1
|
Park BR, Bommireddy R, Chung DH, Kim KH, Subbiah J, Jung YJ, Bhatnagar N, Pack CD, Ramachandiran S, Reddy SJC, Selvaraj P, Kang SM. Hemagglutinin virus-like particles incorporated with membrane-bound cytokine adjuvants provide protection against homologous and heterologous influenza virus challenge in aged mice. Immun Ageing 2023; 20:20. [PMID: 37170231 PMCID: PMC10173218 DOI: 10.1186/s12979-023-00344-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/24/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Current influenza vaccines deliver satisfactory results in young people but are less effective in the elderly. Development of vaccines for an ever-increasing aging population has been an arduous challenge due to immunosenescence that impairs the immune response in the aged, both quantitatively and qualitatively. RESULTS To potentially enhance vaccine efficacy in the elderly, we investigated the immunogenicity and cross-protection of influenza hemagglutinin virus-like particles (HA-VLP) incorporated with glycosylphosphatidylinositol (GPI)-anchored cytokine-adjuvants (GPI-GM-CSF and GPI-IL-12) via protein transfer in aged mice. Lung viral replication against homologous and heterologous influenza viruses was significantly reduced in aged mice after vaccination with cytokine incorporated VLPs (HA-VLP-Cyt) in comparison to HA-VLP alone. Enhanced IFN-γ+CD4+ and IFN-γ+CD8+ T cell responses were also observed in aged mice immunized with HA-VLP-Cyt when compared to HA-VLP alone. CONCLUSIONS Cytokine-adjuvanted influenza HA-VLP vaccine induced enhanced protective response against homologous influenza A virus infection in aged mice. Influenza HA-VLP vaccine with GPI-cytokines also induced enhanced T cell responses correlating with better protection against heterologous infection in the absence of neutralizing antibodies. The results suggest that a vaccination strategy using cytokine-adjuvanted influenza HA-VLPs could be used to enhance protection against influenza A virus in the elderly.
Collapse
Affiliation(s)
- Bo Ryoung Park
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Ramireddy Bommireddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - David Hyunjung Chung
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Ki-Hye Kim
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Jeeva Subbiah
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Noopur Bhatnagar
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | | | | | | | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
2
|
Bommireddy R, Stone S, Bhatnagar N, Kumari P, Munoz LE, Oh J, Kim KH, Berry JTL, Jacobsen KM, Jaafar L, Naing SH, Blackerby AN, der Gaag TV, Wright CN, Lai L, Pack CD, Ramachandiran S, Suthar MS, Kang SM, Kumar M, Reddy SJC, Selvaraj P. Influenza Virus-like Particle-Based Hybrid Vaccine Containing RBD Induces Immunity against Influenza and SARS-CoV-2 Viruses. Vaccines (Basel) 2022; 10:944. [PMID: 35746552 PMCID: PMC9230705 DOI: 10.3390/vaccines10060944] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 02/05/2023] Open
Abstract
Several approaches have produced an effective vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since millions of people are exposed to influenza virus and SARS-CoV-2, it is of great interest to develop a two-in-one vaccine that will be able to protect against infection of both viruses. We have developed a hybrid vaccine for SARS-CoV-2 and influenza viruses using influenza virus-like particles (VLP) incorporated by protein transfer with glycosylphosphatidylinositol (GPI)-anchored SARS-CoV-2 RBD fused to GM-CSF as an adjuvant. GPI-RBD-GM-CSF fusion protein was expressed in CHO-S cells, purified and incorporated onto influenza VLPs to develop the hybrid vaccine. Our results show that the hybrid vaccine induced a strong antibody response and protected mice from both influenza virus and mouse-adapted SARS-CoV-2 challenges, with vaccinated mice having significantly lower lung viral titers compared to naive mice. These results suggest that a hybrid vaccine strategy is a promising approach for developing multivalent vaccines to prevent influenza A and SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Ramireddy Bommireddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.B.); (L.E.M.); (J.T.L.B.)
| | - Shannon Stone
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (P.K.); (M.K.)
| | - Noopur Bhatnagar
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (N.B.); (J.O.); (K.-H.K.); (S.-M.K.)
| | - Pratima Kumari
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (P.K.); (M.K.)
| | - Luis E. Munoz
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.B.); (L.E.M.); (J.T.L.B.)
| | - Judy Oh
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (N.B.); (J.O.); (K.-H.K.); (S.-M.K.)
| | - Ki-Hye Kim
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (N.B.); (J.O.); (K.-H.K.); (S.-M.K.)
| | - Jameson T. L. Berry
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.B.); (L.E.M.); (J.T.L.B.)
| | - Kristen M. Jacobsen
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Lahcen Jaafar
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Swe-Htet Naing
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Allison N. Blackerby
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Tori Van der Gaag
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Chloe N. Wright
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Lilin Lai
- Department of Pediatrics, Emory Vaccine Center, Yerkes Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.L.); (M.S.S.)
| | - Christopher D. Pack
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Sampath Ramachandiran
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Mehul S. Suthar
- Department of Pediatrics, Emory Vaccine Center, Yerkes Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.L.); (M.S.S.)
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (N.B.); (J.O.); (K.-H.K.); (S.-M.K.)
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (S.S.); (P.K.); (M.K.)
| | - Shaker J. C. Reddy
- Metaclipse Therapeutics Corporation, Atlanta, GA 30340, USA; (K.M.J.); (L.J.); (S.-H.N.); (A.N.B.); (T.V.d.G.); (C.N.W.); (C.D.P.); (S.R.); (S.J.C.R.)
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.B.); (L.E.M.); (J.T.L.B.)
| |
Collapse
|
3
|
Pritam M, Singh G, Kumar R, Singh SP. Screening of potential antigens from whole proteome and development of multi-epitope vaccine against Rhizopus delemar using immunoinformatics approaches. J Biomol Struct Dyn 2022; 41:2118-2145. [PMID: 35067195 DOI: 10.1080/07391102.2022.2028676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mucormycosis is a deadly fungal disease mainly caused by Rhizopus oryzae (strain 99-880), also known as Rhizopus delemar. Previously, mucormycosis occurs in immunocompromised patients of diabetes mellitus, cancer, organ transplant, etc. But there was a drastic increase in mucormycosis cases in the ongoing COVID-19 pandemic. Despite several available therapies and antifungal treatments, the mortality rate of mucormycosis is about more than 50%. Currently, there is no vaccine available in the market for mucormycosis that urgently needs to develop a potential vaccine against mucormycosis with high efficacy. In the present study, we have screened 4 genome-derived predicted antigens (GDPA) through sequential filtration of the whole proteome of R. delemar using different benchmarked bioinformatics tools. These 4 GDPA along with 4 randomly selected experimentally reported antigens (ERA) were sourced for prediction of B- and T- cell epitopes and utilized in designing of two potential multi-epitope vaccine candidates which can induce both innate and adaptive immunity against R. delemar. Besides these, comparative immune simulation studies and in silico cloning were performed using L. lactis as an expression system for their possible uses as oral vaccines. This is the first multi-epitope vaccine designed against R. delemar through systematic pipelined reverse vaccinology and immunoinformatic approaches. Although the wet-lab based experimental validation of designed vaccines is required before testing in the preclinical model, the current study will significantly help in reducing the cost of experimentation as well as improving the efficacy of vaccine therapy against mucormycosis and other pathogenic diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manisha Pritam
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Garima Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | | |
Collapse
|
4
|
Creytens S, Pascha MN, Ballegeer M, Saelens X, de Haan CAM. Influenza Neuraminidase Characteristics and Potential as a Vaccine Target. Front Immunol 2021; 12:786617. [PMID: 34868073 PMCID: PMC8635103 DOI: 10.3389/fimmu.2021.786617] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/29/2021] [Indexed: 12/28/2022] Open
Abstract
Neuraminidase of influenza A and B viruses plays a critical role in the virus life cycle and is an important target of the host immune system. Here, we highlight the current understanding of influenza neuraminidase structure, function, antigenicity, immunogenicity, and immune protective potential. Neuraminidase inhibiting antibodies have been recognized as correlates of protection against disease caused by natural or experimental influenza A virus infection in humans. In the past years, we have witnessed an increasing interest in the use of influenza neuraminidase to improve the protective potential of currently used influenza vaccines. A number of well-characterized influenza neuraminidase-specific monoclonal antibodies have been described recently, most of which can protect in experimental challenge models by inhibiting the neuraminidase activity or by Fc receptor-dependent mechanisms. The relative instability of the neuraminidase poses a challenge for protein-based antigen design. We critically review the different solutions that have been proposed to solve this problem, ranging from the inclusion of stabilizing heterologous tetramerizing zippers to the introduction of inter-protomer stabilizing mutations. Computationally engineered neuraminidase antigens have been generated that offer broad, within subtype protection in animal challenge models. We also provide an overview of modern vaccine technology platforms that are compatible with the induction of robust neuraminidase-specific immune responses. In the near future, we will likely see the implementation of influenza vaccines that confront the influenza virus with a double punch: targeting both the hemagglutinin and the neuraminidase.
