1
|
Zhang H, Li Y, Li W, Li Z, Zhou J, Tong D. Surface Display of Cholera Toxin B Subunit Recombinant Escherichia coli Ghosts Further Enhances Resistance to Chlamydia abortus Infection in Mice. Microorganisms 2024; 12:1656. [PMID: 39203498 PMCID: PMC11356887 DOI: 10.3390/microorganisms12081656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
Chlamydia abortus (C. abortus) is an important zoonotic pathogen that seriously endangers the development of animal husbandry. Vaccination is the most effective approach to preventing C. abortus infection. We previously reported a recombinant Escherichia coli ghost (rECG)-based C. abortus vaccine that demonstrated outstanding protective efficacy. In this study, we further attempted to fuse the cholera toxin B subunit (CTB), a widely studied potent mucosal immune adjuvant, with macrophage infectivity potentiator (MIP), a candidate antigen of C. abortus, on the surface of the rECG and explore its protective effect against C. abortus infection. The MIP fusion protein was highly expressed in the rECGs, and the CTB-modified rECGs significantly induced the activation of mouse bone marrow-derived dendritic cells in vitro. Intranasal immunization with rECGs induced a Th1-biased cellular immune response. Compared to the rECGs without CTB, the CTB-modified rECGs induced higher concentrations of IgA in the serum and vaginal wash solution. Moreover, in a mouse infection model, the CTB-modified rECGs significantly improved the clearance efficiency of C. abortus and reduced the pathological damage to the uterus. This study demonstrates that incorporating CTB into rECGs significantly enhances the immunogenic potential of the rECG vaccine and can significantly enhance its protective efficacy against a C. abortus challenge.
Collapse
Affiliation(s)
- Huaiyu Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Yunhui Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Wei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Zhaocai Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Jizhang Zhou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
2
|
Yoshida K, Iyori M, Blagborough AM, Salman AM, Dulal P, Sala KA, Yamamoto DS, Khan SM, Janse CJ, Biswas S, Yoshii T, Yusuf Y, Tokoro M, Hill AVS, Yoshida S. Adenovirus-prime and baculovirus-boost heterologous immunization achieves sterile protection against malaria sporozoite challenge in a murine model. Sci Rep 2018; 8:3896. [PMID: 29497047 PMCID: PMC5832798 DOI: 10.1038/s41598-018-21369-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/02/2018] [Indexed: 12/16/2022] Open
Abstract
With the increasing prevalence of artemisinin-resistant malaria parasites, a highly efficacious and durable vaccine for malaria is urgently required. We have developed an experimental virus-vectored vaccine platform based on an envelope-modified baculovirus dual-expression system (emBDES). Here, we show a conceptually new vaccine platform based on an adenovirus-prime/emBDES-boost heterologous immunization regimen expressing the Plasmodium falciparum circumsporozoite protein (PfCSP). A human adenovirus 5-prime/emBDES-boost heterologous immunization regimen consistently achieved higher sterile protection against transgenic P. berghei sporozoites expressing PfCSP after a mosquito-bite challenge than reverse-ordered or homologous immunization. This high protective efficacy was also achieved with a chimpanzee adenovirus 63-prime/emBDES-boost heterologous immunization regimen against an intravenous sporozoite challenge. Thus, we show that the adenovirus-prime/emBDES-boost heterologous immunization regimen confers sterile protection against sporozoite challenge by two individual routes, providing a promising new malaria vaccine platform for future clinical use.
Collapse
Affiliation(s)
- Kunitaka Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan.,Kanazawa University Graduate School of Medical Sciences, 13 Takara-machi, Kanazawa, 920-0934, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Andrew M Blagborough
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, Imperial College Road, South Kensington, London, SW7 2AZ, UK
| | - Ahmed M Salman
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK.,Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, (LUMC, L4-Q), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Pawan Dulal
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Katarzyna A Sala
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, Imperial College Road, South Kensington, London, SW7 2AZ, UK
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, 329-0431, Tochigi, Japan
| | - Shahid M Khan
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, (LUMC, L4-Q), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, (LUMC, L4-Q), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sumi Biswas
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Tatsuya Yoshii
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Masaharu Tokoro
- Kanazawa University Graduate School of Medical Sciences, 13 Takara-machi, Kanazawa, 920-0934, Japan
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan.
| |
Collapse
|
3
|
Lactobacillus plantarum producing a Chlamydia trachomatis antigen induces a specific IgA response after mucosal booster immunization. PLoS One 2017; 12:e0176401. [PMID: 28467432 PMCID: PMC5415134 DOI: 10.1371/journal.pone.0176401] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/09/2017] [Indexed: 01/23/2023] Open
Abstract
Mucosal immunity is important for the protection against a wide variety of pathogens. Traditional vaccines administered via parenteral routes induce strong systemic immunity, but they often fail to generate mucosal IgA. In contrast, bacteria-based vaccines comprise an appealing strategy for antigen delivery to mucosal sites. Vaginal infection with Chlamydia trachomatis can develop into upper genital tract infections that can lead to infertility. Therefore, the development of an effective vaccine against Chlamydia is a high priority. In the present study, we have explored the use of a common lactic acid bacterium, Lactobacillus plantarum, as a vector for delivery of a C. trachomatis antigen to mucosal sites. The antigen, referred as Hirep2 (H2), was anchored to the surface of L. plantarum cells using an N-terminal lipoprotein anchor. After characterization, the constructed strain was used as an immunogenic agent in mice. We explored a heterologous prime-boost strategy, consisting of subcutaneous priming with soluble H2 antigen co-administered with CAF01 adjuvant, followed by an intranasal boost with H2-displaying L. plantarum. The results show that, when used as a booster, the recombinant L. plantarum strain was able to evoke cellular responses. Most importantly, booster immunization with the Lactobacillus-based vaccine induced generation of antigen-specific IgA in the vaginal cavity.
Collapse
|
4
|
Kessans SA, Linhart MD, Meador LR, Kilbourne J, Hogue BG, Fromme P, Matoba N, Mor TS. Immunological Characterization of Plant-Based HIV-1 Gag/Dgp41 Virus-Like Particles. PLoS One 2016; 11:e0151842. [PMID: 26986483 PMCID: PMC4795674 DOI: 10.1371/journal.pone.0151842] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/04/2016] [Indexed: 02/08/2023] Open
Abstract
It is widely anticipated that a prophylactic vaccine may be needed to control the HIV/AIDS epidemic worldwide. Despite over two decades of research, a vaccine against HIV-1 remains elusive, although a recent clinical trial has shown promising results. Recent studies have focused on highly conserved domains within HIV-1 such as the membrane proximal external region (MPER) of the envelope glycoprotein, gp41. MPER has been shown to play critical roles in mucosal transmission of HIV-1, though this peptide is poorly immunogenic on its own. Here we provide evidence that plant-produced HIV-1 enveloped virus-like particles (VLPs) consisting of Gag and a deconstructed form of gp41 comprising the MPER, transmembrane, and cytoplasmic domains (Dgp41) provides an effective platform to display MPER for use as an HIV vaccine candidate. Prime-boost strategies combining systemic and mucosal priming with systemic boosting using two different vaccine candidates (VLPs and CTB-MPR--a fusion of MPER and the B-subunit of cholera toxin) were investigated in BALB/c mice. Serum antibody responses against both the Gag and gp41 antigens were elicited when systemically primed with VLPs. These responses could be recalled following systemic boosting with VLPs. In addition, mucosal priming with VLPs allowed for a boosting response against Gag and gp41 when boosted with either candidate. Importantly, the VLPs also induced Gag-specific CD4 and CD8 T-cell responses. This report on the immunogenicity of plant-based Gag/Dgp41 VLPs may represent an important milestone on the road towards a broadly efficacious and inexpensive subunit vaccine against HIV-1.
Collapse
Affiliation(s)
- Sarah A. Kessans
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Mark D. Linhart
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Lydia R. Meador
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jacquelyn Kilbourne
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Brenda G. Hogue
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Petra Fromme
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona, United States of America
| | - Nobuyuki Matoba
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Tsafrir S. Mor
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
5
|
Banerjee S, Shi H, Habte HH, Qin Y, Cho MW. Modulating immunogenic properties of HIV-1 gp41 membrane-proximal external region by destabilizing six-helix bundle structure. Virology 2016; 490:17-26. [PMID: 26803471 DOI: 10.1016/j.virol.2016.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/05/2016] [Accepted: 01/09/2016] [Indexed: 01/31/2023]
Abstract
The C-terminal alpha-helix of gp41 membrane-proximal external region (MPER; (671)NWFDITNWLWYIK(683)) encompassing 4E10/10E8 epitopes is an attractive target for HIV-1 vaccine development. We previously reported that gp41-HR1-54Q, a trimeric protein comprised of the MPER in the context of a stable six-helix bundle (6HB), induced strong immune responses against the helix, but antibodies were directed primarily against the non-neutralizing face of the helix. To better target 4E10/10E8 epitopes, we generated four putative fusion intermediates by introducing double point mutations or deletions in the heptad repeat region 1 (HR1) that destabilize 6HB in varying degrees. One variant, HR1-∆10-54K, elicited antibodies in rabbits that targeted W672, I675 and L679, which are critical for 4E10/10E8 recognition. Overall, the results demonstrated that altering structural parameters of 6HB can influence immunogenic properties of the MPER and antibody targeting. Further exploration of this strategy could allow development of immunogens that could lead to induction of 4E10/10E8-like antibodies.
