1
|
Sherwood M, Mitsugi TG, Kaid C, Coke B, Zatz M, Maringer K, Okamoto OK, Ewing RM. Multi-omics analysis reveals key immunogenic signatures induced by oncolytic Zika virus infection of paediatric brain tumour cells. Sci Rep 2025; 15:13090. [PMID: 40240536 PMCID: PMC12003866 DOI: 10.1038/s41598-025-97804-8] [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: 01/08/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Brain tumours disproportionately affect children and are the largest cause of paediatric cancer-related death. Novel therapies that engage the immune system, such as oncolytic viruses (OVs), hold great promise and are desperately needed. Zika virus (ZIKV) infects and destroys aggressive cells from multiple paediatric central nervous system (CNS) tumours. Despite this, the molecular mechanisms underpinning this response are largely unknown. We comprehensively investigate the transcriptomic response of paediatric medulloblastoma and atypical teratoid rhabdoid tumour (ATRT) cells to ZIKV infection. We observe conserved TNF signalling and cytokine signalling-related signatures and show that the TNF-alpha signalling pathway is implicated in oncolysis by reducing the viability of ZIKV-infected brain tumour cells. Our findings highlight TNF-alpha as a potential prognostic marker for oncolytic ZIKV (oZIKV) therapy. Complementing our analysis with a 49-plex ELISA, we demonstrate that ZIKV infection induces a clinically relevant and diverse pro-inflammatory brain tumour cell secretome, including TNF-alpha. We assess publicly available scRNA-Seq data to model how ZIKV-induced secretome paracrine and endocrine signalling may orchestrate the anti-tumoural immune response during oZIKV infection of brain tumours. Our findings significantly contribute to understanding the molecular mechanisms governing oZIKV infection and will help pave the way towards oZIKV therapy.
Collapse
Affiliation(s)
- Matthew Sherwood
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, B85, Life Sciences Building, University Road, Highfield, Southampton, Hants., SO17 1BJ, UK
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
| | - Thiago G Mitsugi
- Centro de Estudos do Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Cidade Universitária, São Paulo, SP, 05508-090, Brazil
| | - Carolini Kaid
- Centro de Estudos do Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Cidade Universitária, São Paulo, SP, 05508-090, Brazil
| | - Brandon Coke
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, B85, Life Sciences Building, University Road, Highfield, Southampton, Hants., SO17 1BJ, UK
| | - Mayana Zatz
- Centro de Estudos do Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Cidade Universitária, São Paulo, SP, 05508-090, Brazil
| | - Kevin Maringer
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| | - Oswaldo K Okamoto
- Centro de Estudos do Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Cidade Universitária, São Paulo, SP, 05508-090, Brazil.
| | - Rob M Ewing
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, B85, Life Sciences Building, University Road, Highfield, Southampton, Hants., SO17 1BJ, UK.
| |
Collapse
|
2
|
Otero JG, Belmonte-Beitia J, Jiménez-Sánchez J. Exploring neuroblastoma's cellular microenvironment: A novel approach using cellular automata to model Celyvir treatment. Comput Biol Med 2025; 188:109782. [PMID: 39946782 DOI: 10.1016/j.compbiomed.2025.109782] [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: 10/15/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025]
Abstract
Neuroblastoma is a significant health concern in children, as it is one of the most common types of cancer among this age group and is associated with poor survival rates. Currently, there are no effective therapies that significantly improve outcomes for these patients. This study explores the efficacy of Celyvir - an advanced therapy comprising mesenchymal stem cells (MSCs) carrying the oncolytic virus ICOVIR 5 - against neuroblastoma, by means of an individual-based model. A probabilistic cellular automaton was developed to implement the dynamic interactions between neuroblastoma cells, T lymphocytes, and the therapeutic agent Celyvir. The model examines various sizes, shapes, and positions of the tumour within a lattice, along with different infection probabilities associated with the action of Celyvir and various treatment schedules. This analysis identifies the most influential infection probabilities according to the cellular automaton model, and demonstrates that different treatment regimens can effectively eradicate the tumour, in contrast to standard clinical approaches. Additionally, Kaplan-Meier curves have been generated to assess different treatment schedules under specific tumour scenarios, highlighting the importance of precise treatment scheduling to optimise therapeutic outcomes. This study provides insights into the potential of Celyvir in neuroblastoma treatment, emphasising the need to understand tumour dynamics and strategically implement treatment schemes to improve clinical outcomes.
Collapse
Affiliation(s)
- José García Otero
- Mathematical Oncology Laboratory (MOLAB), University Castilla-La Mancha, Avda. Camilo José Cela s/n, Ciudad Real, 13071, Castilla-La Mancha, Spain.
| | - Juan Belmonte-Beitia
- Mathematical Oncology Laboratory (MOLAB), University Castilla-La Mancha, Avda. Camilo José Cela s/n, Ciudad Real, 13071, Castilla-La Mancha, Spain.
| | - Juan Jiménez-Sánchez
- Mathematical Oncology Laboratory (MOLAB), University Castilla-La Mancha, Avda. Camilo José Cela s/n, Ciudad Real, 13071, Castilla-La Mancha, Spain; Dipartimento di Scienze Matematiche Giuseppe Luigi Lagrange, Politecnico di Torino, Corso Duca degli Abruzzi, 24, 10129, Turin, Italy.
