1
|
Chen Z, Jiang Y, Cui J, Li W, Han W, Liu G. Elucidating the Mechanism of VVTT Infection Through Machine Learning and Transcriptome Analysis. Int J Mol Sci 2025; 26:1203. [PMID: 39940969 PMCID: PMC11818747 DOI: 10.3390/ijms26031203] [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: 12/17/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
The vaccinia virus (VV) is extensively utilized as a vaccine vector in the treatment of various infectious diseases, cardiovascular diseases, immunodeficiencies, and cancers. The vaccinia virus Tiantan strain (VVTT) has been instrumental as an irreplaceable vaccine strain in the eradication of smallpox in China; however, it still presents significant adverse toxic effects. After the WHO recommended that routine smallpox vaccination be discontinued, the Chinese government stopped the national smallpox vaccination program in 1981. The outbreak of monkeypox in 2022 has focused people's attention on the Orthopoxvirus. However, there are limited reports on the safety and toxic side effects of VVTT. In this study, we employed a combination of transcriptomic analysis and machine learning-based feature selection to identify key genes implicated in the VVTT infection process. We utilized four machine learning algorithms, including random forest (RF), minimum redundancy maximum relevance (MRMR), eXtreme Gradient Boosting (XGB), and least absolute shrinkage and selection operator cross-validation (LASSOCV), for feature selection. Among these, XGB was found to be the most effective and was used for further screening, resulting in an optimal model with an ROC curve of 0.98. Our analysis revealed the involvement of pathways such as spinocerebellar ataxia and the p53 signaling pathway. Additionally, we identified three critical targets during VVTT infection-ARC, JUNB, and EGR2-and further validated these targets using qPCR. Our research elucidates the mechanism by which VVTT infects cells, enhancing our understanding of the smallpox vaccine. This knowledge not only facilitates the development of new and more effective vaccines but also contributes to a deeper comprehension of viral pathogenesis. By advancing our understanding of the molecular mechanisms underlying VVTT infection, this study lays the foundation for the further development of VVTT. Such insights are crucial for strengthening global health security and ensuring a resilient response to future pandemics.
Collapse
Affiliation(s)
- Zhili Chen
- Academy of Military Medical Sciences, Beijing 100850, China
| | - Yongxin Jiang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China (W.H.)
| | - Jiazhen Cui
- Academy of Military Medical Sciences, Beijing 100850, China
| | - Wannan Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China (W.H.)
| | - Weiwei Han
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China (W.H.)
| | - Gang Liu
- Academy of Military Medical Sciences, Beijing 100850, China
| |
Collapse
|
2
|
Li Z, Liu R. Construction and Isolation of Recombinant Vaccinia Virus by Homologous Recombination Using Fluorescent Protein Markers. Methods Mol Biol 2025; 2860:83-95. [PMID: 39621262 DOI: 10.1007/978-1-0716-4160-6_6] [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] [Indexed: 03/26/2025]
Abstract
Genetic modification of vaccinia virus (VACV) is a fundamental and valuable research technique in elucidating the function of VACV genes, as well as the development as vaccine vectors for other infectious diseases, oncolytic therapeutics for cancers, and protein expression systems in mammalian cells. Because of the large size of poxvirus genome and noninfectious feature of the naked viral DNA, construction of recombinant VACV relies on intracellular homologous recombination between transfected DNA and replicating viral DNA in infected cells occurred in VACV infected cells. The efficiency of homologous recombination event for vaccinia virus is relatively low, and recombinant viruses only account for 0.1% of progeny viruses. Therefore, fluorescent protein markers are often included in the transfected DNA to facilitate the selection and screening of recombined viruses. Here we provide a detailed procedure for the design, generation, isolation, and detection of recombinant VACV by homologous recombination using fluorescent protein markers.
Collapse
Affiliation(s)
- Zhichao Li
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ruikang Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China.
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
3
|
Han MM, Fan YK, Zhang Y, Dong ZQ. Advances in herbal polysaccharides-based nano-drug delivery systems for cancer immunotherapy. J Drug Target 2024; 32:311-324. [PMID: 38269853 DOI: 10.1080/1061186x.2024.2309661] [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: 10/13/2023] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
The boom in cancer immunotherapy has provided many patients with a better chance of survival, but opportunities often come with challenges. Single immunotherapy is not good enough to eradicate tumours, and often fails to achieve the desired therapeutic effect because of the low targeting of immunotherapy drugs, and causes more side effects. As a solution to this problem, researchers have developed several nano Drug Delivery Systems (NDDS) to deliver immunotherapeutic agents to achieve good therapeutic outcomes. However, traditional drug delivery systems (DDS) have disadvantages such as poor bioavailability, high cytotoxicity, and difficulty in synthesis, etc. Herbal Polysaccharides (HPS), derived from natural Chinese herbs, inherently possess low toxicity. Furthermore, the biocompatibility, biodegradability, hydrophilicity, ease of modification, and immunomodulatory activities of HPS offer unique advantages in substituting traditional DDS. This review initially addresses the current developments and challenges in immunotherapy. Subsequently, it focuses on the immunomodulatory mechanisms of HPS and their design as nanomedicines for targeted drug delivery in tumour immunotherapy. Our findings reveal that HPS-based nanomedicines exhibit significant potential in enhancing the efficacy of cancer immunotherapy, providing crucial theoretical foundations and practical guidelines for future clinical applications.
Collapse
Affiliation(s)
- Miao-Miao Han
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yi-Kai Fan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| | - Zheng-Qi Dong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| |
Collapse
|
4
|
Ren S, Zhang Y, Gao X, Wang X, Tong L, Wang S, Sun Y, Yin X, Chen H. Platform establishment of the Cre-loxP recombination system for genetic manipulation of the Lumpy skin disease virus. Vet Microbiol 2024; 294:110122. [PMID: 38772074 DOI: 10.1016/j.vetmic.2024.110122] [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: 01/30/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024]
Abstract
Lumpy skin disease virus (LSDV) is a rapidly emerging pathogen in Asia, including China. Genetic manipulation of the LSDV is essential for the elucidation of the pathogenic mechanism and biological function of the LSDV-encoded protein. In this study, we established a platform for the Cre-loxP recombination system under a modified early-late H5 promoter of the VACV for quick construction of the recombinant LSDV virus. The recombinant virus, LSDV-EGFP-ΔTK, was purified and obtained using serial limited dilution and picking the single cells methods. Using the lentiviral package system, a Cre recombinase enzyme stable expression MDBK cell line was established to supply the Cre recombinase for the reporter gene excision. A genetically stable, safe TK gene-deleted LSDV (LSDV-ΔTK) was constructed using homologous recombination and the Cre-loxP system. It was purified using limited dilution in the MDBK-Cre cell line. Establishing the Cre-loxP recombination system will enable sequential deletion of the interested genes from the LSDV genome and genetic manipulation of the LSDV genome, providing technical support and a platform for developing the attenuated LSDV vaccine.
Collapse
Affiliation(s)
- Shanhui Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Yuzhe Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Xiaolong Gao
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai 810016, PR China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Lina Tong
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai 810016, PR China
| | - Shasha Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China.
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China.
| | - Haotai Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China.
| |
Collapse
|
5
|
Agyemang E, Gonneville AN, Tiruvadi-Krishnan S, Lamichhane R. Exploring GPCR conformational dynamics using single-molecule fluorescence. Methods 2024; 226:35-48. [PMID: 38604413 PMCID: PMC11098685 DOI: 10.1016/j.ymeth.2024.03.011] [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: 12/06/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that transmit specific external stimuli into cells by changing their conformation. This conformational change allows them to couple and activate G-proteins to initiate signal transduction. A critical challenge in studying and inferring these structural dynamics arises from the complexity of the cellular environment, including the presence of various endogenous factors. Due to the recent advances in cell-expression systems, membrane-protein purification techniques, and labeling approaches, it is now possible to study the structural dynamics of GPCRs at a single-molecule level both in vitro and in live cells. In this review, we discuss state-of-the-art techniques and strategies for expressing, purifying, and labeling GPCRs in the context of single-molecule research. We also highlight four recent studies that demonstrate the applications of single-molecule microscopy in revealing the dynamics of GPCRs. These techniques are also useful as complementary methods to verify the results obtained from other structural biology tools like cryo-electron microscopy and x-ray crystallography.
Collapse
Affiliation(s)
- Eugene Agyemang
- UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN 37996, USA
| | - Alyssa N Gonneville
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Sriram Tiruvadi-Krishnan
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Rajan Lamichhane
- UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN 37996, USA; Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA.
| |
Collapse
|
6
|
Mirbahari SN, Da Silva M, Zúñiga AIM, Kooshki Zamani N, St-Laurent G, Totonchi M, Azad T. Recent progress in combination therapy of oncolytic vaccinia virus. Front Immunol 2024; 15:1272351. [PMID: 38558795 PMCID: PMC10979700 DOI: 10.3389/fimmu.2024.1272351] [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: 08/03/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
In recent years, oncolytic viruses have emerged as promising agents for treating various cancers. An oncolytic virus is a non-pathogenic virus that, due to genetic manipulation, tends to replicate in and cause lysis of cancerous cells while leaving healthy cells unaffected. Among these viruses, vaccinia virus is an attractive platform for use as an oncolytic platform due to its 190 Kb genome with a high capacity for encoding therapeutic payloads. Combining oncolytic VV therapy with other conventional cancer treatments has been shown to be synergistic and more effective than monotherapies. Additionally, OVV can be used as a vector to deliver therapeutic payloads, alone or in combination with other treatments, to increase overall efficacy. Here, we present a comprehensive analysis of preclinical and clinical studies that have evaluated the efficacy of oncolytic vaccinia viruses in cancer immunotherapy. We discuss the outcomes of these studies, including tumor regression rates, overall survival benefits, and long-term responses. Moreover, we provide insights into the challenges and limitations associated with oncolytic vaccinia virus- based therapies, including immune evasion mechanisms, potential toxicities, and the development of resistance.