Collapse
Affiliation(s)
- Saikat Banerjee
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Heliang Shi
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Habtom H Habte
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Yali Qin
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Michael W Cho
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
6
|
Matoba N. N-Glycosylation of Cholera Toxin B Subunit: Serendipity for Novel Plant-Made Vaccines? FRONTIERS IN PLANT SCIENCE 2015; 6:1132. [PMID: 26732492 PMCID: PMC4686596 DOI: 10.3389/fpls.2015.01132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/29/2015] [Indexed: 05/28/2023]
Abstract
The non-toxic B subunit of cholera toxin (CTB) has attracted considerable interests from vaccinologists due to strong mucosal immunomodulatory effects and potential utility as a vaccine scaffold for heterologous antigens. Along with other conventional protein expression systems, various plant species have been used as production hosts for CTB and its fusion proteins. However, it has recently become clear that the protein is N-glycosylated within the endoplasmic reticulum of plant cells-a eukaryotic post-translational modification that is not present in native CTB. While functionally active aglycosylated variants have been successfully engineered to circumvent potential safety and regulatory issues related to glycosylation, this modification may actually provide advantageous characteristics to the protein as a vaccine platform. Based on data from our recent studies, I discuss the unique features of N-glycosylated CTB produced in plants for the development of novel vaccines.
Collapse
|
7
|
Habte HH, Banerjee S, Shi H, Qin Y, Cho MW. Immunogenic properties of a trimeric gp41-based immunogen containing an exposed membrane-proximal external region. Virology 2015; 486:187-97. [PMID: 26454663 DOI: 10.1016/j.virol.2015.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/09/2015] [Accepted: 09/22/2015] [Indexed: 01/15/2023]
Abstract
The membrane-proximal external region (MPER) of HIV-1 gp41 is an attractive target for vaccine development. Thus, better understanding of its immunogenic properties in various structural contexts is important. We previously described the crystal structure of a trimeric protein complex named gp41-HR1-54Q, which consists of the heptad repeat regions 1 and 2 and the MPER. The protein was efficiently recognized by broadly neutralizing antibodies. Here, we describe its immunogenic properties in rabbits. The protein was highly immunogenic, especially the C-terminal end of the MPER containing 4E10 and 10E8 epitopes ((671)NWFDITNWLWYIK(683)). Although antibodies exhibited strong competition activity against 4E10 and 10E8, neutralizing activity was not detected. Detailed mapping analyses indicated that amino acid residues critical for recognition resided on faces of the alpha helix that are either opposite of or perpendicular to the epitopes recognized by 4E10 and 10E8. These results provide critical information for designing the next generation of MPER-based immunogens.
Collapse
Affiliation(s)
- Habtom H Habte
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Saikat Banerjee
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Heliang Shi
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Yali Qin
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Michael W Cho
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA.
| |
Collapse
|
8
|
Baldauf KJ, Royal JM, Hamorsky KT, Matoba N. Cholera toxin B: one subunit with many pharmaceutical applications. Toxins (Basel) 2015; 7:974-96. [PMID: 25802972 PMCID: PMC4379537 DOI: 10.3390/toxins7030974] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/16/2015] [Indexed: 12/22/2022] Open
Abstract
Cholera, a waterborne acute diarrheal disease caused by Vibrio cholerae, remains prevalent in underdeveloped countries and is a serious health threat to those living in unsanitary conditions. The major virulence factor is cholera toxin (CT), which consists of two subunits: the A subunit (CTA) and the B subunit (CTB). CTB is a 55 kD homopentameric, non-toxic protein binding to the GM1 ganglioside on mammalian cells with high affinity. Currently, recombinantly produced CTB is used as a component of an internationally licensed oral cholera vaccine, as the protein induces potent humoral immunity that can neutralize CT in the gut. Additionally, recent studies have revealed that CTB administration leads to the induction of anti-inflammatory mechanisms in vivo. This review will cover the potential of CTB as an immunomodulatory and anti-inflammatory agent. We will also summarize various recombinant expression systems available for recombinant CTB bioproduction.
Collapse
Affiliation(s)
- Keegan J Baldauf
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Joshua M Royal
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY 42303, USA.
| | - Krystal Teasley Hamorsky
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY 42303, USA.
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Nobuyuki Matoba
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY 42303, USA.
| |
Collapse
|
9
|
Lai RPJ, Hock M, Radzimanowski J, Tonks P, Hulsik DL, Effantin G, Seilly DJ, Dreja H, Kliche A, Wagner R, Barnett SW, Tumba N, Morris L, LaBranche CC, Montefiori DC, Seaman MS, Heeney JL, Weissenhorn W. A fusion intermediate gp41 immunogen elicits neutralizing antibodies to HIV-1. J Biol Chem 2014; 289:29912-26. [PMID: 25160627 PMCID: PMC4208001 DOI: 10.1074/jbc.m114.569566] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 08/01/2014] [Indexed: 11/06/2022] Open
Abstract
The membrane-proximal external region (MPER) of the human immunodeficiency virus, type 1 (HIV-1) envelope glycoprotein subunit gp41 is targeted by potent broadly neutralizing antibodies 2F5, 4E10, and 10E8. These antibodies recognize linear epitopes and have been suggested to target the fusion intermediate conformation of gp41 that bridges viral and cellular membranes. Anti-MPER antibodies exert different degrees of membrane interaction, which is considered to be the limiting factor for the generation of such antibodies by immunization. Here we characterize a fusion intermediate conformation of gp41 (gp41(int)-Cys) and show that it folds into an elongated ∼ 12-nm-long extended structure based on small angle x-ray scattering data. Gp41(int)-Cys was covalently linked to liposomes via its C-terminal cysteine and used as immunogen. The gp41(int)-Cys proteoliposomes were administered alone or in prime-boost regimen with trimeric envelope gp140(CA018) in guinea pigs and elicited high anti-gp41 IgG titers. The sera interacted with a peptide spanning the MPER region, demonstrated competition with broadly neutralizing antibodies 2F5 and 4E10, and exerted modest lipid binding, indicating the presence of MPER-specific antibodies. Although the neutralization potency generated solely by gp140(CA018) was higher than that induced by gp41(int)-Cys, the majority of animals immunized with gp41(int)-Cys proteoliposomes induced modest breadth and potency in neutralizing tier 1 pseudoviruses and replication-competent simian/human immunodeficiency viruses in the TZM-bl assay as well as responses against tier 2 HIV-1 in the A3R5 neutralization assay. Our data thus demonstrate that liposomal gp41 MPER formulation can induce neutralization activity, and the strategy serves to improve breadth and potency of such antibodies by improved vaccination protocols.
Collapse
Affiliation(s)
- Rachel P J Lai
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Miriam Hock
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - Jens Radzimanowski
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - Paul Tonks
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - David Lutje Hulsik
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - Gregory Effantin
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - David J Seilly
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Hanna Dreja
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Alexander Kliche
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Susan W Barnett
- Novartis Vaccines and Diagnostics Inc., Cambridge, Massachusetts 02139
| | - Nancy Tumba
- National Institute for Communicable Diseases, Centre for HIV and Sexually Transmitted Infections, 1 Modderfontein Road, Sandringham 2131, South Africa
| | - Lynn Morris
- National Institute for Communicable Diseases, Centre for HIV and Sexually Transmitted Infections, 1 Modderfontein Road, Sandringham 2131, South Africa
| | - Celia C LaBranche
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, and
| | - David C Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, and
| | - Michael S Seaman
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115
| | - Jonathan L Heeney
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom,
| | - Winfried Weissenhorn
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France,
| |
Collapse
|
10
|
Novel mucosal DNA-MVA HIV vaccination in which DNA-IL-12 plus cholera toxin B subunit (CTB) cooperates to enhance cellular systemic and mucosal genital tract immunity. PLoS One 2014; 9:e107524. [PMID: 25215887 PMCID: PMC4162600 DOI: 10.1371/journal.pone.0107524] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/23/2014] [Indexed: 12/22/2022] Open
Abstract
Induction of local antiviral immune responses at the mucosal portal surfaces where HIV-1 and other viral pathogens are usually first encountered remains a primary goal for most vaccines against mucosally acquired viral infections. Exploring mucosal immunization regimes in order to find optimal vector combinations and also appropriate mucosal adjuvants in the HIV vaccine development is decisive. In this study we analyzed the interaction of DNA-IL-12 and cholera toxin B subunit (CTB) after their mucosal administration in DNA prime/MVA boost intranasal regimes, defining the cooperation of both adjuvants to enhance immune responses against the HIV-1 Env antigen. Our results demonstrated that nasal mucosal DNA/MVA immunization schemes can be effectively improved by the co-delivery of DNA-IL-12 plus CTB inducing elevated HIV-specific CD8 responses in spleen and more importantly in genital tract and genito-rectal draining lymph nodes. Remarkably, these CTL responses were of superior quality showing higher avidity, polyfunctionality and a broader cytokine profile. After IL-12+CTB co-delivery, the cellular responses induced showed an enhanced breadth recognizing with higher efficiency Env peptides from different subtypes. Even more, an in vivo CTL cytolytic assay demonstrated the higher specific CD8 T-cell performance after the IL-12+CTB immunization showing in an indirect manner its potential protective capacity. Improvements observed were maintained during the memory phase where we found higher proportions of specific central memory and T memory stem-like cells T-cell subpopulations. Together, our data show that DNA-IL-12 plus CTB can be effectively employed acting as mucosal adjuvants during DNA prime/MVA boost intranasal vaccinations, enhancing magnitude and quality of HIV-specific systemic and mucosal immune responses.
