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
|
Chakraborty S, Dutta P, Pal A, Chakraborty S, Banik G, Halder P, Gope A, Miyoshi SI, Das S. Intranasal immunization of mice with chimera of Salmonella Typhi protein elicits protective intestinal immunity. NPJ Vaccines 2024; 9:24. [PMID: 38321067 PMCID: PMC10847434 DOI: 10.1038/s41541-024-00812-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/26/2024] [Indexed: 02/08/2024] Open
Abstract
Development of safe, highly effective and affordable enteric fever vaccines is a global health priority. Live, oral typhoid vaccines induce strong mucosal immunity and long-term protection, but safety remains a concern. In contrast, efficacy wears off rapidly for injectable, polysaccharide-based vaccines, which elicit poor mucosal response. We previously reported Salmonella Typhi outer membrane protein, T2544 as a potential candidate for bivalent (S. Typhi and S. Paratyphi A) vaccine development. Here, we show that intranasal immunization with a subunit vaccine (chimera of T2544 and cholera toxin B subunit) induced strong systemic and intestinal mucosal immunity and protection from S. Typhi challenge in a mouse model. CTB-T2544 augmented gut-homing receptor expression on lymphocytes that produced Th1 and Th17 cytokines, secretory IgA in stool that inhibited bacterial motility and epithelial attachment, antibody recall response and affinity maturation with increased number of follicular helper T cells and CD4+ central and effector memory cells.
Collapse
Affiliation(s)
- Suparna Chakraborty
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Pujarini Dutta
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
- Department of Pediatrics, Steele Children's Research Center, University of Arizona, Tuscon, AZ, USA
| | - Ananda Pal
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Swarnali Chakraborty
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - George Banik
- BD Biosciences, INDIA, Smart works Business Center, Victoria Park, 37/2 GN Block, Sector 5, Saltlake City, Kolkata, 700091, India
| | - Prolay Halder
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Animesh Gope
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Shin-Ichi Miyoshi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Collaborative Research Center of Okayama University for Infectious Diseases at Indian Council of Medical Research-National Institute of Cholera and Enteric Diseases, Kolkata, 700010, India
| | - Santasabuj Das
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India.
- ICMR-National Institute of Occupational Health, Meghaninagar, Ahmedabad, 3800016, Gujarat, India.
| |
Collapse
|
3
|
Plant-Derived Recombinant Vaccines against Zoonotic Viruses. Life (Basel) 2022; 12:life12020156. [PMID: 35207444 PMCID: PMC8878793 DOI: 10.3390/life12020156] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Emerging and re-emerging zoonotic diseases cause serious illness with billions of cases, and millions of deaths. The most effective way to restrict the spread of zoonotic viruses among humans and animals and prevent disease is vaccination. Recombinant proteins produced in plants offer an alternative approach for the development of safe, effective, inexpensive candidate vaccines. Current strategies are focused on the production of highly immunogenic structural proteins, which mimic the organizations of the native virion but lack the viral genetic material. These include chimeric viral peptides, subunit virus proteins, and virus-like particles (VLPs). The latter, with their ability to self-assemble and thus resemble the form of virus particles, are gaining traction among plant-based candidate vaccines against many infectious diseases. In this review, we summarized the main zoonotic diseases and followed the progress in using plant expression systems for the production of recombinant proteins and VLPs used in the development of plant-based vaccines against zoonotic viruses.
Collapse
|
4
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|
5
|
Morris DA, Reeves MA, Royal JM, Hamorsky KT, Matoba N. Isolation and detection of a KDEL-tagged recombinant cholera toxin B subunit from Nicotiana benthamiana. Process Biochem 2020; 101:42-49. [PMID: 33304198 DOI: 10.1016/j.procbio.2020.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Here we describe refined methods for the isolation and detection of a KDEL-tagged, plant-produced recombinant cholera toxin B subunit (CTB) that exhibits unique mucosal wound healing activity. The protein was transiently overexpressed in Nicotiana benthamiana, which generates some C-terminal KDEL truncated molecular species that are deficient in epithelial repair activity. With a new CHT chromatographical method described herein, these product-derived impurities were successfully separated from CTB with the intact KDEL sequence, as confirmed by mass spectrometry. In addition, an immunoassay capable of specifically detecting GM1 ganglioside-binding CTB with intact KDEL sequences was developed. Coupled together, these methods will aid in the quality control of KDEL-attached CTB produced in plant-based manufacturing systems towards a novel topical biotherapeutic for the treatment of acute and chronic mucosal inflammation.
