1
|
Yang L, Haeckel A, Beindorff N, Poetzsch SML, Mi H, Ni F, Hojjat H, Brenner W, de Moraes PAD, Guo J, Savic LJ, Schellenberger E. Long circulating XTEN864-HGV-Apoptin fusion protein for selective cancer therapy. Int J Biol Macromol 2025; 306:141679. [PMID: 40037452 DOI: 10.1016/j.ijbiomac.2025.141679] [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: 10/25/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
The virus protein CAV-Apoptin and its homologue HGV-Apoptin selectively kill cancer cells but are not suitable for systemic treatment. The aim was to develop Apoptin-based fusion proteins for intravenous application in cancer therapy, which also contain the hydrophilic polypeptide XTEN, a cleavage site for MMP-2/9, and a TAT peptide for cell penetration. Expression of XTEN864-HGV-Apoptin in E. coli and purification using XTEN as a tag yielded 100 mg protein/L tissue culture. The expression of XTEN864-CAV-Apoptin did not generate a sufficient yield. Cytotoxic effects were assessed using MTT and Annexin A5 assays, whereas cellular uptake was visualized using Cy3.5-XTEN864-HGV-Apoptin. Blood half-life and biodistribution were evaluated with 99mTc-XTEN864-HGV-Apoptin using SPECT-CT and gamma counting. The fusion protein significantly reduced cancer cell growth and induced apoptosis with minimal effects on non-cancerous cells. It accumulates in the nucleus and associates with F-actin. In mice, the protein showed a blood half-life of 0.68 h (fast phase) and 17 h (slow phase), with a tumor/muscle ratio of 9.36 ± 6.22 (SD). In a 4T1 mouse tumor model, it effectively inhibited tumor growth. The cancer-specific cytotoxicity and prolonged circulation of XTEN864-HGV-Apoptin suggest its potential for systemically applicable, biodegradable, and E. coli-producible antitumor drugs.
Collapse
Affiliation(s)
- Liu Yang
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Akvile Haeckel
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicola Beindorff
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Simon Marc Levin Poetzsch
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Honglan Mi
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Fei Ni
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Hamidreza Hojjat
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Winfried Brenner
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany; Charité - Universitätsmedizin Berlin, Department of Nuclear Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Pedro Augusto Dantas de Moraes
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Jing Guo
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Lynn Jeanette Savic
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eyk Schellenberger
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
2
|
Wu J, Lan Z, Li X, He J, Zhang D, Jin T. A novel recombinant adenovirus expressing apoptin and melittin genes kills hepatocellular carcinoma cells and inhibits the growth of ectopic tumor. Invest New Drugs 2024; 42:428-441. [PMID: 38935191 DOI: 10.1007/s10637-024-01453-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
HCC is the most common fatal malignancy. Although surgical resection is the primary treatment strategy, most patients are not eligible for resection due to tumor heterogeneity, underlying liver disease, or comorbidities. Therefore, this study explores the possibility of multi-molecular targeted drug delivery in treating HCC. In this study, we constructed the recombinant adenovirus co-expressing apoptin and melittin (MEL) genes. The inhibitory effect of the recombinant adenovirus on hepatocellular carcinoma cells was detected through experiments on cell apoptosis, migration, invasion, and other factors. The tumor inhibitory effect in vivo was assessed using subcutaneous HCC mice. Results showed that recombinant adenovirus co-expressing anti-tumor genes TAT and apoptin, RGD and MEL can significantly inhibit the proliferation, migration, and invasion of HCC cells by inducing an increase in reactive oxygen species (ROS) levels, upregulation of apoptotic proteins such as Bax, cleaved caspase-3, and cleaved caspase-9, and downregulation of the anti-apoptotic protein Bcl-2. In subcutaneous HCC mice, recombinant adenovirus induced significant apoptosis in tumor, and inhibited tumor growth. In conclusion, recombinant adenovirus co-expressing apoptin and MEL can inhibit the growth and proliferation of tumor cells both in vivo and in vitro.
Collapse
Affiliation(s)
- Jingqiao Wu
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Zhaoyu Lan
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Xin Li
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Jinling He
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Dongchao Zhang
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China.
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China.
- Tianjin Engineering Technology Center of Livestock Pathogen Detection and Genetic Engineering Vaccine, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China.
| | - Tianming Jin
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China.
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China.
- Tianjin Engineering Technology Center of Livestock Pathogen Detection and Genetic Engineering Vaccine, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China.
| |
Collapse
|
3
|
Guterres A, Filho PNS, Moura-Neto V. Breaking Barriers: A Future Perspective on Glioblastoma Therapy with mRNA-Based Immunotherapies and Oncolytic Viruses. Vaccines (Basel) 2024; 12:61. [PMID: 38250874 PMCID: PMC10818651 DOI: 10.3390/vaccines12010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
The use of mRNA-based immunotherapies that leverage the genomes of oncolytic viruses holds significant promise in addressing glioblastoma (GBM), an exceptionally aggressive neurological tumor. We explore the significance of mRNA-based platforms in the area of immunotherapy, introducing an innovative approach to mitigate the risks associated with the use of live viruses in cancer treatment. The ability to customize oncolytic virus genome sequences enables researchers to precisely target specific cancer cells, either through viral genome segments containing structural proteins or through a combination of regions with oncolytic potential. This strategy may enhance treatment effectiveness while minimizing unintended impacts on non-cancerous cells. A notable case highlighted here pertains to advanced findings regarding the application of the Zika virus (ZIKV) in GBM treatment. ZIKV, a member of the family Flaviviridae, shows oncolytic properties against GBM, opening novel therapeutic avenues. We explore intensive investigations of glioblastoma stem cells, recognized as key drivers in GBM initiation, progression, and resistance to therapy. However, a comprehensive elucidation of ZIKV's underlying mechanisms is imperative to pave the way for ZIKV-based clinical trials targeting GBM patients. This investigation into harnessing the potential of oncolytic-virus genomes for mRNA-based immunotherapies underscores its noteworthy implications, potentially paving the way for a paradigm shift in cancer treatment strategies.
Collapse
Affiliation(s)
- Alexandro Guterres
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-360, RJ, Brazil
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-360, RJ, Brazil
| | | | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro 20231-092, RJ, Brazil; (P.N.S.F.)
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil
| |
Collapse
|
4
|
Huang J, Zhang Y, Cheng A, Wang M, Liu M, Zhu D, Chen S, Zhao X, Yang Q, Wu Y, Zhang S, Ou X, Mao S, Gao Q, Sun D, Tian B, Yin Z, Jia R. Duck Circovirus genotype 2 ORF3 protein induces apoptosis through the mitochondrial pathway. Poult Sci 2023; 102:102533. [PMID: 36848756 PMCID: PMC9984893 DOI: 10.1016/j.psj.2023.102533] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/26/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Duck circovirus genotype 2 (DuCV2) belongs to the genus Circovirus, family Circoviridae. It can generally cause lymphocyte atrophy and necrosis in ducks, which leads to immunosuppression. The function of the DuCV2 open reading frame 3 (ORF3) protein in viral pathogenesis in host cells remains unclear. Therefore, a series of studies based on ORF3 of the isolate DuCV GH01 strain (belonging to DuCV2) were carried out in duck embryo fibroblasts (DEFs) in this study. The results showed that the ORF3 protein could induce nuclear shrinkage and fragmentation in DEFs. Chromosomal DNA breakage was observed by TUNEL assay. The expression levels of caspase-related genes showed that ORF3 primarily promoted caspase 3 and caspase 9 expression. Furthermore, the protein expression levels of cleaved caspase 3 and cleaved caspase 9 in DEFs were enhanced by ORF3. Thus, ORF3 may activate the mitochondrial apoptosis pathway. When the 20 amino acid residues at the C-terminus of ORF3 (ORF3ΔC20) were deleted, the apoptosis rates were decreased. Moreover, compared to ORF3, ORF3ΔC20 downregulated the mRNA levels of cytochrome c (Cyt c), poly ADP-ribose polymerase (PARP) and apoptosis protease activating factor 1 (Apaf-1), which are the key molecules in the mitochondrial apoptotic pathway. Further study showed that ORF3ΔC20 could reduce the mitochondrial membrane potential (MMP). This study suggested that the DuCV2 ORF3 protein may primarily activate apoptosis through the mitochondrial pathway in DEFs, and this function is ORF3 C20 dependent.
Collapse
Affiliation(s)
- Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Yanting Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Xumin Ou
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Sai Mao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Qun Gao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Di Sun
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Bin Tian
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
5
|
Yu X, Wang T, Li Y, Li Y, Bai B, Fang J, Han J, Li S, Xiu Z, Liu Z, Yang X, Li Y, Zhu G, Jin N, Shang C, Li X, Zhu Y. Apoptin causes apoptosis in HepG-2 cells via Ca 2+ imbalance and activation of the mitochondrial apoptotic pathway. Cancer Med 2022; 12:8306-8318. [PMID: 36515089 PMCID: PMC10134343 DOI: 10.1002/cam4.5528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/06/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Apoptin is derived from the chicken anemia virus and exhibits specific cytotoxic effects against tumor cells. Herein, we found that Apoptin induced a strong and lasting endoplasmic reticulum (ER) stress response, Ca2+ imbalance, and triggered the mitochondrial apoptotic pathway. The aim of this study was to explore the mechanisms by which Apoptin exhibited anti-tumor effects in HepG-2 cells. METHODS The intracellular levels of calcium (Ca2+ ) were induced by ER stress and determined by electron microscopy, flow cytometry, and fluorescence staining. The mitochondrial injury was determined by mitochondrial membrane potential and electron microscopy. Western blotting was used to investigate the levels of key proteins in ER stress and the apoptotic pathway in mitochondria. The relationship between Ca2+ levels and apoptosis in Apoptin-treated cells was analyzed using a Ca2+ chelator (BAPTA-AM), flow cytometry, and fluorescence staining. We also investigated the in vivo effects of Ca2+ imbalance on the mitochondrial apoptotic pathway using tumor tissues xenografted on nude mice. RESULTS This study showed that Apoptin induced a strong and long- lasting ER stress and injury, which subsequently led to an imbalance of cellular Ca2+ levels, a reduction in the mitochondrial membrane potential, a significant extent image in the mitochondrial structure, and an increase in the expression levels of Smac/Diablo and Cyto-C. CONCLUSIONS In summary, Apoptin induced apoptosis in HepG-2 cells via Ca2+ imbalance and activation of the mitochondrial apoptotic pathway. This study provided a new direction for antitumor research in Apoptin.