| |
Collapse
|
3
|
de Sena Barbosa MG, Messias BR, Tatit RT, de Paula MCG, Júnior VBS, Braga MGB, Santos CVM, Cobos LD, da Silva VO, Figueiredo EG, Rabelo NN, Chaurasia B. Zika virus and brain cancer: Can Zika be an effective treatment for brain cancer? A systematic review. Oncotarget 2024; 15:662-673. [PMID: 39347716 PMCID: PMC11441410 DOI: 10.18632/oncotarget.28647] [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: 06/21/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024] Open
Abstract
INTRODUCTION Many studies have highlighted the use of oncolytic viruses as a new class of therapeutic agents for central nervous system (CNS) tumors, especially glioblastomas (GMB). Zika Virus (ZIKV) proteins targeted to specific stem cells have been studied in vitro and animal models with promising results. MATERIALS AND METHODS A systematic review was evaluated the efficacy and safety of the ZIKV use for CNS tumors treatment. Data were extracted and the in vivo studies were evaluated using the Robins-I tool. We assessed bias in each study using criteria such as selection bias, performance bias, detection bias, attrition bias, reporting bias, and others. According to Cochrane guidelines, bias was classified as high, low, or uncertain. High bias occurred when studies did not meet the criteria. Low bias was assigned when criteria were clearly met. Uncertain bias reflected insufficient information for a clear classification. RESULTS The 14 included studies shown that ZIKV reduced cell viability or inhibited the growth, proliferation of glioma stem cells (GSCs), and Bcl2 expression - which could potentially enhance the effect of chemotherapy/radiotherapy; caused cytopathic effects, induced tumor cell damage, manifested oncolytic properties, and even selectively safely killed GSCs; ultimately, it led to significant tumor remission and enhanced long-term survival through enhanced T-cell response. CONCLUSIONS Although current evidence suggests ZIKV as a promising treatment for CNS tumors and may improve survival when combined with surgery and radiotherapy. Despite limited human evidence, it shows potential benefits. Further research is needed to confirm safety, efficacy, and optimize treatment in humans.
Collapse
Affiliation(s)
| | - Beatriz Rodrigues Messias
- Hospital Israelita Albert Einstein, University of Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
| | - Rafael Trindade Tatit
- Hospital Israelita Albert Einstein, University of Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
| | | | | | | | - Caio Vinícius Marcolino Santos
- Department of Neurosurgery, Nove de Julho University, Campus Vergueiro, São Paulo, Brazil
- Division of Neurosurgery, School of Medicine-University of São Paulo (FMUSP), Hospital das Clínicas/FMUSP, São Paulo, Brazil
| | - Luiza D'Ottaviano Cobos
- Department of Neurosurgery, José do Rosário Vellano University, Alfenas, Minas Gerais, Brazil
- Division of Neurosurgery, School of Medicine-University of São Paulo (FMUSP), Hospital das Clínicas/FMUSP, São Paulo, Brazil
| | | | - Eberval Gadelha Figueiredo
- Division of Neurosurgery, School of Medicine-University of São Paulo (FMUSP), Hospital das Clínicas/FMUSP, São Paulo, Brazil
| | - Nicollas Nunes Rabelo
- Division of Neurosurgery, School of Medicine-University of São Paulo (FMUSP), Hospital das Clínicas/FMUSP, São Paulo, Brazil
| | - Bipin Chaurasia
- Department of Neurosurgery, Neurosurgery Clinic, Birgunj, Nepal
| |
Collapse
|
4
|
Kim D, Puig A, Rabiei F, Hawkins EJ, Hernandez TF, Sung CK. Optimization of SOX2 Expression for Enhanced Glioblastoma Stem Cell Virotherapy. Symmetry (Basel) 2024; 16:1186. [PMID: 40342640 PMCID: PMC12061075 DOI: 10.3390/sym16091186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Abstract
The Zika virus has been shown to infect glioblastoma stem cells via the membrane receptorα v β 5 , which is activated by the stem-specific transcription factor SOX2. Since the expression level of SOX2 is an important predictive marker for successful virotherapy, it is important to understand the fundamental mechanisms of the role of SOX2 in the dynamics of cancer stem cells and Zika viruses. In this paper, we develop a mathematical ODE model to investigate the effects of SOX2 expression levels on Zika virotherapy against glioblastoma stem cells. Our study aimed to identify the conditions under which SOX2 expression level, viral infection, and replication can reduce or eradicate the glioblastoma stem cells. Analytic work on the existence and stability conditions of equilibrium points with respect to the basic reproduction number are provided. Numerical results were in good agreement with analytic solutions. Our results show that critical threshold levels of both SOX2 and viral replication, which change the stability of equilibrium points through population dynamics such as transcritical and Hopf bifurcations, were observed. These critical thresholds provide the optimal conditions for SOX2 expression levels and viral bursting sizes to enhance therapeutic efficacy of Zika virotherapy against glioblastoma stem cells. This study provides critical insights into optimizing Zika virus-based treatment for glioblastoma by highlighting the essential role of SOX2 in viral infection and replication.