Collapse
Affiliation(s)
- Seyedeh Nasim Mirbahari
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Miles Da Silva
- Department of Microbiology and Immunology, University of British Colombia, Vancouver, BC, Canada
- Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| | - Abril Ixchel Muñoz Zúñiga
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Nika Kooshki Zamani
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Gabriel St-Laurent
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Taha Azad
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| |
Collapse
|
7
|
Aggarwal T, Kondabagil K. Assembly and Evolution of Poxviruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:35-54. [PMID: 38801570 DOI: 10.1007/978-3-031-57165-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Poxvirus assembly has been an intriguing area of research for several decades. While advancements in experimental techniques continue to yield fresh insights, many questions are still unresolved. Large genome sizes of up to 380 kbp, asymmetrical structure, an exterior lipid bilayer, and a cytoplasmic life cycle are some notable characteristics of these viruses. Inside the particle are two lateral bodies and a protein wall-bound-biconcave core containing the viral nucleocapsid. The assembly progresses through five major stages-endoplasmic reticulum (ER) membrane alteration and rupture, crescent formation, immature virion formation, genome encapsidation, virion maturation and in a subset of viruses, additional envelopment of the virion prior to its dissemination. Several large dsDNA viruses have been shown to follow a comparable sequence of events. In this chapter, we recapitulate our understanding of the poxvirus morphogenesis process while reviewing the most recent advances in the field. We also briefly discuss how virion assembly aids in our knowledge of the evolutionary links between poxviruses and other Nucleocytoplasmic Large DNA Viruses (NCLDVs).
Collapse
Affiliation(s)
- Tanvi Aggarwal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India
| | - Kiran Kondabagil
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India.
| |
Collapse
|
8
|
Dong X, Wu W, Pan P, Zhang XZ. Engineered Living Materials for Advanced Diseases Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2304963. [PMID: 37436776 DOI: 10.1002/adma.202304963] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/13/2023]
Abstract
Natural living materials serving as biotherapeutics exhibit great potential for treating various diseases owing to their immunoactivity, tissue targeting, and other biological activities. In this review, the recent developments in engineered living materials, including mammalian cells, bacteria, viruses, fungi, microalgae, plants, and their active derivatives that are used for treating various diseases are summarized. Further, the future perspectives and challenges of such engineered living material-based biotherapeutics are discussed to provide considerations for future advances in biomedical applications.
Collapse
Affiliation(s)
- Xue Dong
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Wei Wu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Pei Pan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
9
|
Yang N, Wang Y, Dai P, Li T, Zierhut C, Tan A, Zhang T, Xiang JZ, Ordureau A, Funabiki H, Chen Z, Deng L. Vaccinia E5 is a major inhibitor of the DNA sensor cGAS. Nat Commun 2023; 14:2898. [PMID: 37217469 PMCID: PMC10201048 DOI: 10.1038/s41467-023-38514-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
The DNA sensor cyclic GMP-AMP synthase (cGAS) is critical in host antiviral immunity. Vaccinia virus (VACV) is a large cytoplasmic DNA virus that belongs to the poxvirus family. How vaccinia virus antagonizes the cGAS-mediated cytosolic DNA-sensing pathway is not well understood. In this study, we screened 80 vaccinia genes to identify potential viral inhibitors of the cGAS/Stimulator of interferon gene (STING) pathway. We discovered that vaccinia E5 is a virulence factor and a major inhibitor of cGAS. E5 is responsible for abolishing cGAMP production during vaccinia virus (Western Reserve strain) infection of dendritic cells. E5 localizes to the cytoplasm and nucleus of infected cells. Cytosolic E5 triggers ubiquitination of cGAS and proteasome-dependent degradation via interacting with cGAS. Deleting the E5R gene from the Modified vaccinia virus Ankara (MVA) genome strongly induces type I IFN production by dendritic cells (DCs) and promotes DC maturation, and thereby improves antigen-specific T cell responses.
Collapse
Affiliation(s)
- Ning Yang
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Yi Wang
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Peihong Dai
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tuo Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Christian Zierhut
- Laboratory of Chromosome and Cell Biology, The Rockefeller University, New York, NY, 10065, USA
- The Institute of Cancer Research, London, SW3 6JB, UK
| | - Adrian Tan
- Genomic Resources Core Facility, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Tuo Zhang
- Genomic Resources Core Facility, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Jenny Zhaoying Xiang
- Genomic Resources Core Facility, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hironori Funabiki
- Laboratory of Chromosome and Cell Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Zhijian Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Liang Deng
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
10
|
Yu X, Shi H, Cheng G. Mpox Virus: Its Molecular Evolution and Potential Impact on Viral Epidemiology. Viruses 2023; 15:v15040995. [PMID: 37112975 PMCID: PMC10142743 DOI: 10.3390/v15040995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Mpox (previously known as monkeypox) is an infectious viral illness caused by the mpox virus (MPXV), an orthopoxvirus that belongs to the family Poxviridae. The symptoms of mpox in humans are similar to those of smallpox, although the mortality rate is lower. In recent years, the concern over a potential global pandemic has increased due to reports of mpox spreading across Africa and other parts of the world. Prior to this discovery, mpox was a rare zoonotic disease restricted to endemic regions of Western and Central Africa. The sudden emergence of MPXV cases in multiple regions has raised concerns about its natural evolution. This review aims to provide an overview of previously available information about MPXV, including its genome, morphology, hosts and reservoirs, and virus-host interaction and immunology, as well as to perform phylogenetic analysis on available MPXV genomes, with an emphasis on the evolution of the genome in humans as new cases emerge.
Collapse
Affiliation(s)
- Xi Yu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Huicheng Shi
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| |
Collapse
|
11
|
Wang W, An X, Yan K, Li Q. Construction and Application of Orthogonal T7 Expression System in Eukaryote: An Overview. Adv Biol (Weinh) 2023; 7:e2200218. [PMID: 36464626 DOI: 10.1002/adbi.202200218] [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: 08/06/2022] [Revised: 10/17/2022] [Indexed: 12/12/2022]
Abstract
The T7 system is an orthogonal transcription-system, which is characterized by simplicity, higher efficiency, and higher processivity, and it is used for protein or mRNA synthesis in various biological-systems. In comparison with prokaryotes, the construction of the T7 expression system is still on-going in eukaryotes, but it shows greatly applicable prospects. In the present paper, development of T7 expression system construction in eukaryotes is reviewed, including its construction in animal (mammalian cells, trypanosomatid protozoa, Xenopus oocytes, zebrafish), plant, and microorganism and its application in vaccine production and gene therapy. In addition, the innate challenges of T7 expression system construction in eukaryote and its potential application in vaccine production and gene therapy are discussed.
Collapse
Affiliation(s)
- Wenya Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xiaoyan An
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Kun Yan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Qiang Li
- Key Laboratory for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
12
|
Smith ES, Balch LA, Scrivens M, Shi S, Wang W, Harvey CD, Cornelison AA, Gil-Moore M, Kirk RA, Mueller LL, Hall RL, Howell AP, Reilly CA, Mayer JM, Murante FG, Viggiani KA, Gersz EM, Bussler H, Keefe MR, Evans EE, Paris MJ, Zauderer M. Use of poxvirus display to select antibodies specific for complex membrane antigens. MAbs 2023; 15:2249947. [PMID: 37635331 PMCID: PMC10464538 DOI: 10.1080/19420862.2023.2249947] [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: 02/15/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023] Open
Abstract
Antibody discovery against complex antigens is limited by the availability of a reproducible pure source of concentrated properly folded antigen. We have developed a technology to enable direct incorporation of membrane proteins such as GPCRs and into the membrane of poxvirus. The protein of interest is correctly folded and expressed in the cell-derived viral membrane and does not require any detergents or refolding before downstream use. The poxvirus is selective in which proteins are incorporated into the viral membrane, making the antigen poxvirus an antigenically cleaner target for in vitro panning. Antigen-expressing virus can be readily purified at scale and used for antibody selection using any in vitro display platform.