Collapse
Affiliation(s)
- Xiaoyang Yu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Tongxing Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yue Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Yiquan Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Bing Bai
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Jinbo Fang
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Jicheng Han
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Shanzhi Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Zhiru Xiu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Zirui Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xia Yang
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Yaru Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Guangze Zhu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Ningyi Jin
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Chao Shang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xiao Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yilong Zhu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
6
|
Apoptin mediates mitophagy and endogenous apoptosis by regulating the level of ROS in hepatocellular carcinoma. Cell Commun Signal 2022; 20:134. [PMID: 36050738 PMCID: PMC9438158 DOI: 10.1186/s12964-022-00940-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/17/2022] [Indexed: 12/09/2022] Open
Abstract
Background Apoptin, as a tumor-specific pro-apoptotic protein, plays an important anti-tumoral role, but its mechanism of autophagy activation and the interaction between autophagy and apoptosis have not been accurately elucidated. Here, we studied the mechanism of apoptin-induced apoptosis and autophagy and the interaction between two processes. Methods Using crystal violet staining and the CCK-8 assay, we analyzed the effect of apoptin in the inhibition of liver cancer cells in vitro and analyzed the effect of inhibiting liver cancer in vivo by establishing a nude mouse tumor model. Flow cytometry and fluorescence staining were used to analyze the main types of apoptin-induced apoptosis and autophagy. Subsequently, the relationship between the two events was also analyzed. Flow cytometry was used to analyze the effect of ROS on apoptin-mediated apoptosis and autophagy mediated by apoptin. The effect of ROS on two phenomena was analyzed. Finally, the role of key genes involved in autophagy was analyzed using gene silencing. Results The results showed that apoptin can significantly increase the apoptosis and autophagy of liver cancer cells, and that apoptin can cause mitophagy through the increase in the expression of NIX protein. Apoptin can also significantly increase the level of cellular ROS, involved in apoptin-mediated autophagy and apoptosis of liver cancer cells. The change of ROS may be a key factor causing apoptosis and autophagy. Conclusion The above results indicate that the increase in ROS levels after apoptin treatment of liver cancer cells leads to the loss of mitochondrial transmembrane potential, resulting in endogenous apoptosis and mitophagy through the recruitment of NIX. Therefore, ROS may be a key factor connecting endogenous apoptosis and autophagy induced by apoptin in liver cancer cells. Graphical abstract ![]()
Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00940-1.
Collapse
|
7
|
Liu Z, Li Y, Zhu Y, Li N, Li W, Shang C, Song G, Li S, Cong J, Li T, Xiu Z, Lu J, Ge C, Yang X, Li Y, Sun L, Li X, Jin N. Apoptin induces pyroptosis of colorectal cancer cells via the GSDME-dependent pathway. Int J Biol Sci 2022; 18:717-730. [PMID: 35002520 PMCID: PMC8741846 DOI: 10.7150/ijbs.64350] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Apoptin is a small molecular weight protein encoded by the VP3 gene of chicken anemia virus (CAV). It can induce apoptosis of tumor cells and play anti-tumorigenic functions. In this study, we identified a time-dependent inhibitory role of apoptin on the viability of HCT116 cells. We also demonstrated that apoptin induces pyroptosis through cleaved caspase 3, and with a concomitant cleavage of gasdermin E (GSDME) rather than GSDMD. GSDME knockdown switched the apoptin-induced cell death from pyroptosis to apoptosis in vitro. Furthermore, we demonstrated that the effect of apoptin on GSDME-dependent pyroptosis could be mitigated by caspase-3 and caspase-9 siRNA knockdown. Additionally, apoptin enhanced the intracellular reactive oxygen species (ROS), causing aggregation of the mitochondrial membrane protein Tom20. Moreover, bax and cytochrome c were released to the activating caspase-9, eventually triggering pyroptosis. Therefore, GSDME mediates the apoptin-induced pyroptosis through the mitochondrial apoptotic pathway. Finally, using nude mice xenografted with HCT116 cells, we found that apoptin induces pyroptosis and significantly inhibits tumor growth. Based on this mechanism, apoptin may provide a new strategy for colorectal cancer therapy.
Collapse
Affiliation(s)
- Zirui Liu
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yilong Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Nan Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Wenjie Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Chao Shang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Gaojie Song
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Shanzhi Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Jianan Cong
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Tingyu Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Zhiru Xiu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Jing Lu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Chenchen Ge
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Xia Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Yaru Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Lili Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China.,Department of Head and Neck Surgery, Tumor Hospital of Jilin Province, Changchun, 130012, China
| | - Xiao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Ningyi Jin
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| |
Collapse
|
8
|
Bae Y, Lee J, Kho C, Choi JS, Han J. Apoptin gene delivery by a PAMAM dendrimer modified with a nuclear localization signal peptide as a gene carrier for brain cancer therapy. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:467-478. [PMID: 34448464 PMCID: PMC8405440 DOI: 10.4196/kjpp.2021.25.5.467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 11/30/2022]
Abstract
In this study, we aimed to synthesize PAMAMG3 derivatives (PAMAMG3-KRRR and PAMAMG3-HKRRR), using KRRR peptides as a nuclear localization signal and introduced histidine residues into the KRRR-grafted PAMAMG3 for delivering a therapeutic, carcinoma cell-selective apoptosis gene, apoptin into human primary glioma (GBL-14) cells and human dermal fibroblasts. We examined their cytotoxicity and gene expression using luciferase activity and enhanced green fluorescent protein PAMAMG3 derivatives in both cell lines. We treated cells with PAMAMG3 derivative/apoptin complexes and investigated their intracellular distribution using confocal microscopy. The PAMAMG3-KRRR and PAMAMG3-HKRRR dendrimers were found to escape from endolysosomes into the cytosol. The JC-1 assay, glutathione levels, and Annexin V staining results showed that apoptin triggered cell death in GBL-14 cells. Overall, these findings indicated that the PAMAMG3-HKRRR/apoptin complex is a potential candidate for an effective nonviral gene delivery system for brain tumor therapy in vitro.
Collapse
Affiliation(s)
- Yoonhee Bae
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University, Busan 47392, Korea.,Division of Applied Medicine, Research Institute for Korea Medicine, School of Korean Medicine, Pusan National University, Busan 50612, Korea
| | - Jell Lee
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Changwon Kho
- Division of Applied Medicine, Research Institute for Korea Medicine, School of Korean Medicine, Pusan National University, Busan 50612, Korea
| | - Joon Sig Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University, Busan 47392, Korea
| |
Collapse
|
9
|
Viral Proteins as Emerging Cancer Therapeutics. Cancers (Basel) 2021; 13:cancers13092199. [PMID: 34063663 PMCID: PMC8125098 DOI: 10.3390/cancers13092199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary This review is focused on enlisting viral proteins from different host sources, irrespective of their origin, that may act as future cancer curatives. Unlike the viral proteins that are responsible for tumor progression, these newly emerged viral proteins function as tumor suppressors. Their ability to regulate various cell signaling mechanisms specifically in cancer cells makes them interesting candidates to explore their use in cancer therapy. The discussion about such viral components may provide new insights into cancer treatment in the absence of any adverse effects to normal cells. The study also highlights avian viral proteins as a substitute to human oncolytic viruses for their ability to evade pre-existing immunity. Abstract Viruses are obligatory intracellular parasites that originated millions of years ago. Viral elements cover almost half of the human genome sequence and have evolved as genetic blueprints in humans. They have existed as endosymbionts as they are largely dependent on host cell metabolism. Viral proteins are known to regulate different mechanisms in the host cells by hijacking cellular metabolism to benefit viral replication. Amicable viral proteins, on the other hand, from several viruses can participate in mediating growth retardation of cancer cells based on genetic abnormalities while sparing normal cells. These proteins exert discreet yet converging pathways to regulate events like cell cycle and apoptosis in human cancer cells. This property of viral proteins could be harnessed for their use in cancer therapy. In this review, we discuss viral proteins from different sources as potential anticancer therapeutics.
Collapse
|
10
|
Zhu Y, Li Y, Bai B, Shang C, Fang J, Cong J, Li W, Li S, Song G, Liu Z, Zhao J, Li X, Zhu G, Jin N. Effects of Apoptin-Induced Endoplasmic Reticulum Stress on Lipid Metabolism, Migration, and Invasion of HepG-2 Cells. Front Oncol 2021; 11:614082. [PMID: 33718168 PMCID: PMC7952871 DOI: 10.3389/fonc.2021.614082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/26/2021] [Indexed: 11/13/2022] Open
Abstract
In this study, we investigated the effects of Apoptin-induced endoplasmic reticulum (ER) stress on lipid metabolism, migration and invasion of HepG-2 cells, and preliminarily explored the relationship between endoplasmic reticulum stress, lipid metabolism, migration, and invasion. The effects of Apoptin on ER function and structure in HepG-2 cells were determined by flow cytometry, fluorescence staining and western blotting by assessing the expression levels of ER stress related proteins. The effects of Apoptin on HepG-2 cells' lipid metabolism were determined by western blot analysis of the expression levels of triglyceride, cholesterol, and lipid metabolism related enzymes. The effects of Apoptin on HepG-2 cells' migration and invasion were studied using migration and invasion assays and by Western-blot analysis of the expression of proteins involved in migration and invasion. The in vivo effects of endoplasmic reticulum stress on lipid metabolism, migration and invasion of HepG-2 cells were also investigated by immunohistochemistry analysis of tumor tissues from HepG2 cells xenografted nude mice models. Both in vitro and in vivo experiments showed that Apoptin can cause a strong and lasting ER stress response, damage ER functional structure, significantly change the expression levels of lipid metabolism related enzymes and reduce the migration and invasion abilities of HepG-2 cells. Apoptin can also affect HepG-2 cells' lipid metabolism through endoplasmic reticulum stress and the abnormal expression of enzymes closely related to tumor migration and invasion. These results also showed that lipid metabolism may be one of the main inducements that reduce HepG-2 cells' migration and invasion abilities.