Collapse
MESH Headings
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antigenic Drift and Shift
- Antigens, Viral/immunology
- Antigens, Viral/ultrastructure
- Catalytic Domain/genetics
- Catalytic Domain/immunology
- Cross Protection
- Evolution, Molecular
- Humans
- Immunogenicity, Vaccine
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Alphainfluenzavirus/enzymology
- Alphainfluenzavirus/genetics
- Alphainfluenzavirus/immunology
- Betainfluenzavirus/enzymology
- Betainfluenzavirus/genetics
- Betainfluenzavirus/immunology
- Mutation
- Nanoparticles
- Neuraminidase/administration & dosage
- Neuraminidase/genetics
- Neuraminidase/immunology
- Neuraminidase/ultrastructure
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/ultrastructure
- Viral Proteins/administration & dosage
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Proteins/ultrastructure
Collapse
Affiliation(s)
- Sarah Creytens
- Vlaams Instituut voor Biotechnologie (VIB)-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Mirte N. Pascha
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Marlies Ballegeer
- Vlaams Instituut voor Biotechnologie (VIB)-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- Vlaams Instituut voor Biotechnologie (VIB)-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Cornelis A. M. de Haan
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
5
|
Menne Z, Pliasas VC, Compans RW, Glover S, Kyriakis CS, Skountzou I. Bivalent vaccination with NA1 and NA2 neuraminidase virus-like particles is protective against challenge with H1N1 and H3N2 influenza A viruses in a murine model. Virology 2021; 562:197-208. [PMID: 34375782 PMCID: PMC8479372 DOI: 10.1016/j.virol.2021.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/25/2021] [Accepted: 08/04/2021] [Indexed: 11/29/2022]
Abstract
Neuraminidase (NA) is the second most abundant glycoprotein on the surface of influenza A viruses (IAV). Neuraminidase type 1 (NA1) based virus-like particles (VLPs) have previously been shown to protect against challenge with H1N1 and H3N2 IAV. In this study, we produced neuraminidase type 2 (NA2) VLPs derived from the sequence of the seasonal IAV A/Perth/16/2009. Intramuscular vaccination of mice with NA2 VLPs induced high anti-NA serum IgG levels capable of inhibiting NA activity. NA2 VLP vaccination protected against mortality in a lethal A/Hong Kong/1/1968 (H3N2) virus challenge model, but not against lethal challenge with A/California/04/2009 (H1N1) virus. However, bivalent vaccination with NA1 and NA2 VLPs demonstrated no antigenic competition in anti-NA IgG responses and protected against lethal challenge with H1N1 and H3N2 viruses. Here we demonstrate that vaccination with NA VLPs is protective against influenza challenge and supports focusing on anti-NA responses in the development of future vaccination strategies.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Dose-Response Relationship, Immunologic
- Immunity, Heterologous
- Immunoglobulin G/immunology
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Injections, Intramuscular
- Mice
- Neuraminidase/immunology
- Orthomyxoviridae Infections/prevention & control
- Vaccination/methods
- Vaccines, Combined/administration & dosage
- Vaccines, Combined/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/immunology
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Zach Menne
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA; Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA
| | - Vasilis C Pliasas
- Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Richard W Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA; Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA
| | - Sheniqua Glover
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Constantinos S Kyriakis
- Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA; Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA.
| |
Collapse
|
6
|
Ma J, Wang H, Zheng X, Wu H, Yang S, Xia X. Western equine encephalitis virus virus-like particles from an insect cell-baculovirus system elicit the strong immune responses in mice. Biotechnol J 2021; 16:e2100008. [PMID: 34176228 DOI: 10.1002/biot.202100008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/07/2022]
Abstract
Western equine encephalitis virus (WEEV) causes lethal encephalitis in humans and equines, and it poses a serious public health threat in many countries. Therefore, the development of an efficient vaccine remains an important challenge for the prevention of WEEV infection. This study presents the first description of WEEV virus-like particles (VLPs) generated from insect cells using recombinant baculoviruses. WEEV VLPs with 206 adjuvant could trigger a strong cellular immune response; increase the levels of IL-2, IL-4 and IFN-γ; and induce a high level of neutralizing antibodies against WEEV in mice. These data showed that the insect cell-baculovirus system is suitable for the production of WEEV VLPs and that these VLPs could elicit the strong immunogenicity in mice. These results suggest a new, nonreplicating, and effective vaccine candidate against WEEV infection.
Collapse
Affiliation(s)
- JinZhu Ma
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China.,College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - HuaLei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - XueXing Zheng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - HongXia Wu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - SongTao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - XianZhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| |
Collapse
|
7
|
A knockout cell library of GPI biosynthetic genes for functional studies of GPI-anchored proteins. Commun Biol 2021; 4:777. [PMID: 34162996 PMCID: PMC8222316 DOI: 10.1038/s42003-021-02337-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/19/2021] [Indexed: 11/08/2022] Open
Abstract
Over 100 kinds of proteins are expressed as glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) on the cell surface in mammalian cells. GPI-APs possess unique properties in terms of their intracellular trafficking and association with lipid rafts. Although it is clear that GPI-APs play critical roles in various biological phenomena, it is poorly understood how the GPI moiety contributes to these mechanisms. More than 30 genes are involved in the correct biosynthesis of GPI-APs. We here constructed a cell library in which 32 genes involved in GPI biosynthesis were knocked out in human embryonic kidney 293 cells. Using the cell library, the surface expression and sensitivity to phosphatidylinositol-specific phospholipase C of GPI-APs were analyzed. Furthermore, we identified structural motifs of GPIs that are recognized by a GPI-binding toxin, aerolysin. The cell-based GPI-knockout library could be applied not only to basic researches, but also to applications and methodologies related to GPI-APs.
Collapse
|
8
|
Lei X, Cai X, Yang Y. Genetic engineering strategies for construction of multivalent chimeric VLPs vaccines. Expert Rev Vaccines 2020; 19:235-246. [PMID: 32133886 DOI: 10.1080/14760584.2020.1738227] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Over the past two decades, virus-like particles (VLPs) have been developed as a new generation of vaccines against viral infections. Based on VLPs, chimeric VLPs (chi-VLPs) have been generated through genetic modifications or chemical couplings. For construction of multivalent chi-VLPs vaccines, multiple genetic engineering strategies are continuously being developed. Thus, it is important to provide a summary as reference for researchers in this field.Areas covered: The representative studies on the genetic engineered multivalent chi-VLPs are summarized and mainly focused on chimeric capsid VLPs and chimeric enveloped VLPs. The advantages and limitations of each strategy are also discussed at last, as well as opinions on platform choice and future directions of eVLPs vaccines.Expert opinion: The design of multivalent chi-VLPs vaccines needs to meet the following specifications: 1) the incorporated antigens are suggested to display on the exposed surface of chi-VLPs and do not have excessive adverse effects on the stability of chi-VLPs; 2) the chi-VLPs should elicit protective antibodies against the incorporated antigen as well as the source virus of VLPs. However, there is no requirement of retaining the antigenicity of VLPs when using VLPs solely as carriers for antigens display or drug delivery.
Collapse
Affiliation(s)
- Xinnuo Lei
- Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Research Center of Reverse Vaccinology (RCRV), Hunan Agricultural University, Changsha, Hunan, China.,Laboratory of Functional Proteomics (LFP), College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yi Yang
- Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Research Center of Reverse Vaccinology (RCRV), Hunan Agricultural University, Changsha, Hunan, China.,Laboratory of Functional Proteomics (LFP), College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| |
Collapse
|
9
|
Dong Y, Ma T, Zhang X, Ying Q, Han M, Zhang M, Yang R, Li Y, Wang F, Liu R, Wu X. Incorporation of CD40 ligand or granulocyte-macrophage colony stimulating factor into Hantaan virus (HTNV) virus-like particles significantly enhances the long-term immunity potency against HTNV infection. J Med Microbiol 2019; 68:480-492. [PMID: 30657443 DOI: 10.1099/jmm.0.000897] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Hantavirus infections cause severe haemorrhagic fever with renal syndrome (HFRS) in humans and are associated with high fatality rates. In 2017, numerous outbreaks were reported in China and Germany. This represents a significant public-healthcare issue with no effective HFRS vaccines that offer a long-term immune response. In this study, we investigated the long-term humoral and cellular immune responses and protective immunity of Hantaan virus (HTNV) granulocyte-macrophage colony stimulating factor (GM-CSF) and CD40 ligand (CD40L) virus-like particles (VLPs) in mice. METHODOLOGY GM-CSF and CD40L VLPs were constructed via co-transfection of pCI-S and pCI-M-CD40L, and pCI-S and pCI-M-GM-CSF, into dihydrofolatereductase (dhfr)-deficient Chinese hamster ovary cells, respectively. Mice were immunized with HTNV VLPs 2 weeks apart. The animals were challenged 6 months after immunization. Specific and neutralizing antibodies were assessed by ELISA; IFN-γ was measured by enzyme-linked immunospot (ELISpot) assay and effectiveness by cytotoxic T lymphocyte (CTL) cytotoxicity assays. Nucleic acid loads of HTNV were tested by quantitative real-time PCR and viral antigen was detected via indirect ELISA. Pathological alterations were detected via haematoxylin-eosin staining. RESULTS GM-CSF and CD40L VLPs provided stable, long-term protection with a high titre of neutralizing antibody in mice 6 months after immunization. Furthermore, VLPs increased HTNV-specific cellular immune responses via higher expression of IFN-γ and CTL responses. HTNV challenge assay results showed long-term protection against HFRS. No significant pathological alteration was observed in the organs of mice after immunization. CONCLUSION This is, to the best of our knowledge, the first report demonstrating the long-term potency of HTNV VLP vaccines against HTNV infection and offers new insights into HTNV vaccine development.
Collapse
Affiliation(s)
- Yuhang Dong
- 1Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Tiejun Ma
- 1Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Xiaoxiao Zhang
- 1Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Qikang Ying
- 1Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Mingwei Han
- 2No. 1 Cadet Brigade, Fourth Military Medical University, Xi'an, PR China
| | - Muqi Zhang
- 2No. 1 Cadet Brigade, Fourth Military Medical University, Xi'an, PR China
| | - Rongjin Yang
- 2No. 1 Cadet Brigade, Fourth Military Medical University, Xi'an, PR China
| | - Yuan Li
- 2No. 1 Cadet Brigade, Fourth Military Medical University, Xi'an, PR China
| | - Fang Wang
- 1Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Rongrong Liu
- 1Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Xingan Wu
- 1Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, PR China
| |
Collapse
|
10
|
Virus-Like Particles-Based Mucosal Nanovaccines. NANOVACCINES 2019. [PMCID: PMC7120988 DOI: 10.1007/978-3-030-31668-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Virus-like particles (VLPs) are protein complexes that resemble a virus and constitute highly immunogenic entities as they mimic the pathogen at an important degree. Among nanovaccines, those based on VLPs are the most successful thus far with some formulations already commercialized (e.g., those against hepatitis B and E viruses and human papillomavirus). This chapter highlights the advantages of VLPs-based vaccines, describing approaches for their design and transmittance of the state of the art for mucosal VLPs-based vaccines development. Several candidates have been produced in insect cells, plants, and E. coli and mammalian cells; they have been mainly evaluated in i.n. and oral immunization schemes. i.n. vaccines against the influenza virus and the Norwalk virus are the most advanced applications. For the latter, i.n. formulations are under clinical evaluation. Perspectives for the field comprise the expansion of the use of low-cost platforms such as plants and bacteria, the development of multiepitopic/multivalent vaccines, and computationally designed VLPs. Mucosal VLPs-based vaccines stand as a major promising approach in vaccinology and the initiation of more clinical trials is envisaged in a short time.