Collapse
Affiliation(s)
- Dongwook Kim
- Department of Mathematics, College of Arts and Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Abraham Puig
- Department of Mathematics, College of Arts and Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Faranak Rabiei
- Department of Mathematics, College of Arts and Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Erial J. Hawkins
- Department of Biological and Health Sciences, College of Arts and Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Talia F. Hernandez
- Department of Biological and Health Sciences, College of Arts and Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Chang K. Sung
- Department of Biological and Health Sciences, College of Arts and Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| |
Collapse
|
5
|
Sherwood M, Zhou Y, Sui Y, Wang Y, Skipp P, Kaid C, Gray J, Okamoto K, Ewing RM. Integrated re-analysis of transcriptomic and proteomic datasets reveals potential mechanisms for Zika viral-based oncolytic therapy in neuroblastoma. F1000Res 2024; 12:719. [PMID: 38903860 PMCID: PMC11187533 DOI: 10.12688/f1000research.132627.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
Background Paediatric neuroblastoma and brain tumours account for a third of all childhood cancer-related mortality. High-risk neuroblastoma is highly aggressive and survival is poor despite intensive multi-modal therapies with significant toxicity. Novel therapies are desperately needed. The Zika virus (ZIKV) can access the nervous system and there is growing interest in employing ZIKV as a potential therapy against paediatric nervous system tumours, including neuroblastoma. Methods Here, we perform extensive data mining, integration and re-analysis of ZIKV infection datasets to highlight molecular mechanisms that may govern the oncolytic response in neuroblastoma cells. We collate infection data of multiple neuroblastoma cell lines by different ZIKV strains from a body of published literature to inform the susceptibility of neuroblastoma to the ZIKV oncolytic response. Integrating published transcriptomics, interaction proteomics, dependency factor and compound datasets we propose the involvement of multiple host systems during ZIKV infection. Results Through data mining of published literature, we observed most paediatric neuroblastoma cell lines to be highly susceptible to ZIKV infection and propose the PRVABC59 ZIKV strain to be the most promising candidate for neuroblastoma oncolytic virotherapy. ZIKV induces TNF signalling, lipid metabolism, the Unfolded Protein Response (UPR), and downregulates cell cycle and DNA replication processes. ZIKV infection is dependent on sterol regulatory element binding protein (SREBP)-regulated lipid metabolism and three protein complexes; V-ATPase, ER Membrane Protein Complex (EMC) and mammalian translocon. We propose ZIKV non-structural protein 4B (NS4B) as a likely mediator of ZIKVs interaction with IRE1-mediated UPR, lipid metabolism and mammalian translocon. Conclusions Our work provides a significant understanding of ZIKV infection in neuroblastoma cells, which will facilitate the progression of ZIKV-based oncolytic virotherapy through pre-clinical research and clinical trials.
Collapse
Affiliation(s)
- Matt Sherwood
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, England, SO17 1BJ, UK
| | - Yilu Zhou
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, England, SO17 1BJ, UK
| | - Yi Sui
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, England, SO17 1BJ, UK
| | - Yihua Wang
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, England, SO17 1BJ, UK
| | - Paul Skipp
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, England, SO17 1BJ, UK
| | - Carolini Kaid
- Human Genome and Stem-Cell Center (HUG-CELL), Biosciences Institute, Universidade de Sao Paulo, São Paulo, State of São Paulo, Brazil
| | - Juliet Gray
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, England, UK
| | - Keith Okamoto
- Human Genome and Stem-Cell Center (HUG-CELL), Biosciences Institute, Universidade de Sao Paulo, São Paulo, State of São Paulo, Brazil
| | - Rob M. Ewing
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, England, SO17 1BJ, UK
| |
Collapse
|
6
|
Wang H, Shi P, Shi X, Lv Y, Xie H, Zhao H. Surprising magic of CD24 beyond cancer. Front Immunol 2024; 14:1334922. [PMID: 38313430 PMCID: PMC10834733 DOI: 10.3389/fimmu.2023.1334922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/28/2023] [Indexed: 02/06/2024] Open
Abstract
CD24 has emerged as a molecule of significant interest beyond the oncological arena. Recent studies have unveiled its surprising and diverse roles in various biological processes and diseases. This review encapsulates the expanding spectrum of CD24 functions, delving into its involvement in immune regulation, cancer immune microenvironment, and its potential as a therapeutic target in autoimmune diseases and beyond. The 'magic' of CD24, once solely attributed to cancer, now inspires a new paradigm in understanding its multifunctionality in human health and disease, offering exciting prospects for medical advancements.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Shi
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinyu Shi
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaqing Lv
- Department of Outpatient, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongwei Xie
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Mazar J, Brooks JK, Peloquin M, Rosario R, Sutton E, Longo M, Drehner D, Westmoreland TJ. The Oncolytic Activity of Zika Viral Therapy in Human Neuroblastoma In Vivo Models Confers a Major Survival Advantage in a CD24-dependent Manner. CANCER RESEARCH COMMUNICATIONS 2024; 4:65-80. [PMID: 38214542 PMCID: PMC10775766 DOI: 10.1158/2767-9764.crc-23-0221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/14/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Neuroblastoma is the most common extracranial tumor, accounting for 15% of all childhood cancer-related deaths. The long-term survival of patients with high-risk tumors is less than 40%, and MYCN amplification is one of the most common indicators of poor outcomes. Zika virus (ZIKV) is a mosquito-borne flavivirus associated with mild constitutional symptoms outside the fetal period. Our published data showed that high-risk and recurrent neuroblastoma cells are permissive to ZIKV infection, resulting in cell type-specific lysis. In this study, we assessed the efficacy of ZIKV as an oncolytic treatment for high-risk neuroblastoma using in vivo tumor models. Utilizing both MYCN-amplified and non-amplified models, we demonstrated that the application of ZIKV had a rapid tumoricidal effect. This led to a nearly total loss of the tumor mass without evidence of recurrence, offering a robust survival advantage to the host. Detection of the viral NS1 protein within the tumors confirmed that a permissive infection preceded tissue necrosis. Despite robust titers within the tumor, viral shedding to the host was poor and diminished rapidly, correlating with no detectable side effects to the murine host. Assessments from both primary pretreatment and recurrent posttreatment isolates confirmed that permissive sensitivity to ZIKV killing was dependent on the expression of CD24, which was highly expressed in neuroblastomas and conferred a proliferative advantage to tumor growth. Exploiting this viral sensitivity to CD24 offers the possibility of its use as a prognostic target for a broad population of expressing cancers, many of which have shown resistance to current clinical therapies. SIGNIFICANCE Sensitivity to the tumoricidal effect of ZIKV on high-risk neuroblastoma tumors is dependent on CD24 expression, offering a prognostic marker for this oncolytic therapy in an extensive array of CD24-expressing cancers.