Collapse
Affiliation(s)
- Yilong Zhu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Yiquan Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Bing Bai
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Chao Shang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Jinbo Fang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Jianan Cong
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Wenjie Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Shanzhi Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Gaojie Song
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Zirui Liu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Jin Zhao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xiao Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Guangze Zhu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Ningyi Jin
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Li Y, Zhu Y, Fang J, Li W, Li S, Liu X, Liu Z, Song G, Shang C, Cong J, Bai B, Sun L, Jin N, Li X. Apoptin Regulates Apoptosis and Autophagy by Modulating Reactive Oxygen Species (ROS) Levels in Human Liver Cancer Cells. Front Oncol 2020; 10:1026. [PMID: 32714864 PMCID: PMC7344208 DOI: 10.3389/fonc.2020.01026] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/22/2020] [Indexed: 12/24/2022] Open
Abstract
Apoptin is a protein that specifically induces apoptosis in tumor cells. The anti-tumorigenic functions of Apoptin, including autophagy activation and its interaction with apoptosis, have not been precisely elucidated. Here we investigate the main pathways of apoptin-mediated killing of human liver cancer cells, as well as its putative role in autophagy and apoptosis. The anti-proliferative effect of apoptin in liver cancer cells was analyzed in vitro by crystal violet staining and MTS detection, and also in vivo using a tumor-based model. The main pathway related to apoptin-induced growth inhibition in vitro was evaluated by flow cytometry and fluorescence staining. The relationship between apoptosis and autophagy on apoptin-treating cells was analyzed using apoptosis and autophagy inhibitors, mitochondrial staining, Annexin V-FITC/PI flow detection, LC3 staining, and western blotting. The effect of ROS toward the apoptosis and autophagy of apoptin-treating cells was also evaluated by ROS detection, Annexin V-FITC/PI flow detection, LC3 staining, and western blotting. Inhibition of apoptosis in apoptin-treating liver cancer cells significantly reduced the autophagy levels in vitro. The overall inhibition increased from 12 h and the effect was most obvious at 48 h. Inhibition of autophagy could increase apoptin-induced apoptosis of cells in a time-dependent manner, reaching its peak at 24 h. Apoptin significantly alters ROS levels in liver cancer cells, and this effect is directly related to apoptosis and autophagy. ROS appears to be the key factor linking apoptin-induced autophagy and apoptosis through the mitochondria in liver cancer cells. Therefore, evaluating the interaction between apoptin-induced apoptosis and autophagy is a promising step for the development of alternate tumor therapies.
Collapse
Affiliation(s)
- Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Yilong Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Jinbo Fang
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Wenjie Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Shanzhi Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xing Liu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Zirui Liu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Gaojie Song
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Chao Shang
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Jianan Cong
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Bing Bai
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Lili Sun
- Department of Head and Neck Surgery, Tumor Hospital of Jilin Province, Changchun, China
| | - Ningyi Jin
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xiao Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
14
|
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
|
15
|
Deng H, Yue JK, Zusman BE, Nwachuku EL, Abou-Al-Shaar H, Upadhyayula PS, Okonkwo DO, Puccio AM. B-Cell Lymphoma 2 (Bcl-2) and Regulation of Apoptosis after Traumatic Brain Injury: A Clinical Perspective. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:E300. [PMID: 32570722 PMCID: PMC7353854 DOI: 10.3390/medicina56060300] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
Background and Objectives: The injury burden after head trauma is exacerbated by secondary sequelae, which leads to further neuronal loss. B-cell lymphoma 2 (Bcl-2) is an anti-apoptotic protein and a key modulator of the programmed cell death (PCD) pathways. The current study evaluates the clinical evidence on Bcl-2 and neurological recovery in patients after traumatic brain injury (TBI). Materials and Methods: All studies in English were queried from the National Library of Medicine PubMed database using the following search terms: (B-cell lymphoma 2/Bcl-2/Bcl2) AND (brain injury/head injury/head trauma/traumatic brain injury) AND (human/patient/subject). There were 10 investigations conducted on Bcl-2 and apoptosis in TBI patients, of which 5 analyzed the pericontutional brain tissue obtained from surgical decompression, 4 studied Bcl-2 expression as a biomarker in the cerebrospinal fluid (CSF), and 1 was a prospective randomized trial. Results: Immunohistochemistry (IHC) in 94 adults with severe TBI showed upregulation of Bcl-2 in the pericontusional tissue. Bcl-2 was detected in 36-75% of TBI patients, while it was generally absent in the non-TBI controls, with Bcl-2 expression increased 2.9- to 17-fold in TBI patients. Terminal deoxynucleotidyl transferase-mediated biotinylated dUTP nick-end labeling (TUNEL) positivity for cell death was detected in 33-73% of TBI patients. CSF analysis in 113 TBI subjects (90 adults, 23 pediatric patients) showed upregulation of Bcl-2 that peaked on post-injury day 3 and subsequently declined after day 5. Increased Bcl-2 in the peritraumatic tissue, rising CSF Bcl-2 levels, and the variant allele of rs17759659 are associated with improved mortality and better outcomes on the Glasgow Outcome Score (GOS). Conclusions: Bcl-2 is upregulated in the pericontusional brain and CSF in the acute period after TBI. Bcl-2 has a neuroprotective role as a pro-survival protein in experimental models, and increased expression in patients can contribute to improvement in clinical outcomes. Its utility as a biomarker and therapeutic target to block neuronal apoptosis after TBI warrants further evaluation.
Collapse
Affiliation(s)
- Hansen Deng
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (B.E.Z.); (E.L.N.); (H.A.-A.-S.); (D.O.O.); (A.M.P.)
- Neurotrauma Clinical Trials Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - John K. Yue
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94110, USA;
- Brain and Spinal Injury Center, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
| | - Benjamin E. Zusman
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (B.E.Z.); (E.L.N.); (H.A.-A.-S.); (D.O.O.); (A.M.P.)
| | - Enyinna L. Nwachuku
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (B.E.Z.); (E.L.N.); (H.A.-A.-S.); (D.O.O.); (A.M.P.)
- Neurotrauma Clinical Trials Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Hussam Abou-Al-Shaar
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (B.E.Z.); (E.L.N.); (H.A.-A.-S.); (D.O.O.); (A.M.P.)
| | - Pavan S. Upadhyayula
- Department of Neurological Surgery, University of California Diego, San Diego, CA 92093, USA;
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - David O. Okonkwo
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (B.E.Z.); (E.L.N.); (H.A.-A.-S.); (D.O.O.); (A.M.P.)
- Neurotrauma Clinical Trials Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Ava M. Puccio
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (B.E.Z.); (E.L.N.); (H.A.-A.-S.); (D.O.O.); (A.M.P.)
- Neurotrauma Clinical Trials Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
16
|
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
|
17
|
Maruoka H, Yamazoe R, Takahashi R, Yatsuo K, Ido D, Fuchigami Y, Hoshikawa F, Shimoke K. Molecular mechanism of nur77 gene expression and downstream target genes in the early stage of forskolin-induced differentiation in PC12 cells. Sci Rep 2020; 10:6325. [PMID: 32286359 PMCID: PMC7156746 DOI: 10.1038/s41598-020-62968-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/23/2020] [Indexed: 11/20/2022] Open
Abstract
Forskolin promotes neuronal differentiation of PC12 cells via the PKA-CREB-dependent signaling pathway. Activation of PKA by forskolin phosphorylates CREB, which then binds to CRE sites in numerous gene promoters. However, it is unclear which gene contains the CRE sites responsible for forskolin-induced neuronal differentiation. In this study, we investigated how an immediate early gene, nur77, which has CRE sites in the promoter region, contributes to the early stage of differentiation of forskolin-treated PC12 cells. After treatment with forskolin, expression of Nur77 was upregulated within 1 hr. In addition, knockdown of nur77 inhibited neurite outgrowth induced by forskolin. We also revealed that the specific four CRE sites near the transcriptional start site (TSS) of nur77 were strongly associated with phosphorylated CREB within 1 hr after treatment with forskolin. To analyze the roles of these four sites, reporter assays using the nur77 promoter region were performed. The results showed that nur77 expression was mediated through three of the CRE sites, -242, -222, and -78, and that -78, the nearest of the three to the TSS of nur77, was particularly important. An analysis of neuronal markers controlled by Nur77 after A-CREB-Nur77-Synapsin1 signaling pathway plays a pivotal role in differentiation of forskolin-induced PC12 cells.
Collapse
Affiliation(s)
- Hiroki Maruoka
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Ryosuke Yamazoe
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Ryota Takahashi
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Keisuke Yatsuo
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Daiki Ido
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Yuki Fuchigami
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Fumiya Hoshikawa
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Koji Shimoke
- Laboratory of Neurobiology, Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan.
| |
Collapse
|
18
|
Stem Cell Therapy for Hepatocellular Carcinoma: Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1237:97-119. [PMID: 31728916 DOI: 10.1007/5584_2019_441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of cancer and results in a high mortality rate worldwide. Unfortunately, most cases of HCC are diagnosed in an advanced stage, resulting in a poor prognosis and ineffective treatment. HCC is often resistant to both radiotherapy and chemotherapy, resulting in a high recurrence rate. Although the use of stem cells is evolving into a potentially effective approach for the treatment of cancer, few studies on stem cell therapy in HCC have been published. The administration of stem cells from bone marrow, adipose tissue, the amnion, and the umbilical cord to experimental animal models of HCC has not yielded consistent responses. However, it is possible to induce the apoptosis of cancer cells, repress angiogenesis, and cause tumor regression by administration of genetically modified stem cells. New alternative approaches to cancer therapy, such as the use of stem cell derivatives, exosomes or stem cell extracts, have been proposed. In this review, we highlight these experimental approaches for the use of stem cells as a vehicle for local drug delivery.