Collapse
Affiliation(s)
- David A Morris
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.,Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Micaela A Reeves
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Joshua M Royal
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.,Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Krystal T Hamorsky
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.,Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Medicine, University of Louisville School of Medicine, Louisville, KY
| | - Nobuyuki Matoba
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.,Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
6
|
New GMP manufacturing processes to obtain thermostable HIV-1 gp41 virosomes under solid forms for various mucosal vaccination routes. NPJ Vaccines 2020; 5:41. [PMID: 32435515 PMCID: PMC7235025 DOI: 10.1038/s41541-020-0190-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/28/2020] [Indexed: 01/02/2023] Open
Abstract
The main objective of the MACIVIVA European consortium was to develop new Good Manufacturing Practice pilot lines for manufacturing thermostable vaccines with stabilized antigens on influenza virosomes as enveloped virus-like particles. The HIV-1 gp41-derived antigens anchored in the virosome membrane, along with the adjuvant 3M-052 (TLR7/8 agonist) on the same particle, served as a candidate vaccine for the proof of concept for establishing manufacturing processes, which can be directly applied or adapted to other virosomal vaccines or lipid-based particles. Heat spray-dried powders suitable for nasal or oral delivery, and freeze-dried sublingual tablets were successfully developed as solid dosage forms for mucosal vaccination. The antigenic properties of vaccinal antigens with key gp41 epitopes were maintained, preserving the original immunogenicity of the starting liquid form, and also when solid forms were exposed to high temperature (40 °C) for up to 3 months, with minimal antigen and adjuvant content variation. Virosomes reconstituted from the powder forms remained as free particles with similar size, virosome uptake by antigen-presenting cells in vitro was comparable to virosomes from the liquid form, and the presence of excipients specific to each solid form did not prevent virosome transport to the draining lymph nodes of immunized mice. Virosome integrity was also preserved during exposure to <−15 °C, mimicking accidental freezing conditions. These “ready to use and all-in-one” thermostable needle-free virosomal HIV-1 mucosal vaccines offer the advantage of simplified logistics with a lower dependence on the cold chain during shipments and distribution.
Collapse
|
7
|
Tremouillaux-Guiller J, Moustafa K, Hefferon K, Gaobotse G, Makhzoum A. Plant-made HIV vaccines and potential candidates. Curr Opin Biotechnol 2020; 61:209-216. [PMID: 32058899 DOI: 10.1016/j.copbio.2020.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/19/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
Abstract
Millions of people around the world suffer from heavy social and health burdens related to HIV/AIDS and its associated opportunistic infections. To reduce these burdens, preventive and therapeutic vaccines are required. Effective HIV vaccines have been under investigation for several decades using different animal models. Potential plant-made HIV vaccine candidates have also gained attention in the past few years. In addition to this, broadly neutralizing antibodies produced in plants which can target conserved viral epitopes and neutralize mutating HIV strains have been identified. Numerous epitopes of envelope glycoproteins and capsid proteins of HIV-1 are a part of HIV therapy. Here, we discuss some recent findings aiming to produce anti-HIV-1 recombinant proteins in engineered plants for AIDS prophylactics and therapeutic treatments.