Collapse
|
11
|
Gong Z, Kessans SA, Song L, Dörner K, Lee HH, Meador LR, LaBaer J, Hogue BG, Mor TS, Fromme P. Recombinant expression, purification, and biophysical characterization of the transmembrane and membrane proximal domains of HIV-1 gp41. Protein Sci 2014; 23:1607-18. [PMID: 25155369 DOI: 10.1002/pro.2540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 07/22/2014] [Accepted: 08/20/2014] [Indexed: 11/07/2022]
Abstract
The transmembrane subunit (gp41) of the envelope glycoprotein of HIV-1 associates noncovalently with the surface subunit (gp120) and together they play essential roles in viral mucosal transmission and infection of target cells. The membrane proximal region (MPR) of gp41 is highly conserved and contains epitopes of broadly neutralizing antibodies. The transmembrane (TM) domain of gp41 not only anchors the envelope glycoprotein complex in the viral membrane but also dynamically affects the interactions of the MPR with the membrane. While high-resolution X-ray structures of some segments of the MPR were solved in the past, they represent the post-fusion forms. Structural information on the TM domain of gp41 is scant and at low resolution. Here we describe the design, expression and purification of a protein construct that includes MPR and the transmembrane domain of gp41 (MPR-TMTEV-6His), which reacts with the broadly neutralizing antibodies 2F5 and 4E10 and thereby may represent an immunologically relevant conformation mimicking a prehairpin intermediate of gp41. The expression level of MPR-TMTEV-6His was improved by fusion to the C-terminus of Mistic protein, yielding ∼ 1 mg of pure protein per liter. The isolated MPR-TMTEV-6His protein was biophysically characterized and is a monodisperse candidate for crystallization. This work will enable further investigation into the structure of MPR-TMTEV-6His, which will be important for the structure-based design of a mucosal vaccine against HIV-1.
Collapse
Affiliation(s)
- Zhen Gong
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona, 85287-1604; The Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona, 85287
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Lee HH, Cherni I, Yu H, Fromme R, Doran JD, Grotjohann I, Mittman M, Basu S, Deb A, Dörner K, Aquila A, Barty A, Boutet S, Chapman HN, Doak RB, Hunter MS, James D, Kirian RA, Kupitz C, Lawrence RM, Liu H, Nass K, Schlichting I, Schmidt KE, Seibert MM, Shoeman RL, Spence JCH, Stellato F, Weierstall U, Williams GJ, Yoon C, Wang D, Zatsepin NA, Hogue BG, Matoba N, Fromme P, Mor TS. Expression, purification and crystallization of CTB-MPR, a candidate mucosal vaccine component against HIV-1. IUCRJ 2014; 1:305-17. [PMID: 25295172 PMCID: PMC4174873 DOI: 10.1107/s2052252514014900] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 06/24/2014] [Indexed: 05/03/2023]
Abstract
CTB-MPR is a fusion protein between the B subunit of cholera toxin (CTB) and the membrane-proximal region of gp41 (MPR), the transmembrane envelope protein of Human immunodeficiency virus 1 (HIV-1), and has previously been shown to induce the production of anti-HIV-1 antibodies with antiviral functions. To further improve the design of this candidate vaccine, X-ray crystallography experiments were performed to obtain structural information about this fusion protein. Several variants of CTB-MPR were designed, constructed and recombinantly expressed in Escherichia coli. The first variant contained a flexible GPGP linker between CTB and MPR, and yielded crystals that diffracted to a resolution of 2.3 Å, but only the CTB region was detected in the electron-density map. A second variant, in which the CTB was directly attached to MPR, was shown to destabilize pentamer formation. A third construct containing a polyalanine linker between CTB and MPR proved to stabilize the pentameric form of the protein during purification. The purification procedure was shown to produce a homogeneously pure and monodisperse sample for crystallization. Initial crystallization experiments led to pseudo-crystals which were ordered in only two dimensions and were disordered in the third dimension. Nanocrystals obtained using the same precipitant showed promising X-ray diffraction to 5 Å resolution in femtosecond nanocrystallography experiments at the Linac Coherent Light Source at the SLAC National Accelerator Laboratory. The results demonstrate the utility of femtosecond X-ray crystallography to enable structural analysis based on nano/microcrystals of a protein for which no macroscopic crystals ordered in three dimensions have been observed before.
Collapse
Affiliation(s)
- Ho-Hsien Lee
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Irene Cherni
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| | - HongQi Yu
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Raimund Fromme
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Jeffrey D. Doran
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| | - Ingo Grotjohann
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Michele Mittman
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| | - Shibom Basu
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Arpan Deb
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| | - Katerina Dörner
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Andrew Aquila
- Center for Free-Electron Laser Science, DESY, Notkestrasse 85, 22607 Hamburg, Germany
| | - Anton Barty
- Center for Free-Electron Laser Science, DESY, Notkestrasse 85, 22607 Hamburg, Germany
| | - Sébastien Boutet
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Henry N. Chapman
- Center for Free-Electron Laser Science, DESY, Notkestrasse 85, 22607 Hamburg, Germany
- University of Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - R. Bruce Doak
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Mark S. Hunter
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Daniel James
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Richard A. Kirian
- Center for Free-Electron Laser Science, DESY, Notkestrasse 85, 22607 Hamburg, Germany
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Christopher Kupitz
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Robert M. Lawrence
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| | - Haiguang Liu
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Karol Nass
- Center for Free-Electron Laser Science, DESY, Notkestrasse 85, 22607 Hamburg, Germany
- University of Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Ilme Schlichting
- Max-Planck-Institut für medizinische Forschung, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Kevin E. Schmidt
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - M. Marvin Seibert
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Robert L. Shoeman
- Max-Planck-Institut für medizinische Forschung, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - John C. H. Spence
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Francesco Stellato
- Center for Free-Electron Laser Science, DESY, Notkestrasse 85, 22607 Hamburg, Germany
| | - Uwe Weierstall
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Garth J. Williams
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Chunhong Yoon
- Center for Free-Electron Laser Science, DESY, Notkestrasse 85, 22607 Hamburg, Germany
- European XFEL GmbH, Albert-Einstein-Ring 19, 22761 Hamburg, Germany
| | - Dingjie Wang
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Nadia A. Zatsepin
- Department of Physics, Arizona State University, PO Box 871504, Tempe, AZ 85287-1504, USA
| | - Brenda G. Hogue
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| | - Nobuyuki Matoba
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| | - Petra Fromme
- Department of Chemistry and Biochemistry, Arizona State University, PO Box 871604, Tempe, AZ 85287-1604, USA
| | - Tsafrir S. Mor
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, PO Box 874501, Tempe, AZ 85287-5401, USA
| |
Collapse
|
13
|
Kessans SA, Linhart MD, Matoba N, Mor T. Biological and biochemical characterization of HIV-1 Gag/dgp41 virus-like particles expressed in Nicotiana benthamiana. PLANT BIOTECHNOLOGY JOURNAL 2013; 11:681-90. [PMID: 23506331 PMCID: PMC3688661 DOI: 10.1111/pbi.12058] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/10/2013] [Accepted: 01/27/2013] [Indexed: 05/29/2023]
Abstract
The transmembrane HIV-1 envelope protein gp41 has been shown to play critical roles in the viral mucosal transmission and infection of CD4⁺ cells. Gag is a structural protein configuring the enveloped viral particles and has been suggested to constitute a target of the cellular immunity that may control viral load. We hypothesized that HIV enveloped virus-like particles (VLPs) consisting of Gag and a deconstructed form of gp41 comprising the membrane proximal external, transmembrane and cytoplasmic domains (dgp41) could be expressed in plants. To this end, plant-optimized HIV-1 genes were constructed and expressed in Nicotiana benthamiana by stable transformation, or transiently using a Tobamovirus-based expression system or a combination of both. Our results of biophysical, biochemical and electron microscopy characterization demonstrates that plant cells could support not only the formation of enveloped HIV-1 Gag VLPs, but also the accumulation of VLPs that incorporated dgp41. These findings provide further impetus for the journey towards a broadly efficacious and inexpensive subunit vaccine against HIV-1.