Collapse
|
11
|
Keshavarz M, Mirzaei H, Salemi M, Momeni F, Mousavi MJ, Sadeghalvad M, Arjeini Y, Solaymani-Mohammadi F, Sadri Nahand J, Namdari H, Mokhtari-Azad T, Rezaei F. Influenza vaccine: Where are we and where do we go? Rev Med Virol 2018; 29:e2014. [PMID: 30408280 DOI: 10.1002/rmv.2014] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022]
Abstract
The alarming rise of morbidity and mortality caused by influenza pandemics and epidemics has drawn attention worldwide since the last few decades. This life-threatening problem necessitates the development of a safe and effective vaccine to protect against incoming pandemics. The currently available flu vaccines rely on inactivated viral particles, M2e-based vaccine, live attenuated influenza vaccine (LAIV) and virus like particle (VLP). While inactivated vaccines can only induce systemic humoral responses, LAIV and VLP vaccines stimulate both humoral and cellular immune responses. Yet, these vaccines have limited protection against newly emerging viral strains. These strains, however, can be targeted by universal vaccines consisting of conserved viral proteins such as M2e and capable of inducing cross-reactive immune response. The lack of viral genome in VLP and M2e-based vaccines addresses safety concern associated with existing attenuated vaccines. With the emergence of new recombinant viral strains each year, additional effort towards developing improved universal vaccine is warranted. Besides various types of vaccines, microRNA and exosome-based vaccines have been emerged as new types of influenza vaccines which are associated with new and effective properties. Hence, development of a new generation of vaccines could contribute to better treatment of influenza.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Salemi
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Fatemeh Momeni
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Mousavi
- Department of Immunology and Allergy, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.,Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Sadeghalvad
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Arjeini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Solaymani-Mohammadi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haideh Namdari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Ren Z, Zhao Y, Liu J, Ji X, Meng L, Wang T, Sun W, Zhang K, Sang X, Yu Z, Li Y, Feng N, Wang H, Yang S, Yang Z, Wang Z, Gao Y, Xia X. Inclusion of membrane-anchored LTB or flagellin protein in H5N1 virus-like particles enhances protective responses following intramuscular and oral immunization of mice. Vaccine 2018; 36:5990-5998. [PMID: 30172635 DOI: 10.1016/j.vaccine.2018.08.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/10/2018] [Accepted: 08/18/2018] [Indexed: 12/24/2022]
Abstract
We previously demonstrated that intramuscular immunization with virus-like particles (VLPs) composed of the haemagglutinin (HA), neuraminidase (NA), and matrix (M1) proteins of A/meerkat/Shanghai/SH-1/2012 (clade 2.3.2.1) protected mice from lethal challenge with viruses from other H5 HPAI clades. The inclusion of additional proteins that can serve as immunological adjuvants in VLPs may enhance adaptive immune responses following vaccination, and oral vaccines may represent the safest choice. Here, we report the generation of H5N1 VLPs composed of the viral HA, NA, and M1 proteins and membrane-anchored forms of the Escherichia coli heat-labile enterotoxin B subunit protein (LTB) or the Toll-like receptor 5 ligand flagellin (Flic). Mice intramuscularly or orally immunized with VLPs containing LTB or Flic generated greater humoural and cellular immune responses than those administered H5N1 VLPs without LTB or Flic. Intramuscular immunization with VLPs protected mice from lethal challenge with homologous or heterologous H5N1 viruses irrespective of whether the VLPs additionally included LTB or Flic. In contrast, oral immunization of mice with LTB- or Flic-VLPs conferred substantial protection against lethal challenge with both homologous and heterologous H5N1 influenza viruses, whereas mice immunized orally with VLPs lacking LTB and Flic universally succumbed to infection. Mice immunized orally with LTB- or Flic-VLPs showed 10-fold higher virus-specific IgG titres than mice immunized with H5N1-VLPs lacking LTB or Flic. Collectively, these results indicate that the inclusion of immunostimulatory proteins, such as LTB and Flic, in VLP-based vaccines may represent a promising new approach for the control of current H5N1 HPAI outbreaks by eliciting higher humoural and cellular immune responses and conferring improved cross-clade protection.
Collapse
Affiliation(s)
- Zhiguang Ren
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China.
| | - Yongkun Zhao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Jing Liu
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianliang Ji
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Lingnan Meng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Tiecheng Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Weiyang Sun
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Kun Zhang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Xiaoyu Sang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Zhijun Yu
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Yuanguo Li
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Na Feng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Hualei Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Songtao Yang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Zhengyan Yang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China
| | - Zhizeng Wang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China
| | - Yuwei Gao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| | - Xianzhu Xia
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| |
Collapse
|
13
|
Liu J, Ren Z, Wang H, Zhao Y, Wilker PR, Yu Z, Sun W, Wang T, Feng N, Li Y, Wang H, Ji X, Li N, Yang S, He H, Qin C, Gao Y, Xia X. Influenza virus-like particles composed of conserved influenza proteins and GPI-anchored CCL28/GM-CSF fusion proteins enhance protective immunity against homologous and heterologous viruses. Int Immunopharmacol 2018; 63:119-128. [PMID: 30081250 DOI: 10.1016/j.intimp.2018.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/01/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022]
Abstract
Influenza viruses cause significant morbidity and mortality and pose a substantial threat to public health. Vaccination represents the principle means of preventing influenza virus infection. Current vaccine approaches are hindered by the need to routinely reformulate vaccine compositions in an effort to account for the progressive antigenic changes that occur as influenza viruses circulate in the human population. In this study, we evaluated chimeric virus-like particle (cVLP) vaccines containing conserved elements of influenza proteins (HL5M2e (HA stem gene with 5M2e gene inserted) and NP), with or without glycosylphosphatidylinositol-anchored CCL28 (GPI-CCL28) and/or GM-CSF (GPI-GM-CSF) fusion proteins as molecular adjuvants. cVLPs elicited strong humoral and cellular immune responses against homologous and heterologous viruses, and improved survival following lethal challenge with both homologous and heterologous viruses. Inclusion of GPI-anchored adjuvants in cVLP vaccines augmented the generation of influenza-specific humoral and cellular immune responses in mice in comparison to the non-adjuvanted cVLP vaccines. VLPs containing GPI-anchored adjuvants reduced morbidity and improved survival to lethal challenge with homologous and heterologous influenza viruses. This work suggests that VLP vaccines incorporating conserved influenza virus proteins and GPI-anchored molecular adjuvants may serve as a platform for a broadly protective "universal" influenza vaccine.
Collapse
Affiliation(s)
- Jing Liu
- Comparative Medicine Center, Peking Union Medical College (PUMC) and Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Zhiguang Ren
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medicine, Kaifeng 475004, China; Henan University, Kaifeng, Hennan Province, China
| | - Hongmei Wang
- Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Disease Research Center, College of Life Science, Shandong Normal University, Shandong Province 250014, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Peter R Wilker
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Zhijun Yu
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, Jinan 250023, China
| | - Weiyang Sun
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Yuanguo Li
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Hualei Wang
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Xianliang Ji
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Hongbin He
- Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Disease Research Center, College of Life Science, Shandong Normal University, Shandong Province 250014, China
| | - Chuan Qin
- Comparative Medicine Center, Peking Union Medical College (PUMC) and Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| | - Xianzhu Xia
- Comparative Medicine Center, Peking Union Medical College (PUMC) and Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; Key Laboratory of Jilin Province for Zoonosis, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province 130122, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
14
|
Incorporation of CD40 ligand enhances the immunogenicity of tumor‑associated calcium signal transducer 2 virus‑like particles against lung cancer. Int J Mol Med 2018; 41:3671-3679. [PMID: 29568866 DOI: 10.3892/ijmm.2018.3570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/05/2018] [Indexed: 11/05/2022] Open
Abstract
The cell surface glycoprotein Trop‑2 is overexpressed in various types of cancer, including in lung cancer, and has recently been used as an effective immunotherapeutic target. CD40 ligand (CD40L), a tumor necrosis factor superfamily member, is a promising immune adjuvant. Human immunodeficiency virus (HIV) gag‑based virus‑like particles (VLPs) are highly immunogenic, and foreign antigens can be incorporated onto their membrane envelope for cancer vaccine development. In the present study, a HIV gag‑based VLP strategy and Bac‑to‑Bac system were utilized to construct Trop‑2, CD40L and gag recombinant baculoviruses, which were then used to infect TN5 cells in order to form Trop‑2 VLPs or Trop‑2‑CD40L VLPs. These VLPs were characterized using transmission electron microscopy and western blot analysis methods. VLPs incorporating murine Trop‑2 only or incorporating Trop‑2 and CD40L were used to immunize C57BL/6 mice. Immunized mice demonstrated high humoral and cellular immunity responses, whereas the Trop‑2‑CD40L VLPs led to higher immune responses in comparison with Trop‑2 only VLPs. Immunization with Trop‑2‑CD40L VLPs also reduced tumor growth more effectively compared with Trop‑2 VLPs. Furthermore, Trop‑2‑CD40L VLP immunization increased the survival rate of Lewis tumor‑bearing mice more significantly when compared with Trop‑2 only VLPs. In conclusion, the present study provided a novel vaccine design by combination of a tumor antigen and an immune adjuvant based on a VLP strategy, which may be potentially applied as an alternative immunotherapeutic option in the treatment of lung cancer.