Collapse
Affiliation(s)
| | | | | | | | - Wei Wang
- Research, Vaccinex, Inc, Rochester, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Sah R, Mohanty A, Hada V, Singh P, Govindaswamy A, Siddiq A, Reda A, Dhama K. The Emergence of Monkeypox: A Global Health Threat. Cureus 2022; 14:e29304. [DOI: 10.7759/cureus.29304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/05/2022] Open
|
14
|
Mun SJ, Cho E, Kim JS, Yang CS. Pathogen-derived peptides in drug targeting and its therapeutic approach. J Control Release 2022; 350:716-733. [PMID: 36030988 DOI: 10.1016/j.jconrel.2022.08.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 02/06/2023]
Abstract
Peptides, short stretches of amino acids or small proteins that occupy a strategic position between proteins and amino acids, are readily accessible by chemical and biological methods. With ideal properties for forming high-affinity and specific interactions with host target proteins, they have established an important niche in the drug development spectrum complementing small molecule and biological therapeutics. Among the most successful biomedicines in use today, peptide-based drugs show great promise. This, coupled with recent advances in synthetic and nanochemical biology, has led to the creation of tailor-made peptide therapeutics for improved biocompatibility. This review presents an overview of the latest research on pathogen-derived, host-cell-interacting peptides. It also highlights strategies for using peptide-based therapeutics that address cellular transport challenges through the introduction of nanoparticles that serve as platforms to facilitate the delivery of peptide biologics and therapeutics for treating various inflammatory diseases. Finally, this paper describes future perspectives, specific pathogen-based peptides that can enhance specificity, efficiency, and capacity in functional peptide-based therapeutics, which are in the spotlight as new treatment alternatives for various diseases, and also presents verified sequences and targets that can increase chemical and pharmacological value.
Collapse
Affiliation(s)
- Seok-Jun Mun
- Department of Bionano Technology, Hanyang University, Seoul 04673, Republic of Korea; Center for Bionano Intelligence Education and Research, Ansan 15588, Republic of Korea
| | - Euni Cho
- Department of Bionano Technology, Hanyang University, Seoul 04673, Republic of Korea; Center for Bionano Intelligence Education and Research, Ansan 15588, Republic of Korea
| | - Jae-Sung Kim
- Department of Bionano Technology, Hanyang University, Seoul 04673, Republic of Korea; Institute of Natural Science & Technology, Hanyang University, Ansan 15588, Republic of Korea
| | - Chul-Su Yang
- Center for Bionano Intelligence Education and Research, Ansan 15588, Republic of Korea; Department of Molecular and Life Science, Hanyang University, Ansan 15588, Republic of Korea.
| |
Collapse
|
15
|
Schnierle BS. Monkeypox Goes North: Ongoing Worldwide Monkeypox Infections in Humans. Viruses 2022; 14:1874. [PMID: 36146681 PMCID: PMC9503176 DOI: 10.3390/v14091874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
In the late 1970s, global vaccination programs resulted in the eradication of smallpox. The Monkeypox virus (MPXV), which is closely related to the smallpox-inducing variola virus, was previously endemic only in Sub-Saharan Africa but is currently spreading worldwide. Only older people who have been vaccinated against smallpox are expected to be sufficiently protected against poxviruses. Here I will summarize current knowledge about the virus, the disease caused by MPXV infections, and strategies to limit its spread.
Collapse
Affiliation(s)
- Barbara S Schnierle
- Section AIDS and Newly Emerging Pathogens, Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany
| |
Collapse
|
16
|
Mahmood F, Xu R, Awan MUN, Song Y, Han Q, Xia X, Zhang J. PDIA3: Structure, functions and its potential role in viral infections. Biomed Pharmacother 2021; 143:112110. [PMID: 34474345 DOI: 10.1016/j.biopha.2021.112110] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
The catalysis of disulphide (SS) bonds is the most important characteristic of protein disulphide isomerase (PDI) family. Catalysis occurs in the endoplasmic reticulum, which contains many proteins, most of which are secretory in nature and that have at least one s-s bond. Protein disulphide isomerase A3 (PDIA3) is a member of the PDI family that acts as a chaperone. PDIA3 is highly expressed in response to cellular stress, and also intercept the apoptotic cellular death related to endoplasmic reticulum (ER) stress, and protein misfolding. PDIA3 expression is elevated in almost 70% of cancers and its expression has been linked with overall low cell invasiveness, survival and metastasis. Viral diseases present a significant public health threat. The presence of PDIA3 on the cell surface helps different viruses to enter the cells and also helps in replication. Therefore, inhibitors of PDIA3 have great potential to interfere with viral infections. In this review, we summarize what is known about the basic structure, functions and role of PDIA3 in viral infections. The review will inspire studies of pathogenic mechanisms and drug targeting to counter viral diseases.
Collapse
Affiliation(s)
- Faisal Mahmood
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Ruixian Xu
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Maher Un Nisa Awan
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Yuzhu Song
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Qinqin Han
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Xueshan Xia
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China.
| | - Jinyang Zhang
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China.
| |
Collapse
|
17
|
Amundsen MM, Tartor H, Andersen K, Sveinsson K, Thoen E, Gjessing MC, Dahle MK. Mucosal and Systemic Immune Responses to Salmon Gill Poxvirus Infection in Atlantic Salmon Are Modulated Upon Hydrocortisone Injection. Front Immunol 2021; 12:689302. [PMID: 34177946 PMCID: PMC8221106 DOI: 10.3389/fimmu.2021.689302] [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: 03/31/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
Salmon Gill Poxvirus Disease (SGPVD) has emerged as a cause of acute mortality in Atlantic salmon (Salmo salar L.) presmolts in Norwegian aquaculture. The clinical phase of the disease is associated with apoptotic cell death in the gill epithelium causing acute respiratory distress, followed by proliferative changes in the regenerating gill in the period after the disease outbreak. In an experimental SGPV challenge trial published in 2020, acute disease was only seen in fish injected with hydrocortisone 24 h prior to infection. SGPV-mediated mortality in the hydrocortisone-injected group was associated with more extensive gill pathology and higher SGPV levels compared to the group infected with SGPV only. In this study based on the same trial, SGPV gene expression and the innate and adaptive antiviral immune response was monitored in gills and spleen in the presence and absence of hydrocortisone. Whereas most SGPV genes were induced from day 3 along with the interferon-regulated innate immune response in gills, the putative SGPV virulence genes of the B22R family were expressed already one day after SGPV exposure, indicating a potential role as early markers of SGPV infection. In gills of the hydrocortisone-injected fish infected with SGPV, MX expression was delayed until day 10, and then expression skyrocketed along with the viral peak, gill pathology and mortality occurring from day 14. A similar expression pattern was observed for Interferon gamma (IFNγ) and granzyme A (GzmA) in the gills, indicating a role of acute cytotoxic cell activity in SGPVD. Duplex in situ hybridization demonstrated effects of hydrocortisone on the number and localization of GzmA-containing cells, and colocalization with SGPV infected cells in the gill. SGPV was generally not detected in spleen, and gill infection did not induce any corresponding systemic immune activity in the absence of stress hormone injection. However, in fish injected with hydrocortisone, IFNγ and GzmA gene expression was induced in spleen in the days prior to acute mortality. These data indicate that suppressed mucosal immune response in the gills and the late triggered systemic immune response in the spleen following hormonal stress induction may be the key to the onset of clinical SGPVD.
Collapse
Affiliation(s)
- Marit M Amundsen
- Department of Fish Health, Norwegian Veterinary Institute, Ås, Norway
| | - Haitham Tartor
- Department of Fish Health, Norwegian Veterinary Institute, Ås, Norway
| | - Kathrine Andersen
- Department of Fish Health, Norwegian Veterinary Institute, Ås, Norway
| | | | - Even Thoen
- Department of Fish Health, Norwegian Veterinary Institute, Ås, Norway.,Patogen, Ålesund, Norway
| | - Mona C Gjessing
- Department of Fish Health, Norwegian Veterinary Institute, Ås, Norway
| | - Maria K Dahle
- Department of Fish Health, Norwegian Veterinary Institute, Ås, Norway.,The Norwegian College of Fishery Science, UiT - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
18
|
Aggio JB, Krmeská V, Ferguson BJ, Wowk PF, Rothfuchs AG. Vaccinia Virus Infection Inhibits Skin Dendritic Cell Migration to the Draining Lymph Node. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:776-784. [PMID: 33419767 PMCID: PMC7851745 DOI: 10.4049/jimmunol.2000928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
There is a paucity of information on dendritic cell (DC) responses to vaccinia virus (VACV), including the traffic of DCs to the draining lymph node (dLN). In this study, using a mouse model of infection, we studied skin DC migration in response to VACV and compared it with the tuberculosis vaccine Mycobacterium bovis bacille Calmette-Guérin (BCG), another live attenuated vaccine administered via the skin. In stark contrast to BCG, skin DCs did not relocate to the dLN in response to VACV. Infection with UV-inactivated VACV or modified VACV Ankara promoted DC movement to the dLN, indicating that interference with skin DC migration requires replication-competent VACV. This suppressive effect of VACV was capable of mitigating responses to a secondary challenge with BCG in the skin, ablating DC migration, reducing BCG transport, and delaying CD4+ T cell priming in the dLN. Expression of inflammatory mediators associated with BCG-triggered DC migration were absent from virus-injected skin, suggesting that other pathways invoke DC movement in response to replication-deficient VACV. Despite adamant suppression of DC migration, VACV was still detected early in the dLN and primed Ag-specific CD4+ T cells. In summary, VACV blocks skin DC mobilization from the site of infection while retaining the ability to access the dLN to prime CD4+ T cells.