Collapse
|
19
|
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
|
20
|
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
|
21
|
Dai Y, Jin W, Cheng L, Yu C, Chen C, Ni H. Nur77 is a promoting factor in traumatic brain injury-induced nerve cell apoptosis. Biomed Pharmacother 2018; 108:774-782. [PMID: 30248546 DOI: 10.1016/j.biopha.2018.09.091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 10/28/2022] Open
Abstract
Traumatic brain injury (TBI) poses a serious threat to human health. TBI has a high mortality rate, resulting in a great burden on the affected individual's family as well as society as a whole. The incidence of craniocerebral fractures continues to rise as both the economy and transportation options grow, making it imperative that the mortality and disability rate of craniocerebral trauma be reduced. Nur77 is a transcription factor of the nuclear receptor superfamily. Following stimulation of extracellular apoptosis, Nur77 is involved in a variety of diseases as a powerful pro-apoptotic molecule. Here, we determined the effect and mechanism of Nur77 in TBI-induced nerve cell apoptosis in vitro and in vivo. We found that Nur77 and Bcl-2 protein expression increased as nerve cell apoptosis increased in TBI tissues. Furthermore, inhibition of Nur77 improved nerve cell injury by regulation of Bcl-2 and downstream pathways in vitro and in vivo.
Collapse
Affiliation(s)
- Yuxiang Dai
- Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Wei Jin
- Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Longyang Cheng
- Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Chen Yu
- Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Cheng Chen
- Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Hongbin Ni
- Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
22
|
Hou Z, Mao J, Lu Y, Li L. rApoptin induces apoptosis in human breast cancer cells via phosphorylation of Nur77 and Akt. Biochem Biophys Res Commun 2018; 498:221-227. [PMID: 29501489 DOI: 10.1016/j.bbrc.2018.02.204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 02/28/2018] [Indexed: 12/13/2022]
Abstract
Breast cancer is the leading cause of cancer incidence and cancer-related mortality among women and is becoming a major public health problem around the world. The current study aims to investigate the possible role and mechanism of recombinant Apoptin (rApoptin), a potential anticancer candidate that minimally impacts normal cells, in the breast cancer cell proliferation and apoptosis in vitro and in vivo. We found that rApoptin could effectively inhibit the proliferation and apoptosis in MCF-7 and MDA-MB-231 cells in vitro, which was further confirmed by flow cytometry analysis. Apoptin partially inhibited MCF-7 cell xenograft tumor development in vivo. Furthermore, we found via western blot that rApoptin-induced apoptosis in MCF-7 and MDA-MB-231 cells was associated with the phosphorylation of Nur77 (p-Nur77) and Akt (p-Akt). In addition, compared with the control groups, rApoptin-treated tissues showed significantly higher expression of Bax and Cyt c while Bcl-2 expression was decreased by rApoptin treatment. Together, our results are the first to demonstrate that rApoptin was able to effectively induce breast cancer cell apoptosis both in vitro and in vivo and that this activity could be regulated by the phosphorylation of Nur77 and Akt and the mitochondrial pathway. Our findings highlight the potential application of rApoptin as a breast cancer treatment.
Collapse
Affiliation(s)
- Zhenhuan Hou
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Jun Mao
- Department of Pathology, Dalian Medical University, Dalian 116044, China; Key Laboratory of Tumor Stem Cell Research of Liaoning Province, Dalian Medical University, Dalian 116044, China; Teaching Laboratory of Morphology, Dalian Medical University, Dalian 116044, China
| | - Ying Lu
- Key Laboratory of Tumor Stem Cell Research of Liaoning Province, Dalian Medical University, Dalian 116044, China; Teaching Laboratory of Morphology, Dalian Medical University, Dalian 116044, China
| | - Lianhong Li
- Department of Pathology, Dalian Medical University, Dalian 116044, China; Key Laboratory of Tumor Stem Cell Research of Liaoning Province, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
23
|
Cao HD, Yang YX, Lü L, Liu SN, Wang PL, Tao XH, Wang LJ, Xiang TX. Attenuated Salmonella Typhimurium Carrying TRAIL and VP3 Genes Inhibits the Growth of Gastric Cancer Cells in Vitro and in Vivo. TUMORI JOURNAL 2018; 96:296-303. [DOI: 10.1177/030089161009600218] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) and apoptin (VP3) of chicken anemia virus can selectively induce apoptosis in human tumor cell lines by two different pathways. Salmonella not only delivers functional genes to mammalian cells but also possesses antitumor activity and therefore could be adopted as a novel vector for anticancer therapy. Materials and methods TRAIL and VP3 genes were cloned into a pBudCE4.1 vector and delivered by attenuated Salmonella typhimurium into gastric cancer cells, and their expression and antitumor effects in nude mice were monitored by Western blot, fluorescence microscopy, MTT assay, TUNEL staining, and immunohistochemistry. Results pBud-VP3 and pBud-TRAIL-VP3 plasmids were constructed to express TRAIL and apoptin in gastric cancer cells, leading to inhibition of cancer cell proliferation after 48 hours (P <0.05). TRAIL and VP3 genes in pBudCE4.1 vector were also successfully delivered by attenuated S. typhimurium into gastric cancer cells in vivo, in which both TRAIL and apoptin were expressed. In vivo data indicated that S. typhimurium carring pBud-TRAIL-VP3 induced significant cell growth inhibition and tumor regression (P <0.05). Moreover, expression of TRAIL and apoptin increased the expression of caspase-3 and caspase-9, resulting in enhanced apoptosis. Conclusion Delivery of TRAIL and VP3 genes by attenuated S. typhimurium can significantly inhibit the growth of gastric cancer cells in vitro and in vivo.
Collapse
Affiliation(s)
- Hong-Dan Cao
- Department of Gastroenterology, the First Affiliated Hospital, Chongqing Medical University, Chongqing
| | - Yin-Xue Yang
- Department of Surgery, the Affiliated Hospital of Ning Xia Medical College, Ning Xia, China
| | - Lin Lü
- Department of Gastroenterology, the First Affiliated Hospital, Chongqing Medical University, Chongqing
| | - Shao-Ning Liu
- Department of Gastroenterology, the First Affiliated Hospital, Chongqing Medical University, Chongqing
| | - Pi-Long Wang
- Department of Gastroenterology, the First Affiliated Hospital, Chongqing Medical University, Chongqing
| | - Xiao-Hong Tao
- Department of Gastroenterology, the First Affiliated Hospital, Chongqing Medical University, Chongqing
| | - Li-Juan Wang
- Department of Gastroenterology, the First Affiliated Hospital, Chongqing Medical University, Chongqing
| | - Ting-Xiu Xiang
- Experimental Research Center, the First Affiliated Hospital, Chongqing Medical University, Chongqing
| |
Collapse
|
24
|
Agha Amiri S, Shahhosseini S, Zarei N, Khorasanizadeh D, Aminollahi E, Rezaie F, Zargari M, Azizi M, Khalaj V. A novel anti-CD22 scFv-apoptin fusion protein induces apoptosis in malignant B-cells. AMB Express 2017; 7:112. [PMID: 28582973 PMCID: PMC5457376 DOI: 10.1186/s13568-017-0410-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/17/2022] Open
Abstract
CD22 marker is a highly internalizing antigen which is located on the surface of B-cells and is being used as a promising target for treatment of B cell malignancies. Monoclonal antibodies targeting CD22 have been introduced and some are currently under investigation in clinical trials. Building on the success of antibody drug conjugates, we developed a fusion protein consisting of a novel anti-CD22 scFv and apoptin and tested binding and therapeutic effects in lymphoma cells. The recombinant protein was expressed in E. coli and successfully purified and refolded. In vitro binding analysis by immunofluorescence and flow cytometry demonstrated that the recombinant protein specifically binds to CD22 positive Raji cells but not to CD22 negative Jurkat cells. The cytotoxic properties of scFv–apoptin were assessed by an MTT assay and Annexin V/PI flow cytometry analysis and showed that the recombinant protein induced apoptosis preferentially in Raji cells with no detectable effects in Jurkat cells. Our findings indicated that the recombinant anti-CD22 scFv–apoptin fusion protein could successfully cross the cell membrane and induce apoptosis with high specificity, make it as a promising molecule for immunotherapy of B-cell malignancies.
Collapse
|
25
|
Fountzilas C, Patel S, Mahalingam D. Review: Oncolytic virotherapy, updates and future directions. Oncotarget 2017; 8:102617-102639. [PMID: 29254276 PMCID: PMC5731986 DOI: 10.18632/oncotarget.18309] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022] Open
Abstract
Oncolytic viruses (OVs) are viral strains that can infect and kill malignant cells while spare their normal counterparts. OVs can access cells through binding to receptors on their surface or through fusion with the plasma membrane and establish a lytic cycle in tumors, while leaving normal tissue essentially unharmed. Multiple viruses have been investigated in humans for the past century. IMLYGIC™ (T-VEC/Talimogene Laherparepvec), a genetically engineered Herpes Simplex Virus, is the first OV approved for use in the United States and the European Union for patients with locally advanced or non-resectable melanoma. Although OVs have a favorable toxicity profile and are impressively active anticancer agents in vitro and in vivo the majority of OVs have limited clinical efficacy as a single agent. While a virus-induced antitumor immune response can enhance oncolysis, when OVs are used systemically, the antiviral immune response can prevent the virus reaching the tumor tissue and having a therapeutic effect. Intratumoral administration can provide direct access to tumor tissue and be beneficial in reducing side effects. Immune checkpoint stimulation in tumor tissue has been noted after OV therapy and can be a natural response to viral-induced oncolysis. Also for immune checkpoint inhibition to be effective in treating cancer, an immune response to tumor neoantigens and an inflamed tumor microenvironment are required, both of which treatment with an OV may provide. Therefore, direct and indirect mechanisms of tumor killing provide rationale for clinical trials investigating the combination of OVs other forms of cancer therapy, including immune checkpoint inhibition.