Collapse
Affiliation(s)
- Sarah A Kessans
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
| | - Mark D Linhart
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
| | - Nobuyuki Matoba
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
- Owensboro Cancer Research ProgramOwensboro, KY, USA
- James Graham Brown Cancer Center and Department of Pharmacology & Toxicology, University of Louisville School of MedicineLouisville, KY, USA
| | - Tsafrir Mor
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
| |
Collapse
|
14
|
Hamorsky KT, Kouokam JC, Bennett LJ, Baldauf KJ, Kajiura H, Fujiyama K, Matoba N. Rapid and scalable plant-based production of a cholera toxin B subunit variant to aid in mass vaccination against cholera outbreaks. PLoS Negl Trop Dis 2013; 7:e2046. [PMID: 23505583 PMCID: PMC3591335 DOI: 10.1371/journal.pntd.0002046] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 12/18/2012] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Cholera toxin B subunit (CTB) is a component of an internationally licensed oral cholera vaccine. The protein induces neutralizing antibodies against the holotoxin, the virulence factor responsible for severe diarrhea. A field clinical trial has suggested that the addition of CTB to killed whole-cell bacteria provides superior short-term protection to whole-cell-only vaccines; however, challenges in CTB biomanufacturing (i.e., cost and scale) hamper its implementation to mass vaccination in developing countries. To provide a potential solution to this issue, we developed a rapid, robust, and scalable CTB production system in plants. METHODOLOGY/PRINCIPAL FINDINGS In a preliminary study of expressing original CTB in transgenic Nicotiana benthamiana, the protein was N-glycosylated with plant-specific glycans. Thus, an aglycosylated CTB variant (pCTB) was created and overexpressed via a plant virus vector. Upon additional transgene engineering for retention in the endoplasmic reticulum and optimization of a secretory signal, the yield of pCTB was dramatically improved, reaching >1 g per kg of fresh leaf material. The protein was efficiently purified by simple two-step chromatography. The GM1-ganglioside binding capacity and conformational stability of pCTB were virtually identical to the bacteria-derived original B subunit, as demonstrated in competitive enzyme-linked immunosorbent assay, surface plasmon resonance, and fluorescence-based thermal shift assay. Mammalian cell surface-binding was corroborated by immunofluorescence and flow cytometry. pCTB exhibited strong oral immunogenicity in mice, inducing significant levels of CTB-specific intestinal antibodies that persisted over 6 months. Moreover, these antibodies effectively neutralized the cholera holotoxin in vitro. CONCLUSIONS/SIGNIFICANCE Taken together, these results demonstrated that pCTB has robust producibility in Nicotiana plants and retains most, if not all, of major biological activities of the original protein. This rapid and easily scalable system may enable the implementation of pCTB to mass vaccination against outbreaks, thereby providing better protection of high-risk populations in developing countries.
Collapse
Affiliation(s)
- Krystal Teasley Hamorsky
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - J. Calvin Kouokam
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Lauren J. Bennett
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
| | - Keegan J. Baldauf
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Hiroyuki Kajiura
- International Center for Biotechnology, Osaka University, Osaka, Japan
| | - Kazuhito Fujiyama
- International Center for Biotechnology, Osaka University, Osaka, Japan
| | - Nobuyuki Matoba
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| |
Collapse
|
15
|
|
16
|
Abstract
PURPOSE OF REVIEW The major target groups for an HIV vaccine include breastfeeding infants and adolescents. Differential immune maturity in these age groups may significantly impact vaccine efficacy, and should be taken into account when developing vaccines. Here we review these differences, with an emphasis on the immune response to vaccines for HIV and other pathogens. Recommendations for potential adaptation of current HIV vaccines are also made. RECENT FINDINGS An effective neonatal vaccine needs to be immunogenic in the presence of maternal antibody, and must induce cytotoxic T-lymphocyte responses, neutralizing antibody responses, both systemic and mucosal. There is renewed hope in the possibility of stimulating neutralizing antibodies with HIV vaccination. DNA vaccines are promising for neonates, but will need appropriate boosting. Certain adjuvants and vector delivery systems are more suitable for neonates. Adolescents may have stronger immune responses to HIV vaccines than adults, and will also require induction of mucosal neutralizing humoral and cellular immunity. SUMMARY Some current HIV vaccine strategies may need adaptation for neonates and suitable product development should be accelerated. Vaccines could induce better responses in adolescents and therefore should not be discarded prematurely. Development of vaccines that have potential for these age groups is an urgent global priority.
Collapse
|
17
|
Cox JH, Ferrari MG, Earl P, Lane JR, Jagodzinski LL, Polonis VR, Kuta EG, Boyer JD, Ratto-Kim S, Eller LA, Pham DT, Hart L, Montefiori D, Ferrari G, Parrish S, Weiner DB, Moss B, Kim JH, Birx D, VanCott TC. Inclusion of a CRF01_AE HIV envelope protein boost with a DNA/MVA prime-boost vaccine: Impact on humoral and cellular immunogenicity and viral load reduction after SHIV-E challenge. Vaccine 2012; 30:1830-40. [PMID: 22234262 PMCID: PMC3324265 DOI: 10.1016/j.vaccine.2011.12.131] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 12/21/2011] [Accepted: 12/28/2011] [Indexed: 01/13/2023]
Abstract
The current study assessed the immunogenicity and protective efficacy of various prime-boost vaccine regimens in rhesus macaques using combinations of recombinant DNA (rDNA), recombinant MVA (rMVA), and subunit gp140 protein. The rDNA and rMVA vectors were constructed to express Env from HIV-1 subtype CRF01_AE and Gag-Pol from CRF01_AE or SIVmac 239. One of the rMVAs, MVA/CMDR, has been recently tested in humans. Immunizations were administered at months 0 and 1 (prime) and months 3 and 6 (boost). After priming, HIV env-specific serum IgG was detected in monkeys receiving gp140 alone or rMVA but not in those receiving rDNA. Titers were enhanced in these groups after boosting either with gp140 alone or with rMVA plus gp140. The groups that received the rDNA prime developed env-specific IgG after boosting with rMVA with or without gp140. HIV Env-specific serum IgG binding antibodies were elicited more frequently and of higher titer, and breadth of neutralizing antibodies was increased with the inclusion of the subunit Env boost. T cell responses were measured by tetramer binding to Gag p11c in Mamu-A*01 macaques, and by IFN-γ ELISPOT assay to SIV-Gag. T cell responses were induced after vaccination with the highest responses seen in macaques immunized with rDNA and rMVA. Macaques were challenged intravenously with a novel SHIV-E virus (SIVmac239 Gag-Pol with an HIV-1 subtype E-Env CAR402). Post challenge with SHIV-E, antibody titers were boosted in all groups and peaked at 4 weeks. Robust T cell responses were seen in all groups post challenge and in macaques immunized with rDNA and rMVA a clear boosting of responses was seen. A greater than two-log drop in RNA copies/ml at peak viremia and earlier set point was achieved in macaques primed with rDNA, and boosted with rMVA/SHIV-AE plus gp140. Post challenge viremia in macaques immunized with other regimens was not significantly different to that of controls. These results demonstrate that a gp140 subunit and inclusion of SIV Gag-Pol may be critical for control of SHIV post challenge.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- Antibodies, Neutralizing/blood
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Products, gag/immunology
- Gene Products, pol/immunology
- HIV Antibodies/blood
- HIV-1/immunology
- Immunity, Cellular
- Immunity, Humoral
- Immunization, Secondary
- Immunoglobulin G/blood
- Macaca mulatta
- Male
- Simian Immunodeficiency Virus/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Load
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- env Gene Products, Human Immunodeficiency Virus/immunology
Collapse
|
18
|
Giuliano EA, Finn K. Characterization of membranous (M) cells in normal feline conjunctiva-associated lymphoid tissue (CALT). Vet Ophthalmol 2012; 14 Suppl 1:60-6. [PMID: 21923825 DOI: 10.1111/j.1463-5224.2011.00917.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To characterize conjunctival lymphoid nodules obtained from the nictitans of healthy cats to determine if the follicle-associated epithelium (FAE) of conjunctiva-associated lymphoid tissue (CALT) in this species contains membranous (M)-cells analogous to those described in other regions of mucosa-associated lymphoid tissue (MALT). METHODS Lymphoid follicles from nictitan bulbar surfaces of 10 healthy cats (20 eyes total) were examined. Nictitans from five cats were harvested immediately post-mortem and a minimum of 12 lymphoid nodules from each third eyelid were isolated using a Zeiss operating microscope. At least three lymphoid follicles from each eye were examined using light microscopy (LM), transmission electron microscopy (TEM), and scanning electron microscopy (SEM) using standard fixation and embedding protocols. Nictitan-lymphoid follicles from another five healthy cats were processed for immunohistochemistry to characterize the distribution of T- and B-lymphocytes present beneath the FAE. RESULTS The FAE overlying CALT from 10 healthy cats demonstrated morphology characteristic of M-cells including attenuated apical cell surface with blunted microvilli and microfolds, invaginated basolateral membrane forming a cytoplasmic pocket, and diminished distance between the apical and pocket membrane. Immunohistochemistry of lymphoid tissue subtending the FAE demonstrated B-cell dependent regions in the germinal centers surrounded by T-cell dependent interfollicular zones. CONCLUSIONS Healthy feline CALT contains morphologic features analogous to those described in other regions of MALT. Documentation of feline conjunctival M-cells is of clinical relevance in the study of primary infectious, allergic, and autoimmune ocular diseases, as well as a potential means of vaccination or drug delivery.