Collapse
Affiliation(s)
- Juliana Bernardi Aggio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | - Veronika Krmeská
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Brian J Ferguson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Pryscilla Fanini Wowk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | | |
Collapse
|
19
|
Matía A, Lorenzo MM, Blasco R. Tools for the targeted genetic modification of poxvirus genomes. Curr Opin Virol 2020; 44:183-190. [PMID: 33242829 DOI: 10.1016/j.coviro.2020.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
The potential of viruses as biotechnology platforms is becoming more appealing due to technological advances in synthetic biology techniques and to the increasing accessibility of means to manipulate virus genomes. Among viral systems, poxviruses, and their prototype member Vaccinia Virus, are one of the outstanding choices for different biotechnological and medical applications based on heterologous gene expression, recombinant vaccines or oncolytic viruses. The refinement of genetic engineering methods on Vaccinia Virus over the last decades have contributed to facilitate the manipulation of the genomes of poxviruses, and may aid in the improvement of virus variants designed for different goals through reverse genetic approaches. Targeted genetic changes are usually performed by homologous recombination with the viral genome. In addition to the classic approach, recent methodological advances that may assist new strategies for the mutation or edition of poxvirus genomes are reviewed.
Collapse
Affiliation(s)
- Alejandro Matía
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain
| | - María M Lorenzo
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain
| | - Rafael Blasco
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain.
| |
Collapse
|
20
|
Affiliation(s)
- Éva Nagy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| |
Collapse
|
21
|
Boldogkői Z, Moldován N, Balázs Z, Snyder M, Tombácz D. Long-Read Sequencing – A Powerful Tool in Viral Transcriptome Research. Trends Microbiol 2019; 27:578-592. [DOI: 10.1016/j.tim.2019.01.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/21/2019] [Accepted: 01/30/2019] [Indexed: 12/16/2022]
|
22
|
Forsyth KS, Addison MM, Eisenlohr LC. Recombinant Poxviruses: Versatile Tools for Immunological Assays. Methods Mol Biol 2019; 1988:217-248. [PMID: 31147943 DOI: 10.1007/978-1-4939-9450-2_16] [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] [Indexed: 06/09/2023]
Abstract
The study of antigen processing and presentation is critical to our understanding of the mechanisms that govern immune surveillance. A typical requirement of assays designed to examine antigen processing and presentation is the de novo biosynthesis of a model antigen. Historically, Vaccinia virus, a poxvirus closely related to Cowpox virus, has enjoyed widespread use for this purpose. Recombinant poxvirus-based expression has a number of advantages over other systems. Poxviruses accommodate the insertion of large pieces of recombinant DNA into their genome, and recombination and selection are relatively efficient. Poxviruses readily infect a variety of cell types, and they drive rapid and high levels of antigen expression. Additionally, they can be utilized in a variety of assays to study both MHC class I restricted and MHC class II restricted antigen processing and presentation. Ultimately, the numerous advantages of poxvirus recombinants have made the Vaccinia expression system a mainstay in the study of processing and presentation over the past two decades. In an attempt to address one shortcoming of Vaccinia virus while simultaneously retaining the benefits inherent to poxviruses, our laboratory has begun to engineer recombinant Ectromelia viruses. Ectromelia virus, or mousepox, is a natural pathogen of murine cells and performing experiments in the context of a natural host-pathogen relationship may elucidate unknown factors that influence epitope generation and host response. This chapter will describe several recombinant poxvirus system protocols used to study both MHC class I and class II antigen processing and presentation, as well as provide insight and troubleshooting techniques to improve the reproducibility and fidelity of these experiments.
Collapse
Affiliation(s)
- Katherine S Forsyth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary M Addison
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laurence C Eisenlohr
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Children's Hospital of Philadelphia, PA, USA.
| |
Collapse
|
23
|
Ibrahim N, Traktman P. Assessing the Structure and Function of Vaccinia Virus Gene Products by Transient Complementation. Methods Mol Biol 2019; 2023:131-141. [PMID: 31240675 DOI: 10.1007/978-1-4939-9593-6_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Poxviruses are large, complex dsDNA viruses that are highly unusual in replicating solely within the cytoplasm of the infected cell. The most infamous poxvirus was variola virus, the etiological agent of smallpox; today, poxviruses remain of biomedical significance, both as pathogens and as recombinant vaccines and oncolytic therapies. Vaccinia virus is the prototypic poxvirus for experimental analysis. The 195 kb dsDNA genome contains >200 genes that encode proteins involved in such processes as viral entry, gene expression, genome replication and maturation, virion assembly, virion egress, and immune evasion.Molecular genetic analysis has been instrumental in the study of the structure and function of many viral gene products. Temperature-sensitive (ts) mutants have been especially useful in this endeavor; inducible recombinants and deletion mutants are now also important tools. Once a phenotype is observed following the repression, deletion, or inactivation of a particular gene product, the technique of transient complementation becomes central for further study.Simply put, transient complementation involves the transient expression of a variety of alleles of a given viral gene within infected cells, and the evaluation of which of these alleles can "complement" or "rescue" the phenotype caused by the loss of the endogenous allele. This analysis leads to the identification of key domains, motifs, and sites of posttranslational modification. Subcellular localization and protein:protein interactions can also be evaluated in these studies. The development of a reliable toolbox of vectors encoding viral promoters of different temporal classes, and the use of a variety of epitope tags, has greatly enhanced the utility of this experimental approach for poxvirus research.
Collapse
Affiliation(s)
- Nouhou Ibrahim
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Paula Traktman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
24
|
Kieser Q, Paszkowski P, Lin J, Evans D, Noyce R. Visualizing Poxvirus Replication and Recombination Using Live-Cell Imaging. Methods Mol Biol 2019; 2023:221-235. [PMID: 31240681 DOI: 10.1007/978-1-4939-9593-6_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A modernized version of an old saying goes that "If a picture is worth a thousand words, then a video is worth a million." Although made with reference to "YouTube", the quotation also has relevance for microbiologists when one considers how modern microscopes can be used to track biological fluorophores for hours without bleaching or phototoxicity. Confocal fluorescence microscopy provides a powerful tool for capturing dynamic processes within a cellular context that are better understood when viewed using time-lapse videos. In our laboratory we have long been interested in the links between poxvirus DNA replication and recombination and, since these are cytoplasmic viruses, such DNA-dependent processes are easily imaged throughout the virus life cycle without interference from signals coming from nuclear DNA. In this chapter we outline methods that can be used to follow the movement and replication of vaccinia virus DNA, and to also detect the products of poxvirus-catalyzed recombination reactions. We describe how to use the bacteriophage lambda DNA-binding protein, cro, as a way of labeling DNA within a cell when it is conjugated to fluorescent proteins. When used in conjunction with other fluorescent reagents, new labeling technologies, and tagged reporter constructs, these approaches can generate visually appealing and highly informative insights into diverse aspects of vaccinia virus biology.
Collapse
Affiliation(s)
- Quinten Kieser
- Department of Medical Microbiology and Immunology and Li Ka Shing Institute of Virology, 6020 Katz Group Centre, University of Alberta, Edmonton, AB, Canada
| | - Patrick Paszkowski
- Department of Medical Microbiology and Immunology and Li Ka Shing Institute of Virology, 6020 Katz Group Centre, University of Alberta, Edmonton, AB, Canada
| | - James Lin
- Department of Medical Microbiology and Immunology and Li Ka Shing Institute of Virology, 6020 Katz Group Centre, University of Alberta, Edmonton, AB, Canada
| | - David Evans
- Department of Medical Microbiology and Immunology and Li Ka Shing Institute of Virology, 6020 Katz Group Centre, University of Alberta, Edmonton, AB, Canada
| | - Ryan Noyce
- Department of Medical Microbiology and Immunology and Li Ka Shing Institute of Virology, 6020 Katz Group Centre, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
25
|
Full Genome Sequence of the Western Reserve Strain of Vaccinia Virus Determined by Third-Generation Sequencing. GENOME ANNOUNCEMENTS 2018; 6:6/11/e01570-17. [PMID: 29545308 PMCID: PMC5854772 DOI: 10.1128/genomea.01570-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The vaccinia virus is a large, complex virus belonging to the Poxviridae family. Here, we report the complete, annotated genome sequence of the neurovirulent Western Reserve laboratory strain of this virus, which was sequenced on the Pacific Biosciences RS II and Oxford Nanopore MinION platforms.
Collapse
|
26
|
Chea LS, Amara RR. Immunogenicity and efficacy of DNA/MVA HIV vaccines in rhesus macaque models. Expert Rev Vaccines 2017; 16:973-985. [PMID: 28838267 DOI: 10.1080/14760584.2017.1371594] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Despite 30 years of research on HIV, a vaccine to prevent infection and limit disease progression remains elusive. The RV144 trial showed moderate, but significant protection in humans and highlighted the contribution of antibody responses directed against HIV envelope as an important immune correlate for protection. Efforts to further build upon the progress include the use of a heterologous prime-boost regimen using DNA as the priming agent and the attenuated vaccinia virus, Modified Vaccinia Ankara (MVA), as a boosting vector for generating protective HIV-specific immunity. Areas covered: In this review, we summarize the immunogenicity of DNA/MVA vaccines in non-human primate models and describe the efficacy seen in SIV infection models. We discuss immunological correlates of protection determined by these studies and potential approaches for improving the protective immunity. Additionally, we describe the current progress of DNA/MVA vaccines in human trials. Expert commentary: Efforts over the past decade have provided the opportunity to better understand the dynamics of vaccine-induced immune responses and immune correlates of protection against HIV. Based on what we have learned, we outline multiple areas where the field will likely focus on in the next five years.