Collapse
Affiliation(s)
- Christos Fountzilas
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sukeshi Patel
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | |
Collapse
|
26
|
Unexpected heat shock element binding ability and tumor-killing activity of the combinatorial function domain of apoptin. Anticancer Drugs 2017; 28:401-409. [PMID: 28045701 DOI: 10.1097/cad.0000000000000471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Apoptin, derived from the chicken anemia virus, has been found to exert tumor-preferential apoptotic activity. It is a potential anticancer agent with direct clinical applications. However, if this viral protein were to be used as a new drug, it might also induce a strong immune response, causing toxic side effects. In a previous study, our group showed that TAT-apoptin downregulates the stress expression of heat shock protein 70 by competing with heat shock factor protein 1 in binding to the heat shock element (HSE) of the promoter region of heat shock protein 70, thus inducing specific apoptosis in HepG2 cells. In this study, we investigated the HSE-binding properties of the minimal functional region of apoptin. We showed that apoptin's nuclear localization signals 1 and nuclear localization signals 2 represented functional regions that could bind with HSE and that this binding capacity was increased by polymers formed through the introduction of a leucine-rich stretch. Our data also showed that truncated combinatorial apoptin peptide has greater tumor-specific cell-killing activity and could be a potential antitumor agent.
Collapse
|
27
|
Lai GH, Lien YY, Lin MK, Cheng JH, Tzen JT, Sun FC, Lee MS, Chen HJ, Lee MS. VP2 of Chicken Anaemia Virus Interacts with Apoptin for Down-regulation of Apoptosis through De-phosphorylated Threonine 108 on Apoptin. Sci Rep 2017; 7:14799. [PMID: 29093508 PMCID: PMC5665943 DOI: 10.1038/s41598-017-14558-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/11/2017] [Indexed: 12/18/2022] Open
Abstract
Chicken anaemia virus (CAV) is an important contagious agent that causes immunosuppressive disease in chickens. CAV Apoptin is a nucleoplasmic shuffling protein that induces apoptosis in chicken lymphoblastoid cells. In the present study, confocal microscopy revealed co-localisation of expressed CAV non-structural protein VP2 with Apoptin in the nucleus of MDCC-MSB1 cells and the nucleoplasmic compartment of CHO-K1 cells. In vitro pull-down and ex vivo biomolecular fluorescent complementation (BiFC) assays further showed that the VP2 protein directly interacts with Apoptin. Transient co-expression of VP2 and Apoptin in MDCC-MSB1 cells significantly decreased the rate of apoptosis compared with that in cells transfected with the Apoptin gene alone. In addition, the phosphorylation status of threonine 108 (Thr108) of Apoptin was found to decrease upon interaction with VP2. Although dephosphorylated Thr108 did not alter the subcellular distribution of Apoptin in the nucleus of MDCC-MSB1 cells, it did suppress apoptosis. These findings provide the first evidence that VP2 directly interacts with Apoptin in the nucleus to down-regulate apoptosis through alterations in the phosphorylation status of the latter. This information will be useful to further elucidate the underlying mechanism of viral replication in the CAV life cycle.
Collapse
Affiliation(s)
- Guan-Hua Lai
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 40402, Taiwan
| | - Yi-Yang Lien
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 91201, Taiwan
| | - Ming-Kuem Lin
- Department of Chinese Pharmaceutical Science and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan
| | - Jai-Hong Cheng
- Center for Shockwave Medicine and Tissue Engineering, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Jason Tc Tzen
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 40402, Taiwan
| | - Fang-Chun Sun
- Department of Bioresources, Da-Yeh University, Changhua, 515, Taiwan
| | - Meng-Shiunn Lee
- Research Assistance Center, Show Chwan Memorial Hospital, Changhua, 500, Taiwan
| | - Hsi-Jien Chen
- Department of Safety, Health and Environmental Engineering, Ming Chi University of Technology, New Taipei, 24301, Taiwan
| | - Meng-Shiou Lee
- Department of Chinese Pharmaceutical Science and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
28
|
Liu J, Wang GH, Duan YH, Dai Y, Bao Y, Hu M, Zhou YQ, Li M, Jiang F, Zhou H, Yao XS, Zhang XK. Modulation of the Nur77-Bcl-2 apoptotic pathway by p38α MAPK. Oncotarget 2017; 8:69731-69745. [PMID: 29050237 PMCID: PMC5642512 DOI: 10.18632/oncotarget.19227] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/05/2017] [Indexed: 02/02/2023] Open
Abstract
Orphan nuclear receptor Nur77 promotes apoptosis by targeting mitochondria through interaction with Bcl-2, an event that converts Bcl-2 from a survival to killer. However, how the Nur77-Bcl-2 apoptotic pathway is regulated remains largely unknown. In this study, we examined the regulation of the Nur77-Bcl-2 pathway by CCE9, a xanthone compound. Our results demonstrated that the apoptotic effect of CCE9 depended on its induction of Nur77 expression, cytoplasmic localization, and mitochondrial targeting. The activation of the Nur77-Bcl-2 pathway by CCE9 was associated with its activation of p38α MAPK. Inhibition of p38α MAPK activation by knocking down or knocking out p38α MAPK impaired the effect of CCE9 on inducing apoptosis and the expression and cytoplasmic localization of Nur77. In addition, CCE9 activation of p38α MAPK resulted in Bcl-2 phosphorylation and Bcl-2 interaction with Nur77, whereas inhibition of p38α MAPK activation or expression suppressed the interaction. Moreover, mutating Ser87 and Thr56 in the loop of Bcl-2, which are known to be phosphorylated by p38α MAPK, impaired the ability Bcl-2 to interact with Nur77. Together, our results reveal a profound role of p38α MAPK in regulating the Nur77-Bcl-2 apoptotic pathway through its modulation of Nur77 expression, Bcl-2 phosphorylation, and their interaction.
Collapse
Affiliation(s)
- Jie Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Guang-Hui Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Ying-Hui Duan
- Institutes of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou, China
| | - Yi Dai
- Institutes of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou, China
| | - Yuzhou Bao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Mengjie Hu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Yu-Qi Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Mingyu Li
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Fuquan Jiang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Xin-Sheng Yao
- Institutes of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou, China
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China.,Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| |
Collapse
|
29
|
Kumari A, Singh KP, Mandal A, Paswan RK, Sinha P, Das P, Ali V, Bimal S, Lal CS. Intracellular zinc flux causes reactive oxygen species mediated mitochondrial dysfunction leading to cell death in Leishmania donovani. PLoS One 2017; 12:e0178800. [PMID: 28586364 PMCID: PMC5460814 DOI: 10.1371/journal.pone.0178800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023] Open
Abstract
Leishmaniasis caused by Leishmania parasite is a global threat to public health and one of the most neglected tropical diseases. Therefore, the discovery of novel drug targets and effective drug is a major challenge and an important goal. Leishmania is an obligate intracellular parasite that alternates between sand fly and human host. To survive and establish infections, Leishmania parasites scavenge and internalize nutrients from the host. Nevertheless, host cells presents mechanism like nutrient restriction to inhibit microbial growth and control infection. Zinc is crucial for cellular growth and disruption in its homeostasis hinders growth and survival in many cells. However, little is known about the role of zinc in Leishmania growth and survival. In this study, the effect of zinc on the growth and survival of L.donovani was analyzed by both Zinc-depletion and Zinc-supplementation using Zinc-specific chelator N, N, N', N'–tetrakis (2-pyridylmethyl) ethylenediamine (TPEN) and Zinc Sulfate (ZnSO4). Treatment of parasites with TPEN rather than ZnSO4 had significantly affected the growth in a dose- and time-dependent manner. The pre-treatment of promastigotes with TPEN resulted into reduced host-parasite interaction as indicated by decreased association index. Zn depletion resulted into flux in intracellular labile Zn pool and increased in ROS generation correlated with decreased intracellular total thiol and retention of plasma membrane integrity without phosphatidylserine exposure in TPEN treated promastigotes. We also observed that TPEN-induced Zn depletion resulted into collapse of mitochondrial membrane potential which is associated with increase in cytosolic calcium and cytochrome-c. DNA fragmentation analysis showed increased DNA fragments in Zn-depleted cells. In summary, intracellular Zn depletion in the L. donovani promastigotes led to ROS-mediated caspase-independent mitochondrial dysfunction resulting into apoptosis-like cell death. Therefore, cellular zinc homeostasis in Leishmania can be explored for new drug targets and chemotherapeutics to control Leishmanial growth and disease progression.
Collapse
Affiliation(s)
- Anjali Kumari
- Division of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Krishn Pratap Singh
- Laboratory of Molecular Biochemistry and Cell Biology, Division of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Abhishek Mandal
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Ranjeet Kumar Paswan
- Division of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Preeti Sinha
- Division of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Pradeep Das
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Vahab Ali
- Laboratory of Molecular Biochemistry and Cell Biology, Division of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Sanjiva Bimal
- Division of Immunology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
| | - Chandra Shekhar Lal
- Division of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of medical Research), Agamkuan, Patna, Bihar, India
- * E-mail:
| |
Collapse
|
30
|
Apoptin Gene Delivery by the Functionalized Polyamidoamine (PAMAM) Dendrimer Modified with Ornithine Induces Cell Death of HepG2 Cells. Polymers (Basel) 2017; 9:polym9060197. [PMID: 30970874 PMCID: PMC6432117 DOI: 10.3390/polym9060197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 02/02/2023] Open
Abstract
The use of tumor-specific therapeutic agents is a promising option for efficient and safe nonviral gene transfer in gene therapy. In this study, we describe the efficacy of polyamidoamine (PAMAM)-based nonviral gene delivery carriers, namely, an ornithine conjugated PAMAM (PAMAM-O) dendrimer in delivering apoptin, a tumor-specific killer gene, into human hepatocellular carcinoma (HepG2 cells) and dermal fibroblasts. We analyzed the transfection efficiency by the luciferase assay and assessed cell viability in both cell types. The transfection efficiency of the PAMAM-O dendrimer was found to be higher than that of the PAMAM dendrimer. Moreover, the cytotoxicity of the PAMAM-O dendrimer was very low. We treated both cell types with a polyplex of PAMAM-O dendrimer with apoptin, and analyzed its cellular uptake and localization by confocal microscopy. Cell cycle distribution, tetramethylrhodamine, ethyl ester (TMRE) analysis, and transmission electron microscopy imaging showed that apoptin induced cell death in HepG2 cells. We therefore demonstrated that a PAMAM-O/apoptin polyplex can be used as an effective therapeutic strategy in cancer owing to its effectiveness as a suitable nonviral gene vector for gene therapy.