Collapse
Affiliation(s)
- Elizabeth A Giuliano
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA.
| | | |
Collapse
|
19
|
Naz RK. Female genital tract immunity: distinct immunological challenges for vaccine development. J Reprod Immunol 2012; 93:1-8. [DOI: 10.1016/j.jri.2011.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 09/27/2011] [Accepted: 09/30/2011] [Indexed: 10/14/2022]
|
20
|
Wang J, Xu L, Tong P, Chen YH. Mucosal antibodies induced by tandem repeat of 2F5 epitope block transcytosis of HIV-1. Vaccine 2011; 29:8542-8. [PMID: 21939723 DOI: 10.1016/j.vaccine.2011.09.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 08/31/2011] [Accepted: 09/09/2011] [Indexed: 11/19/2022]
Abstract
Induction of mucosal antibodies to prevent HIV infection is an important strategy for the HIV-1 prophylaxis. Here we report an epitope-vaccine based antigen that was able to elicit mucosal antibodies capable of blocking HIV-1 transcytosis. Because the ELDKWA epitope of neutralizing antibody 2F5 plays a crucial role in transcytosis, a series of immunogens that contain tandem copies of ELDKWA were prepared. Mice were immunized with these immunogens intranasally, and received intraperitoneal+intranasal boosters. The immunogens that contained more ELDKWA epitopes elicited higher level of mucosal ELDKWA-epitope specific IgAs and systemic IgGs. Although the antisera from the immunized mice exhibited mild neutralizing potency to HIV-1 isolates HXB2 and JRFL, the affinity purified mucosal ELDKWA-epitope specific antibodies could block the transcytosis of cell-free CNE3 (a primary isolate of subtype CRF01_AE) in human tight epithelial models.
Collapse
Affiliation(s)
- Ji Wang
- School of Life Sciences, Tsinghua University, People's Republic of China
| | | | | | | |
Collapse
|
21
|
Matoba N, Shah NR, Mor TS. Humoral immunogenicity of an HIV-1 envelope residue 649-684 membrane-proximal region peptide fused to the plague antigen F1-V. Vaccine 2011; 29:5584-90. [PMID: 21693158 PMCID: PMC3152316 DOI: 10.1016/j.vaccine.2011.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 05/09/2011] [Accepted: 06/07/2011] [Indexed: 12/13/2022]
Abstract
The membrane-proximal region spanning residues 649-684 of the HIV-1 envelope protein gp41 (MPR₆₄₉₋₆₈₄) is an attractive vaccine target for humoral immunity that blocks viral transcytosis across the mucosal epithelia. However, induction of high-titer MPR₆₄₉₋₆₈₄-specific antibodies remains a challenging task. To explore potential solutions for this challenge, we tested a new translational fusion protein comprising the plague F1-V antigen and MPR₆₄₉₋₆₈₄ (F1-V-MPR₆₄₉₋₆₈₄). We employed systemic immunization for initial feasibility analyses. Despite strong immunogenicity demonstrated for the immunogen, repeated systemic immunizations of mice with F1-V-MPR₆₄₉₋₆₈₄ hardly induced MPR₆₄₉₋₆₈₄-specific IgG. In contrast, a single immunization with F1-V-MPR₆₄₉₋₆₈₄ mounted a significant anti-MPR₆₄₉₋₆₈₄ IgG response in animals that were primed with another MPR₆₄₉₋₆₈₄ fusion protein based on the cholera toxin B subunit. Additional boost immunizations with F1-V-MPR₆₄₉₋₆₈₄ recalled and maintained the antibody response and expanded the number of specific antibody-secreting B cells. Thus, while F1-V-MPR₆₄₉₋₆₈₄ alone was not sufficiently immunogenic to induce detectable levels of MPR₆₄₉₋₆₈₄-specific antibodies, these results suggest that prime-boost immunization using heterologous antigen-display platforms may overcome the poor humoral immunogenicity of MPR₆₄₉₋₆₈₄ for the induction of durable humoral immunity. Further studies are warranted to evaluate the feasibility of this strategy in mucosal immunization. Lastly, our findings add to a growing body of evidence in support of this strategy for immunogen design for poorly immunogenic epitopes besides the MPR of HIV-1's transmembrane envelope protein.
Collapse
Affiliation(s)
- Nobuyuki Matoba
- Corresponding authors: Nobuyuki Matoba, Tel: 270 691-5955; Fax: 270 685-5684; ; Tsafrir S. Mor, Tel: 480 727-7405; Fax: 480 727-7615;
| | - Namrata Rahul Shah
- Center for Infectious Diseases and Vaccinology at the Biodesign Institute and School of Life Sciences, P.O. Box 874501, Arizona State University, Tempe, AZ 85287-4501, USA
| | - Tsafrir S Mor
- Center for Infectious Diseases and Vaccinology at the Biodesign Institute and School of Life Sciences, P.O. Box 874501, Arizona State University, Tempe, AZ 85287-4501, USA
| |
Collapse
|
22
|
Girard MP, Osmanov S, Assossou OM, Kieny MP. Human immunodeficiency virus (HIV) immunopathogenesis and vaccine development: a review. Vaccine 2011; 29:6191-218. [PMID: 21718747 DOI: 10.1016/j.vaccine.2011.06.085] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/20/2011] [Accepted: 06/22/2011] [Indexed: 02/08/2023]
Abstract
The development of a safe, effective and globally affordable HIV vaccine offers the best hope for the future control of the HIV-1 pandemic. Since 1987, scores of candidate HIV-1 vaccines have been developed which elicited varying degrees of protective responses in nonhuman primate models, including DNA vaccines, subunit vaccines, live vectored recombinant vaccines and various prime-boost combinations. Four of these candidate vaccines have been tested for efficacy in human volunteers, but, to the exception of the recent RV144 Phase III trial in Thailand, which elicited a modest but statistically significant level of protection against infection, none has shown efficacy in preventing HIV-1 infection or in controlling virus replication and delaying progression of disease in humans. Protection against infection was observed in the RV144 trial, but intensive research is needed to try to understand the protective immune mechanisms at stake. Building-up on the results of the RV144 trial and deciphering what possibly are the immune correlates of protection are the top research priorities of the moment, which will certainly accelerate the development of an highly effective vaccine that could be used in conjunction with other HIV prevention and treatment strategies. This article reviews the state of the art of HIV vaccine development and discusses the formidable scientific challenges met in this endeavor, in the context of a better understanding of the immunopathogenesis of the disease.
Collapse
Affiliation(s)
- Marc P Girard
- University Paris 7, French National Academy of Medicine, 39 rue Seignemartin, FR 69008 Lyon, France.
| | | | | | | |
Collapse
|
23
|
Hong B, Song XT, Rollins L, Berry L, Huang XF, Chen SY. Mucosal and systemic anti-HIV immunity controlled by A20 in mouse dendritic cells. J Clin Invest 2011; 121:739-51. [PMID: 21206085 DOI: 10.1172/jci42656] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 11/03/2010] [Indexed: 11/17/2022] Open
Abstract
Both mucosal and systemic immune responses are required for preventing or containing HIV transmission and chronic infection. However, currently described vaccination approaches are largely ineffective in inducing both mucosal and systemic responses. In this study, we found that the ubiquitin-editing enzyme A20--an inducible feedback inhibitor of the TNFR, RIG-I, and TLR signaling pathways that broadly controls the maturation, cytokine production, and immunostimulatory potency of DCs--restricted systemically immunized DCs to induce both robust mucosal and systemic HIV-specific cellular and humoral responses. Mechanistic studies revealed that A20 regulated DC production of retinoic acid and proinflammatory cytokines, inhibiting the expression of gut-homing receptors on T and B cells. Furthermore, A20-silenced, hyperactivated DCs exhibited an enhanced homing capacity to draining and gut-associated lymphoid tissues (GALTs) after systemic administration. Thus, this study provides insights into the role of A20 in innate immunity. This work may allow the development of an efficient HIV vaccination strategy that is capable of inducing both robust systemic and mucosal anti-HIV cellular and humoral responses.