Collapse
Affiliation(s)
- Lynette Siv Chea
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| | - Rama Rao Amara
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| |
Collapse
|
27
|
Pavot V, Sebastian S, Turner AV, Matthews J, Gilbert SC. Generation and Production of Modified Vaccinia Virus Ankara (MVA) as a Vaccine Vector. Methods Mol Biol 2017; 1581:97-119. [PMID: 28374245 DOI: 10.1007/978-1-4939-6869-5_6] [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] [Indexed: 12/23/2022]
Abstract
The smallpox vaccine based on the vaccinia virus was successfully used to eradicate smallpox, but although very effective, it was a very reactogenic vaccine and responsible for the deaths of one to two people per million vaccinated. Modified Vaccinia virus Ankara (MVA) is an attenuated derivative, also used in the smallpox eradication campaign and now being developed as a recombinant viral vector to produce vaccines against infectious diseases and cancer. MVA can encode one or more foreign antigens and thus can function as a multivalent vaccine. The vector can be used at biosafety level 1, has intrinsic adjuvant properties, and induces humoral and cellular immune responses. Many clinical trials of these new vaccines have been conducted, and the safety of MVA is now well documented. Immunogenicity is influenced by the dose and vaccination regimen, and information on the efficacy of MVA-vectored vaccines is now beginning to accumulate. In this chapter, we provide protocols for generation, isolation, amplification, and purification of recombinant MVA for preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Vincent Pavot
- The Jenner Institute, University of Oxford, Research Bldg., Old Road Campus, ORCRB, Oxford, OX3 7DQ, UK
| | - Sarah Sebastian
- The Jenner Institute, University of Oxford, Research Bldg., Old Road Campus, ORCRB, Oxford, OX3 7DQ, UK
| | - Alison V Turner
- The Jenner Institute, University of Oxford, Research Bldg., Old Road Campus, ORCRB, Oxford, OX3 7DQ, UK
| | - Jake Matthews
- The Jenner Institute, University of Oxford, Research Bldg., Old Road Campus, ORCRB, Oxford, OX3 7DQ, UK
| | - Sarah C Gilbert
- The Jenner Institute, University of Oxford, Research Bldg., Old Road Campus, ORCRB, Oxford, OX3 7DQ, UK.
| |
Collapse
|
28
|
Abstract
ATP-binding cassette (ABC) proteins form a large and ubiquitous family, most members of which are membrane-associated primary transporters. Plant genomes code for a particularly large number of these ABC proteins, with more than 120 genes present in both Arabidopsis thaliana and Oryza sativa (rice). Although plant ABC transporters were initially identified as detoxifiers, sequestering xenobitotics into the vacuole, they were later found to be involved in a wide range of essential physiological processes. Currently, the exact substrates transported by most of these transporters are still unknown and we therefore cannot exclude that a single substrate (e.g. a hormone) is responsible for the diversity of physiological roles. This gap in our knowledge is mainly due to the fact that only a few studies have used direct methods to identify the substrates of these membrane transporters. To address this issue, transport assays involving isolated cells, vesicular membranes or reconstituted liposomes are essential. In this review, we will highlight the importance of the direct biochemical characterization of plant ABC transporters and give some insights into the current status of the homologous and heterologous expression of such proteins.
Collapse
|
29
|
Li H, Hwang Y, Perry K, Bushman F, Van Duyne GD. Structure and Metal Binding Properties of a Poxvirus Resolvase. J Biol Chem 2016; 291:11094-104. [PMID: 27013661 DOI: 10.1074/jbc.m115.709139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 11/06/2022] Open
Abstract
Poxviruses replicate their linear genomes by forming concatemers that must be resolved into monomeric units to produce new virions. A viral resolvase cleaves DNA four-way junctions extruded at the concatemer junctions to produce monomeric genomes. This cleavage reaction is required for viral replication, so the resolvase is an attractive target for small molecule inhibitors. To provide a platform for understanding resolvase mechanism and designing inhibitors, we have determined the crystal structure of the canarypox virus (CPV) resolvase. CPV resolvase is dimer of RNase H superfamily domains related to Escherichia coli RuvC, with an active site lined by highly conserved acidic residues that bind metal ions. There are several intriguing structural differences between resolvase and RuvC, and a model of the CPV resolvase·Holliday junction complex provides insights into the consequences of these differences, including a plausible explanation for the weak sequence specificity exhibited by the poxvirus enzymes. The model also explains why the poxvirus resolvases are more promiscuous than RuvC, cleaving a variety of branched, bulged, and flap-containing substrates. Based on the unique active site structure observed for CPV resolvase, we have carried out a series of experiments to test divalent ion usage and preferences. We find that the two resolvase metal binding sites have different preferences for Mg(2+) versus Mn(2+) Optimal resolvase activity is maintained with 5 μm Mn(2+) and 100 μm Mg(2+), concentrations that are well below those required for either metal alone. Together, our findings provide biochemical insights and structural models that will facilitate studying poxvirus replication and the search for efficient poxvirus inhibitors.
Collapse
Affiliation(s)
- Huiguang Li
- From the Department of Biochemistry & Biophysics, the Graduate Group in Biochemistry and Molecular Biophysics, and
| | - Young Hwang
- the Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kay Perry
- the Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, and the Argonne National Laboratory, Argonne, Illinois 60439
| | - Frederic Bushman
- the Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104,
| | | |
Collapse
|
30
|
Breitbach CJ, Parato K, Burke J, Hwang TH, Bell JC, Kirn DH. Pexa-Vec double agent engineered vaccinia: oncolytic and active immunotherapeutic. Curr Opin Virol 2015; 13:49-54. [DOI: 10.1016/j.coviro.2015.03.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/16/2015] [Accepted: 03/19/2015] [Indexed: 01/21/2023]
|
31
|
Valiente-Echeverría F, Hermoso MA, Soto-Rifo R. RNA helicase DDX3: at the crossroad of viral replication and antiviral immunity. Rev Med Virol 2015; 25:286-99. [PMID: 26174373 DOI: 10.1002/rmv.1845] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/08/2015] [Accepted: 06/08/2015] [Indexed: 12/17/2022]
Abstract
Asp-Glu-Ala-Asp (DEAD)-box polypeptide 3, or DDX3, belongs to the DEAD-box family of ATP-dependent RNA helicases and is known to play different roles in RNA metabolism ranging from transcription to nuclear export, translation, and assembly of stress granules. In addition, there is growing evidence that DDX3 is a component of the innate immune response against viral infections. As such, DDX3 has been shown to play roles both upstream and downstream of I-kappa beta kinase ε (IKKε)/TANK-binding kinase 1, leading to IFN-β production. Interestingly, several RNA viruses, including human threats such as HIV-1 and hepatitis C virus, hijack DDX3 to accomplish various steps of their replication cycles. Thus, it seems that viruses have evolved to exploit DDX3's functions while threatening the innate immune response. Understanding this interesting dichotomy in DDX3 function will help us not only to improve our knowledge of virus-host interactions but also to develop novel antiviral drugs targeting the multifaceted roles of DDX3 in viral replication.
Collapse
Affiliation(s)
- Fernando Valiente-Echeverría
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Marcela A Hermoso
- Innate Immunity Laboratory, Immunology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ricardo Soto-Rifo
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
32
|
Kidokoro M, Shida H. Vaccinia Virus LC16m8∆ as a Vaccine Vector for Clinical Applications. Vaccines (Basel) 2014; 2:755-71. [PMID: 26344890 PMCID: PMC4494248 DOI: 10.3390/vaccines2040755] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/16/2014] [Accepted: 09/28/2014] [Indexed: 01/14/2023] Open
Abstract
The LC16m8 strain of vaccinia virus, the active ingredient in the Japanese smallpox vaccine, was derived from the Lister/Elstree strain. LC16m8 is replication-competent and has been administered to over 100,000 infants and 3,000 adults with no serious adverse reactions. Despite this outstanding safety profile, the occurrence of spontaneously-generated large plaque-forming virulent LC16m8 revertants following passage in cell culture is a major drawback. We identified the gene responsible for the reversion and deleted the gene (B5R) from LC16m8 to derive LC16m8Δ. LC16m8∆ is non-pathogenic in immunodeficient severe combined immunodeficiency (SCID) mice, genetically-stable and does not reverse to a large-plaque phenotype upon passage in cell culture, even under conditions in which most LC16m8 populations are replaced by revertants. Moreover, LC16m8∆ is >500-fold more effective than the non-replicating vaccinia virus (VV), Modified Vaccinia Ankara (MVA), at inducing murine immune responses against pathogenic VV. LC16m8∆, which expresses the SIV gag gene, also induced anti-Gag CD8⁺ T-cells more efficiently than MVA and another non-replicating VV, Dairen I minute-pock variants (DIs). Moreover, LC16m8∆ expressing HIV-1 Env in combination with a Sendai virus vector induced the production of anti-Env antibodies and CD8⁺ T-cells. Thus, the safety and efficacy of LC16m8∆ mean that it represents an outstanding platform for the development of human vaccine vectors.