Collapse
Affiliation(s)
| | | | | | - Goabaone Gaobotse
- Department of Biological Sciences & Biotechnology, Botswana International University of Science & Technology, Botswana
| | - Abdullah Makhzoum
- Department of Biological Sciences & Biotechnology, Botswana International University of Science & Technology, Botswana.
| |
Collapse
|
8
|
Ji J, Sundquist J, Sundquist K. Cholera Vaccine Use Is Associated With a Reduced Risk of Death in Patients With Colorectal Cancer: A Population-Based Study. Gastroenterology 2018; 154:86-92.e1. [PMID: 28923497 DOI: 10.1053/j.gastro.2017.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/01/2017] [Accepted: 09/11/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Cholera toxin can act as a modulator of the immune response with anti-inflammatory effects; it reduces development of colon polyps in mouse models of colorectal cancer (CRC). We performed a population-based study to determine whether, in patients with a diagnosis of CRC, subsequent administration of the cholera vaccine (killed Vibrio cholerae O1 whole cells and recombinant cholera toxin B subunit) affects mortality. METHODS We identified patients from the Swedish Cancer Register who were diagnosed with CRC from July 2005 through December 2012. These patients were linked to the Swedish Prescribed Drug Register to retrieve cholera vaccine use. We used Cox regression analysis to calculate the hazard ratio (HR) of death from CRC and overall mortality in patients with post-diagnostic use of cholera vaccine compared with matched controls. RESULTS A total of 175 patients were diagnosed with CRC and given a prescription for the cholera vaccine after their cancer diagnosis. Compared with propensity score-matched controls and adjusted for confounding factors, patients with CRC who received the cholera vaccine had a decreased risk of death from CRC (HR, 0.53; 95% CI, 0.29-0.99) and a decreased risk of death overall (HR, 0.59; 95% CI, 0.37-0.94). The decrease in mortality with cholera vaccination was largely observed, irrespective of patient age or tumor stage at diagnosis or sex. CONCLUSIONS In a population-based study, we associated administration of the cholera vaccine after CRC diagnosis with decreased risk of death from CRC and overall mortality.
Collapse
Affiliation(s)
- Jianguang Ji
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden.
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
9
|
Meador LR, Kessans SA, Kilbourne J, Kibler KV, Pantaleo G, Roderiguez ME, Blattman JN, Jacobs BL, Mor TS. A heterologous prime-boosting strategy with replicating Vaccinia virus vectors and plant-produced HIV-1 Gag/dgp41 virus-like particles. Virology 2017; 507:242-256. [PMID: 28458036 PMCID: PMC5529300 DOI: 10.1016/j.virol.2017.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/24/2017] [Accepted: 04/06/2017] [Indexed: 12/22/2022]
Abstract
Showing modest efficacy, the RV144 HIV-1 vaccine clinical trial utilized a non-replicating canarypox viral vector and a soluble gp120 protein boost. Here we built upon the RV144 strategy by developing a novel combination of a replicating, but highly-attenuated Vaccinia virus vector, NYVAC-KC, and plant-produced HIV-1 virus-like particles (VLPs). Both components contained the full-length Gag and a membrane anchored truncated gp41 presenting the membrane proximal external region with its conserved broadly neutralizing epitopes in the pre-fusion conformation. We tested different prime/boost combinations of these components in mice and showed that the group primed with NYVAC-KC and boosted with both the viral vectors and plant-produced VLPs have the most robust Gag-specific CD8 T cell responses, at 12.7% of CD8 T cells expressing IFN-γ in response to stimulation with five Gag epitopes. The same immunization group elicited the best systemic and mucosal antibody responses to Gag and dgp41 with a bias towards IgG1.
Collapse
Affiliation(s)
- Lydia R Meador
- Ira A. Fulton School of Engineering, Arizona State University, Tempe, AZ, USA; Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Sarah A Kessans
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Jacquelyn Kilbourne
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Karen V Kibler
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland; Swiss Vaccine Research Institute, Lausanne, Switzerland
| | | | - Joseph N Blattman
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Bertram L Jacobs
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| | - Tsafrir S Mor
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
10
|
Deb A, Johnson WA, Kline AP, Scott BJ, Meador LR, Srinivas D, Martin-Garcia JM, Dörner K, Borges CR, Misra R, Hogue BG, Fromme P, Mor TS. Bacterial expression, correct membrane targeting and functional folding of the HIV-1 membrane protein Vpu using a periplasmic signal peptide. PLoS One 2017; 12:e0172529. [PMID: 28225803 PMCID: PMC5321405 DOI: 10.1371/journal.pone.0172529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/06/2017] [Indexed: 12/04/2022] Open
Abstract
Viral protein U (Vpu) is a type-III integral membrane protein encoded by Human Immunodeficiency Virus-1 (HIV- 1). It is expressed in infected host cells and plays several roles in viral progeny escape from infected cells, including down-regulation of CD4 receptors. But key structure/function questions remain regarding the mechanisms by which the Vpu protein contributes to HIV-1 pathogenesis. Here we describe expression of Vpu in bacteria, its purification and characterization. We report the successful expression of PelB-Vpu in Escherichia coli using the leader peptide pectate lyase B (PelB) from Erwinia carotovora. The protein was detergent extractable and could be isolated in a very pure form. We demonstrate that the PelB signal peptide successfully targets Vpu to the cell membranes and inserts it as a type I membrane protein. PelB-Vpu was biophysically characterized by circular dichroism and dynamic light scattering experiments and was shown to be an excellent candidate for elucidating structural models.
