His-apoptin served as a bait for capturing apoptin-associated proteins in both a hepatoma carcinoma cell line (HepG2) and a human fetal liver cell line (L-02). The captured proteins obtained from a pull-down assay were separated by two-dimensional gel electrophoresis. Mass spectrometry was employed to detect the effect of HSPA9 overexpression (one of the interacting proteins with apoptin in vitro) and downregulation of HSPA9 on HepG2 cells. The data revealed that HSPA9 overexpression resulted in partial distribution of apoptin in the cytoplasm. Notably, HSPA9 overexpression markedly decreased the apoptosis rate of HepG2 cells from 41.2 to 31.7%, while the downregulation of HSPA9 using small interfering RNA significantly enhanced the apoptosis of HepG2 cells. Our results suggest new insights into the localization mechanism of apoptin which is tightly associated with HSPA9 overexpression and its crucial role in cellular apoptosis both in a tumor cell line (HepG2) and a normal cell line (L-02). These findings shed new light on the elucidation of the underlying mechanism of anticancer action of apoptin.
Collapse
Affiliation(s)
- Chuanmei Peng
- Clinical Laboratory of Yanan Hospital of Kunming, and Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650051, PR China
| | | | | | | | | | | |
Collapse
|
36
|
Livin promotes progression of breast cancer through induction of epithelial-mesenchymal transition and activation of AKT signaling. Cell Signal 2013; 25:1413-22. [PMID: 23524337 DOI: 10.1016/j.cellsig.2013.03.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 03/11/2013] [Indexed: 10/27/2022]
Abstract
The inhibitor of apoptosis proteins (IAP) are closely correlated with proliferation, apoptosis, motility, and metastasis. Livin is the most recently identified IAP, and its role in breast progression remains unknown. In our study, analyses of 50 patients with breast cancer revealed that the positive expression rate of Livin was higher in breast cancer tissues (62%) relative to that in adjacent (35%) and normal tissues (25%). Livin expression in breast cancer correlated with the clinical stage and axillary lymph node metastasis and could be used as a prognostic marker. Our in vitro experiment revealed that Livin was highly expressed in high-invasive MDA-MB-231 cells as compared to low-invasive cells (MCF-7). Suppression of Livin by short-hairpin RNA reduced the Livin expression of MDA-MB-231 cells and subsequently inhibited tumor cell growth, proliferation, and colony formation and induced tumor cell apoptosis, motility, migration, and invasion. Overexpression of Livin in MCF7 cells resulted in increased migration and invasion capabilities of the cells without affecting proliferation and apoptosis. In addition, epithelial-mesenchymal transition (EMT) was induced by Livin expression in breast cancer cell lines. The high level of phosphorylated AKT in MDA-MB-231 cells was suppressed by Livin knockdown. Further, Livin-induced migration and invasion could be abolished by either the application of the phosphoinositide-3-kinase inhibitor LY294002 or knockdown of AKT expression using small-interfering RNA. In conclusion, Livin serves as an independent prognostic indicator for breast cancer. Livin expression promotes breast cancer metastasis through the activation of AKT signaling and induction of EMT in breast cancer cells both in vitro and in vivo.
Collapse
|
37
|
ZHANG MUCHUN, WANG JINHUI, LI CHANG, HU NINGNING, WANG KAI, JI HUIFAN, HE DONGYUN, QUAN CHENGSHI, LI XIAO, JIN NINGYI, LI YULIN. Potent growth-inhibitory effect of a dual cancer-specific oncolytic adenovirus expressing apoptin on prostate carcinoma. Int J Oncol 2013; 42:1052-60. [DOI: 10.3892/ijo.2013.1783] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 12/17/2012] [Indexed: 11/06/2022] Open
|
38
|
ROS-mediated activation of AKT induces apoptosis via pVHL in prostate cancer cells. Mol Cell Biochem 2013; 376:63-71. [PMID: 23315288 DOI: 10.1007/s11010-012-1549-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/14/2012] [Indexed: 12/29/2022]
Abstract
Reactive oxygen species (ROS) play a central role in oxidative stress, which leads to the onset of diseases, such as cancer. Furthermore, ROS contributes to the delicate balance between tumor cell survival and death. However, the mechanisms by which tumor cells decide to elicit survival or death signals during oxidative stress are not completely understood. We have previously reported that ROS enhanced tumorigenic functions in prostate cancer cells, such as transendothelial migration and invasion, which depended on CXCR4 and AKT signaling. Here, we report a novel mechanism by which ROS facilitated cell death through activation of AKT. We initially observed that ROS enhanced the expression of phosphorylated AKT (p-AKT) in 22Rv1 human prostate cancer cells. The tumor suppressor PTEN, a negative regulator of AKT signaling, was rendered catalytically inactive through oxidation by ROS, although the expression levels remained consistent. Despite these events, cells still underwent apoptosis. Further investigation into apoptosis revealed that expression of the tumor suppressor pVHL increased, and contains a target site for p-AKT phosphorylation. pVHL and p-AKT associated in vitro, and knockdown of pVHL rescued HIF1α expression and the cells from apoptosis. Collectively, our study suggests that in the context of oxidative stress, p-AKT facilitated apoptosis by inducing pVHL function.