Collapse
Affiliation(s)
- Minoru Kidokoro
- Department of Virology III, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| | - Hisatoshi Shida
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Japan.
| |
Collapse
|
33
|
Acres B, Bonnefoy JY. Clinical development of MVA-based therapeutic cancer vaccines. Expert Rev Vaccines 2014; 7:889-93. [DOI: 10.1586/14760584.7.7.889] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
34
|
Sui Y, Gordon S, Franchini G, Berzofsky JA. Nonhuman primate models for HIV/AIDS vaccine development. ACTA ACUST UNITED AC 2013; 102:12.14.1-12.14.30. [PMID: 24510515 DOI: 10.1002/0471142735.im1214s102] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The development of HIV vaccines has been hampered by the lack of an animal model that can accurately predict vaccine efficacy. Chimpanzees can be infected with HIV-1 but are not practical for research. However, several species of macaques are susceptible to the simian immunodeficiency viruses (SIVs) that cause disease in macaques, which also closely mimic HIV in humans. Thus, macaque-SIV models of HIV infection have become a critical foundation for AIDS vaccine development. Here we examine the multiple variables and considerations that must be taken into account in order to use this nonhuman primate (NHP) model effectively. These include the species and subspecies of macaques, virus strain, dose and route of administration, and macaque genetics, including the major histocompatibility complex molecules that affect immune responses, and other virus restriction factors. We illustrate how these NHP models can be used to carry out studies of immune responses in mucosal and other tissues that could not easily be performed on human volunteers. Furthermore, macaques are an ideal model system to optimize adjuvants, test vaccine platforms, and identify correlates of protection that can advance the HIV vaccine field. We also illustrate techniques used to identify different macaque lymphocyte populations and review some poxvirus vaccine candidates that are in various stages of clinical trials. Understanding how to effectively use this valuable model will greatly increase the likelihood of finding a successful vaccine for HIV.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,These authors contributed equally
| | - Shari Gordon
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,These authors contributed equally
| | - Genoveffa Franchini
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,These authors contributed equally
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,These authors contributed equally
| |
Collapse
|
35
|
Wennier ST, Brinkmann K, Steinhäußer C, Mayländer N, Mnich C, Wielert U, Dirmeier U, Hausmann J, Chaplin P, Steigerwald R. A novel naturally occurring tandem promoter in modified vaccinia virus ankara drives very early gene expression and potent immune responses. PLoS One 2013; 8:e73511. [PMID: 23951355 PMCID: PMC3741161 DOI: 10.1371/journal.pone.0073511] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/22/2013] [Indexed: 01/06/2023] Open
Abstract
Modified vaccinia virus Ankara (MVA) has been shown to be suitable for the generation of experimental vaccines against cancer and infectious diseases, eliciting strong humoral and cellular immune responses. In viral vectored vaccines, strong recombinant antigen expression and timing of expression influence the quantity and quality of the immune response. Screening of synthetic and native poxvirus promoters for strong protein expression in vitro and potent immune responses in vivo led to the identification of the MVA13.5L promoter, a unique and novel naturally occurring tandem promoter in MVA composed of two 44 nucleotide long repeated motifs, each containing an early promoter element. The MVA13.5L gene is highly conserved across orthopoxviruses, yet its function is unknown. The unique structure of its promoter is not found for any other gene in the MVA genome and is also conserved in other orthopoxviruses. Comparison of the MVA13.5L promoter activity with synthetic poxviral promoters revealed that the MVA13.5L promoter produced higher levels of protein early during infection in HeLa cells and particularly in MDBK cells, a cell line in which MVA replication stops at an early stage before the expression of late genes. Finally, a recombinant antigen expressed under the control of this novel promoter induced high antibody titers and increased CD8 T cell responses in homologous prime-boost immunization compared to commonly used promoters. In particular, the recombinant antigen specific CD8 T cell responses dominated over the immunodominant B8R vector-specific responses after three vaccinations and even more during the memory phase. These results have identified the native MVA13.5L promoter as a new potent promoter for use in MVA vectored preventive and therapeutic vaccines.
Collapse
Affiliation(s)
- Sonia T. Wennier
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | - Kay Brinkmann
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | | | - Nicole Mayländer
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | - Claudia Mnich
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | - Ursula Wielert
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | - Ulrike Dirmeier
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | - Jürgen Hausmann
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | - Paul Chaplin
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
| | - Robin Steigerwald
- Infectious Disease Division, Bavarian Nordic GmbH, Martinsried, Germany
- * E-mail:
| |
Collapse
|
36
|
Genome-wide profiling of human cap-independent translation-enhancing elements. Nat Methods 2013; 10:747-50. [PMID: 23770754 PMCID: PMC3731418 DOI: 10.1038/nmeth.2522] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/02/2013] [Indexed: 11/23/2022]
Abstract
We report an in vitro selection strategy to identify RNA sequences that mediate cap-independent translation initiation. This method entails the mRNA display of trillions of genomic fragments, selection for translation initiation, and high-throughput deep sequencing. We identified >12,000 translation enhancing elements (TEEs) in the human genome, generated a high-resolution map of human TEE bearing regions (TBRs), and validated the function of a subset of sequences in vitro and in cells.
Collapse
|
37
|
Siciliano NA, Huang L, Eisenlohr LC. Recombinant poxviruses: versatile tools for immunological assays. Methods Mol Biol 2013; 960:219-245. [PMID: 23329491 DOI: 10.1007/978-1-62703-218-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The study of antigen processing and presentation is critical to our understanding of the mechanisms that govern immune surveillance. A typical requirement of assays designed to examine antigen processing and presentation is the de novo biosynthesis of a model antigen. Historically, Vaccinia virus (VACV), a poxvirus closely related to Cowpox, has enjoyed widespread use for this purpose. Recombinant poxvirus-based expression has a number of advantages over other systems. Poxviruses accommodate the insertion of large pieces of recombinant DNA into their genome, and recombination and selection are relatively efficient. Poxviruses readily infect a variety of cell types, and they drive rapid and high levels of antigen expression. Additionally, they can be utilized in a variety of assays to study both MHC class I-restricted and MHC class II-restricted antigen processing and presentation. Ultimately, the numerous advantages of poxvirus recombinants have made the Vaccinia expression system a mainstay in the study of processing and presentation over the past two decades. In an attempt to address one shortcoming of VACV while simultaneously retaining the benefits inherent to poxviruses, our laboratory has begun to engineer recombinant Ectromelia viruses. Ectromelia virus, or mousepox, is a natural pathogen of murine cells and performing experiments in the context of a natural host-pathogen relationship may elucidate unknown factors that influence epitope generation and host response. This chapter describes several recombinant poxvirus system protocols used to study both MHC class I and class II antigen processing and presentation, as well as provides insight and troubleshooting techniques to improve the reproducibility and fidelity of these experiments.
Collapse
Affiliation(s)
- Nicholas A Siciliano
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lan Huang
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Laurence C Eisenlohr
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
38
|
O'Connell RJ, Kim JH, Corey L, Michael NL. Human immunodeficiency virus vaccine trials. Cold Spring Harb Perspect Med 2012; 2:a007351. [PMID: 23209178 PMCID: PMC3543076 DOI: 10.1101/cshperspect.a007351] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
More than 2 million AIDS-related deaths occurred globally in 2008, and more than 33 million people are living with HIV/AIDS. Despite promising advances in prevention, an estimated 2.7 million new HIV infections occurred in that year, so that for every two patients placed on combination antiretroviral treatment, five people became infected. The pandemic poses a formidable challenge to the development, progress, and stability of global society 30 years after it was recognized. Experimental preventive HIV-1 vaccines have been administered to more than 44,000 human volunteers in more than 187 separate trials since 1987. Only five candidate vaccine strategies have been advanced to efficacy testing. The recombinant glycoprotein (rgp)120 subunit vaccines, AIDSVAX B/B and AIDSVAX B/E, and the Merck Adenovirus serotype (Ad)5 viral-vector expressing HIV-1 Gag, Pol, and Nef failed to show a reduction in infection rate or lowering of postinfection viral set point. Most recently, a phase III trial that tested a heterologous prime-boost vaccine combination of ALVAC-HIV vCP1521 and bivalent rgp120 (AIDSVAX B/E) showed 31% efficacy in protection from infection among community-risk Thai participants. A fifth efficacy trial testing a DNA/recombinant(r) Ad5 prime-boost combination is currently under way. We review the clinical trials of HIV vaccines that have provided insight into human immunogenicity or efficacy in preventing HIV-1 infection.