Collapse
Affiliation(s)
- Arpan Deb
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - William A. Johnson
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Alexander P. Kline
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Boston J. Scott
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, 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
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Dustin Srinivas
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jose M. Martin-Garcia
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Katerina Dörner
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Chad R. Borges
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Personal Diagnostics, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Rajeev Misra
- School of Life Sciences, 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
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Petra Fromme
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
- The Biodesign Center for Applied Structural Discovery, 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
- The Biodesign Center for Immunotherapy, Vaccines, and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
11
|
Wiedinger K, Pinho D, Bitsaktsis C. Utilization of cholera toxin B as a mucosal adjuvant elicits antibody-mediated protection against S. pneumoniae infection in mice. THERAPEUTIC ADVANCES IN VACCINES 2017; 5:15-24. [PMID: 28344805 DOI: 10.1177/2051013617691041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/07/2016] [Indexed: 11/16/2022]
Abstract
BACKGOUND The introduction of the pneumococcal conjugate and polysaccharide vaccines have been valuable tools for combating invasive pneumococcal infection in children and healthy adults. Despite the available vaccination strategies, pneumococcal pneumonia and associated diseases continue to cause substantial morbidity and mortality, particularly in individuals with chronic disease and ageing populations. Next-generation pneumococcal vaccines will need to be highly immunogenic across patient populations providing both mucosal and systemic protective immunity. Mucosal immunization is an effective strategy for stimulating the immune response at the site of pathogen entry while increasing systemic immunity. In this study we utilized intranasal immunization with pneumococcal surface protein A (PspA), in combination with the mucosal adjuvant cholera toxin B (CTB), to characterize the immune components providing protection against S. pneumoniae challenge. METHODS Mice were immunized intranasally with CTB and PspA individually, and in combination, followed by lethal bacterial challenge with S. pneumoniae, strain A66.1. Animals were monitored for survival and tested for lung bacterial burden, cytokine production as well as S. pneumoniae-specific antibody titer in mouse sera. The primary immunological contributor to the observed protection was confirmed by cytokine neutralization and serum passive transfer. RESULTS The combination of CTB and PspA provided complete protection against bacterial challenge, which coincided with a significant decrease in lung bacterial burden. Increases in the T-helper (Th) 1 cytokines, interferon (IFN)-γ and interleukin (IL)-2 were observed in the lung 24 h post-challenge while decreases in proinflammatory mediators IL-6 and tumor necrosis factor (TNF)-α were also recorded at the same time point. The adjuvanted PspA immunization induced significant titers of S. pneumoniae-specific antibody in the serum of mice prior to infection. Serum adoptive transfer passively protected animals against subsequent challenge while IFN-γ neutralization had no impact on the outcome of immunization, suggesting a primary role for antibody-mediated protection in the context of this immunization strategy. CONCLUSION Mucosal immunization with CTB and PspA induced a local cellular immune response and systemic humoral immunity which resulted in effective reduction of pulmonary bacterial burden and complete protection against S. pneumoniae challenge. While induction of the pleiotropic cytokine IFN-γ likely contributes to control of infection through activation of effector pathways, it was not required for protection. Instead, immunization with PspA and CTB-induced S. pneumoniae-specific antibodies in the serum prior to infection that were sufficient to protect against mucosal challenge.