Collapse
Affiliation(s)
- Bangxing Hong
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | |
Collapse
|
24
|
Pinkhasov J, Alvarez ML, Pathangey LB, Tinder TL, Mason HS, Walmsley AM, Gendler SJ, Mukherjee P. Analysis of a cholera toxin B subunit (CTB) and human mucin 1 (MUC1) conjugate protein in a MUC1-tolerant mouse model. Cancer Immunol Immunother 2010; 59:1801-11. [PMID: 20824430 PMCID: PMC3256590 DOI: 10.1007/s00262-010-0906-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 08/06/2010] [Indexed: 10/19/2022]
Abstract
Since epithelial mucin 1 (MUC1) is associated with several adenocarcinomas at the mucosal sites, it is pertinent to test the efficacy of a mucosally targeted vaccine formulation. The B subunit of the Vibrio cholerae cholera toxin (CTB) has great potential to act as a mucosal carrier for subunit vaccines. In the present study we evaluated whether a MUC1 tandem repeat (TR) peptide chemically linked to CTB would break self-antigen tolerance in the transgenic MUC1-tolerant mouse model (MUC1.Tg) through oral or parenteral immunizations. We report that oral immunization with the CTB-MUC1 conjugate along with mucosal adjuvant, unmethylated CpG oligodeoxynucleotide (ODN) and interleukin-12 (IL-12) did not break self-antigen tolerance in MUC1.Tg mice, but induced a strong humoral response in wild-type C57BL/6 mice. However, self-antigen tolerance in the MUC1.Tg mouse model was broken after parenteral immunizations with different doses of the CTB-MUC1 conjugate protein and with the adjuvant CpG ODN co-delivered with CTB-MUC1. Importantly, mice immunized systemically with CpG ODN alone and with CTB-MUC1 exhibited decreased tumor burden when challenged with a mammary gland tumor cell line that expresses human MUC1.
Collapse
Affiliation(s)
- Julia Pinkhasov
- Center for Infectious Diseases and Vaccinology (CIDV), The Biodesign Institute at Arizona State University, 1001 South McAllister Avenue, Tempe, AZ 85287-4501 USA
| | - M. Lucrecia Alvarez
- Center for Infectious Diseases and Vaccinology (CIDV), The Biodesign Institute at Arizona State University, 1001 South McAllister Avenue, Tempe, AZ 85287-4501 USA
| | - Latha B. Pathangey
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Scottsdale, AZ USA
| | - Teresa L. Tinder
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Scottsdale, AZ USA
| | - Hugh S. Mason
- Center for Infectious Diseases and Vaccinology (CIDV), The Biodesign Institute at Arizona State University, 1001 South McAllister Avenue, Tempe, AZ 85287-4501 USA
| | - Amanda M. Walmsley
- Center for Infectious Diseases and Vaccinology (CIDV), The Biodesign Institute at Arizona State University, 1001 South McAllister Avenue, Tempe, AZ 85287-4501 USA
| | - Sandra J. Gendler
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Scottsdale, AZ USA
| | - Pinku Mukherjee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Scottsdale, AZ USA
- Department of Immunology, Cellular Immunology Laboratory, Mayo Clinic Scottsdale, 13400 E. Shea Blvd, Scottsdale, AZ 85255 USA
- Present Address: Irwin Belk Endowed Professor of Cancer Research, Department of Biology, University of North Carolina Charlotte, 9201 University City Blvd, Charlotte, NC 28223 USA
| |
Collapse
|
25
|
Jain S, Patrick AJ, Rosenthal KL. Multiple tandem copies of conserved gp41 epitopes incorporated in gag virus-like particles elicit systemic and mucosal antibodies in an optimized heterologous vector delivery regimen. Vaccine 2010; 28:7070-80. [DOI: 10.1016/j.vaccine.2010.08.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/17/2010] [Accepted: 08/02/2010] [Indexed: 10/19/2022]
|
26
|
Yu M, Vajdy M. Mucosal HIV transmission and vaccination strategies through oral compared with vaginal and rectal routes. Expert Opin Biol Ther 2010; 10:1181-95. [PMID: 20624114 PMCID: PMC2904634 DOI: 10.1517/14712598.2010.496776] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE OF THE FIELD There are currently over thirty million people infected with HIV and there are no vaccines available to prevent HIV infections or disease. The genitourinary, rectal and oral mucosa are the mucosal HIV transmission routes. An effective vaccine that can induce both systemic and local mucosal immunity is generally accepted as a major means of protection against mucosal HIV transmission and AIDS. WHAT THE READER WILL GAIN Structure and cells that comprise the oral, vaginal and rectal mucosa pertaining to HIV transmission and vaccination strategies through each mucosal route to prevent mucosal and systemic infection will be discussed. AREAS COVERED IN THIS REVIEW Covering publications from 1980s through 2010, mucosal transmission of HIV and current and previous approaches to vaccinations are discussed. TAKE HOME MESSAGE Although oral transmission of HIV is far less common than vaginal and rectal transmissions, infections through this route do occur through oral sex as well as vertically from mother to child. Mucosal vaccination strategies against oral and other mucosal HIV transmissions are under intensive research but the lack of consensus on immune correlates of protection and lack of safe and effective mucosal adjuvants and delivery systems hamper progress towards a licensed vaccine.
Collapse
Affiliation(s)
- Mingke Yu
- EpitoGenesis, Inc., Walnut Creek, CA 94598, USA
| | | |
Collapse
|
27
|
Totrov M, Jiang X, Kong XP, Cohen S, Krachmarov C, Salomon A, Williams C, Seaman MS, Abagyan R, Cardozo T, Gorny MK, Wang S, Lu S, Pinter A, Zolla-Pazner S. Structure-guided design and immunological characterization of immunogens presenting the HIV-1 gp120 V3 loop on a CTB scaffold. Virology 2010; 405:513-23. [PMID: 20663531 DOI: 10.1016/j.virol.2010.06.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 06/04/2010] [Accepted: 06/14/2010] [Indexed: 11/24/2022]
Abstract
V3 loop is a major neutralizing determinant of the HIV-1 gp120. Using 3D structures of cholera toxin B subunit (CTB), complete V3 in the gp120 context, and V3 bound to a monoclonal antibody (mAb), we designed two V3-scaffold immunogen constructs (V3-CTB). The full-length V3-CTB presenting the complete V3 in a structural context mimicking gp120 was recognized by the large majority of our panel of 24 mAbs. The short V3-CTB presenting a V3 fragment in the conformation observed in the complex with the 447-52D Fab, exhibited high-affinity binding to this mAb. The immunogens were evaluated in rabbits using DNA-prime/protein-boost protocol. Boosting with the full-length V3-CTB induced high anti-V3 titers in sera that potently neutralize multiple HIV virus strains. The short V3-CTB was ineffective. The results suggest that very narrow antigenic profile of an immunogen is associated with poor Ab response. An immunogen with broader antigenic activity elicits robust Ab response.
Collapse
Affiliation(s)
- Maxim Totrov
- Molsoft LLC, 3366 N Torrey Pines Ct., La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Immune responses elicited in mice with recombinant Lactococcus lactis expressing F4 fimbrial adhesin FaeG by oral immunization. Vet Res Commun 2010; 34:491-502. [PMID: 20532816 DOI: 10.1007/s11259-010-9418-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2010] [Indexed: 10/19/2022]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major pathogenic agent causing piglet diarrhea. The major subunit and adhesin FaeG of F4(+) ETEC is an important virulence factor with strong immunogenicity. To determine whether Lactococcus lactis (L. lactis) could effectively deliver FaeG to the mucosal immune system, recombinant L. lactis expressing FaeG was constructed, and immune responses in mice following oral route delivery of recombinant L. lactis were explored. The production of FaeG expressed in L. lactis was up to approximately 10% of soluble whole-cell proteins, and recombinant FaeG (rFaeG) possessed good immunoreactivity by Western blot analysis. Oral immunization with recombinant L. lactis expressing FaeG induced F4-specific mucosal and systemic immune responses in the mice. In addition, high dose recombinant L. lactis or co-administration of high dose recombinant L. lactis with CTB enhanced the immune responses. These results suggested that L. lactis expressing FaeG was a promising candidate vaccine against ETEC.