Collapse
|
39
|
Guan GF, Zhao M, Liu LM, Jin CS, Sun K, Zhang DJ, Yu DJ, Cao HW, Lu YQ, Wen LJ. Salmonella typhimurium mediated delivery of Apoptin in human laryngeal cancer. Int J Med Sci 2013; 10:1639-48. [PMID: 24155656 PMCID: PMC3805922 DOI: 10.7150/ijms.6960] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/30/2013] [Indexed: 01/26/2023] Open
Abstract
An effective cancer therapeutic should target tumours specifically with limited systemic toxicity. Here, we transformed an attenuated Salmonella typhimurium (S. typhimurium) with an Apoptin expressing plasmid into a human laryngeal carcinoma cell line. The expression of the inserted gene was measured using fluorescence and immunoblotting assays. The attenuated S. typhimurium-mediated Apoptin significantly decreased cytotoxicity and strongly increased cell apoptosis through the activation of caspase-3. The process was mediated by Bax, cytochrome c and caspase-9. A syngeneic nude murine tumour model was used to determine the anti-tumour effects of the recombinant bacteria in vivo. Systemic injection of the recombinant bacteria with and without re-dosing caused significant tumour growth delay and reduced tumour microvessel density, thereby extending host survival. Our findings indicated that the use of recombinant Salmonella typhimurium as an Apoptin expression vector has potential cancer therapeutic benefits.
Collapse
Affiliation(s)
- Guo-fang Guan
- 1. Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chaabane W, User SD, El-Gazzah M, Jaksik R, Sajjadi E, Rzeszowska-Wolny J, Los MJ. Autophagy, apoptosis, mitoptosis and necrosis: interdependence between those pathways and effects on cancer. Arch Immunol Ther Exp (Warsz) 2012; 61:43-58. [PMID: 23229678 DOI: 10.1007/s00005-012-0205-y] [Citation(s) in RCA: 212] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 09/06/2012] [Indexed: 12/21/2022]
Abstract
Cell death is a fundamental ingredient of life. Thus, not surprisingly more than one form of cell death exists. Several excellent reviews on various forms of cell death have already been published but manuscripts describing interconnection and interdependence between such processes are uncommon. Here, what follows is a brief introduction on all three classical forms of cell death, followed by a more detailed insight into the role of p53, the master regulator of apoptosis, and other forms of cell death. While discussing p53 and also the role of caspases in cell death forms, we offer insight into the interplay between autophagy and apoptosis, or necrosis, where autophagy may initially serve pro-survival functions. The review moves further to present some details about less researched forms of programmed cell death, namely necroptosis, necrosis and mitoptosis. These "mixed" forms of cell death allow us to highlight the interconnected nature of cell death forms, particularly apoptosis and necrosis. The interdependence between apoptosis, autophagy and necrosis, and their significance for cancer development and treatment are also analyzed in further parts of the review. In the concluding parts, the afore-mentioned issues will be put in perspective for the development of novel anti-cancer therapies.