Collapse
|
12
|
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
|
13
|
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
|
14
|
Shen R, Achenbach J, Shen Y, Palaia J, Rahkola JT, Nick HJ, Smythies LE, McConnell M, Fowler MG, Smith PD, Janoff EN. Mother-to-Child HIV-1 Transmission Events Are Differentially Impacted by Breast Milk and Its Components from HIV-1-Infected Women. PLoS One 2015; 10:e0145150. [PMID: 26680219 PMCID: PMC4683070 DOI: 10.1371/journal.pone.0145150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/01/2015] [Indexed: 11/21/2022] Open
Abstract
Breast milk is a vehicle of infection and source of protection in post-natal mother-to-child HIV-1 transmission (MTCT). Understanding the mechanism by which breast milk limits vertical transmission will provide critical insight into the design of preventive and therapeutic approaches to interrupt HIV-1 mucosal transmission. However, characterization of the inhibitory activity of breast milk in human intestinal mucosa, the portal of entry in postnatal MTCT, has been constrained by the limited availability of primary mucosal target cells and tissues to recapitulate mucosal transmission ex vivo. Here, we characterized the impact of skimmed breast milk, breast milk antibodies (Igs) and non-Ig components from HIV-1-infected Ugandan women on the major events of HIV-1 mucosal transmission using primary human intestinal cells and tissues. HIV-1-specific IgG antibodies and non-Ig components in breast milk inhibited the uptake of Ugandan HIV-1 isolates by primary human intestinal epithelial cells, viral replication in and transport of HIV-1- bearing dendritic cells through the human intestinal mucosa. Breast milk HIV-1-specific IgG and IgA, as well as innate factors, blocked the uptake and transport of HIV-1 through intestinal mucosa. Thus, breast milk components have distinct and complementary effects in reducing HIV-1 uptake, transport through and replication in the intestinal mucosa and, therefore, likely contribute to preventing postnatal HIV-1 transmission. Our data suggests that a successful preventive or therapeutic approach would require multiple immune factors acting at multiple steps in the HIV-1 mucosal transmission process.
Collapse
Affiliation(s)
- Ruizhong Shen
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail: (ENJ); (RS)
| | - Jenna Achenbach
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
| | - Yue Shen
- Department of Biological Sciences, Auburn University, Auburn, Alabama United States of America
| | - Jana Palaia
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
| | - Jeremy T. Rahkola
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
- Denver Veterans Affairs Medical Center, Denver, Colorado, United States of America
| | - Heidi J. Nick
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lesley E. Smythies
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Michelle McConnell
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mary G. Fowler
- The Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - Phillip D. Smith
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Veterans Affairs Medical Center, Birmingham, Alabama, United States of America
| | - Edward N. Janoff
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
- Denver Veterans Affairs Medical Center, Denver, Colorado, United States of America
- * E-mail: (ENJ); (RS)
| |
Collapse
|
15
|
Cholera Toxin Subunit B as Adjuvant--An Accelerator in Protective Immunity and a Break in Autoimmunity. Vaccines (Basel) 2015; 3:579-96. [PMID: 26350596 PMCID: PMC4586468 DOI: 10.3390/vaccines3030579] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/07/2015] [Accepted: 07/20/2015] [Indexed: 11/16/2022] Open
Abstract
Cholera toxin subunit B (CTB) is the nontoxic portion of cholera toxin. Its affinity to the monosialotetrahexosylganglioside (GM1) that is broadly distributed in a variety of cell types including epithelial cells of the gut and antigen presenting cells, macrophages, dendritic cells, and B cells, allows its optimal access to the immune system. CTB can easily be expressed on its own in a variety of organisms, and several approaches can be used to couple it to antigens, either by genetic fusion or by chemical manipulation, leading to strongly enhanced immune responses to the antigens. In autoimmune diseases, CTB has the capacity to evoke regulatory responses and to thereby dampen autoimmune responses, in several but not all animal models. It remains to be seen whether the latter approach translates to success in the clinic, however, the versatility of CTB to manipulate immune responses in either direction makes this protein a promising adjuvant for vaccine development.