Collapse
Affiliation(s)
- Joseph Mazar
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
- Burnett School of Biological Sciences, The University of Central Florida College of Medicine, Orlando, Florida
| | | | | | - Rosa Rosario
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
| | - Emma Sutton
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
| | - Matthew Longo
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
- Burnett School of Biological Sciences, The University of Central Florida College of Medicine, Orlando, Florida
| | - Dennis Drehner
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
| | - Tamarah J. Westmoreland
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
- Burnett School of Biological Sciences, The University of Central Florida College of Medicine, Orlando, Florida
| |
Collapse
|
8
|
Morovati S, Mohammadi A, Masoudi R, Heidari AA, Asad Sangabi M. The power of mumps virus: Matrix protein activates apoptotic pathways in human colorectal cell lines. PLoS One 2023; 18:e0295819. [PMID: 38091318 PMCID: PMC10718445 DOI: 10.1371/journal.pone.0295819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
New therapeutic approaches can significantly impact the control of colorectal cancer (CRC), which is increasing worldwide. In this study, we investigated the potential of targeting viral proteins to combat cancer cells. Specifically, we examined the anticancer potential of the matrix (M) protein of the mumps virus Hoshino strain in SW480 CRC cell lines. To begin, we individually transfected SW480 cells with pcDNA3 plasmids containing the mumps virus M gene. We then investigated the percentage of cell death, caspase activity, and the expression levels of genes involved in apoptosis pathways. Following this, we performed bioinformatics analysis on the M protein to identify any similarities with Bcl-2 family members and their viral homologs. Our diagnostic methods showed that treatment with the mumps M protein induced apoptosis and upregulated the expression and activity of pro-apoptotic proteins in SW480 CRC cells compared to the control and vector groups. Based on our bioinformatics studies, we proposed that the BH3 motif in the M protein may trigger apoptosis in CRC cells by interacting with cellular Bax. Overall, our study showed for the first time that the mumps virus M protein could be considered as a targeted treatment for CRC by inducing apoptotic pathways.
Collapse
Affiliation(s)
- Solmaz Morovati
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Mohammadi
- Department of Pathobiology, Division of Virology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ramin Masoudi
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amir Ali Heidari
- Department of Clinical Sciences, Division of Aquatic Animal Health and Diseases, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mehdi Asad Sangabi
- Department of Pathobiology, Division of Virology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
9
|
Nazarenko AS, Vorovitch MF, Biryukova YK, Pestov NB, Orlova EA, Barlev NA, Kolyasnikova NM, Ishmukhametov AA. Flaviviruses in AntiTumor Therapy. Viruses 2023; 15:1973. [PMID: 37896752 PMCID: PMC10611215 DOI: 10.3390/v15101973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 10/29/2023] Open
Abstract
Oncolytic viruses offer a promising approach to tumor treatment. These viruses not only have a direct lytic effect on tumor cells but can also modify the tumor microenvironment and activate antitumor immunity. Due to their high pathogenicity, flaviviruses have often been overlooked as potential antitumor agents. However, with recent advancements in genetic engineering techniques, an extensive history with vaccine strains, and the development of new attenuated vaccine strains, there has been a renewed interest in the Flavivirus genus. Flaviviruses can be genetically modified to express transgenes at acceptable levels, and the stability of such constructs has been greatly improving over the years. The key advantages of flaviviruses include their reproduction cycle occurring entirely within the cytoplasm (avoiding genome integration) and their ability to cross the blood-brain barrier, facilitating the systemic delivery of oncolytics against brain tumors. So far, the direct lytic effects and immunomodulatory activities of many flaviviruses have been widely studied in experimental animal models across various types of tumors. In this review, we delve into the findings of these studies and contemplate the promising potential of flaviviruses in oncolytic therapies.
Collapse
Affiliation(s)
- Alina S. Nazarenko
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Mikhail F. Vorovitch
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Yulia K. Biryukova
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Nikolay B. Pestov
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Ekaterina A. Orlova
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Nickolai A. Barlev
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Nadezhda M. Kolyasnikova
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Aydar A. Ishmukhametov
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| |
Collapse
|
10
|
Mundhra S, Bondre VP. Higher replication potential of West Nile virus governs apoptosis induction in human neuroblastoma cells. Apoptosis 2023:10.1007/s10495-023-01844-2. [PMID: 37186273 DOI: 10.1007/s10495-023-01844-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2023] [Indexed: 05/17/2023]
Abstract
The extent of neuronal cell damage caused by West Nile virus (WNV) infection governs the disease severity ranging from mild, febrile illness to fatal encephalitis. Availability of naturally occurring genetic variants is helpful to study viral factors governing differential pathogenesis. During WNV infection, apoptosis serves as a virulence determinant positively contributing to viral pathogenesis. We investigated the levels of apoptosis induced by a low neurovirulent WNV lineage 5 strain 804994 and a high neurovirulent lineage 1 strain 68856 in human neuroblastoma cells, IMR-32. Our investigations clearly show the correlation between higher multiplication capacities of 68856 with higher levels of cytopathology induced by apoptosis. We observed activation of both the extrinsic and intrinsic apoptotic pathways during WNV infection. Infection with higher neurovirulent strain resulted in higher upregulation of pro-apoptotic proteins including death receptors (DR), adaptor protein, BH3-only regulatory proteins and higher cleavage of initiator caspases of both pathways. These results suggest that the virulence of a WNV strain may correlate with its higher replication fitness and ability to cause more cellular damage.