Collapse
Affiliation(s)
- Wiem Chaabane
- Division of Cell Biology, Department Clinical and Experimental Medicine (IKE), and Integrative Regenerative Medicine Center (IGEN), Linköping University, Cell Biology Building, Linköping, Sweden
| | | | | | | | | | | | | |
Collapse
|
41
|
Apoptin selectively induces the apoptosis of tumor cells by suppressing the transcription of HSP70. Tumour Biol 2012. [DOI: 10.1007/s13277-012-0585-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
42
|
Taebunpakul P, Sayan BS, Flinterman M, Klanrit P, Gäken J, Odell EW, Melino G, Tavassoli M. Apoptin induces apoptosis by changing the equilibrium between the stability of TAp73 and ΔNp73 isoforms through ubiquitin ligase PIR2. Apoptosis 2012; 17:762-76. [PMID: 22484480 DOI: 10.1007/s10495-012-0720-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Apoptin, a protein derived from the chicken anaemia virus, induces cell death in various cancer cells but shows little or no cytotoxicity in normal cells. The mechanism of apoptin-induced cell death is currently unknown but it appears to induce apoptosis independent of p53 status. Here we show that p73, a p53 family member, is important in apoptin-induced apoptosis. In p53 deficient and/or mutated cells, apoptin induced the expression of TAp73 leading to the induction of apoptosis. Knockdown of p73 using siRNA resulted in a significant reduction in apoptin-induced cytotoxicity. The p53 and p73 pro-apoptotic target PUMA plays an important role in apoptin-induced cell death as knockdown of PUMA significantly reduced cell sensitivity to apoptin. Importantly, apoptin expression resulted in a marked increase in TAp73 protein stability. Investigation into the mechanisms of TAp73 stability showed that apoptin induced the expression of the ring finger domain ubiquitin ligase PIR2 which is involved in the degradation of the anti-apoptotic ∆Np73 isoform. Collectively, our results suggest a novel mechanism of apoptin-induced apoptosis through increased TAp73 stability and induction of PIR2 resulting in the degradation of ∆Np73 and activation of pro-apoptotic targets such as PUMA causing cancer cell death.
Collapse
Affiliation(s)
- P Taebunpakul
- Head and Neck Oncology Group, King's College London Dental Institute, Floor 28 Tower Wing, Guy's Hospital Campus, London, SE1 9RT, UK
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Human Gyrovirus Apoptin shows a similar subcellular distribution pattern and apoptosis induction as the chicken anaemia virus derived VP3/Apoptin. Cell Death Dis 2012; 3:e296. [PMID: 22495351 PMCID: PMC3358012 DOI: 10.1038/cddis.2012.34] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The chicken anaemia virus-derived protein Apoptin/VP3 (CAV-Apoptin) has the important ability to induce tumour-selective apoptosis in a variety of human cancer cells. Recently the first human Gyrovirus (HGyV) was isolated from a human skin swab. It shows significant structural and organisational resemblance to CAV and encodes a homologue of CAV-Apoptin/VP3. Using overlapping primers we constructed a synthetic human Gyrovirus Apoptin (HGyV-Apoptin) fused to green fluorescent protein in order to compare its apoptotic function in various human cancer cell lines to CAV-Apoptin. HGyV-Apoptin displayed a similar subcellular expression pattern as observed for CAV-Apoptin, marked by translocation to the nucleus of cancer cells, although it is predominantly located in the cytosol of normal human cells. Furthermore, expression of either HGyV-Apoptin or CAV-Apoptin in several cancer cell lines triggered apoptosis at comparable levels. These findings indicate a potential anti-cancer role for HGyV-Apoptin.
Collapse
|
44
|
Akt: a double-edged sword in cell proliferation and genome stability. JOURNAL OF ONCOLOGY 2012; 2012:951724. [PMID: 22481935 PMCID: PMC3317191 DOI: 10.1155/2012/951724] [Citation(s) in RCA: 217] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 12/29/2011] [Indexed: 01/31/2023]
Abstract
The Akt family of serine/threonine protein kinases are key regulators of multiple aspects of cell behaviour, including proliferation, survival, metabolism, and tumorigenesis. Growth-factor-activated Akt signalling promotes progression through normal, unperturbed cell cycles by acting on diverse downstream factors involved in controlling the G1/S and G2/M transitions. Remarkably, several recent studies have also implicated Akt in modulating DNA damage responses and genome stability. High Akt activity can suppress ATR/Chk1 signalling and homologous recombination repair (HRR) via direct phosphorylation of Chk1 or TopBP1 or, indirectly, by inhibiting recruitment of double-strand break (DSB) resection factors, such as RPA, Brca1, and Rad51, to sites of damage. Loss of checkpoint and/or HRR proficiency is therefore a potential cause of genomic instability in tumor cells with high Akt. Conversely, Akt is activated by DNA double-strand breaks (DSBs) in a DNA-PK- or ATM/ATR-dependent manner and in some circumstances can contribute to radioresistance by stimulating DNA repair by nonhomologous end joining (NHEJ). Akt therefore modifies both the response to and repair of genotoxic damage in complex ways that are likely to have important consequences for the therapy of tumors with deregulation of the PI3K-Akt-PTEN pathway.