Collapse
Affiliation(s)
- Robert J O'Connell
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | | | | |
Collapse
|
39
|
Lorente E, Infantes S, Abia D, Barnea E, Beer I, García R, Lasala F, Jiménez M, Mir C, Morreale A, Admon A, López D. A viral, transporter associated with antigen processing (TAP)-independent, high affinity ligand with alternative interactions endogenously presented by the nonclassical human leukocyte antigen E class I molecule. J Biol Chem 2012; 287:34895-34903. [PMID: 22927436 PMCID: PMC3471699 DOI: 10.1074/jbc.m112.362293] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 08/10/2012] [Indexed: 01/05/2023] Open
Abstract
The transporter associated with antigen processing (TAP) enables the flow of viral peptides generated in the cytosol by the proteasome and other proteases to the endoplasmic reticulum, where they complex with nascent human leukocyte antigen (HLA) class I. Later, these peptide-HLA class I complexes can be recognized by CD8(+) lymphocytes. Cancerous cells and infected cells in which TAP is blocked, as well as individuals with unusable TAP complexes, are able to present peptides on HLA class I by generating them through TAP-independent processing pathways. Here, we identify a physiologically processed HLA-E ligand derived from the D8L protein in TAP-deficient vaccinia virus-infected cells. This natural high affinity HLA-E class I ligand uses alternative interactions to the anchor motifs previously described to be presented on nonclassical HLA class I molecules. This octameric peptide was also presented on HLA-Cw1 with similar binding affinity on both classical and nonclassical class I molecules. In addition, this viral peptide inhibits HLA-E-mediated cytolysis by natural killer cells. Comparison between the amino acid sequences of the presenting HLA-E and HLA-Cw1 alleles revealed a shared structural motif in both HLA class molecules, which could be related to their observed similar cross-reactivity affinities. This motif consists of several residues located on the floor of the peptide-binding site. These data expand the role of HLA-E as an antigen-presenting molecule.
Collapse
Affiliation(s)
- Elena Lorente
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Susana Infantes
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - David Abia
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Ilan Beer
- IBM Haifa Research Lab, Haifa 31905, Israel
| | - Ruth García
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Fátima Lasala
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Mercedes Jiménez
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Carmen Mir
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Antonio Morreale
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Arie Admon
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Daniel López
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain.
| |
Collapse
|
40
|
Elena Gómez C, Perdiguero B, García-Arriaza J, Esteban M. Poxvirus vectors as HIV/AIDS vaccines in humans. Hum Vaccin Immunother 2012; 8:1192-207. [PMID: 22906946 PMCID: PMC3579898 DOI: 10.4161/hv.20778] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The RV144 phase III clinical trial with the combination of the poxvirus vector ALVAC and the HIV gp120 protein has taught us that a vaccine against HIV/AIDS is possible but further improvements are still needed. Although the HIV protective effect of RV144 was modest (31.2%), these encouraging results reinforce the use of poxvirus vectors as HIV/AIDS vaccine candidates. In this review we focus on the prophylactic clinical studies thus far performed with the more widely studied poxvirus vectors, ALVAC, MVA, NYVAC and fowlpox expressing HIV antigens. We describe the characteristics of each vector administered either alone or in combination with other vectors, with emphasis on the immune parameters evaluated in healthy volunteers, percentage of responders and triggering of humoral and T cell responses. Some of these immunogens induced broad, polyfunctional and long-lasting CD4(+) and CD8(+) T cell responses to HIV-1 antigens in most volunteers, with preference for effector memory T cells, and neutralizing antibodies, immune parameters that might be relevant in protection. Finally, we consider improvements in immunogenicity of the poxvirus vectors by the selective deletion of viral immunomodulatory genes and insertion of host range genes in the poxvirus genome. Overall, the poxvirus vectors have proven to be excellent HIV/AIDS vaccine candidates, with distinct behavior among them, and the future implementation will be dictated by their optimized immune profile in clinical trials.
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC); Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC); Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC); Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC); Madrid, Spain
| |
Collapse
|
41
|
Lace MJ, Ushikai M, Yamakawa Y, Anson JR, Ishiji T, Turek LP, Haugen TH. The truncated C-terminal E2 (E2-TR) protein of bovine papillomavirus (BPV) type-1 is a transactivator that modulates transcription in vivo and in vitro in a manner distinct from the E2-TA and E8^E2 gene products. Virology 2012; 429:99-111. [DOI: 10.1016/j.virol.2012.03.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 01/09/2012] [Accepted: 03/30/2012] [Indexed: 10/28/2022]
|
42
|
Rojas JJ, Thorne SH. Theranostic potential of oncolytic vaccinia virus. Theranostics 2012; 2:363-73. [PMID: 22509200 PMCID: PMC3326721 DOI: 10.7150/thno.3724] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/18/2012] [Indexed: 11/17/2022] Open
Abstract
Biological cancer therapies, such as oncolytic, or replication-selective viruses have advantages over traditional therapeutics as they can employ multiple different mechanisms to target and destroy cancers (including direct cell lysis, immune activation and vascular collapse). This has led to their rapid recent clinical development. However this also makes their pre-clinical and clinical study complex, as many parameters may affect their therapeutic potential and so defining reason for treatment failure or approaches that might enhance their therapeutic activity can be complicated. The ability to non-invasively image viral gene expression in vivo both in pre-clinical models and during clinical testing will considerably enhance the speed of oncolytic virus development as well as increasing the level and type of useful data produced from these studies. Further, subsequent to future clinical approval, imaging of reporter gene expression might be used to evaluate the likelihood of response to oncolytic viral therapy prior to changes in tumor burden. Here different reporter genes used in conjunction with oncolytic viral therapy are described, along with the imaging modalities used to measure their expression, while their applications both in pre-clinical and clinical testing are discussed. Possible future applications for reporter gene expression from oncolytic viruses in the phenotyping of tumors and the personalizing of treatment regimens are also discussed.
Collapse
|
43
|
Liu Q, Huang W, Nie J, Zhu R, Gao D, Song A, Meng S, Xu X, Wang Y. A novel high-throughput vaccinia virus neutralization assay and preexisting immunity in populations from different geographic regions in China. PLoS One 2012; 7:e33392. [PMID: 22438922 PMCID: PMC3306400 DOI: 10.1371/journal.pone.0033392] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 02/08/2012] [Indexed: 11/25/2022] Open
Abstract
Background Pre-existing immunity to Vaccinia Tian Tan virus (VTT) resulting from a large vaccination campaign against smallpox prior to the early 1980s in China, has been a major issue for application of VTT-vector based vaccines. It is essential to establish a sensitive and high-throughput neutralization assay to understand the epidemiology of Vaccinia-specific immunity in current populations in China. Methodology/Principal Findings A new anti-Vaccinia virus (VACV) neutralization assay that used the attenuated replication-competent VTT carrying the firefly luciferase gene of Photinus pyralis (rTV-Fluc) was established and standardized for critical parameters that included the choice of cell line, viral infection dose, and the infection time. The current study evaluated the maintenance of virus-specific immunity after smallpox vaccination by conducting a non-randomized, cross-sectional analysis of antiviral antibody-mediated immune responses in volunteers examined 30–55 years after vaccination. The rTV-Fluc neutralization assay was able to detect neutralizing antibodies (NAbs) against Vaccinia virus without the ability to differentiate strains of Vaccinia virus. We showed that the neutralizing titers measured by our assay were similar to those obtained by the traditional plaque reduction neutralization test (PRNT). Using this assay, we found a low prevalence of NAb to VTT (7.6%) in individuals born before 1980 from Beijing and Anhui provinces in China, and when present, anti-VTT NAb titers were low. No NAbs were detected in all 222 samples from individuals born after 1980. There was no significant difference observed for titer or prevalence by gender, age range and geographic origin. Conclusion A simplified, sensitive, standardized, reproducible, and high-throughput assay was developed for the quantitation of NAbs against different Vaccinia strains. The current study provides useful insights for the future development of VTT-based vaccination in Beijing and Anhui provinces of China.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Cell Biology, National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing, China
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Weijin Huang
- Department of Cell Biology, National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing, China
| | - Jianhui Nie
- Department of Cell Biology, National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing, China
| | - Rong Zhu
- Department of Cell Biology, National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing, China
| | | | - Aijing Song
- Department of Cell Biology, National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing, China
| | - Shufang Meng
- Department of Cell Biology, National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing, China
| | - Xuemei Xu
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Youchun Wang
- Department of Cell Biology, National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing, China
- * E-mail:
| |
Collapse
|
44
|
Raafat N, Sadowski-Cron C, Mengus C, Heberer M, Spagnoli GC, Zajac P. Preventing vaccinia virus class-I epitopes presentation by HSV-ICP47 enhances the immunogenicity of a TAP-independent cancer vaccine epitope. Int J Cancer 2012; 131:E659-69. [PMID: 22116674 DOI: 10.1002/ijc.27362] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 11/08/2011] [Indexed: 11/06/2022]
Abstract
Herpes simplex virus protein ICP47, encoded by US12 gene, strongly downregulates major histocompatibility complex (MHC) class-I antigen restricted presentation by blocking transporter associated with antigen processing (TAP) protein. To decrease viral vector antigenic immunodominance and MHC class-I driven clearance, we engineered recombinant vaccinia viruses (rVV) expressing ICP47 alone (rVV-US12) or together with endoplasmic reticulum (ER)-targeted Melan-A/MART-1(27-35) model tumor epitope (rVV-MUS12). In this study, we show that antigen presenting cells (APC), infected with rVV-US12, display a decreased ability to present TAP dependent MHC class-I restricted viral antigens to CD8+ T-cells. While HLA class-I cell surface expression is strongly downregulated, other important immune related molecules such as CD80, CD44 and, most importantly, MHC class-II are unaffected. Characterization of rVV-MUS12 infected cells demonstrates that over-expression of a TAP-independent peptide, partially compensates for ICP47 induced surface MHC class-I downregulation (30% vs. 70% respectively). Most importantly, in conditions where clearance of infected APC by virus-specific CTL represents a limiting factor, a significant enhancement of CTL responses to the tumor epitope can be detected in cultures stimulated with rVV-MUS12, as compared to those stimulated by rVV-MART alone. Such reagents could become of high relevance in multiple boost protocols required for cancer immunotherapy, to limit vector-specific responsiveness.