Collapse
Affiliation(s)
- Sonal Mundhra
- Encephalitis Group, ICMR-National Institute of Virology, Pashan - Sus Road, Pune, Maharashtra, 411021, India
| | - Vijay P Bondre
- Encephalitis Group, ICMR-National Institute of Virology, Pashan - Sus Road, Pune, Maharashtra, 411021, India.
| |
Collapse
|
11
|
CD24 Expression Dampens the Basal Antiviral State in Human Neuroblastoma Cells and Enhances Permissivity to Zika Virus Infection. Viruses 2022; 14:v14081735. [PMID: 36016357 PMCID: PMC9416398 DOI: 10.3390/v14081735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
Zika virus (ZIKV) exhibits distinct selectivity for infection of various cells and tissues, but how host cellular factors modulate varying permissivity remains largely unknown. Previous studies showed that the neuroblastoma cell line SK-N-AS (expressing low levels of cellular protein CD24) was highly restricted for ZIKV infection, and that this restriction was relieved by ectopic expression of CD24. We tested the hypothesis that CD24 expression allowed ZIKV replication by suppression of the antiviral response. SK-N-AS cells expressing an empty vector (termed CD24-low cells) showed elevated basal levels of phosphorylated STAT1, IRF-1, IKKE, and NFκB. In response to exogenously added type I interferon (IFN-I), CD24-low cells had higher-level induction of antiviral genes and activity against two IFN-I-sensitive viruses (VSV and PIV5-P/V) compared to SK-N-AS cells with ectopic CD24 expression (termed CD24-high cells). Media-transfer experiments showed that the inherent antiviral state of CD24-low cells was not dependent on a secreted factor such as IFN-I. Transcriptomics analysis revealed that CD24 expression decreased expression of genes involved in intracellular antiviral pathways, including IFN-I, NFκB, and Ras. Our findings that CD24 expression in neuroblastoma cells represses intracellular antiviral pathways support the proposal that CD24 may represent a novel biomarker in cancer cells for susceptibility to oncolytic viruses.
Collapse
|
12
|
de la Nava D, Selvi KM, Alonso MM. Immunovirotherapy for Pediatric Solid Tumors: A Promising Treatment That is Becoming a Reality. Front Immunol 2022; 13:866892. [PMID: 35493490 PMCID: PMC9043602 DOI: 10.3389/fimmu.2022.866892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy has seen tremendous strides in the last decade, acquiring a prominent position at the forefront of cancer treatment since it has been proven to be efficacious for a wide variety of tumors. Nevertheless, while immunotherapy has changed the paradigm of adult tumor treatment, this progress has not yet been translated to the pediatric solid tumor population. For this reason, alternative curative therapies are urgently needed for the most aggressive pediatric tumors. In recent years, oncolytic virotherapy has consolidated as a feasible strategy for cancer treatment, not only for its tumor-specific effects and safety profile but also for its capacity to trigger an antitumor immune response. This review will summarize the current status of immunovirotherapy to treat cancer, focusing on pediatric solid malignancies. We will revisit previous basic, translational, and clinical research and discuss advances in overcoming the existing barriers and limitations to translate this promising therapeutic as an every-day cancer treatment for the pediatric and young adult populations.
Collapse
Affiliation(s)
- Daniel de la Nava
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
- Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Kadir Mert Selvi
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
- Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marta M. Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
- Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
13
|
Apoptosis during ZIKA Virus Infection: Too Soon or Too Late? Int J Mol Sci 2022; 23:ijms23031287. [PMID: 35163212 PMCID: PMC8835863 DOI: 10.3390/ijms23031287] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
Cell death by apoptosis is a major cellular response in the control of tissue homeostasis and as a defense mechanism in the case of cellular aggression such as an infection. Cell self-destruction is part of antiviral responses, aimed at limiting the spread of a virus. Although it may contribute to the deleterious effects in infectious pathology, apoptosis remains a key mechanism for viral clearance and the resolution of infection. The control mechanisms of cell death processes by viruses have been extensively studied. Apoptosis can be triggered by different viral determinants through different pathways as a result of virally induced cell stresses and innate immune responses. Zika virus (ZIKV) induces Zika disease in humans, which has caused severe neurological forms, birth defects, and microcephaly in newborns during the last epidemics. ZIKV also surprised by revealing an ability to persist in the genital tract and in semen, thus being sexually transmitted. Mechanisms of diverting antiviral responses such as the interferon response, the role of cytopathic effects and apoptosis in the etiology of the disease have been widely studied and debated. In this review, we examined the interplay between ZIKV infection of different cell types and apoptosis and how the virus deals with this cellular response. We illustrate a duality in the effects of ZIKV-controlled apoptosis, depending on whether it occurs too early or too late, respectively, in neuropathogenesis, or in long-term viral persistence. We further discuss a prospective role for apoptosis in ZIKV-related therapies, and the use of ZIKV as an oncolytic agent.
Collapse
|
14
|
Chen XT, Dai SY, Zhan Y, Yang R, Chen DQ, Li Y, Zhou EQ, Dong R. Progress of oncolytic virotherapy for neuroblastoma. Front Pediatr 2022; 10:1055729. [PMID: 36467495 PMCID: PMC9716318 DOI: 10.3389/fped.2022.1055729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022] Open
Abstract
As a neuroendocrine tumor derived from the neural crest, neuroblastoma (NB) is the most common extracranial solid tumor in children. The prognosis in patients with low- and intermediate-risk NB is favorable while that in high-risk patients is often detrimental. However, the management of the considerably large proportion of high-risk patients remains challenging in clinical practice. Among various new approaches, oncolytic virus (OV) therapy offers great advantages in tumor treatment, especially for high-risk NB. Genetic modified OVs can target NB specifically without affecting normal tissue and avoid the widespread drug resistance issue in anticancer monotherapy. Meanwhile, its safety profile provides great potential in combination therapy with chemo-, radio-, and immunotherapy. The therapeutic efficacy of OV for NB is impressive from bench to bedside. The effectiveness and safety of OVs have been demonstrated and reported in studies on children with NB. Furthermore, clinical trials on some OVs (Celyvir, Pexa-Vec (JX-594) and Seneca Valley Virus (NTX-010)) have reported great results. This review summarizes the latest evidence in the therapeutic application of OVs in NB, including those generated in cell lines, animal models and clinical trials.