Collapse
|
15
|
Vzorov AN, Uryvaev LV. Requirements for the Induction of Broadly Neutralizing Antibodies against HIV-1 by Vaccination. Mol Biol 2017. [DOI: 10.1134/s0026893317060176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
16
|
Cheng LF, Wang F, Zhang L, Yu L, Ye W, Liu ZY, Ying QK, Wu XA, Xu ZK, Zhang FL. Incorporation of GM-CSF or CD40L Enhances the Immunogenicity of Hantaan Virus-Like Particles. Front Cell Infect Microbiol 2016; 6:185. [PMID: 28066721 PMCID: PMC5167722 DOI: 10.3389/fcimb.2016.00185] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/29/2016] [Indexed: 12/22/2022] Open
Abstract
A safe and effective Hantaan virus (HTNV) vaccine is highly desirable because HTNV causes an acute and often fatal disease (hemorrhagic fever with renal syndrome, HFRS). Since the immunity of the inactivated vaccine is weak and the safety is poor, HTNV virus-like particles (VLPs) offer an attractive and safe alternative. These particles lack the viral genome but are perceived by the immune system as virus particles. We hypothesized that adding immunostimulatory signals to VLPs would enhance their efficacy. To accomplish this enhancement, we generated chimeric HTNV VLPs containing glycosylphosphatidylinositol (GPI)-anchored granulocyte macrophage colony-stimulating factor (GM-CSF) or CD40 ligand (CD40L) and investigated their biological activity in vitro. The immunization of mice with chimeric HTNV VLPs containing GM-CSF or CD40L induced stronger humoral immune responses and cellular immune responses compared to the HTNV VLPs and Chinese commercial inactivated hantavirus vaccine. Chimeric HTNV VLPs containing GM-CSF or CD40L also protected mice from an HTNV challenge. Altogether, our results suggest that anchoring immunostimulatory molecules into HTNV VLPs can be a potential approach for the control and prevention of HFRS.
Collapse
Affiliation(s)
- Lin-Feng Cheng
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Fang Wang
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Liang Zhang
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Lan Yu
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Wei Ye
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Zi-Yu Liu
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Qi-Kang Ying
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Xing-An Wu
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Zhi-Kai Xu
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| | - Fang-Lin Zhang
- Department of Microbiology, Fourth Military Medical University Xi'an, China
| |
Collapse
|
17
|
Heider S, Dangerfield JA, Metzner C. Biomedical applications of glycosylphosphatidylinositol-anchored proteins. J Lipid Res 2016; 57:1778-1788. [PMID: 27542385 PMCID: PMC5036375 DOI: 10.1194/jlr.r070201] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Indexed: 01/13/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) use a unique posttranslational modification to link proteins to lipid bilayer membranes. The anchoring structure consists of both a lipid and carbohydrate portion and is highly conserved in eukaryotic organisms regarding its basic characteristics, yet highly variable in its molecular details. The strong membrane targeting property has made the anchors an interesting tool for biotechnological modification of lipid membrane-covered entities from cells through extracellular vesicles to enveloped virus particles. In this review, we will take a closer look at the mechanisms and fields of application for GPI-APs in lipid bilayer membrane engineering and discuss their advantages and disadvantages for biomedicine.
Collapse
Affiliation(s)
- Susanne Heider
- Institute of Virology, University of Veterinary Medicine, 1210 Vienna, Austria
| | | | - Christoph Metzner
- Institute of Virology, University of Veterinary Medicine, 1210 Vienna, Austria.
| |
Collapse
|
18
|
Mohan T, Kim J, Berman Z, Wang S, Compans RW, Wang BZ. Co-delivery of GPI-anchored CCL28 and influenza HA in chimeric virus-like particles induces cross-protective immunity against H3N2 viruses. J Control Release 2016; 233:208-19. [PMID: 27178810 DOI: 10.1016/j.jconrel.2016.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 12/20/2022]
Abstract
Influenza infection typically initiates at respiratory mucosal surfaces. Induction of immune responses at the sites where pathogens initiate replication is crucial for the prevention of infection. We studied the adjuvanticity of GPI-anchored CCL28 co-incorporated with influenza HA-antigens in chimeric virus-like particles (cVLPs), in boosting strong protective immune responses through an intranasal (i.n.) route in mice. We compared the immune responses to that from influenza VLPs without CCL28, or physically mixed with soluble CCL28 at systemic and various mucosal compartments. The cVLPs containing GPI-CCL28 showed in-vitro chemotactic activity towards spleen and lung cells expressing CCR3/CCR10 chemokine receptors. The cVLPs induced antigen specific endpoint titers and avidity indices of IgG in sera and IgA in tracheal, lung, and intestinal secretions, significantly higher (4-6 fold) than other formulations. Significantly higher (3-5 fold) hemagglutination inhibition titers and high serum neutralization against H3N2 viruses were also detected with CCL28-containing VLPs compared to other groups. The CCL28-containing VLPs showed complete and 80% protection, when vaccinated animals were challenged with A/Aichi/2/1968/H3N2 (homologous) and A/Philippines/2/1982/H3N2 (heterologous) viruses, respectively. Thus, GPI-anchored CCL28 in influenza VLPs act as a strong immunostimulator at both systemic and mucosal sites, boosting significant cross-protection in animals against heterologous viruses across a large distance.
Collapse
Affiliation(s)
- Teena Mohan
- Department of Microbiology and Immunology, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA 30322, USA; Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Jongrok Kim
- Department of Microbiology and Immunology, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA 30322, USA
| | - Zachary Berman
- Department of Microbiology and Immunology, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA 30322, USA; Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Shelly Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA 30322, USA
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA 30322, USA
| | - Bao-Zhong Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA 30322, USA; Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
19
|
Vassilieva EV, Kalluri H, McAllister D, Taherbhai MT, Esser ES, Pewin WP, Pulit-Penaloza JA, Prausnitz MR, Compans RW, Skountzou I. Improved immunogenicity of individual influenza vaccine components delivered with a novel dissolving microneedle patch stable at room temperature. Drug Deliv Transl Res 2016; 5:360-71. [PMID: 25895053 DOI: 10.1007/s13346-015-0228-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Prevention of seasonal influenza epidemics and pandemics relies on widespread vaccination coverage to induce protective immunity. In addition to a good antigenic match with the circulating viruses, the effectiveness of individual strains represented in the trivalent vaccines depends on their immunogenicity. In this study, we evaluated the immunogenicity of H1N1, H3N2, and B seasonal influenza virus vaccine strains delivered individually with a novel dissolving microneedle patch and the stability of this formulation during storage at 25 °C. Our data demonstrate that all strains retained their antigenic activity after incorporation in the dissolving patches as measured by single radial diffusion (SRID) assay and immune responses to vaccination in BALB/c mice. After a single immunization, all three antigens delivered with microneedle patches induced superior neutralizing antibody titers compared to intramuscular immunization. Cutaneous antigen delivery was especially beneficial for the less immunogenic B strain. Mice immunized with dissolving microneedle patches encapsulating influenza A/Brisbane/59/07 (H1N1) vaccine were fully protected against lethal challenge by homologous mouse-adapted influenza virus. All vaccine components retained activity during storage at room temperature for at least 3 months as measured in vitro by SRID assay and in vivo by mouse immunization studies. Our data demonstrate that dissolving microneedle patches are a promising advance for influenza cutaneous vaccination due to improved immune responses using less immunogenic influenza antigens and enhanced stability.
Collapse
Affiliation(s)
- Elena V Vassilieva
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Vzorov AN, Compans RW. VLP vaccines and effects of HIV-1 Env protein modifications on their antigenic properties. Mol Biol 2016. [DOI: 10.1134/s0026893316030110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
21
|
Shirbaghaee Z, Bolhassani A. Different applications of virus-like particles in biology and medicine: Vaccination and delivery systems. Biopolymers 2016; 105:113-32. [PMID: 26509554 PMCID: PMC7161881 DOI: 10.1002/bip.22759] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/25/2015] [Accepted: 10/25/2015] [Indexed: 12/17/2022]
Abstract
Virus-like particles (VLPs) mimic the whole construct of virus particles devoid of viral genome as used in subunit vaccine design. VLPs can elicit efficient protective immunity as direct immunogens compared to soluble antigens co-administered with adjuvants in several booster injections. Up to now, several prokaryotic and eukaryotic systems such as insect, yeast, plant, and E. coli were used to express recombinant proteins, especially for VLP production. Recent studies are also generating VLPs in plants using different transient expression vectors for edible vaccines. VLPs and viral particles have been applied for different functions such as gene therapy, vaccination, nanotechnology, and diagnostics. Herein, we describe VLP production in different systems as well as its applications in biology and medicine.
Collapse
Affiliation(s)
- Zeinab Shirbaghaee
- Department of Hepatitis and AIDSPasteur Institute of IranTehranIran
- Department of Immunology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Azam Bolhassani
- Department of Hepatitis and AIDSPasteur Institute of IranTehranIran
| |
Collapse
|
22
|
Feng H, Zhang H, Deng J, Wang L, He Y, Wang S, Seyedtabaei R, Wang Q, Liu L, Galipeau J, Compans RW, Wang BZ. Incorporation of a GPI-anchored engineered cytokine as a molecular adjuvant enhances the immunogenicity of HIV VLPs. Sci Rep 2015; 5:11856. [PMID: 26150163 PMCID: PMC4493578 DOI: 10.1038/srep11856] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 05/22/2015] [Indexed: 12/15/2022] Open
Abstract
HIV vaccines should elicit immune responses at both the mucosal portals of entry to block transmission and systemic compartments to clear disseminated viruses. Co-delivery of mucosal adjuvants has been shown to be essential to induce effective mucosal immunity by non-replicating vaccines. A novel cytokine, GIFT4, engineered by fusing GM-CSF and interleukin-4, was previously found to simulate B cell proliferation and effector function. Herein a membrane-anchored form of GIFT4 was constructed by fusing a glycolipid (GPI)-anchoring sequence and incorporated into Env-enriched HIV virus-like particles (VLPs) as a molecular adjuvant. Guinea pigs were immunized with the resulting HIV VLPs through an intramuscular priming-intranasal boosting immunization route. The GIFT4-containing VLPs induced higher levels of systemic antibody responses with significantly increased binding avidity and improved neutralizing breadth and potency to a panel of selected strains, as well as higher levels of IgG and IgA at several mucosal sites. Thus, the novel GPI-GIFT4-containging VLPs have the potential to be developed into a prophylactic HIV vaccine. Incorporation of GPI-anchored GIFT4 into VLPs as a molecular adjuvant represents a novel approach to increase their immunogenicity.