Collapse
Affiliation(s)
- Nermin Raafat
- Department of Biomedicine, Oncology group, Institute of Surgical Research and Hospital Management, University of Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
45
|
Gavrilov BK, Rogers K, Fernandez-Sainz IJ, Holinka LG, Borca MV, Risatti GR. Effects of glycosylation on antigenicity and immunogenicity of classical swine fever virus envelope proteins. Virology 2011; 420:135-45. [DOI: 10.1016/j.virol.2011.08.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 08/15/2011] [Accepted: 08/31/2011] [Indexed: 11/24/2022]
|
46
|
Multiple viral ligands naturally presented by different class I molecules in transporter antigen processing-deficient vaccinia virus-infected cells. J Virol 2011; 86:527-41. [PMID: 22031944 DOI: 10.1128/jvi.05737-11] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The transporter associated with antigen processing (TAP) delivers the viral proteolytic products generated by the proteasome in the cytosol to the endoplasmic reticulum lumen that are subsequently recognized by cytotoxic T lymphocytes (CTLs). However, several viral epitopes have been identified in TAP-deficient models. Using mass spectrometry to analyze complex human leukocyte antigen (HLA)-bound peptide pools isolated from large numbers of TAP-deficient vaccinia virus-infected cells, we identified 11 ligands naturally presented by four different HLA-A, HLA-B, and HLA-C class I molecules. Two of these ligands were presented by two different HLA class I alleles, and, as a result, 13 different HLA-peptide complexes were formed simultaneously in the same vaccinia virus-infected cells. In addition to the high-affinity ligands, one low-affinity peptide restricted by each of the HLA-A, HLA-B, and HLA-C class I molecules was identified. Both high- and low-affinity ligands generated long-term memory CTL responses to vaccinia virus in an HLA-A2-transgenic mouse model. The processing and presentation of two vaccinia virus-encoded HLA-A2-restricted antigens took place via proteasomal and nonproteasomal pathways, which were blocked in infected cells with chemical inhibitors specific for different subsets of metalloproteinases. These data have implications for the study of the effectiveness of early empirical vaccination with cowpox virus against smallpox disease.
Collapse
|
47
|
Intravenous delivery of a multi-mechanistic cancer-targeted oncolytic poxvirus in humans. Nature 2011; 477:99-102. [PMID: 21886163 DOI: 10.1038/nature10358] [Citation(s) in RCA: 426] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 07/05/2011] [Indexed: 12/14/2022]
Abstract
The efficacy and safety of biological molecules in cancer therapy, such as peptides and small interfering RNAs (siRNAs), could be markedly increased if high concentrations could be achieved and amplified selectively in tumour tissues versus normal tissues after intravenous administration. This has not been achievable so far in humans. We hypothesized that a poxvirus, which evolved for blood-borne systemic spread in mammals, could be engineered for cancer-selective replication and used as a vehicle for the intravenous delivery and expression of transgenes in tumours. JX-594 is an oncolytic poxvirus engineered for replication, transgene expression and amplification in cancer cells harbouring activation of the epidermal growth factor receptor (EGFR)/Ras pathway, followed by cell lysis and anticancer immunity. Here we show in a clinical trial that JX-594 selectively infects, replicates and expresses transgene products in cancer tissue after intravenous infusion, in a dose-related fashion. Normal tissues were not affected clinically. This platform technology opens up the possibility of multifunctional products that selectively express high concentrations of several complementary therapeutic and imaging molecules in metastatic solid tumours in humans.
Collapse
|
48
|
Zou J, Huang XX, Yin GW, Ding Y, Liu XY, Wang H, Chen QJ, Suo X. Evaluation of Toxoplasma gondii as a live vaccine vector in susceptible and resistant hosts. Parasit Vectors 2011; 4:168. [PMID: 21871123 PMCID: PMC3177786 DOI: 10.1186/1756-3305-4-168] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 08/28/2011] [Indexed: 11/10/2022] Open
Abstract
Background Toxoplasma gondii has been shown to trigger strong cellular immune responses to heterologous antigens expressed by the parasite in the inbred mouse model [1]. We studied the immune response induced by T. gondii as an effective vaccine vector in chickens and rabbits. Results T. gondii RH strain was engineered to express the yellow fluorescent protein (YFP) in the cytoplasm. A subcutaneous injection of the transgenic T. gondii YFP in chickens afforded partial protection against the infection of transgenic E. tenella YFP. T. gondii YFP induced low levels of antibodies to YFP in chickens, suggesting that YFP specific cellular immune response was probably responsible for the protective immunity against E. tenella YFP infection. The measurement of T-cell response and IFN-γ production further confirmed that YFP specific Th1 mediated immune response was induced by T. gondii YFP in immunized chickens. The transgenic T. gondii stimulated significantly higher YFP specific IgG titers in rabbits than in chickens, suggesting greater immunogenicity in a T. gondii susceptible species than in a resistant species. Priming with T. gondii YFP and boosting with the recombinant YFP can induce a strong anti-YFP antibody response in both animal species. Conclusions Our findings suggest that T. gondii can be used as an effective vaccine vector and future research should focus on exploring avirulent no cyst-forming strains of T. gondii as a live vaccine vector in animals.
Collapse
Affiliation(s)
- Jun Zou
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Paris RM, Kim JH, Robb ML, Michael NL. Prime-boost immunization with poxvirus or adenovirus vectors as a strategy to develop a protective vaccine for HIV-1. Expert Rev Vaccines 2010; 9:1055-69. [PMID: 20822348 DOI: 10.1586/erv.10.106] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Challenges in the development of an effective HIV-1 vaccine are myriad with significant hurdles posed by viral diversity, the lack of a human correlate of protection and difficulty in creating immunogens capable of eliciting broadly neutralizing antibodies. The implicit requirement for novel approaches to these problems has resulted in vaccine candidates designed to elicit cellular and/or humoral immune responses, to include recombinant DNA, viral and bacterial vectors, and subunit proteins. Here, we review data from clinical studies primarily of poxvirus and adenovirus vector vaccines, used in a heterologous prime-boost combination strategy. Currently, this strategy appears to hold the most promise for an effective vaccine based on results from immunogenicity testing and nonhuman primate challenge models, as well as the modest efficacy recently observed in the Thai prime-boost trial.
Collapse
Affiliation(s)
- Robert M Paris
- US Military HIV Research Program (MHRP), Department of Retrovirology, Armed Forces Research Institute of Medical Sciences, 315/6 Rajvithi Road, Bangkok, 10400, Thailand.
| | | | | | | |
Collapse
|
50
|
Abstract
BACKGROUND Recombinant vaccinia virus (rVV) strains expressing the immunomodulatory cholera toxin B subunit (CTB) fused to the autoantigen glutamic acid decarboxylase (GAD) or the immunosuppressive cytokine interleukin-10 (IL-10) were independently able to generate only low levels of immune suppression of type 1 diabetes mellitus (T1DM). Here we suggest that a vaccinia virus (VV)-mediated combination of CTB::GAD fusion and IL-10 proteins promises a effective and durable immunotherapeutic strategy for T1DM. METHODS To explore this hypothesis, a CTB::GAD fusion gene was co-delivered with a gene encoding IL-10 by rVV infection (rVV-CTB::GAD + rVV-IL10) into 5-7-week-old non-obese diabetic (NOD) mice. The mice were assessed for vaccine protection against development of hyperglycemia from 12 to 64 weeks of age by assessment of pancreatic inflammation (insulitis) and splenocyte-secreted interferon-gamma and IL-10 cytokine levels. RESULTS By 36 weeks of age, from 54% to 80% of the mice in the negative control animal groups (either mock-infected or inoculated with unrelated plasmid or VV) had developed hyperglycemia. Similarly, no statistically significant improvement in protection against diabetes onset was achieved by inoculation with VV expressing CTB::GAD or IL-10 independently. Surprisingly, only 20% of mice co-inoculated with rVV-CTB::GAD + rVV-IL10 developed hyperglycemia by 28 weeks of age. Other treatment groups developed hyperglycemia by 32-36 weeks. After 36 weeks, diabetes incidence no longer increased in any groups until the end of experiment at 64 weeks of age. Histological analysis of pancreatic tissues of hyperglycemic mice revealed high levels of intra-islet insulitis. Analysis of insulitis at termination of the experiment showed that euglycemic mice co-inoculated with VV expressing CTB::GAD and IL-10 had more effectively reduced inflammation in comparison with the other groups. CONCLUSIONS A combinatorial vaccination strategy based on VV co-delivery of genes encoding the immunoenhanced autoantigen CTB::GAD and the anti-inflammatory cytokine IL-10 can maintain effective and durable euglycemia and immunological homeostasis in NOD mice with prediabetes.
Collapse
Affiliation(s)
- Béla Dénes
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
- Department of Immunology, Central Veterinary Institute, Budapest, Hungary
| | - István Fodor
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
| | - William H.R. Langridge
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
| |
Collapse
|