Collapse
|
29
|
Phillips TE, Sharp J, Rodgers K, Liu H. M cell-targeted ocular immunization: effect on immunoglobulins in tears, feces, and serum. Invest Ophthalmol Vis Sci 2009; 51:1533-9. [PMID: 19892871 DOI: 10.1167/iovs.09-4491] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE This study investigates whether antigen-sampling M cells, present in the follicle-associated epithelium (FAE) above organized conjunctiva-associated lymphoid tissue in rabbits, bind and retro-transport secretory IgA (sIgA) from the tear film. The hypothesis that IgA-mediated uptake of antigens promotes local and systemic production of immunoglobulins was tested. METHODS sIgA binding and retro-translocation by M cells was characterized by immunocytochemistry. Immunoglobulin concentrations in tears, feces and serum were measured using enzyme-linked immunoassays (ELISA) after topical and systemic immunization with either goat IgG anti-rabbit IgA or nonspecific goat IgG. RESULTS Endogenous sIgA was found associated with the apical membrane of conjunctival M cells. Exogenous anti-IgA immunoglobulins were translocated across M cells. Significant levels of sIgA against goat IgG were present in tears of pre-immune animals. Topical application of either goat IgG specific for rabbit IgA or nonspecific goat IgG led to similar increases in antigen-specific IgA in tear, feces, and serum. The antigen-specific IgG response in tears mirrored the serum response for both immunogens consistent with transudation of this immunoglobulin. The IgM response in tears and serum was weak for both immunogens. Systemic immunization did not sustain or enhance the local mucosal IgA responses. CONCLUSIONS Conjunctival M cells bind and translocate sIgA from the tear film. Topical conjunctival immunization leads to generation of antigen-specific immunoglobulins from both local and distant mucosae and in serum. Natural antibodies, present in the tear film before immunization, may have contributed to similar immune responses to goat anti-rabbit IgA and nonspecific goat IgG.
Collapse
Affiliation(s)
- Thomas E Phillips
- Division of Biological Sciences, University of Missouri, Columbia, Missouri 65211-7400, USA.
| | | | | | | |
Collapse
|
30
|
Li S, Zheng W, KuoLee R, Hirama T, Henry M, Makvandi-Nejad S, Fjällman T, Chen W, Zhang J. Pentabody-mediated antigen delivery induces antigen-specific mucosal immune response. Mol Immunol 2009; 46:1718-26. [DOI: 10.1016/j.molimm.2009.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 01/29/2009] [Accepted: 02/02/2009] [Indexed: 11/30/2022]
|
31
|
Abstract
HIV vaccine research is at a crossroads carefully contemplating on the next path. The unexpected results of the Merck vaccine trial, while providing a stunning blow to a field in dire need of a protective vaccine, has also raised several fundamental questions regarding the candidate immunogen itself, preexisting immunity to vaccine vectors, surrogate assays and animal models used for assessing preclinical protective responses, as well as relevant endpoints to be measured in a clinical trial. As a result, the research community is faced with the daunting task of identifying novel vaccine concepts and products to continue the search. This review highlights and addresses some of the scientific and practical concerns.
Collapse
|
32
|
Broad neutralization of human immunodeficiency virus type 1 (HIV-1) elicited from human rhinoviruses that display the HIV-1 gp41 ELDKWA epitope. J Virol 2009; 83:5087-100. [PMID: 19279101 DOI: 10.1128/jvi.00184-09] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In efforts to develop AIDS vaccine components, we generated combinatorial libraries of recombinant human rhinoviruses that display the well-conserved ELDKWA epitope of the membrane-proximal external region of human immunodeficiency virus type 1 (HIV-1) gp41. The broadly neutralizing human monoclonal antibody 2F5 was used to select for viruses whose ELDKWA conformations resemble those of HIV. Immunization of guinea pigs with different chimeras, some boosted with ELDKWA-based peptides, elicited antibodies capable of neutralizing HIV-1 pseudoviruses of diverse subtypes and coreceptor usages. These recombinant immunogens are the first reported that elicit broad, albeit modest, neutralization of HIV-1 using an ELDKWA-based epitope and are among the few reported that elicit broad neutralization directed against any recombinant HIV epitope, providing a critical advance in developing effective AIDS vaccine components.
Collapse
|
33
|
Matoba N, Kajiura H, Cherni I, Doran JD, Bomsel M, Fujiyama K, Mor TS. Biochemical and immunological characterization of the plant-derived candidate human immunodeficiency virus type 1 mucosal vaccine CTB-MPR. PLANT BIOTECHNOLOGY JOURNAL 2009; 7:129-45. [PMID: 19037902 DOI: 10.1111/j.1467-7652.2008.00381.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Plants are potentially the most economical platforms for the large-scale production of recombinant proteins. Thus, plant-based expression of subunit human immunodeficiency virus type 1 (HIV-1) vaccines provides an opportunity for their global use against the acquired immunodeficiency syndrome pandemic. CTB-MPR(649-684)[CTB, cholera toxin B subunit; MPR, membrane proximal (ectodomain) region of gp41] is an HIV-1 vaccine candidate that has been shown previously to induce antibodies that block a pathway of HIV-1 mucosal transmission. In this article, the molecular characterization of CTB-MPR(649-684) expressed in transgenic Nicotiana benthamiana plants is reported. Virtually all of the CTB-MPR(649-684) proteins expressed in the selected line were shown to have assembled into pentameric, GM1 ganglioside-binding complexes. Detailed biochemical analyses on the purified protein revealed that it was N-glycosylated, predominantly with high-mannose-type glycans (more than 75%), as predicted from a consensus asparagine-X-serine/threonine (Asn-X-Ser/Thr) N-glycosylation sequon on the CTB domain and an endoplasmic reticulum retention signal attached at the C-terminus of the fusion protein. Despite this modification, the plant-expressed protein retained the nanomolar affinity to GM1 ganglioside and the critical antigenicity of the MPR(649-684) moiety. Furthermore, the protein induced mucosal and serum anti-MPR(649-684) antibodies in mice after mucosal prime-systemic boost immunization. Our data indicate that plant-based expression can be a viable alternative for the production of this subunit HIV-1 vaccine candidate.
Collapse
Affiliation(s)
- Nobuyuki Matoba
- Center for Infectious Diseases and Vaccinology at the Biodesign Institute and School of Life Sciences, PO Box 874501, Arizona State University, Tempe, AZ 85287-4501, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Coûtant J, Yu H, Clément M, Alfsen A, Toma F, Curmi PA, Bomsel M. Both lipid environment and pH are critical for determining physiological solution structure of 3‐D‐conserved epitopes of the HIV‐1 gp41‐MPER peptide P1. FASEB J 2008; 22:4338-51. [DOI: 10.1096/fj.08-113142] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jérôme Coûtant
- Structure Activité des Biomolécules Normales et PathologiquesINSERM/UEVE U829 Université d'EvryEvryFrance
| | - Huifeng Yu
- Entrée Muqueuse du VIH et Immunite Muqueuse, Departement de Biologie CellulaireInstitut Cochin, Université Paris Descartes, CNRS UMR 8104ParisFrance
- INSERM U567ParisFrance
| | - Marie‐Jeanne Clément
- Structure Activité des Biomolécules Normales et PathologiquesINSERM/UEVE U829 Université d'EvryEvryFrance
| | - Annette Alfsen
- Entrée Muqueuse du VIH et Immunite Muqueuse, Departement de Biologie CellulaireInstitut Cochin, Université Paris Descartes, CNRS UMR 8104ParisFrance
- INSERM U567ParisFrance
| | - Flavio Toma
- Structure Activité des Biomolécules Normales et PathologiquesINSERM/UEVE U829 Université d'EvryEvryFrance
| | - Patrick A. Curmi
- Structure Activité des Biomolécules Normales et PathologiquesINSERM/UEVE U829 Université d'EvryEvryFrance
| | - Morgane Bomsel
- Entrée Muqueuse du VIH et Immunite Muqueuse, Departement de Biologie CellulaireInstitut Cochin, Université Paris Descartes, CNRS UMR 8104ParisFrance
- INSERM U567ParisFrance
| |
Collapse
|
35
|
Edwards KA, Duan F, Baeumner AJ, March JC. Fluorescently labeled liposomes for monitoring cholera toxin binding to epithelial cells. Anal Biochem 2008; 380:59-67. [DOI: 10.1016/j.ab.2008.05.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Accepted: 05/18/2008] [Indexed: 11/16/2022]
|
36
|
Dorosko SM, Ayres SL, Connor RI. Induction of HIV-1 MPR(649-684)-specific IgA and IgG antibodies in caprine colostrum using a peptide-based vaccine. Vaccine 2008; 26:5416-22. [PMID: 18708113 DOI: 10.1016/j.vaccine.2008.07.079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 07/28/2008] [Accepted: 07/29/2008] [Indexed: 12/21/2022]
Abstract
Induction of antigen-specific antibodies against HIV-1 in colostrum and milk may help prevent breast milk transmission of the virus. A peptide vaccine against the HIV-1 gp41 membrane proximal region (MPR(649-684)) was evaluated as proof-of-principle in a caprine model. Pregnant Alpine/Saanen goats were immunized with MPR(649-684) peptide conjugated to KLH using alum adjuvant. Immunizations were intramuscular, intranasal, and in the supramammary lymph node region. Samples collected after parturition demonstrated the presence of MPR(649-684)-specific antibodies in colostrum and serum. These results support the concept that a peptide vaccine can effectively induce MPR(649-684)-specific sIgA and IgG in the colostrum of a lactating species.