Collapse
|
31
|
Khairallah A, Farag AA, Johar D, Bernstein L. Endocrine Imbalance Associated With Proteome Changes in Diabetes. J Cell Biochem 2017; 118:3569-3576. [PMID: 28419534 DOI: 10.1002/jcb.26071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 04/14/2017] [Indexed: 11/06/2022]
Abstract
The dynamics of cellular metabolism involves rapid interactions between proteins and nucleic acids, proteins and proteins, and signaling. These involve the interactions with respect to the sulfur bond, noncovalent electrostatic interactions, protein structure stabilization and protein-ligand binding, weak electrostatic interactions in proteins, oxygen radicals that initiate a change in conformation and a chain of events. We review a development in molecular medicine that is a very promising work in progress. We also review the current and future research methods involving mitochondria. Long-term effects of diabetes include glycation of proteins, for example, glycohemoglobin (HbA1c), increased risk of cardiovascular diseases, atherosclerosis, retinopathy, nephropathy, and neurological dysfunctions. Tissues are exposed to significant quantities of highly reactive chemical species including nitric oxide • NO and reactive oxygen species ROS over months to years, to an extent generated by mitochondrial activities. The reactions of • NO can be broadly discussed with reference to three main processes which control their fate in biological systems: (1) diffusion and intra-cellular consumption; (2) autooxidation to form nitrous anhydride N2 O3 ; and (3) reaction with superoxide O2• - to form peroxynitrite ONOO-. Reactive nitrogen species produced by macrophages and neutrophils in the interstitial space, with emphasis on • NO, N2 O3 , ONOO-, and nitrogen dioxide radicals • NO2 generate protein and DNA damage. Serum thiol (-SH) groups act as an important extracellular scavenger of peroxides and are therefore helpful in protecting the surrounding tissues. The events described here are a homeostatic endocrine imbalance that is associated with proteostasis. The advances we have seen in untangling this web of interactions are sure to continue at a breathtaking pace. J. Cell. Biochem. 118: 3569-3576, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ahmed Khairallah
- Pharmacology Department, Medical Research Division, National Research Center, Dokki, Cairo, Egypt
| | | | - Dina Johar
- Faculty of Women for Arts, Sciences and Education, Department of Biochemistry and Nutrition, Ain Shams University, Heliopolis, Cairo, Egypt.,Rady College of Medicine, Max Rady Faculty of Health Sciences, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Larry Bernstein
- Triplex Consulting, 54 Firethorn Lane, Northampton, Massachusetts
| |
Collapse
|
32
|
Bae Y, Rhim HS, Lee S, Ko KS, Han J, Choi JS. Apoptin Gene Delivery by the Functionalized Polyamidoamine Dendrimer Derivatives Induces Cell Death of U87-MG Glioblastoma Cells. J Pharm Sci 2017; 106:1618-1633. [PMID: 28188727 DOI: 10.1016/j.xphs.2017.01.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/18/2017] [Accepted: 01/27/2017] [Indexed: 11/08/2022]
Abstract
Malignant glioma is the most common and aggressive form of primary brain tumor in adults. In this study, we describe the efficacy of nonviral gene delivery carriers, histidine- and arginine- or histidine- and lysine-grafted polyamidoamine (PAMAM) dendrimers (PAMAM-H-R and PAMAM-H-K), in delivering a therapeutic and a tumor-selective killer gene, apoptin, using human glioma cells (U87-MG) and newborn human dermal fibroblast cells. We analyzed transfection efficiency using luciferase and a plasmid DNA encoding for enhanced green fluorescent protein and assessed cell viability in both cells. The results show that transfection efficiency of PAMAM-H-R and PAMAM-H-K was greatly increased compared with that of native PAMAM. Moreover, among PAMAM derivatives, cytotoxicity of PAMAM-H-K was very low. We treated both cells with complexes of PAMAM-H-R or PAMAM-H-K and apoptin and analyzed their cellular uptake by flow cytometry and localization by confocal microscopy. Furthermore, cell cycle distribution, caspase 3 activity assay, and JC-1 analysis showed cell death induced by apoptin in U87-MG cells. The present study demonstrates that a PAMAM-H-R/apoptin complex is an effective gene carrier system in glioma cell culture.
Collapse
Affiliation(s)
- Yoonhee Bae
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Hyang-Shuk Rhim
- Department of Biomedical Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea; Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seulgi Lee
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Korea
| | - Kyung Soo Ko
- Department of Internal Medicine, Sanggye Paik Hospital, Cardiovascular and Metabolic Disease Center, Inje University, Seoul, Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea.
| | - Joon Sig Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Korea.
| |
Collapse
|
33
|
Yang E, Li X, Jin N. The chimeric multi-domain proteins mediating specific DNA transfer for hepatocellular carcinoma treatment. Cancer Cell Int 2016; 16:80. [PMID: 27752239 PMCID: PMC5062862 DOI: 10.1186/s12935-016-0351-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 09/20/2016] [Indexed: 01/13/2023] Open
Abstract
AIM This study was aimed to evaluate the therapeutic efficiency of a non-virus based specific chimeric multi-domain DNA transferred with apoptin in human hepatocellular carcinoma (HCC) HepG-2 cells in vitro and in mice H22 cells in vivo. METHODS We firstly constructed the multi-domain recombinant chimeric proteins based on recombinant proteins [G (yeast GAL4), NG (none GAL4), TG (GAL4 + Tat protein) and TNG (Tat protein)] and pUAS-Apoptin plasmid, and transfected them into human HepG-2 cells. The antitumor effect of this multi-domain recombinant chimeric proteins to HCC cells were detected by MTT assay, AO/EB staining, DAPI staining and Annexin V assay. In order to find the pathway of cell apoptosis, the Caspase (1, 3, 6 and 8) activity was detected. We then constructed the H22 liver cancer mice model and analyzed the anti-tumor rate and mice survival rate after treated with G/pUAS-Apoptin NG/pUAS-Apoptin TG/pUAS-Apoptin, and TNG/pUAS-Apoptin. RESULTS MTT results showed that the Tat protein (TG and TNG) significantly induced cell death in a time dependent manner. AO/EB, DAPI, Annexin V and Caspases assay results indicated that the Caspase 1, 3, 6 and 8 were highly expressed in TG/pUAS-Apoptin, and TNG/pUAS-Apoptin treated mouse groups. The antitumor rate and survival rate in TG/pUAS-Apoptin, and TNG/pUAS-Apoptin treated mouse groups were higher than in the other groups. CONCLUSION The Tat-apoptin is a potential anti-tumor agent for HCC treatment with remarkable anti-tumor efficacy and high safety based on non-virus gene transfer system. The anti-tumor function may be associated with high expression of Caspase 1, 3, 6 and 8.
Collapse
Affiliation(s)
- Encheng Yang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 China
| | - Xiao Li
- Institute of Military Veterinary, Academy of Military Medical Sciences of PLA, #666 Liuying West Road, Jingyue District, Changchun, 130122 Jilin province China
| | - Ningyi Jin
- Institute of Military Veterinary, Academy of Military Medical Sciences of PLA, #666 Liuying West Road, Jingyue District, Changchun, 130122 Jilin province China
| |
Collapse
|
34
|
Bae Y, Green ES, Kim GY, Song SJ, Mun JY, Lee S, Park JI, Park JS, Ko KS, Han J, Choi JS. Dipeptide-functionalized polyamidoamine dendrimer-mediated apoptin gene delivery facilitates apoptosis of human primary glioma cells. Int J Pharm 2016; 515:186-200. [PMID: 27732896 DOI: 10.1016/j.ijpharm.2016.09.083] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 09/23/2016] [Accepted: 09/29/2016] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiform (GBM) is the most frequent and aggressive form of brain tumors in adults. However, the development of more efficient and safe nonviral vector gene therapy represents a promising therapeutic approach, using a tumor-specific killer gene, named apoptin. In this study, we describe the efficacy of non-viral gene delivery vectors, the amino acid-conjugated PAMAM derivatives (PAMAM-H-R and PAMAM-H-K) in delivering a therapeutic gene, displaying affinity toward human primary glioma cells (GBL-14 cells) and dermal fibroblasts. We analyzed transfection efficiency, using luciferase (Luci) and a pDNA encoding for enhanced fluorescent protein (EGFP), and cytotoxicity in both cells. The results show that transfection efficiency of PAMAM-H-R improved compared to native PAMAM dendrimer, but cytotoxicity of PAMAM-H-R and PAMAM-H-K were very low. We treated both cells with a polyplex formation of PAMAM-H-R or PAMAM-H-K/apoptin, and analyzed their cellular uptake and localization by flow cytometry and confocal microscopy. Furthermore, we analyzed the endosomal escape effect using TEM images, and found that PAMAM-H-R showed very fast escape from endosome to the cytosol. Caspase 3 activity assay, cell cycle distribution, and JC-1 analysis showed apoptosis induced by apoptin in GBL-14 cells. This indicates that PAMAM-H-R can be a potential nonviral vector gene delivery carrier for brain tumor therapy. The present study demonstrates that PAMAM-H-R/apoptin gene polyplex can be used as an effective therapeutic candidate for GBM due to its selective induction of apoptosis in primary glioma cells as a potential nonviral gene delivery carrier for brain tumor therapy.