Collapse
|
45
|
Zhang KJ, Qian J, Wang SB, Yang Y. Targeting Gene-Viro-Therapy with AFP driving Apoptin gene shows potent antitumor effect in hepatocarcinoma. J Biomed Sci 2012; 19:20. [PMID: 22321574 PMCID: PMC3311074 DOI: 10.1186/1423-0127-19-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 02/09/2012] [Indexed: 12/18/2022] Open
Abstract
Background Gene therapy and viral therapy are used for cancer therapy for many years, but the results are less than satisfactory. Our aim was to construct a new recombinant adenovirus which is more efficient to kill hepatocarcinoma cells but more safe to normal cells. Methods By using the Cancer Targeting Gene-Viro-Therapy strategy, Apoptin, a promising cancer therapeutic gene was inserted into the double-regulated oncolytic adenovirus AD55 in which E1A gene was driven by alpha fetoprotein promoter along with a 55 kDa deletion in E1B gene to form AD55-Apoptin. The anti-tumor effects and safety were examined by western blotting, virus yield assay, real time polymerase chain reaction, 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay, Hoechst33342 staining, Fluorescence-activated cell sorting, xenograft tumor model, Immunohistochemical assay, liver function analysis and Terminal deoxynucleotidyl transferase mediated dUTP Nick End Labeling assay. Results The recombinant virus AD55-Apoptin has more significant antitumor effect for hepatocelluar carcinoma cell lines (in vitro) than that of AD55 and even ONYX-015 but no or little impair on normal cell lines. Furthermore, it also shows an obvious in vivo antitumor effect on the Huh-7 liver carcinoma xenograft in nude mice with bigger beginning tumor volume till about 425 mm3 but has no any damage on the function of liver. The induction of apoptosis is involved in AD55-Apoptin induced antitumor effects. Conclusion The AD55-Apoptin can be a potential anti-hepatoma agent with remarkable antitumor efficacy as well as higher safety in cancer targeting gene-viro-therapy system.
Collapse
Affiliation(s)
- Kang-Jian Zhang
- State key Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, The Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | |
Collapse
|
46
|
Modeling of molecular interaction between apoptin, BCR-Abl and CrkL--an alternative approach to conventional rational drug design. PLoS One 2012; 7:e28395. [PMID: 22253690 PMCID: PMC3254606 DOI: 10.1371/journal.pone.0028395] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 11/07/2011] [Indexed: 12/02/2022] Open
Abstract
In this study we have calculated a 3D structure of apoptin and through modeling and docking approaches, we show its interaction with Bcr-Abl oncoprotein and its downstream signaling components, following which we confirm some of the newly-found interactions by biochemical methods. Bcr-Abl oncoprotein is aberrantly expressed in chronic myelogenous leukaemia (CML). It has several distinct functional domains in addition to the Abl kinase domain. The SH3 and SH2 domains cooperatively play important roles in autoinhibiting its kinase activity. Adapter molecules such as Grb2 and CrkL interact with proline-rich region and activate multiple Bcr-Abl downstream signaling pathways that contribute to growth and survival. Therefore, the oncogenic effect of Bcr-Abl could be inhibited by the interaction of small molecules with these domains. Apoptin is a viral protein with well-documented cancer-selective cytotoxicity. Apoptin attributes such as SH2-like sequence similarity with CrkL SH2 domain, unique SH3 domain binding sequence, presence of proline-rich segments, and its nuclear affinity render the molecule capable of interaction with Bcr-Abl. Despite almost two decades of research, the mode of apoptin's action remains elusive because 3D structure of apoptin is unavailable. We performed in silico three-dimensional modeling of apoptin, molecular docking experiments between apoptin model and the known structure of Bcr-Abl, and the 3D structures of SH2 domains of CrkL and Bcr-Abl. We also biochemically validated some of the interactions that were first predicted in silico. This structure-property relationship of apoptin may help in unlocking its cancer-selective toxic properties. Moreover, such models will guide us in developing of a new class of potent apoptin-like molecules with greater selectivity and potency.
Collapse
|
47
|
Mechanisms of Apoptin-induced cell death. Med Oncol 2011; 29:2985-91. [DOI: 10.1007/s12032-011-0119-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 11/09/2011] [Indexed: 12/22/2022]
|
48
|
Lanz HL, Florea BI, Noteborn MHM, Backendorf C. Development and application of an in vitro apoptin kinase assay. Anal Biochem 2011; 421:68-74. [PMID: 22080040 DOI: 10.1016/j.ab.2011.10.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 10/07/2011] [Accepted: 10/17/2011] [Indexed: 10/16/2022]
Abstract
Apoptin, a protein derived from chicken anemia virus (CAV), induces apoptosis selectively in human tumor cells as compared with normal cells. This activity depends on phosphorylation and relocation of apoptin to the nucleus of cancer cells. Here, we describe an in vitro kinase assay that allows the biochemical characterization of apoptin kinase activity in tumor cells. The kinase phosphorylates apoptin in a strictly ATP-dependent fashion and in a broad salt range. The kinase activity is present constitutively in both cytoplasm and nucleus of various human tumor cells. Q-column chromatography showed that both cytoplasmic and nuclear fractions have identical fractionation characteristics, suggesting that the same kinase is present in both cellular compartments. Kinase activity derived from positive Q-column fractions bound to amylose-maltose-binding protein (MBP)-apoptin and could be eluted with ATP only in the presence of the cofactor Mg(2+). Apparently, unphosphorylated apoptin interacts with the kinase and is released only after phosphorylation has occurred, proving that our assay recognizes the genuine apoptin kinase. This is further corroborated by the finding that apoptin is phosphorylated in vitro at positions Thr108 and Thr107, in concert with earlier in vivo observations. Our assay excludes cyclin-dependent kinase 2 (CDK2) and protein kinase C beta (PKC-β), previously nominated by two separate studies as being the genuine apoptin kinase.