Collapse
Affiliation(s)
- Xiao-Tong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Shu-Yang Dai
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Yong Zhan
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Ran Yang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - De-Qian Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Yi Li
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - En-Qing Zhou
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, China
| |
Collapse
|
15
|
Zwernik SD, Adams BH, Raymond DA, Warner CM, Kassam AB, Rovin RA, Akhtar P. AXL receptor is required for Zika virus strain MR-766 infection in human glioblastoma cell lines. Mol Ther Oncolytics 2021; 23:447-457. [PMID: 34901388 PMCID: PMC8626839 DOI: 10.1016/j.omto.2021.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 12/28/2022] Open
Abstract
Recent reports have shown that Zika virus (ZIKV) has oncolytic potential against human glioblastoma (GBM); however, the mechanisms underlying its tropism and cell entry are not completely understood. The receptor tyrosine kinase AXL has been identified as an entry receptor for ZIKV in a cell-type-specific manner. Interestingly, AXL is frequently overexpressed in GBM patients. Using commercially available GBM cell lines, we first show that cells expressing AXL are permissive for ZIKV infection, while cells that do not express AXL are not. Furthermore, inhibition of AXL kinase using R428 and antibody blockade of AXL receptor strongly attenuated virus entry in GBM cell lines. Additionally, CRISPR knockout of the AXL gene in GBM cell lines completely abolished ZIKV infection, significantly inhibited viral replication, and significantly reduced apoptosis compared with parental lines. Lastly, introduction of AXL receptor into non-expressing cell lines renders the cells susceptible to ZIKV infection. Together, these findings demonstrate that ZIKV entry into GBM cells in vitro is mediated by the AXL receptor and that following cell entry, productive infection is cytotoxic. Thus, ZIKV is a potential oncolytic virus for GBM.
Collapse
Affiliation(s)
- Samuel D Zwernik
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Beau H Adams
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Daniel A Raymond
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Catherine M Warner
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Amin B Kassam
- Aurora Neuroscience Innovation Institute, Advocate Aurora Health, Milwaukee, WI 53215, USA
| | - Richard A Rovin
- Aurora Neuroscience Innovation Institute, Advocate Aurora Health, Milwaukee, WI 53215, USA
| | - Parvez Akhtar
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| |
Collapse
|
16
|
Jan MW, Su HL, Chang TH, Tsai KJ. Characterization of Pathogenesis and Inflammatory Responses to Experimental Parechovirus Encephalitis. Front Immunol 2021; 12:753683. [PMID: 34899705 PMCID: PMC8654935 DOI: 10.3389/fimmu.2021.753683] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.
Collapse
Affiliation(s)
- Ming-Wei Jan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, Agriculture Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
| | - Tsung-Hsien Chang
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
17
|
Mlera L, Offerdahl DK, Dorward DW, Carmody A, Chiramel AI, Best SM, Bloom ME. The liver X receptor agonist LXR 623 restricts flavivirus replication. Emerg Microbes Infect 2021; 10:1378-1389. [PMID: 34162308 PMCID: PMC8259867 DOI: 10.1080/22221751.2021.1947749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The vector-borne flaviviruses (VBFVs) are well known for causing great misery and death in humans worldwide. The VBFVs include those transmitted by mosquitos, such as Zika virus (ZIKV), dengue virus; and those transmitted by ticks including the tick-borne flavivirus serocomplex and Powassan virus (POWV). Two of our recent reports showed that intracranial POWV infection in the reservoir host, Peromyscus leucopus, was restricted and caused no overt clinical disease. Several modes of analyses suggested activation of the LXR pathway. Activation of the LXR pathway leads to increased efflux of cholesterol from cells and consequent disturbances in membrane biogenesis. Because VBFV replication is dependent on membrane biogenesis, we evaluated the effect of an LXR agonist (LXR623) on POWV and ZIKV infection and observed that the compound impaired permissive replication of both viruses in a human neuroblastoma SK-N-SH cell line. The LXR agonist resulted in failure of the viruses to induce ER expansion and elaborate vesicle formation, suggesting that the efflux of cholesterol was part of the antiviral mechanism. We also observed that the LXR agonist contributed to the mechanism of virus suppression by increased expression of mRNAs encoding for the antiviral cytokines CXCL10, RANTES and IFN1β. In sharp contrast, a LXR antagonist (GSK2033) had no significant effect on VBFV replication. We conclude that LXR623 impairs flavivirus replication by stimulating cellular antiviral factors.