Collapse
|
16
|
Rubio-Infante N, Govea-Alonso DO, Romero-Maldonado A, García-Hernández AL, Ilhuicatzi-Alvarado D, Salazar-González JA, Korban SS, Rosales-Mendoza S, Moreno-Fierros L. A Plant-Derived Multi-HIV Antigen Induces Broad Immune Responses in Orally Immunized Mice. Mol Biotechnol 2015; 57:662-74. [PMID: 25779638 DOI: 10.1007/s12033-015-9856-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Multi-HIV, a multiepitopic protein derived from both gp120 and gp41 envelope proteins of the human immunodeficiency virus (HIV), has been proposed as a vaccine prototype capable of inducing broad immune responses, as it carries various B and T cell epitopes from several HIV strains. In this study, the immunogenic properties of a Multi-HIV expressed in tobacco chloroplasts are evaluated in test mice. BALB/c mice orally immunized with tobacco-derived Multi-HIV have elicited antibody responses, including both the V3 loop of gp120 and the ELDKWA epitope of gp41. Based on splenocyte proliferation assays, stimulation with epitopes of the C4, V3 domain of gp120, and the ELDKWA domain of gp41 elicits positive cellular responses. Furthermore, specific interferon gamma production is observed in both CD4+ and CD8+ T cells stimulated with HIV peptides. These results demonstrate that plant-derived Multi-HIV induces T helper-specific responses. Altogether, these findings illustrate the immunogenic potential of plant-derived Multi-HIV in an oral immunization scheme. The potential of this low-cost immunization approach and its implications on HIV/AIDS vaccine development are discussed.
Collapse
Affiliation(s)
- Néstor Rubio-Infante
- Inmunidad en Mucosas, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, 54090, Tlalnepantla, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Roques E, Lessard M, Archambault D. The Cholera Toxin B Subunit (CTB) Fused to the Porcine Arterivirus Matrix M and GP5 Envelope Proteins Fails to Enhance the GP5-Specific Antibody Response in Pigs Immunized with Adenovectors. Mol Biotechnol 2015; 57:701-8. [PMID: 25801418 DOI: 10.1007/s12033-015-9861-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is an arterivirus of the Arteriviridae family. As the current commercial vaccines are incompletely protective effective against PRRSV infection, we developed a vaccine strategy using replicating but non-disseminating adenovectors (rAdVs) expressing the PRRSV M matrix protein in fusion with the neutralizing major epitope-carrying GP5 envelope protein (Roques et al. in Vet Res 44:17, 2013). Although production of GP5-specific antibodies (Abs) was observed, no PRRSV-specific neutralizing Abs (NAbs) were induced in pigs given the rAdVs expressing M-GP5 or M-GP5m (GP5m being a mutant form of GP5). Nevertheless, partial protection was observed in the M-GP5m-rAdV-inoculated pigs experimentally infected with PRRSV. Here, we determined the impact of the cholera toxin B subunit (CTB, known for its adjuvant effect) in fusion with the C-terminus of M-GP5m on the Ab response to PRRSV. Three-week-old pigs were immunized twice both intramuscularly and intranasally at 3-week intervals with rAdV-expressing the green fluorescent protein (rAdV-GFP), rAdV-M-GP5m, or rAdV-M-GP5m-CTB. Pigs immunized with rAdV-M-GP5m showed a high level of serum GP5-specific Abs (as determined by an indirect ELISA). In contrast, CTB in fusion with M-GP5m had an unexpected severe negative impact on GP5-specific Ab production. PRRSV-specific NAbs could not be detected in any pigs of all groups.