Collapse
Affiliation(s)
- Henriëtte L Lanz
- Department of Molecular Genetics, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | | | | | | |
Collapse
|
49
|
Chen K, Luo Z, Zheng SJ. Gallus Heat shock cognate protein 70, a novel binding partner of Apoptin. Virol J 2011; 8:324. [PMID: 21707973 PMCID: PMC3138435 DOI: 10.1186/1743-422x-8-324] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 06/27/2011] [Indexed: 11/15/2022] Open
Abstract
Background Chicken anemia virus (CAV) infection of newly hatched chickens induces generalized lymphoid atrophy and causes immunosuppressive. VP3, also known as Apoptin, is non-structural protein of CAV. Apoptin specifically induces apoptosis in transformed or tumor cells but not in normal cells. In particular, there are no known cellular homologues of Apoptin hindering genetic approaches to elucidate its cellular function. Although a number of Apoptin-interacting molecules have been identified, the molecular mechanism underlying Apoptin's action is still poorly understood. To learn more about the molecular mechanism of Apoptin's action, we searched for Apoptin associated proteins. Results Using yeast two-hybrid and colony-life filter approaches we got five positive yeast clones. Through sequencing and BLASTed against NCBI, one of the clones was confirmed containing Gallus heat shock cognate protein 70 (Hsc70). Hsc70 gene was clone into pRK5-Flag plasmid, coimmunoprecipitation assay show both exogenous Hsc70 and endogenous Hsc70 can interact with Apoptin. Truncated Apoptin expression plasmids were made and coimmunoprecipitation were performed, the results show the binding domain of Apoptin with Hsc70 is located between amino acids 30-60. Truncated expression plasmids of Hsc70 were also constructed and coimmunoprecipitation were performed, the results show the peptide-binding and variable domains of Hsc70 are responsible for the binding to Apoptin. Confocal assays were performed and results show that under physiological condition Hsc70 is predominantly distributed in cytoplasm, whereas Hsc70 is translocated into the nuclei and colocalized with Apoptin in the presence of Apoptin in DF-1 cell. Functional studies show that Apoptin markedly down-regulate the mRNA level of RelA/p65 in DF-1 cell. To explore the effect of Hsc70 on Apoptin-mediated RelA/p65 gene expression, we have searched two Hsc70 RNAi sequences, and found that all of them dramatically inhibited the expression of endogenous Hsc70, with the #2 Hsc70 RNAi sequence being the most effective. Knockdown of Hsc70 show Apoptin-inhibited RelA/p65 expression was abolished. Our data demonstrate that Hsc70 is responsible for the down-regulation of Apoptin induced RelA/p65 gene expression. Conclusion We identified Gallus Hsc70 as an Apoptin binding protein and showed the translocation of Hsc70 into the nuclei of DF-1 cells treated with Apoptin. Hsc70 regulates RelA/p65 gene expression induced by Apoptin.
Collapse
Affiliation(s)
- Kun Chen
- State Key Laboratory of Agrobiotechnology, Beijing 100193, China
| | | | | |
Collapse
|
50
|
Identification of the first human gyrovirus, a virus related to chicken anemia virus. J Virol 2011; 85:7948-50. [PMID: 21632766 DOI: 10.1128/jvi.00639-11] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have identified in a skin swab sample from a healthy donor a new virus that we have named human gyrovirus (HGyV) because of its similarity to the chicken anemia virus (CAV), the only previously known member of the Gyrovirus genus. In particular, this virus encodes a homolog of the CAV apoptin, a protein that selectively induces apoptosis in cancer cells. By PCR screening, HGyV was found in 5 of 115 other nonlesional skin specimens but in 0 of 92 bronchoalveolar lavages or nasopharyngeal aspirates and in 0 of 92 fecal samples.
Collapse
|