Collapse
Affiliation(s)
- Luwanika Mlera
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - Danielle K Offerdahl
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - David W Dorward
- Microscopy Unit, Research Technologies Branch, NIAID/NIH, Hamilton, MT, USA
| | - Aaron Carmody
- Research Technologies Branch, NIAID/NIH, Hamilton, MT, USA
| | - Abhilash I Chiramel
- Innate Immunity and Pathogenesis Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - Sonja M Best
- Innate Immunity and Pathogenesis Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - Marshall E Bloom
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| |
Collapse
|
18
|
Mallepalli S, Gupta MK, Vadde R. Neuroblastoma: An Updated Review on Biology and Treatment. Curr Drug Metab 2020; 20:1014-1022. [PMID: 31878853 DOI: 10.2174/1389200221666191226102231] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neuroblastoma (NB) is the second leading extracranial solid tumors of early childhood and clinically characterized by the presence of round, small, monomorphic cells with excess nuclear pigmentation (hyperchromasia).Owing to a lack of definitive treatment against NB and less survival rate in high-risk patients, there is an urgent requirement to understand molecular mechanisms associated with NB in a better way, which in turn can be utilized for developing drugs towards the treatment of NB in human. OBJECTIVES In this review, an approach was adopted to understand major risk factors, pathophysiology, the molecular mechanism associated with NB, and various therapeutic agents that can serve as drugs towards the treatment of NB in humans. CONCLUSION Numerous genetic (e.g., MYCN amplification), perinatal, and gestational factors are responsible for developing NB. However, no definite environmental or parental exposures responsible for causing NB have been confirmed to date. Though intensive multimodal treatment approaches, namely, chemotherapy, surgery & radiation, may help in improving the survival rate in children, these approaches have several side effects and do not work efficiently in high-risk patients. However, recent studies suggested that numerous phytochemicals, namely, vincristine, and matrine have a minimal side effect in the human body and may serve as a therapeutic drug during the treatment of NB. Most of these phytochemicals work in a dose-dependent manner and hence must be prescribed very cautiously. The information discussed in the present review will be useful in the drug discovery process as well as treatment and prevention on NB in humans.
Collapse
Affiliation(s)
- Suresh Mallepalli
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa-516003, A.P., India
| | - Manoj Kumar Gupta
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa-516003, A.P., India
| | - Ramakrishna Vadde
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa-516003, A.P., India
| |
Collapse
|
19
|
The Propagation and Quantification of Two Emerging Oncolytic Viruses: Vesicular Stomatitis (VSV) and Zika (ZIKV). Methods Mol Biol 2019. [PMID: 31776931 DOI: 10.1007/978-1-0716-0203-4_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Abstract
Developments in genetic engineering have allowed researchers and clinicians to begin harnessing viruses to target and kill cancer cells, either through direct lysis or through recruitment of antiviral immune responses. Two powerful viruses in the fight against cancer are the single-stranded RNA viruses vesicular stomatitis virus and Zika virus. Here, we describe methods to propagate and titer these two viruses. We also describe a simple cell-killing assay to begin testing modified viruses for increased potential killing of glioblastoma cells.
Collapse
|
20
|
Ye W, Liang F, Ying C, Zhang M, Feng D, Jiang X. Downregulation of microRNA-3934-5p induces apoptosis and inhibits the proliferation of neuroblastoma cells by targeting TP53INP1. Exp Ther Med 2019; 18:3729-3736. [PMID: 31616506 PMCID: PMC6781830 DOI: 10.3892/etm.2019.8007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 03/21/2019] [Indexed: 12/30/2022] Open
Abstract
Neuroblastoma is the most common pediatric extracranial solid tumour in the world. miRNAs are a group of endogenous small non-coding RNAs that act on target genes to serve critical roles in many biological processes. Presently, the expression and role of miR-3934-5p in neuroblastoma remains unclear. Therefore, the aim of the present study was to investigate the expression of miR-3934-5p in neuroblastoma tissues and cell lines and to assess the role of miR-3934-5p in neuroblastoma. In the current study, the results revealed that miR-3934-5p was significantly upregulated in neuroblastoma tissues and cell lines. The data also identified TP53INP1 as a direct target gene of miR-3934-5p, which was negatively regulated by miR-3934-5p. The present study further demonstrated that TP53INP1 was downregulated in both neuroblastoma tissues and cell lines. Furthermore, the results of the current study indicate that miR-3934-5p downregulation may induce apoptosis and inhibit neuroblastoma cell viability. However, these effects were reversed via TP53INP1-siRNA. Data from the current study indicates that the miR-3934-5p/TP53INP1 axis may be a novel therapeutic target for neuroblastoma treatment.
Collapse
Affiliation(s)
- Wei Ye
- Department of Neurology, Jianou Municipal Hospital, Jianou, Fujian 353100, P.R. China
| | - Fulv Liang
- Department of Urology, The Third Hospital of Xiamen, Xiamen, Fujian 361000, P.R. China
| | - Chen Ying
- Department of Urology, Haicang Hospital of Xiamen, Xiamen, Fujian 361026, P.R. China
| | - Maolin Zhang
- Department of Surgery, Xiapu County Hospital, Xiapu County, Ningde, Fujian 355100, P.R. China
| | - Dongbo Feng
- Department of Sports Medicine, The Central Hospital of Yongzhou, Yongzhou, Hunan 425000, P.R. China
| | - Xinyu Jiang
- Department of General Surgery, Xiamen Maternity and Child Health Care Hospital, Xiamen, Fujian 361000, P.R. China
| |
Collapse
|
21
|
Fox CR, Parks GD. Histone Deacetylase Inhibitors Enhance Cell Killing and Block Interferon-Beta Synthesis Elicited by Infection with an Oncolytic Parainfluenza Virus. Viruses 2019; 11:E431. [PMID: 31083335 PMCID: PMC6563284 DOI: 10.3390/v11050431] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/28/2022] Open
Abstract
Previous results have shown that infection with the cytoplasmic-replicating parainfluenza virus 5 mutant P/V-CPI- sensitizes cells to DNA damaging agents, resulting in the enhanced killing of airway cancer cells. Here, we have tested the hypothesis that histone deacetylase (HDAC) inhibitors can also act with P/V-CPI- infection to enhance cancer cell killing. Using human small cell lung cancer and laryngeal cancer cell lines, 10 HDAC inhibitors were tested for their effect on viability of P/V-CPI- infected cells. HDAC inhibitors such as scriptaid enhanced caspase-3/7, -8 and -9 activity induced by P/V-CPI- and overall cell toxicity. Scriptaid-mediated enhanced killing was eliminated in lung cancer cells that were engineered to express a protein which sequesters double stranded RNA. Scriptaid also enhanced cancer cell killing by two other negative strand RNA viruses - the La Crosse virus and vesicular stomatitis virus. Scriptaid treatment enhanced the spread of the P/V-CPI- virus through a population of cancer cells, and suppressed interferon-beta induction through blocking phosphorylation and nuclear translocation of Interferon Regulatory Factor 3 (IRF-3). Taken together, these data support a role for combinations of a cytoplasmic-replicating RNA virus such as the P/V-CPI- mutant along with chemotherapeutic agents.