Collapse
Affiliation(s)
- Hao Feng
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Han Zhang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Jiusheng Deng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Li Wang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Yuan He
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Shelly Wang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Roheila Seyedtabaei
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Qing Wang
- Department of Bioengineering, Henan University of Technology, Zhengzhou 450052, China
| | - Laiting Liu
- Department of Bioengineering, Henan University of Technology, Zhengzhou 450052, China
| | - Jacques Galipeau
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Bao-Zhong Wang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
23
|
Rynda-Apple A, Patterson DP, Douglas T. Virus-like particles as antigenic nanomaterials for inducing protective immune responses in the lung. Nanomedicine (Lond) 2015; 9:1857-68. [PMID: 25325241 DOI: 10.2217/nnm.14.107] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The lung is a major entry point for many of the most detrimental pathogens to human health. The onslaught of pathogens encountered by the lung is counteracted by protective immune responses that are generated locally, which can be stimulated through vaccine strategies to prevent pathogen infections. Here, we discuss the use of virus-like particles (VLPs), nonpathogen derivatives of viruses or protein cage structures, to construct new vaccines exploiting the lung as a site for immunostimulation. VLPs are unique in their ability to be engineered with near molecular level detail and knowledge of their composition and structure. A summary of research in developing VLP-based vaccines for the lung is presented that suggests promising results for future vaccine development.
Collapse
|
24
|
Abstract
Virus-like particles (VLPs) are an effective means of establishing both prophylactic and therapeutic immunity against their source virus or heterologous antigens. The particulate nature and repetitive structure of VLPs makes them ideal for stimulating potent immune responses. Epitopes delivered by VLPs can be presented on MHC-II for stimulation of a humoral immune response, or cross-presented onto MHC-I leading to cell-mediated immunity. VLPs as particulate subunit vaccine carriers are showing promise in preclinical and clinical trials for the treatment of many conditions including cancer, autoimmunity, allergies and addiction. Supporting the delivery of almost any form of antigenic material, VLPs are ideal candidate vectors for development of future vaccines.
Collapse
|
25
|
Wang S, Liu H, Zhang X, Qian F. Intranasal and oral vaccination with protein-based antigens: advantages, challenges and formulation strategies. Protein Cell 2015; 6:480-503. [PMID: 25944045 PMCID: PMC4491048 DOI: 10.1007/s13238-015-0164-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/10/2015] [Indexed: 02/06/2023] Open
Abstract
Most pathogens initiate their infections at the human mucosal surface. Therefore, mucosal vaccination, especially through oral or intranasal administration routes, is highly desired for infectious diseases. Meanwhile, protein-based antigens provide a safer alternative to the whole pathogen or DNA based ones in vaccine development. However, the unique biopharmaceutical hurdles that intranasally or orally delivered protein vaccines need to overcome before they reach the sites of targeting, the relatively low immunogenicity, as well as the low stability of the protein antigens, require thoughtful and fine-tuned mucosal vaccine formulations, including the selection of immunostimulants, the identification of the suitable vaccine delivery system, and the determination of the exact composition and manufacturing conditions. This review aims to provide an up-to-date survey of the protein antigen-based vaccine formulation development, including the usage of immunostimulants and the optimization of vaccine delivery systems for intranasal and oral administrations.
Collapse
Affiliation(s)
- Shujing Wang
- Department of Pharmacology and Pharmaceutical Sciences, School of Medicine and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | | | | | | |
Collapse
|
26
|
Patel JM, Kim MC, Vartabedian VF, Lee YN, He S, Song JM, Choi HJ, Yamanaka S, Amaram N, Lukacher A, Montemagno CD, Compans RW, Kang SM, Selvaraj P. Protein transfer-mediated surface engineering to adjuvantate virus-like nanoparticles for enhanced anti-viral immune responses. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1097-107. [PMID: 25752855 DOI: 10.1016/j.nano.2015.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/28/2015] [Accepted: 02/16/2015] [Indexed: 12/29/2022]
Abstract
UNLABELLED Recombinant virus-like nanoparticles (VLPs) are a promising nanoparticle platform to develop safe vaccines for many viruses. Herein, we describe a novel and rapid protein transfer process to enhance the potency of enveloped VLPs by decorating influenza VLPs with exogenously added glycosylphosphatidylinositol-anchored immunostimulatory molecules (GPI-ISMs). With protein transfer, the level of GPI-ISM incorporation onto VLPs is controllable by varying incubation time and concentration of GPI-ISMs added. ISM incorporation was dependent upon the presence of a GPI-anchor and incorporated proteins were stable and functional for at least 4weeks when stored at 4°C. Vaccinating mice with GPI-granulocyte macrophage colony-stimulating factor (GM-CSF)-incorporated-VLPs induced stronger antibody responses and better protection against a heterologous influenza virus challenge than unmodified VLPs. Thus, VLPs can be enriched with ISMs by protein transfer to increase the potency and breadth of the immune response, which has implications in developing effective nanoparticle-based vaccines against a broad spectrum of enveloped viruses. FROM THE CLINICAL EDITOR The inherent problem with current influenza vaccines is that they do not generate effective cross-protection against heterologous viral strains. In this article, the authors described the development of virus-like nanoparticles (VLPs) as influenza vaccines with enhanced efficacy for cross-protection, due to an easy protein transfer modification process.
Collapse
Affiliation(s)
- Jaina M Patel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Vincent F Vartabedian
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yu-Na Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sara He
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jae-Min Song
- Department of Global Medical Science, Sungshin Women's University, Seoul, Korea
| | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
| | - Satoshi Yamanaka
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nikhil Amaram
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Anna Lukacher
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Carlo D Montemagno
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
27
|
Qi Y, Kang H, Zheng X, Wang H, Gao Y, Yang S, Xia X. Incorporation of membrane-anchored flagellin or Escherichia coli heat-labile enterotoxin B subunit enhances the immunogenicity of rabies virus-like particles in mice and dogs. Front Microbiol 2015; 6:169. [PMID: 25784906 PMCID: PMC4347500 DOI: 10.3389/fmicb.2015.00169] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/13/2015] [Indexed: 01/20/2023] Open
Abstract
Rabies remains an important worldwide public health threat, so safe, effective, and affordable vaccines are still being sought. Virus-like particle-based vaccines targeting various viral pathogens have been successfully produced, licensed, and commercialized. Here, we designed and constructed two chimeric rabies virus-like particles (cRVLPs) containing rabies virus (RABV) glycoprotein (G), matrix (M) protein, and membrane-anchored flagellin (EVLP-F) or Escherichia coli heat-labile enterotoxin B subunit (EVLP-L) as molecular adjuvants to enhance the immune response against rabies. The immunogenicity and potential of cRVLPs as novel rabies vaccine were evaluated by intramuscular vaccination in mouse and dog models. Mouse studies demonstrated that both EVLP-F and EVLP-L induced faster and larger virus-neutralizing antibodies (VNAs) responses and elicited greater numbers of CD4+ and CD8+ T cells secreting IFN-γ or IL-4 compared with a standard rabies VLP (sRVLP) containing only G and M. Moreover, cRVLPs recruited and/or activated more B cells and dendritic cells in inguinal lymph nodes. EVLP-F induced a strong, specific IgG2a response but not an IgG1 response, suggesting the activation of Th1 class immunity; in contrast, Th2 class immunity was observed with EVLP-L. The significantly enhanced humoral and cellular immune responses induced by cRVLPs provided complete protection against lethal challenge with RABV. Most importantly, dogs vaccinated with EVLP-F or EVLP-L exhibited increased VNA titers in sera and enhanced IFN-γ and IL-4 secretion from peripheral blood mononuclear cells. Taken together, these results illustrate that when incorporated into sRVLP, membrane-anchored flagellin, and heat-labile enterotoxin B subunit possess strong adjuvant activity. EVLP-F and EVLP-L induce significantly enhanced RABV-specific humoral and cellular immune responses in both mouse and dog. Therefore, these cRVLPs may be developed as safe and more efficacious rabies vaccine candidate for animals.
Collapse
Affiliation(s)
- Yinglin Qi
- College of Veterinary Medicine, Jilin University Changchun, China ; Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Hongtao Kang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China ; College of Veterinary Medicine, South China Agricultural University Guangzhou, China
| | - Xuexing Zheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Hualei Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Yuwei Gao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Songtao Yang
- College of Veterinary Medicine, Jilin University Changchun, China ; Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin University Changchun, China ; Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| |
Collapse
|
28
|
Yang DK, Nah JJ, Kim HH, Song JY. Inactivated genotype 1 Japanese encephalitis vaccine for swine. Clin Exp Vaccine Res 2014; 3:212-9. [PMID: 25003095 PMCID: PMC4083074 DOI: 10.7774/cevr.2014.3.2.212] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 04/14/2014] [Accepted: 04/20/2014] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Japanese encephalitis is a reproductive disorder caused by Japanese encephalitis virus (JEV) in swine. Recent genotype (G) shift phenomenon (G3 to G1) in the Asia-wide has posed a challenge for proper prevention by the current vaccine strain. Thus, new kinds of JEV G1 vaccines with enhanced immunogenicity have been required for pigs. MATERIALS AND METHODS Recombinant porcine granulocyte monocyte-colony stimulating factor (reporGM-CSF) protein was expressed in Spodoptera frugiperda (Sf-9) cells using baculovirus expression system. Two kinds of trials with inactivated JEV vaccines containing IMS1313 adjuvant (Seppic, France) were prepared with or without reporGM-CSF protein. Safety and immunogenicity of the pigs inoculated with the JEV vaccines via intramuscular route was evaluated for 28 days after inoculation. RESULTS Mice, guinea pigs, and fattening pigs inoculated with the inactivated vaccine showed no signs for 14 and 21 days. Both hemagglutination inhibition and plaque reduction neutralizing antibody titers were significantly higher in pigs immunized with the vaccine containing reporGM-CSF protein after boosting. However, on the side of vaccine efficacy, most mice (87%) immunized with the inactivated JEV vaccine survived after virulent JEV challenge. Whereas the group with the vaccine containing reporGM-CSF protein showed lower protective effects than the vaccine alone for the biological activity of the GM-CSF depending on species specific. CONCLUSION Our data indicate that animals inoculated with the JEV vaccines was safe and pigs inoculated with inactivated JEV vaccine containing reporGM-CSF protein showed higher humoral immune responses than that of inactivated JEV vaccine without reporGM-CSF protein.