Collapse
Affiliation(s)
- Stephanie M Dorosko
- Department of Microbiology & Immunology, Dartmouth Medical School, 1 Medical Center Drive, Lebanon, NH 03756, USA.
| | | | | |
Collapse
|
37
|
The membrane-proximal external region of the human immunodeficiency virus type 1 envelope: dominant site of antibody neutralization and target for vaccine design. Microbiol Mol Biol Rev 2008; 72:54-84, table of contents. [PMID: 18322034 DOI: 10.1128/mmbr.00020-07] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Enormous efforts have been made to produce a protective vaccine against human immunodeficiency virus type 1; there has been little success. However, the identification of broadly neutralizing antibodies against epitopes on the highly conserved membrane-proximal external region (MPER) of the gp41 envelope protein has delineated this region as an attractive vaccine target. Furthermore, emerging structural information on the MPER has provided vaccine designers with new insights for building relevant immunogens. This review describes the current state of the field regarding (i) the structure and function of the gp41 MPER; (ii) the structure and binding mechanisms of the broadly neutralizing antibodies 2F5, 4E10, and Z13; and (iii) the development of an MPER-targeting vaccine. In addition, emerging approaches to vaccine design are presented.
Collapse
|
38
|
Yu H, Alfsen A, Tudor D, Bomsel M. The binding of HIV-1 gp41 membrane proximal domain to its mucosal receptor, galactosyl ceramide, is structure-dependent. Cell Calcium 2008; 43:73-82. [PMID: 17553560 DOI: 10.1016/j.ceca.2007.04.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2006] [Revised: 04/04/2007] [Indexed: 01/19/2023]
Abstract
The peptide of HIV-1 envelope gp41 (a.a 628-683), referred to herein as P5, contains P1, a conserved galactose-specific lectin domain for binding the mucosal HIV-1-receptor, galactosyl ceramide (GalCer), as shown earlier, and a potential calcium-binding site (a.a 628-648). P1 contains contiguous epitopes recognized by the broadly neutralizing antibodies 2F5, 4E10, Z13. However, similar neutralizing antibodies could not be raised in animal model using immunogens based on these epitopes. We now show that the structure of both P5 and P1 peptides, as measured by circular dichroism, differs according to their environment: aqueous or lipidic, and as a function of calcium concentration. P5, but not P1, binds to calcium with a low binding affinity constant in the order of 2.5x10(4). Calcium binding results in a conformational change of P5, leading in turn to a decrease in affinity for GalCer. Hence, the affinity of the gp41-lectin site for the galactose harbored by the mucosal HIV-1 receptor GalCer is modulated by the peptide secondary and tertiary structure and the local environment. Therefore, definition of the conformation of this novel extended gp41 membrane proximal region, containing the conserved peptide P1 and the Ca(2+) binding site, could help designing an immunogen efficient at inducing neutralizing anti-HIV-1 antibodies.
Collapse
Affiliation(s)
- Huifeng Yu
- Entrée Muqueuse du VIH et Immunité Muqueuse, Département de Biologie Cellulaire, Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), 22 rue Méchain, 75014 Paris, France
| | | | | | | |
Collapse
|
39
|
Bock R. Plastid biotechnology: prospects for herbicide and insect resistance, metabolic engineering and molecular farming. Curr Opin Biotechnol 2007; 18:100-6. [PMID: 17169550 DOI: 10.1016/j.copbio.2006.12.001] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 11/15/2006] [Accepted: 12/01/2006] [Indexed: 10/23/2022]
Abstract
Transgene expression from the chloroplast (plastid) genome offers several attractions to plant biotechnologists, including high-level accumulation of foreign proteins, transgene stacking in operons and a lack of epigenetic interference with the stability of transgene expression. In addition, the technology provides an environmentally benign method of plant genetic engineering, because plastids and their genetic information are maternally inherited in most crops and thus are largely excluded from pollen transmission. During the past few years, researchers in both the public and private sectors have begun to explore possible areas of application of plastid transformation in plant biotechnology as a viable alternative to conventional nuclear transgenic technologies. Recent proof-of-concept studies highlight the potential of plastid genome engineering for the expression of resistance traits, the production of biopharmaceuticals and metabolic pathway engineering in plants.
Collapse
Affiliation(s)
- Ralph Bock
- Max-Planck-Institut für Molekulare Pflanzenphysiologie, Am Mühlenberg 1, D-14476 Potsdam-Golm, Germany.
| |
Collapse
|
40
|
Magérus-Chatinet A, Yu H, Garcia S, Ducloux E, Terris B, Bomsel M. Galactosyl ceramide expressed on dendritic cells can mediate HIV-1 transfer from monocyte derived dendritic cells to autologous T cells. Virology 2007; 362:67-74. [PMID: 17234232 DOI: 10.1016/j.virol.2006.11.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 09/22/2006] [Accepted: 11/28/2006] [Indexed: 10/23/2022]
Abstract
Mucosa, comprising epithelial and dendritic cells, are the major sites for Human Immunodeficiency Virus type 1 (HIV-1) transmission. There, DCs can capture incoming HIV-1 and in turn transfer virus to CD4(+) T lymphocytes in a two-phase process, thereby initiating HIV-1 dissemination. We show that the glycosphingolipid Galactosyl Ceramide (GalCer), acting as mucosal epithelial receptor for HIV-1, was expressed by human monocyte derived immature DCs (iDCs), human primary DCs isolated from blood and mucosal tissue and in situ on mucosal tissue and acts as HIV-1-gp41 receptor. Blocking both GalCer and CD4 with specific mAbs results in a >95% transfer inhibition of HIV-1 from human monocyte-derived iDCs to autologous resting T cells. GalCer interaction with HIV-1 controls the early infection-independent phase of HIV-1 transfer to T cells. Thus, GalCer appears as an initial receptor for HIV-1, common to both mucosal epithelial cells and iDCs.
Collapse
Affiliation(s)
- Aude Magérus-Chatinet
- Entrée Muqueuse du VIH et immunité muqueuse (Mucosal entry of HIV-1 and mucosal immunity), Departement de Biologie Cellulaire (Cell Biology Department), Institut Cochin, CNRS, INSERM, Université René Descartes, 22 rue Mechain, 75014 Paris, France
| | | | | | | | | | | |
Collapse
|
41
|
Youm JW, Won YS, Jeon JH, Ryu CJ, Choi YK, Kim HC, Kim BD, Joung H, Kim HS. Oral immunogenicity of potato-derived HBsAg middle protein in BALB/c mice. Vaccine 2006; 25:577-84. [PMID: 16945456 DOI: 10.1016/j.vaccine.2006.05.131] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Revised: 05/23/2006] [Accepted: 05/24/2006] [Indexed: 11/29/2022]
Abstract
The antibodies to preS2 synthetic peptides have been probed to neutralize hepatitis B virus (HBV), and also the addition of preS2 sequence could enhance the antibody response compared with a conventional vaccine in the non- and low responders. Previously, we generated transgenic potatoes expressing middle protein, which contains additional 55 amino acid preS2 region at the N-terminus of the S protein, of HBV to determine the feasibility of developing a plant-delivered HBV vaccine. In this study, we monitored the immune response after induction of immunoglobulin by boosting and assessed the efficacy of the mucosal immune response with regard to generate IgA antibodies. The HBsAg middle protein expressed in our transgenic potatoes was well immunized at low antigenic quantities in mice and the induced anti-S or anti-preS2 antibodies were sustained for the whole period without decrease. Orally delivery of plant-derived HBsAg middle protein to mice resulted in fecal anti-S or anti-preS2 as well as serum IgG. In addition, we used antibodies induced from the immunized mice with the potato-derived rHBsAg in competition assay as competitors to confirm the binding ability of preS2 antibodies to surface antigen of hepatitis virus. Anti-preS2 antibodies induced from immunized mice with transgenic potatoes effectively competed with anti-preS2 murine antibody H8 as expected. From these results, the inclusion of preS2 antigen to HBV plant vaccine may provide additional protective immunity in the HBV prevention.
Collapse
Affiliation(s)
- Jung-Won Youm
- Plant Genomics Research Center, KRIBB, 52 Oun-Dong, Yusung-Gu, Daejon 305-333, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|