Collapse
Affiliation(s)
- Candace R Fox
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| | - Griffith D Parks
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| |
Collapse
|
22
|
Bernatchez JA, Coste M, Beck S, Wells GA, Luna LA, Clark AE, Zhu Z, Hecht D, Rich JN, Sohl CD, Purse BW, Siqueira-Neto JL. Activity of Selected Nucleoside Analogue ProTides against Zika Virus in Human Neural Stem Cells. Viruses 2019; 11:v11040365. [PMID: 31010044 PMCID: PMC6521205 DOI: 10.3390/v11040365] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 01/27/2023] Open
Abstract
Zika virus (ZIKV), an emerging flavivirus that causes neurodevelopmental impairment to fetuses and has been linked to Guillain-Barré syndrome continues to threaten global health due to the absence of targeted prophylaxis or treatment. Nucleoside analogues are good examples of efficient anti-viral inhibitors, and prodrug strategies using phosphate masking groups (ProTides) have been employed to improve the bioavailability of ribonucleoside analogues. Here, we synthesized and tested a small library of 13 ProTides against ZIKV in human neural stem cells. Strong activity was observed for 2′-C-methyluridine and 2′-C-ethynyluridine ProTides with an aryloxyl phosphoramidate masking group. Substitution of a 2-(methylthio) ethyl phosphoramidate for the aryloxyl phosphoramidate ProTide group of 2′-C-methyluridine completely abolished antiviral activity of the compound. The aryloxyl phosphoramidate ProTide of 2′-C-methyluridine outperformed the hepatitis C virus (HCV) drug sofosbuvir in suppression of viral titers and protection from cytopathic effect, while the former compound’s triphosphate active metabolite was better incorporated by purified ZIKV NS5 polymerase over time. These findings suggest both a nucleobase and ProTide group bias for the anti-ZIKV activity of nucleoside analogue ProTides in a disease-relevant cell model.
Collapse
Affiliation(s)
- Jean A Bernatchez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Michael Coste
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA 92182, USA.
| | - Sungjun Beck
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Grace A Wells
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA 92182, USA.
| | - Lucas A Luna
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA 92182, USA.
| | - Alex E Clark
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Zhe Zhu
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, USA.
- Department of Medicine, Division of Regenerative Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - David Hecht
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA 92182, USA.
- Department of Chemistry, Southwestern College, Chula Vista, CA 91910, USA.
| | - Jeremy N Rich
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, USA.
- Department of Medicine, Division of Regenerative Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Christal D Sohl
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA 92182, USA.
| | - Byron W Purse
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA 92182, USA.
- The Viral Information Institute, San Diego State University, San Diego, CA 92182, USA.
| | - Jair L Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
23
|
Svyatchenko VA, Razumov IA, Protopopova EV, Demina AV, Solovieva OI, Zavjalov EL, Loktev VB. Zika virus has an oncolytic activity against human glioblastoma U87 cells. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj18.448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma is a highly lethal brain cancer. Virotherapy with the use of oncolytic viruses has since recently been regarded as a promising approach for the clinic treatment of human glioblastomas. The purpose of this work was to perform a primary evaluation of the Zika virus as a potential oncolytic agent against glioblastomas.In vitroexperiments showed that the Zika virus strain MR 766 is able to selectively infect and lyse neoplastic cells of the human glioblastoma cell line U87 MG. The selectivity index (SI, the ratio of infectious titer for tumor cells to titer on normal untransformed cells) was 2·102. The selectivity of the replicative activity of Zika virus in relation to U87 MG glioblastoma cells was additionally confrmed by indirect immunofluorescence. Using the model of immunodefcient SCID mice with subcutaneous xenografts of human glioblastoma U87 MG, a strong antitumor activity of the Zika virus under a course (daily for 4 days) of intratumoral administration of 5·105 TCID50 of Zika virus was shown. Treatment with Zika virus resulted in more than a 10fold reduction in mean volumes of tumors. The tumor growth inhibition index was 92.63 %. Recurrences (metastases) of tumor regrowth were not registered within 64 days of observation. This result demonstrated the prospect of further indepth studies of the Zika virus as a potential oncolytic agent against human glioblastomas.
Collapse
Affiliation(s)
| | - I. A. Razumov
- State Research Center of Virology and Biotechnology “Vector”; Institute of Cytology and Genetics, SB RAS
| | | | - A. V. Demina
- State Research Center of Virology and Biotechnology “Vector”
| | | | | | - V. B. Loktev
- State Research Center of Virology and Biotechnology “Vector”; Institute of Cytology and Genetics, SB RAS
| |
Collapse
|