Collapse
Affiliation(s)
- Dong-Kun Yang
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| | - Jin-Ju Nah
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| | - Ha-Hyun Kim
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| | - Jae-Young Song
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| |
Collapse
|
29
|
Ma J, Wang H, Zheng X, Xue X, Wang B, Wu H, Zhang K, Fan S, Wang T, Li N, Zhao Y, Gao Y, Yang S, Xia X. CpG/Poly (I:C) mixed adjuvant priming enhances the immunogenicity of a DNA vaccine against eastern equine encephalitis virus in mice. Int Immunopharmacol 2014; 19:74-80. [PMID: 24440303 DOI: 10.1016/j.intimp.2014.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 01/06/2014] [Accepted: 01/06/2014] [Indexed: 12/26/2022]
Abstract
Eastern equine encephalitis virus (EEEV) poses a serious public health threat in many countries. Therefore, developing efficient vaccine against EEEV remains an important challenge in the field of disease control. To identify immunogenic proteins in EEEV, we constructed an expression vector containing the protein coding genes C, E3, E2, 6k, and E1 (pcDNA3.1-C-E). After verifying the target gene expression in 293 T cells, we immunized BALB/c mice with the pcDNA3.1-C-E vector as a DNA vaccine in conjunction with either CpG or poly (I:C) or a mixture of both adjuvants and monitored various aspects of the immune response. After two immunizations, the mice vaccinated with antigen plus mixed CpG/poly (I:C) adjuvant exhibited significantly stronger IFN-gamma responses and generated high-level CD4(+) cell responses for the cytokines IL-2, IL-4, and IFN-γ and CD8(+) T cell responses for the cytokines IL-2 and IFN-γ compared to the mice vaccinated with the corresponding antigen plus CpG or poly(I:C) alone. In addition, the higher antibody titers against EEEV effectively neutralized the EEEV pseudoviruses in the group immunized with antigen plus mixed CpG/poly (I:C) adjuvant after tertiary immunization. This study demonstrates that the pcDNA3.1-C-E plasmids in conjunction with mixed CpG/poly (I:C) adjuvant priming maximize the cellular immune response and specific antibody generation in mice. Moreover, this mixed adjuvant priming provides a promising strategy for enhancing the immune effectiveness of a DNA vaccine against EEEV.
Collapse
Affiliation(s)
- JinZhu Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, Heilongjiang Province, China; The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China; College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Province, China
| | - HuaLei Wang
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - XueXing Zheng
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - XiangHong Xue
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - BeiYan Wang
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Province, China
| | - HongXia Wu
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - Kun Zhang
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - ShengTao Fan
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - TieCheng Wang
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - Nan Li
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - YongKun Zhao
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - YuWei Gao
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China
| | - SongTao Yang
- The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China.
| | - XianZhu Xia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, Heilongjiang Province, China; The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun 130122, Jilin Province, China.
| |
Collapse
|
30
|
Ungaro F, Conte C, Quaglia F, Tornesello ML, Buonaguro FM, Buonaguro L. VLPs and particle strategies for cancer vaccines. Expert Rev Vaccines 2013; 12:1173-1193. [PMID: 24124878 DOI: 10.1586/14760584.2013.836909] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Effective delivery of tumor antigens to APCs is one of the key steps for eliciting a strong and durable immune response to tumors. Several cancer vaccines have been evaluated in clinical trials, based on soluble peptides, but results have not been fully satisfactory. To improve immunogenicity particles provide a valid strategy to display and/or incorporate epitopes which can be efficiently targeted to APCs for effective induction of adaptive immunity. In the present review, we report some leading technologies for developing particulate vaccines employed in cancer immunotherapy, highlighting the key parameters for a rational design to elicit both humoral and cellular responses.
Collapse
Affiliation(s)
- Francesca Ungaro
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131, Napoli, Italy
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
In this study, we describe a versatile, flexible, and quick method to label different families of enveloped viruses with glycosylphosphatidylinositol-modified green fluorescent protein, termed fluorescence molecular painting (FMP). As an example for a potential application, we investigated virus attachment by means of flow cytometry to determine if viral binding behavior may be analyzed after FMP of enveloped viruses. Virus attachment was inhibited by using either dextran sulfate or by blocking attachment sites with virus pre-treatment. Results from the FMP-flow cytometry approach were verified by immunoblotting and enzyme-linked immunosorbent assay. Since the modification strategy is applicable to a broad range of proteins and viruses, variations of this method may be useful in a range of research and applied applications from bio-distribution studies to vaccine development and targeted infection for gene delivery.
Collapse
Affiliation(s)
- Christoph Metzner
- Institute of Virology, University of Veterinary Medicine, Vienna, Veterinarplatz 1, 1210 Vienna, Austria
| | - Feliks Kochan
- Institute of Virology, University of Veterinary Medicine, Vienna, Veterinarplatz 1, 1210 Vienna, Austria
| | - John A. Dangerfield
- Institute of Virology, University of Veterinary Medicine, Vienna, Veterinarplatz 1, 1210 Vienna, Austria
- Anovasia Pte Ltd, 20 Biopolis Way, #05-518 Centros, Singapore, 138668 Singapore
| |
Collapse
|
32
|
Kang SM, Kim MC, Compans RW. Virus-like particles as universal influenza vaccines. Expert Rev Vaccines 2013; 11:995-1007. [PMID: 23002980 DOI: 10.1586/erv.12.70] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Current influenza vaccines are primarily targeted to induce immunity to the influenza virus strain-specific hemagglutinin antigen and are not effective in controlling outbreaks of new pandemic viruses. An approach for developing universal vaccines is to present highly conserved antigenic epitopes in an immunogenic conformation such as virus-like particles (VLPs) together with an adjuvant to enhance the vaccine immunogenicity. In this review, the authors focus on conserved antigenic targets and molecular adjuvants that were presented in VLPs. Conserved antigenic targets that include the hemagglutinin stalk domain, the external domain of influenza M2 and neuraminidase are discussed in addition to molecular adjuvants that are engineered to be incorporated into VLPs in a membrane-anchored form.
Collapse
Affiliation(s)
- Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| | | | | |
Collapse
|
33
|
Buonaguro L, Tagliamonte M, Visciano ML, Tornesello ML, Buonaguro FM. Developments in virus-like particle-based vaccines for HIV. Expert Rev Vaccines 2013; 12:119-127. [PMID: 23414404 DOI: 10.1586/erv.12.152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Virus-like particles (VLPs) hold great promise for the development of effective and affordable vaccines. VLPs, indeed, are suitable for presentation and efficient delivery to antigen-presenting cells of linear as well as conformational antigens. This will ultimately result in a crosspresentation with both MHC class I and II molecules to prime CD4(+) T-helper and CD8(+) cytotoxic T cells. This review describes an update on the development and use of VLPs as vaccine approaches for HIV.
Collapse
Affiliation(s)
- Luigi Buonaguro
- Department of Experimental Oncology, Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori 'Fond Pascale', Via Mariano Semmola 142, 80131 Napoli, Italy
| | | | | | | | | |
Collapse
|
34
|
Kushnir N, Streatfield SJ, Yusibov V. Virus-like particles as a highly efficient vaccine platform: diversity of targets and production systems and advances in clinical development. Vaccine 2012; 31:58-83. [PMID: 23142589 PMCID: PMC7115575 DOI: 10.1016/j.vaccine.2012.10.083] [Citation(s) in RCA: 417] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 10/13/2012] [Accepted: 10/25/2012] [Indexed: 12/16/2022]
Abstract
Virus-like particles (VLPs) are a class of subunit vaccines that differentiate themselves from soluble recombinant antigens by stronger protective immunogenicity associated with the VLP structure. Like parental viruses, VLPs can be either non-enveloped or enveloped, and they can form following expression of one or several viral structural proteins in a recombinant heterologous system. Depending on the complexity of the VLP, it can be produced in either a prokaryotic or eukaryotic expression system using target-encoding recombinant vectors, or in some cases can be assembled in cell-free conditions. To date, a wide variety of VLP-based candidate vaccines targeting various viral, bacterial, parasitic and fungal pathogens, as well as non-infectious diseases, have been produced in different expression systems. Some VLPs have entered clinical development and a few have been licensed and commercialized. This article reviews VLP-based vaccines produced in different systems, their immunogenicity in animal models and their status in clinical development.
Collapse
Affiliation(s)
- Natasha Kushnir
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | | | | |
Collapse
|
35
|
Delivery of subunit influenza vaccine to skin with microneedles improves immunogenicity and long-lived protection. Sci Rep 2012; 2:357. [PMID: 22500210 PMCID: PMC3324956 DOI: 10.1038/srep00357] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 03/22/2012] [Indexed: 11/23/2022] Open
Abstract
Influenza infection represents a major socio-economic burden worldwide. Novel delivery methods can render influenza vaccination easier and more acceptable by the public, and importantly confer protection equal or superior to that induced by conventional systemic administration. An attractive target for vaccine delivery is the skin. Recent studies have demonstrated improved immune responses after transdermal delivery of inactivated influenza virus with microneedle patches. Here we show that immunization with a licensed influenza subunit vaccine coated on metal microneedles can activate both humoral and cellular arms of the immune response and confer improved long-term protection in the mouse model when compared to the conventional systemic route of delivery. These results demonstrate the promising potential of microneedle delivery of licensed influenza subunit vaccines, that could be beneficial in increasing vaccine coverage and protection and reducing influenza-related mortality worldwide.
Collapse
|
36
|
Buonaguro L, Tagliamonte M, Tornesello ML, Buonaguro FM. Developments in virus-like particle-based vaccines for infectious diseases and cancer. Expert Rev Vaccines 2012; 10:1569-83. [PMID: 22043956 DOI: 10.1586/erv.11.135] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Virus-like particles hold great promise for the development of effective and affordable vaccines. Indeed, virus-like particles are suitable for presentation and efficient delivery of linear as well as conformational antigens to antigen-presenting cells. This will ultimately result in optimal B-cell activation and cross-presentation with both MHC class I and II molecules to prime CD4(+) T-helper as well as CD8(+) cytotoxic T cells. This article provides an update on the development and use of virus-like particles as vaccine approaches for infectious diseases and cancer.