Collapse
Affiliation(s)
- Yoonhee Bae
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 614-735, Republic of Korea
| | - Eric S Green
- Salt Lake Community College, Salt Lake City, UT, USA
| | - Goo-Young Kim
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Su Jeong Song
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Ji Young Mun
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Republic of Korea
| | - Sunray Lee
- Cell Engineering for Origin Research Center 46-21, Susong-dog, Jongno-gu, Seoul 110-140, Republic of Korea
| | - Jong-Il Park
- Chungnam National University School of Medicine, Daejeon 301-747, Republic of Korea
| | - Jong-Sang Park
- School of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Kyung Soo Ko
- Department of Internal Medicine, Sanggye Paik Hospital, Cardiovascular and Metabolic Disease Center, Inje University, Seoul 139-707, Republic of Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 614-735, Republic of Korea.
| | - Joon Sig Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Republic of Korea.
| |
Collapse
|
35
|
Zhang J, Hou L, Wu X, Zhao D, Wang Z, Hu H, Fu Y, He J. Inhibitory effect of genetically engineered mesenchymal stem cells with Apoptin on hepatoma cells in vitro and in vivo. Mol Cell Biochem 2016; 416:193-203. [PMID: 27142531 DOI: 10.1007/s11010-016-2707-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/19/2016] [Indexed: 01/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive tumor and has become one of the most frequent causes of cancer death in the world. The rate of post-operative recurrence and metastasis are still high even though after surgical resection. It is a difficult problem with extraordinary importance for the clinical treatment. So stem cell therapy becomes one of the anti-tumor biotherapy methods which is exploring. Due to the feature of homing to tumor site and immunosuppressive, mesenchymal stem cells (MSCs) have the capacity of gene treatment to tumor as a vehicle. Apoptin derived from chicken anemia virus is one kind of protein with an inherent ability to lyse cancer cells while leaving normal cells unharmed. Adenovirus (Ad) vectors can be modified to deliver therapeutic genes with the advantages of low toxicity and high transfer capacity. Now it has not been reported that combining MSCs and Adenovirus with Apoptin are used in HCC treatment. This study intends to construct recombinant adenovirus which expresses Apoptin and then infects human bone marrow MSCs, and explore the migration of MSCs to the hepatoma cells and inhibitory effect of genetically engineered mesenchymal stem cells with Apoptin on hepatoma cells in vitro and in vivo. Our research successfully established the recombinant Ad which was constructed by Ad system, and obtained MSCs which could secrete Apoptin. We found that both the modified MSCs with Apoptin and their conditional medium significantly inhibited the proliferation of liver cancer cells HepG2, which provided a novel means and experimental basis for stem cell treatment for HCC. This study tries to search for a stem cell therapy for cancers, which will provide a new approach and experimental basis for the clinical treatment of cancer. At the same time, this research will also provide experimental basis for a novel in vivo drug delivery system through stem cells as vehicle, which will resolve immune rejection induced by repeated applications of drug directly delivered by Ad vectors and reduce the high cost of a large-scale production and purification of exogenous drugs.
Collapse
Affiliation(s)
- Jingsi Zhang
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| | - Lingling Hou
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China.
| | - Xiaoyan Wu
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| | - Diandian Zhao
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| | - Ziling Wang
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| | - Honggang Hu
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| | - Yuanhui Fu
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| | - Jinsheng He
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, People's Republic of China
| |
Collapse
|
36
|
Gupta SK, Tiwari AK, Gandham RK, Sahoo AP. Combined administration of the apoptin gene and poly (I:C) induces potent anti-tumor immune response and inhibits growth of mouse mammary tumors. Int Immunopharmacol 2016; 35:163-173. [PMID: 27064544 DOI: 10.1016/j.intimp.2016.03.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/25/2016] [Accepted: 03/28/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Many viral proteins exhibit selective cytotoxicity for tumor cells without affecting the normal diploid cells. The apoptin protein of chicken infectious anemia virus is one of such proteins, which has been shown to kill tumor cells specifically. However, an effective cancer treatment strategy also requires assistance from the immune system. Recently, poly (I:C) has been shown to be an effective cancer vaccine adjuvant. AIM In this study, we assessed the anti-tumor potential of apoptin gene transfer alone and in combination with poly (I:C) in a 4T1 mouse mammary tumor model. METHODS 4T1 cells were used to induce mammary tumor in Balb/c mice. Mice bearing tumors were divided into 6 groups, and each group received six intratumoral injections during a period of one month. After the last immunization, the animals were sacrificed, and peripheral blood, spleen, lungs, liver, heart, kidney and tumor tissues were collected for immunological, molecular and pathological analysis. RESULTS We report that intratumoral administration of apoptin plasmid along with poly (I:C) not only significantly inhibited the growth of mammary tumor, but also induced a potent anti-tumor immune response as indicated by the increase in blood CD4+, CD8+ cells and infiltration of immune cells in the tumor tissue. Further, blood serum analysis of the cytokines revealed increased secretion of Th1 cytokines (IFN-γ and IL-2). CONCLUSIONS The results of our study demonstrate that the inclusion of poly (I:C) significantly enhanced the anti-tumor activity of apoptin mainly by inducing a potent anti-tumor immune response. Therefore, we report the use of apoptin and poly (I:C) combination as a novel and powerful strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Shishir Kumar Gupta
- Molecular Biology Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP, India.
| | - Ashok K Tiwari
- Molecular Biology Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP, India.
| | - Ravi Kumar Gandham
- Molecular Biology Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP, India
| | - A P Sahoo
- Molecular Biology Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP, India
| |
Collapse
|
37
|
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
|
38
|
Zhang X, Zhou H, Su Y. Targeting truncated RXRα for cancer therapy. Acta Biochim Biophys Sin (Shanghai) 2016; 48:49-59. [PMID: 26494413 DOI: 10.1093/abbs/gmv104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/24/2015] [Indexed: 01/08/2023] Open
Abstract
Retinoid X receptor-alpha (RXRα), a unique member of the nuclear receptor superfamily, is a well-established drug target, representing one of the most important targets for pharmacologic interventions and therapeutic applications for cancer. However, how RXRα regulates cancer cell growth and how RXRα modulators suppress tumorigenesis are poorly understood. Altered expression and aberrant function of RXRα are implicated in the development of cancer. Previously, several studies had demonstrated the presence of N-terminally truncated RXRα (tRXRα) proteins resulted from limited proteolysis of RXRα in tumor cells. Recently, we discovered that overexpression of tRXRα can promote tumor growth by interacting with tumor necrosis factor-alpha-induced phosphoinositide 3-kinase and NF-κB signal transduction pathways. We also identified nonsteroidal anti-inflammatory drug Sulindac and analogs as effective inhibitors of tRXRα activities via a unique binding mechanism. This review discusses the emerging roles of tRXRα and modulators in the regulation of cancer cell survival and death as well as inflammation and our recent understanding of tRXRα regulation by targeting the alternate binding sites on its surface.
Collapse
Affiliation(s)
- Xiaokun Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| | - Hu Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ying Su
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| |
Collapse
|
39
|
Nur77 is involved in graft infiltrating T lymphocyte apoptosis in rat cardiac transplantation model. Pathol Res Pract 2015; 211:633-40. [DOI: 10.1016/j.prp.2015.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 04/05/2015] [Accepted: 04/17/2015] [Indexed: 12/12/2022]
|
40
|
Jangamreddy JR, Panigrahi S, Lotfi K, Yadav M, Maddika S, Tripathi AK, Sanyal S, Łos MJ. Mapping of apoptin-interaction with BCR-ABL1, and development of apoptin-based targeted therapy. Oncotarget 2015; 5:7198-211. [PMID: 25216532 PMCID: PMC4196195 DOI: 10.18632/oncotarget.2278] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Majority of chronic myeloid leukemia patients experience an adequate therapeutic effect from imatinib however, 26-37% of patients discontinue imatinib therapy due to a suboptimal response or intolerance. Here we investigated derivatives of apoptin, a chicken anemia viral protein with selective toxicity towards cancer cells, which can be directed towards inhibiting multiple hyperactive kinases including BCR-ABL1. Our earlier studies revealed that a proline-rich segment of apoptin interacts with the SH3 domain of fusion protein BCR-ABL1 (p210) and acts as a negative regulator of BCR-ABL1 kinase and its downstream targets. In this study we show for the first time, the therapeutic potential of apoptin-derived decapeptide for the treatment of CML by establishing the minimal region of apoptin interaction domain with BCR-ABL1. We further show that the apoptin decapeptide is able to inhibit BCR-ABL1 down stream target c-Myc with a comparable efficacy to full-length apoptin and Imatinib. The synthetic apoptin is able to inhibit cell proliferation in murine (32Dp210), human cell line (K562), and ex vivo in both imatinib-resistant and imatinib sensitive CML patient samples. The apoptin based single or combination therapy may be an additional option in CML treatment and eventually be feasible as curative therapy.