Collapse
Affiliation(s)
- Luigi Buonaguro
- Molecular Biology and Viral Oncology, Department of Experimental Oncology, Istituto Nazionale Tumori Fond Pascale, Via Mariano Semmola 142, 80131 Napoli, Italy.
| | | | | | | |
Collapse
|
37
|
CD40L-containing virus-like particle as a candidate HIV-1 vaccine targeting dendritic cells. J Acquir Immune Defic Syndr 2011; 56:393-400. [PMID: 21239998 DOI: 10.1097/qai.0b013e31820b844e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The central role of dendritic cell (DC) in mounting an immune response to a novel antigen is now well established. We sought to demonstrate the use of a particular vaccine strategy based on directing HIV-1 Gag proteins to DCs in conjunction with an activation signal. CD40L was expressed on the surface of virus-like particles (VLPs) to target HIV-1 Gag antigens to the CD40 receptor on DCs, whereas CD40L-CD40 interaction would also result in cellular activation. Multiple CD40L VLP constructs were made and evaluated in vitro and in vivo. Indeed, one VLP that expressed CD40L to the highest level showed greatest capacity to activate DCs in vitro. Correspondingly, this CD40L-VLP also proved to be most immunogenic in mice in raising both humoral and cellular responses to HIV-1 Gag. Confirmatory studies were performed to demonstrate the increased immunogenicity of CD40L-VLP is no longer observed when tested in CD40-/- mice. Our findings lend support to the belief that vaccine strategies that both target and activate DCs could yield a superior immune response.
Collapse
|
38
|
|
39
|
Melchers M, Matthews K, de Vries RP, Eggink D, van Montfort T, Bontjer I, van de Sandt C, David K, Berkhout B, Moore JP, Sanders RW. A stabilized HIV-1 envelope glycoprotein trimer fused to CD40 ligand targets and activates dendritic cells. Retrovirology 2011; 8:48. [PMID: 21689404 PMCID: PMC3141652 DOI: 10.1186/1742-4690-8-48] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 06/20/2011] [Indexed: 12/22/2022] Open
Abstract
Background One reason why subunit protein and DNA vaccines are often less immunogenic than live-attenuated and whole-inactivated virus vaccines is that they lack the co-stimulatory signals provided by various components of the more complex vaccines. The HIV-1 envelope glycoprotein complex (Env) is no exception to this rule. Other factors that limit the induction of neutralizing antibodies against HIV-1 lie in the structure and instability of Env. We have previously stabilized soluble trimeric mimics of Env by introducing a disulfide bond between gp120 and gp41 and adding a trimer stabilizing mutation in gp41 (SOSIP.R6 gp140). Results We further stabilized the SOSIP.R6 gp140 using a GCN4-based isoleucine zipper motif, creating SOSIP.R6-IZ gp140. In order to target SOSIP.R6-IZ to immune cells, including dendritic cells, while at the same time activating these cells, we fused SOSIP.R6-IZ to the active domain of CD40 ligand (CD40L), which may serve as a 'cis-adjuvant'. The Env component of the SOSIP.R6-IZ-CD40L fusion construct bound to CD4 and neutralizing antibodies, while the CD40L moiety interacted with CD40. Furthermore, the chimeric molecule was able to signal efficiently through CD40 and induce maturation of human dendritic cells. Dendritic cells secreted IL-6, IL-10 and IL-12 in response to stimulation by SOSIP.R6-IZ-CD40L and were able to activate naïve T cells. Conclusions Chimeric HIV-1 gp140 - CD40L trimers can target and activate dendritic cells. Targeting and activating immune cells using CD40L and other 'cis-adjuvants' may improve subunit protein vaccine immunogenicity for HIV-1 and other infectious diseases.
Collapse
Affiliation(s)
- Mark Melchers
- Laboratory of Experimental Virology, Department of Medical Microbiology Center for Infection and Immunity Amsterdam, Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gupta S, Termini JM, Niu L, Kanagavelu SK, Schmidtmayerova H, Snarsky V, Kornbluth RS, Stone GW. EBV LMP1, a viral mimic of CD40, activates dendritic cells and functions as a molecular adjuvant when incorporated into an HIV vaccine. J Leukoc Biol 2011; 90:389-98. [PMID: 21586676 DOI: 10.1189/jlb.0211068] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
HIV-1 does not significantly activate cellular immunity, which has made it difficult to use attenuated forms of HIV-1 as a vaccine. In contrast, EBV induces robust T cell responses in most infected individuals, perhaps as this virus contains LMP1, a viral mimic of CD40, which is a key activating molecule for DCs and macrophages. Consequently, studies were conducted using LMP1 and LMP1-CD40, a related construct formed by replacing the intracellular signaling domain of LMP1 with that of CD40. Upon electroporation into DCs, LMP1 and LMP1-CD40 mRNAs were sufficient to up-regulate costimulatory molecules and proinflammatory cytokines, indicating that these molecules can function in isolation as adjuvant-like molecules. As a first step toward an improved HIV vaccine, LMP1 and LMP1-CD40 were introduced into a HIV-1 construct to produce virions encoding these proteins. Transduction of DCs and macrophages with these viruses induced morphological changes and up-regulated costimulatory molecules and cytokine production by these cells. HIV-LMP1 enhanced the antigen-presenting function of DCs, as measured in an in vitro immunization assay. Taken together, these data show that LMP1 and LMP1-CD40 are portable gene cassettes with strong adjuvant properties that can be introduced into viruses such as HIV, which by themselves, are insufficient to induce protective cellular immunity.
Collapse
Affiliation(s)
- Sachin Gupta
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Urban JH, Merten CA. Retroviral display in gene therapy, protein engineering, and vaccine development. ACS Chem Biol 2011; 6:61-74. [PMID: 21171610 DOI: 10.1021/cb100285n] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The display and analysis of proteins expressed on biological surfaces has become an attractive tool for the study of molecular interactions in enzymology, protein engineering, and high-throughput screening. Among the growing number of established display systems, retroviruses offer a unique and fully mammalian platform for the expression of correctly folded and post-translationally modified proteins in the context of cell plasma membrane-derived particles. This is of special interest for therapeutic applications such as gene therapy and vaccine development and also offers advantages for the engineering of mammalian proteins toward customized binding affinities and catalytic activities. This review critically summarizes the basic concepts and applications of retroviral display and analyses its benefits in comparison to other display techniques.
Collapse
Affiliation(s)
- Johannes H. Urban
- Duke Translational Research Institute and Department of Surgery, Duke University Medical Center, MSRBII, 106 Research Drive, Durham, North Carolina 27710, United States
| | - Christoph A. Merten
- Genome Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, Heidelberg 69117, Germany
| |
Collapse
|
42
|
Song JM, Wang BZ, Park KM, Van Rooijen N, Quan FS, Kim MC, Jin HT, Pekosz A, Compans RW, Kang SM. Influenza virus-like particles containing M2 induce broadly cross protective immunity. PLoS One 2011; 6:e14538. [PMID: 21267073 PMCID: PMC3022578 DOI: 10.1371/journal.pone.0014538] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 11/25/2010] [Indexed: 12/29/2022] Open
Abstract
Background Current influenza vaccines based on the hemagglutinin protein are strain specific and do not provide good protection against drifted viruses or emergence of new pandemic strains. An influenza vaccine that can confer cross-protection against antigenically different influenza A strains is highly desirable for improving public health. Methodology/Principal Findings To develop a cross protective vaccine, we generated influenza virus-like particles containing the highly conserved M2 protein in a membrane-anchored form (M2 VLPs), and investigated their immunogenicity and breadth of cross protection. Immunization of mice with M2 VLPs induced anti-M2 antibodies binding to virions of various strains, M2 specific T cell responses, and conferred long-lasting cross protection against heterologous and heterosubtypic influenza viruses. M2 immune sera were found to play an important role in providing cross protection against heterosubtypic virus and an antigenically distinct 2009 pandemic H1N1 virus, and depletion of dendritic and macrophage cells abolished this cross protection, providing new insight into cross-protective immune mechanisms. Conclusions/Significance These results suggest that presenting M2 on VLPs in a membrane-anchored form is a promising approach for developing broadly cross protective influenza vaccines.
Collapse
Affiliation(s)
- Jae-Min Song
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Bao-Zhong Wang
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kyoung-Mi Park
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nico Van Rooijen
- Department of Molecular Cell Biology, Vrije Universiteit Medisch Centrum, Amsterdam, The Netherlands
| | - Fu-Shi Quan
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Min-Chul Kim
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Hyun-Tak Jin
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Richard W. Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (RWC); (SMK)
| | - Sang-Moo Kang
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (RWC); (SMK)
| |
Collapse
|
43
|
Zeshan B, Mushtaq MH, Wang X, Li W, Jiang P. Protective immune responses induced by in ovo immunization with recombinant adenoviruses expressing spike (S1) glycoprotein of infectious bronchitis virus fused/co-administered with granulocyte-macrophage colony stimulating factor. Vet Microbiol 2010; 148:8-17. [PMID: 20850939 DOI: 10.1016/j.vetmic.2010.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 07/29/2010] [Accepted: 08/06/2010] [Indexed: 11/25/2022]
Abstract
Infectious bronchitis virus (IBV) causes tremendous economic losses associated with production inefficiencies and mortality in poultry industry worldwide. In the present report, the recombinant adenoviruses expressing chicken granulocyte-macrophage colony stimulating factor (GM-CSF) and S1 gene of nephropathogenic IBV were constructed and characterized. Then, the immunological efficacy and protection against homologous IBV challenge were assessed in specific pathogen free (SPF) chickens. The results showed that the chickens vaccinated in ovo with rAd-S1, rAd-GM-S1 (GM-CSF fused with S1 using glycine linkers) and rAd-GM-CSF plus rAd-S1 (co-administered) developed specific anti-IBV HI antibodies. Moreover, the fusion of the GM-CSF markedly increased spleen cell proliferation and IFN-γ production while mild increased in IL-4 production, which demonstrated the enhancement of cell-mediated immune responses. Following challenge with IBV, the chickens in the group vaccinated with rAd-S1 fused or co-administered with GM-CSF had fewer nephropathic lesions and showed 100% protection as compared to that of rAd-S1 alone which showed 70% protection. It indicated that the single dose in ovo vaccination of the GM-CSF fused or co-administered with S1 of IBV could enhance significantly the humoral, cellular immune responses and provide complete protection against nephropathogenic IBV challenge. This finding may provide basic information for effective in ovo vaccines design against IBV.
Collapse
Affiliation(s)
- Basit Zeshan
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | | | |
Collapse
|
44
|
Zhang R, Zhang S, Li M, Chen C, Yao Q. Incorporation of CD40 ligand into SHIV virus-like particles (VLP) enhances SHIV-VLP-induced dendritic cell activation and boosts immune responses against HIV. Vaccine 2010; 28:5114-27. [PMID: 20471443 DOI: 10.1016/j.vaccine.2010.03.079] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 03/22/2010] [Accepted: 03/30/2010] [Indexed: 11/29/2022]
Abstract
Engagement of CD40 with CD40L induces dendritic cell (DC) maturation and activation, thereby promoting immune responses. The objective of this study was to investigate whether immunization with chimeric CD40L/SHIV virus-like particles (CD40L/SHIV-VLP) could enhance immune responses to SIV Gag and HIV Env proteins by directly activating DCs. We found that CD83, CD40, and CD86 were significantly up-regulated and significantly increased cytokines production were observed after hCD40L/SHIV-VLP treatment in human CD14(+) monocyte-derived DCs as compared to SHIV-VLP treatment. Mice immunized with mCD40L/SHIV-VLP showed more than a two-fold increase in HIV Env-specific IgG antibody production, an increase in SIV Gag and HIV Env-specific IFN-gamma and IL-4 producing cells, and an increase in HIV Env-specific cytotoxic activity compared to that in SHIV-VLP immunized mice. Furthermore, multifunctional CD4(+) Th1 cells, which simultaneously produce IFN-gamma, IL-2 and TNF-alpha triple cytokines, and CD8(+) T-cells, which produce IFN-gamma were elevated in the mCD40L/SHIV-VLP immunized group. These data demonstrate that chimeric CD40L/SHIV-VLP potently induce DC activation and enhance the magnitude of both humoral and cellular immune responses to the SIV Gag and HIV Env proteins in the mouse model. Therefore, incorporation of CD40L into VLP may represent a novel strategy to develop effective HIV vaccines.
Collapse
Affiliation(s)
- Rongxin Zhang
- Molecular Surgeon Research Center, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, United States
| | | | | | | | | |
Collapse
|
45
|
Skountzou I, Martin MDP, Wang B, Ye L, Koutsonanos D, Weldon W, Jacob J, Compans RW. Salmonella flagellins are potent adjuvants for intranasally administered whole inactivated influenza vaccine. Vaccine 2009; 28:4103-12. [PMID: 19654062 DOI: 10.1016/j.vaccine.2009.07.058] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 07/01/2009] [Accepted: 07/19/2009] [Indexed: 12/24/2022]
Abstract
Bacterial flagellins are potent inducers of innate immune responses in the mouse lung because they bind to TLR5 expressed on the apical surfaces of airway epithelial cells. TLR engagement leads to the initiation of a signaling cascade that results in a pro-inflammatory response with subsequent up-regulation of several cytokines and leads to adaptive immune responses. We examined the ability of two soluble flagellins, a monomeric flagellin expressed in Escherichia coli and a highly purified polymeric flagellin directly isolated from Salmonella, to enhance the efficacy of influenza vaccines in mice. Here we demonstrate that both flagellins co-administered intranasally with inactivated A/PR/8/34 (PR8) virus induced robust increases of systemic influenza-specific IgA and IgG titers and resulted in a more comprehensive humoral response as indicated by the increase of IgG2a and IgG2b subclass responses. Groups immunized with the adjuvanted vaccines were fully protected against high dose lethal challenge by homologous virus whereas inactivated PR8 alone conferred only partial protection. Finally we show that shortly after immunization the adjuvanted vaccines induced a dramatic increase in pro-inflammatory cytokines in the lung, resulting in extensive lung infiltration by granulocytes and monocytes/macrophages. Our results reveal a promising perspective for the use of both soluble monomeric and polymeric flagellin as mucosal vaccine adjuvants to improve protection against influenza epidemics as well as a range of other infectious diseases.
Collapse
Affiliation(s)
- Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang X, Li J, Jiang P, Li Y, Zeshan B, Cao J, Wang X. GM-CSF fused with GP3 and GP5 of porcine reproductive and respiratory syndrome virus increased the immune responses and protective efficacy against virulent PRRSV challenge. Virus Res 2009; 143:24-32. [DOI: 10.1016/j.virusres.2009.02.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Revised: 02/25/2009] [Accepted: 02/27/2009] [Indexed: 11/26/2022]
|
47
|
Kang SM, Song JM, Quan FS, Compans RW. Influenza vaccines based on virus-like particles. Virus Res 2009; 143:140-6. [PMID: 19374929 DOI: 10.1016/j.virusres.2009.04.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 04/03/2009] [Accepted: 04/07/2009] [Indexed: 12/14/2022]
Abstract
The simultaneous expression of structural proteins of virus can produce virus-like particles (VLPs) by a self-assembly process in a viral life cycle even in the absence of genomic material. Taking an advantage of structural and morphological similarities of VLPs to native virions, VLPs have been suggested as a promising platform for new viral vaccines. In the light of a pandemic threat, influenza VLPs have been recently developed as a new generation of non-egg based cell culture-derived vaccine candidates against influenza infection. Animals vaccinated with VLPs containing hemagglutinin (HA) or HA and neuraminidase (NA) were protected from morbidity and mortality resulting from lethal influenza infections. Influenza VLPs serve as an excellent model system of an enveloped virus for understanding the properties of VLPs in inducing protective immunity. In this review, we briefly describe the characteristics of influenza VLPs assembled with a lipid bilayer containing glycoproteins, and summarize the current progress on influenza VLPs as an alternative vaccine candidate against seasonal as well as pandemic influenza viruses. In addition, the protective immune correlates induced by vaccination with influenza VLPs are discussed.
Collapse
Affiliation(s)
- Sang-Moo Kang
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|
48
|
Koutsonanos DG, Martin MDP, Zarnitsyn VG, Sullivan SP, Compans RW, Prausnitz MR, Skountzou I. Transdermal influenza immunization with vaccine-coated microneedle arrays. PLoS One 2009; 4:e4773. [PMID: 19274084 PMCID: PMC2651574 DOI: 10.1371/journal.pone.0004773] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 02/12/2009] [Indexed: 11/25/2022] Open
Abstract
Background Influenza is a contagious disease caused by a pathogenic virus, with outbreaks all over the world and thousands of hospitalizations and deaths every year. Due to virus antigenic drift and short-lived immune responses, annual vaccination is required. However, vaccine coverage is incomplete, and improvement in immunization is needed. The objective of this study is to investigate a novel method for transdermal delivery using metal microneedle arrays (MN) coated with inactivated influenza virus to determine whether this route is a simpler and safer approach than the conventional immunization, capable to induce robust immune responses and confer protection against lethal virus challenge. Methodology/Principal Findings Inactivated A/Aichi/2/68 (H3N2) influenza virus was coated on metal microneedle arrays and applied to mice as a vaccine in the caudal dorsal skin area. Substantial antibody titers with hemagglutination inhibition activity were detected in sera collected two and four weeks after a single vaccine dose. Challenge studies in mice with 5×LD50 of mouse adapted Aichi virus demonstrated complete protection. Microneedle vaccination induced a broad spectrum of immune responses including CD4+ and CD8+ responses in the spleen and draining lymph node, a high frequency of antigen-secreting cells in the lung and induction of virus-specific memory B-cells. In addition, the use of MN showed a dose-sparing effect and a strong Th2 bias when compared to an intramuscular (IM) reference immunization. Conclusions/Significance The present results show that delivery of inactivated influenza virus through the skin using metal microneedle arrays induced strong humoral and cellular immune responses capable of conferring protection against virus challenge as efficiently as intramuscular immunization, which is the standard vaccination route. In view of the convenience of delivery and the potential for self-administration, vaccine-coated metal microneedles may provide a novel and highly effective immunization method.
Collapse
Affiliation(s)
- Dimitrios G. Koutsonanos
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Maria del Pilar Martin
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Vladimir G. Zarnitsyn
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Sean P. Sullivan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Richard W. Compans
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mark R. Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail: (MRP); (IS)
| | - Ioanna Skountzou
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (MRP); (IS)
| |
Collapse
|
49
|
Abstract
There is an urgent need to develop novel approaches for vaccination against emerging pathogenic avian influenza viruses as a priority for pandemic preparedness. Influenza virus-like particles (VLPs) have been suggested and developed as a new generation of non-egg-based cell culture-derived vaccine candidates against influenza infection. Influenza VLPs are formed by a self-assembly process incorporating structural proteins into budding particles composed of the hemagglutinin (HA), neuraminidase (NA) and M1 proteins, and may include additional influenza proteins such as M2. Animals vaccinated with VLPs were protected from morbidity and mortality resulting from lethal influenza infections. The protective mechanism of influenza VLP vaccines was similar to that of the currently licensed influenza vaccines inducing neutralizing antibodies and hemagglutination inhibition activities. Current studies demonstrate that influenza VLP approaches can be a promising alternative approach to developing a vaccine for pandemic influenza viruses. The first human clinical trial of a recombinant pandemic-like H5N1 influenza VLP vaccine was initiated in July 2007 (Bright et al., unpublished).
Collapse
|
50
|
Rafts, anchors and viruses — A role for glycosylphosphatidylinositol anchored proteins in the modification of enveloped viruses and viral vectors. Virology 2008; 382:125-31. [DOI: 10.1016/j.virol.2008.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 09/12/2008] [Accepted: 09/18/2008] [Indexed: 12/18/2022]
|