Collapse
Affiliation(s)
- Jaganmohan R Jangamreddy
- Dept. Clinical & Experimental Medicine, Integrative Regenerative Med. Center (IGEN), Linköping University, Sweden. Authors contributed equally
| | - Soumya Panigrahi
- Dept. Medicine/ Infectious Diseases, Case Western Reserve University, Cleveland, OH 44106, USA. Authors contributed equally
| | - Kourosh Lotfi
- Dept. of Medical and Health Sciences, Linköping University, Department of Hematology, County Council of Östergötland, Linköping, Sweden
| | - Manisha Yadav
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Rd, Lucknow 226031, UP, India
| | - Subbareddy Maddika
- Laboratory of Cell Death & Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| | - Anil Kumar Tripathi
- Department of Clinical Hematology and Medical Oncology, King George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Sabyasachi Sanyal
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Rd, Lucknow 226031, UP, India
| | - Marek J Łos
- Dept. Clinical & Experimental Medicine, Integrative Regenerative Med. Center (IGEN), Linköping University, Sweden. Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
41
|
Lezhnin YN, Kravchenko YE, Frolova EI, Chumakov PM, Chumakov SP. Oncotoxic proteins in cancer therapy: Mechanisms of action. Mol Biol 2015. [DOI: 10.1134/s0026893315020077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Du J, Zhang Y, Xu C, Xu X. Apoptin-modified human mesenchymal stem cells inhibit growth of lung carcinoma in nude mice. Mol Med Rep 2015; 12:1023-9. [PMID: 25816208 PMCID: PMC4438975 DOI: 10.3892/mmr.2015.3501] [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: 03/12/2014] [Accepted: 02/13/2015] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) represent a novel carrier for gene therapy and apoptin is a potential tumor-selective apoptosis-inducing protein. In the present study, the anti-tumoral effect of MSCs modified with apoptin against lung carcinoma was evaluated. Apoptin protein was expressed in a prokaryotic expression system and purified by affinity chromatography. Subsequently, anti-apoptin antibody was prepared by immunizing BALB/c mice with purified apoptin protein. Human MSCs were isolated, amplified and transduced with lentiviral vectors encoding full-length apoptin, in which the secretory signal and protein transduction sequence were added into the amino terminus to assist apoptin in entering into target cells. The differentiation and apoptin expression of apoptin-modified MSCs were confirmed. Subsequently, the anti-tumor effect of apoptin-modified MSCs was measured in vitro and in vivo. Following modification with apoptin, MSCs retained their differentiation capacity, and successfully synthesized and secreted apoptin, which entered target cells and selectively induced lung cancer cell apoptosis through activating caspase-3. The percentage of tumor cells with activated caspase-3 in the apoptin-modified MSCs group was markedly higher than that in the MSCs group (16.5±2.9% at 24 h and 27.3±2.0% at 48 h vs. 3.4±1.1% at 24 h and 2.2±0.6% at 48 h). When injected into nude mice, apoptin-modified MSCs inhibited the growth of lung carcinoma compared with that in the control groups (0.14±0.02 g vs. 0.21±0.04 g vs. 0.31±0.05 g, P<0.05). The results of the present study provided preclinical support of apoptin-based cancer therapy with MSCs as cellular vehicles.
Collapse
Affiliation(s)
- Jingchun Du
- Department of Laboratory Medicine, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Yanling Zhang
- Department of Laboratory Medicine, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Chun Xu
- Department of Clinical Immunology, School of Kingmed Diagnostics, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Xia Xu
- Department of Clinical Immunology, School of Kingmed Diagnostics, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| |
Collapse
|
43
|
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
|
44
|
Zhou Y, Zhao W, Xie G, Huang M, Hu M, Jiang X, Zeng D, Liu J, Zhou H, Chen H, Wang GH, Zhang XK. Induction of Nur77-dependent apoptotic pathway by a coumarin derivative through activation of JNK and p38 MAPK. Carcinogenesis 2014; 35:2660-9. [PMID: 25187486 DOI: 10.1093/carcin/bgu186] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Coumarins are plant-derived natural products with a broad range of known pharmacological activities including anticancer effects. However, the molecular mechanisms by which this class of promising compounds exerts their anticancer effects remain largely unknown. We report here that a furanocoumarin named apaensin could effectively induce apoptosis of cancer cells through its activation of Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Apoptosis induction by apaensin in cancer cells was suppressed by chemical inhibitors of JNK and p38 MAPK. Inhibition of the expression of orphan nuclear receptor Nur77 by small interfering RNA (siRNA) approach also abrogated the death effect of apaensin. Molecular analysis demonstrated that JNK activation was required for the nuclear export of Nur77, a known apoptotic event in cancer cells. Although p38 MAPK activation was not involved in Nur77 nuclear export, it was essential for Nur77 mitochondrial targeting through induction of Nur77 interaction with Bcl-2, which is also known to convert Bcl-2 from an antiapoptotic to a proapoptotic molecule. Together, our results identify a new natural product that targets orphan nuclear receptor Nur77 through its unique activation of JNK and p38 MAPK and provide insight into the complex regulation of the Nur77-Bcl-2 apoptotic pathway.
Collapse
Affiliation(s)
- Yuqi Zhou
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Wei Zhao
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Guobin Xie
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Mingfeng Huang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Mengjie Hu
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Xin Jiang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Dequan Zeng
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Jie Liu
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Hu Zhou
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Haifeng Chen
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Guang-Hui Wang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Xiao-Kun Zhang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
45
|
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
|
46
|
Celastrol induces apoptosis in gefitinib-resistant non-small cell lung cancer cells via caspases-dependent pathways and Hsp90 client protein degradation. Molecules 2014; 19:3508-22. [PMID: 24662070 PMCID: PMC6271537 DOI: 10.3390/molecules19033508] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/26/2014] [Accepted: 03/12/2014] [Indexed: 12/17/2022] Open
Abstract
Celastrol, a triterpene extracted from the Chinese herb Tripterygium wilfordii, has been shown to have multiple bioactivities. Although among these activities, its anti-cancer effects have attracted the most attention, the effect of celastrol on gefitinib-resistant non-small cell lung cancer (NSCLC) cells is not clearly known. Here, we examined the potency of celastrol in three different NSCLC cell lines. We explored its treatment mechanism in two gefitinib-resistant NSCLC cell lines (H1650 and H1975). Our data demonstrated that celastrol exerted its apoptotic effect in a dose- and time-dependent manner. Also, the mitochondria membrane potential was gradually lost and the ratio of Bax/Bcl-2 increased after the treatment of celastrol, both of which are indicators of mitochondria membrane integrity. Although the caspases were activated, the treatment with pan-caspase inhibitor could partially inhibit the level of apoptosis. Moreover, the protein level of Hsp90 client proteins, EGFR and AKT, was measured. Interestingly, both client proteins were remarkably down-regulated after the treatment of celastrol. Taken together, our data showed that celastrol may be developed as a promising agent for treating gefitinib-resistant NSCLCs by inducing apoptosis through caspase-dependent pathways and Hsp90 client protein degradation.
Collapse
|
47
|
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
|
48
|
λ Phage nanobioparticle expressing apoptin efficiently suppress human breast carcinoma tumor growth in vivo. PLoS One 2013; 8:e79907. [PMID: 24278212 PMCID: PMC3838365 DOI: 10.1371/journal.pone.0079907] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 10/02/2013] [Indexed: 12/21/2022] Open
Abstract
Using phages is a novel field of cancer therapy and phage nanobioparticles (NBPs) such as λ phage could be modified to deliver and express genetic cassettes into eukaryotic cells safely in contrast with animal viruses. Apoptin, a protein from chicken anemia virus (CAV) has the ability to specifically induce apoptosis only in carcinoma cells. We presented a safe method of breast tumor therapy via the apoptin expressing λ NBPs. Here, we constructed a λ ZAP-CMV-apoptin recombinant NBP and investigated the effectiveness of its apoptotic activity on BT-474, MDA-MB-361, SKBR-3, UACC-812 and ZR-75 cell lines that over-expressing her-2 marker. Apoptosis was evaluated via annexin-V fluorescent iso-thiocyanate/propidium iodide staining, flow-cytometric method and TUNEL assay. Transfection with NBPs carrying λ ZAP-CMV-apoptin significantly inhibited growth of all the breast carcinoma cell lines in vitro. Also nude mice model implanted BT-474 human breast tumor was successfully responded to the systemic and local injection of untargeted recombinant λ NBPs. The results presented here reveal important features of recombinant λ nanobioparticles to serve as safe delivery and expression platform for human cancer therapy.
Collapse
|
49
|
Pennant WA, An S, Gwak SJ, Choi S, Banh DT, Nguyen ABL, Song HY, Ha Y, Park JS. Local non-viral gene delivery of apoptin delays the onset of paresis in an experimental model of intramedullary spinal cord tumor. Spinal Cord 2013; 52:3-8. [DOI: 10.1038/sc.2013.106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 07/18/2013] [Accepted: 08/06/2013] [Indexed: 12/25/2022]
|
50
|
Ji X, Li J, Zhu L, Cai J, Zhang J, Qu Y, Zhang H, Liu B, Zhao R, Zhu Z. CHD1L promotes tumor progression and predicts survival in colorectal carcinoma. J Surg Res 2013; 185:84-91. [PMID: 23746766 DOI: 10.1016/j.jss.2013.05.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 04/17/2013] [Accepted: 05/02/2013] [Indexed: 11/24/2022]
Abstract
BACKGROUND To evaluate the expression of chromodomain helicase/adenosine triphosphatase DNA binding protein 1-like gene (CHD1L) in colorectal carcinoma (CRC) and its clinical significance. Its oncogenic ability was also investigated. MATERIALS AND METHODS CHD1L amplification and overexpression were detected by fluorescence in situ hybridization, real-time reverse transcriptase-polymerase chain reaction, and immunohistochemistry in 86 patients with CRC. The correlation between the clinical characteristics and prognosis was also determined. To evaluate the tumorigenic ability of CHD1L, it was cloned into expression vector pcDNA3.1(+) and transfected into CRC cell line SW1116. Next, the changes in the biologic behavior of the CRC cells, including cell proliferation, adhesion, migration, and invasion, were examined. Apoptosis and the cell cycle of the CRC cells were detected using flow cytometry. RESULTS We have demonstrated that CHD1L is frequently amplified and overexpressed in CRC. Overexpression of CHD1L correlated with a large tumor size, deep tumor invasion, and a high histologic grade. It also conferred worse disease-free survival. CHD1L-transfected cells possessed a strong oncogenic ability, increasing the tumorigenicity in nude mice, which could be effectively suppressed by small interfering RNA against CHD1L. Functional studies showed that overexpression of CHD1L could promote G1/S-phase cells and inhibit apoptosis. CONCLUSIONS Our results suggest that CHD1L is the target oncogene within the 1q21 amplicon and plays a pivotal role in CRC pathogenesis.
Collapse
Affiliation(s)
- Xiaopin Ji
- Shanghai Key Laboratory of Gastric Neoplasms, Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|