Collapse
Affiliation(s)
- Elodie Roques
- Department of Biological Sciences, University of Québec at Montréal, Succursale Centre-Ville, P.O. Box 8888, Montreal, QC, H3C 3P8, Canada
| | | | | |
Collapse
|
18
|
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: 132] [Impact Index Per Article: 13.2] [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
|
19
|
Hamorsky KT, Kouokam JC, Jurkiewicz JM, Nelson B, Moore LJ, Husk AS, Kajiura H, Fujiyama K, Matoba N. N-glycosylation of cholera toxin B subunit in Nicotiana benthamiana: impacts on host stress response, production yield and vaccine potential. Sci Rep 2015; 5:8003. [PMID: 25614217 PMCID: PMC4303877 DOI: 10.1038/srep08003] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 12/16/2014] [Indexed: 01/01/2023] Open
Abstract
Plant-based transient overexpression systems enable rapid and scalable production of subunit vaccines. Previously, we have shown that cholera toxin B subunit (CTB), an oral cholera vaccine antigen, is N-glycosylated upon expression in transgenic Nicotiana benthamiana. Here, we found that overexpression of aglycosylated CTB by agroinfiltration of a tobamoviral vector causes massive tissue necrosis and poor accumulation unless retained in the endoplasmic reticulum (ER). However, the re-introduction of N-glycosylation to its original or an alternative site significantly relieved the necrosis and provided a high CTB yield without ER retention. Quantitative gene expression analysis of PDI, BiP, bZIP60, SKP1, 26Sα proteasome and PR1a, and the detection of ubiquitinated CTB polypeptides revealed that N-glycosylation significantly relieved ER stress and hypersensitive response, and facilitated the folding/assembly of CTB. The glycosylated CTB (gCTB) was characterized for potential vaccine use. Glycan profiling revealed that gCTB contained approximately 38% plant-specific glycans. gCTB retained nanomolar affinity to GM1-ganglioside with only marginal reduction of physicochemical stability and induced an anti-cholera holotoxin antibody response comparable to native CTB in a mouse oral immunization study. These findings demonstrated gCTB's potential as an oral immunogen and point to a potential role of N-glycosylation in increasing recombinant protein yields in plants.
Collapse
Affiliation(s)
- Krystal Teasley Hamorsky
- 1] Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY, USA [2] Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - J Calvin Kouokam
- 1] Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY, USA [2] Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jessica M Jurkiewicz
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY, USA
| | - Bailey Nelson
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY, USA
| | - Lauren J Moore
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY, USA
| | - Adam S Husk
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY, USA
| | - Hiroyuki Kajiura
- The International Center for Biotechnology, Osaka University, Osaka, Japan
| | - Kazuhito Fujiyama
- The International Center for Biotechnology, Osaka University, Osaka, Japan
| | - Nobuyuki Matoba
- 1] Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY, USA [2] Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
20
|
Mucosal Vaccines from Plant Biotechnology. Mucosal Immunol 2015. [PMCID: PMC7158328 DOI: 10.1016/b978-0-12-415847-4.00065-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of plants for production of recombinant proteins has evolved over the past 25 years. The first plant-based vaccines were expressed in stably transgenic plants, with the idea to conveniently deliver “edible vaccines” by ingestion of the antigen-containing plant material. These systems provided a proof of concept that oral delivery of vaccines in crude plant material could stimulate antigen-specific serum and mucosal antibodies. Transgenic grains like rice in particular provide a stable and robust vehicle for antigen delivery. However, some issues exist with stably transgenic plants, including relatively low expression levels and regulatory issues. Thus, many recent studies use transient expression with plant viral vectors to achieve rapid high expression in Nicotiana benthamiana, followed by purification of antigen and intranasal delivery for effective stimulation of mucosal immune responses.
Collapse
|
21
|
Abstract
Plant-made or "biofarmed" viral vaccines are some of the earliest products of the technology of plant molecular farming, and remain some of the brightest prospects for the success of this field. Proofs of principle and of efficacy exist for many candidate viral veterinary vaccines; the use of plant-made viral antigens and of monoclonal antibodies for therapy of animal and even human viral disease is also well established. This review explores some of the more prominent recent advances in the biofarming of viral vaccines and therapies, including the recent use of ZMapp for Ebolavirus infection, and explores some possible future applications of the technology.
Collapse
Affiliation(s)
- Edward P Rybicki
- Biopharming Research Unit, Department of Molecular & Cell Biology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Private Bag X3, Rondebosch, 7701, Cape Town, South Africa.
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Gorantala J, Grover S, Rahi A, Chaudhary P, Rajwanshi R, Sarin NB, Bhatnagar R. Generation of protective immune response against anthrax by oral immunization with protective antigen plant-based vaccine. J Biotechnol 2014; 176:1-10. [PMID: 24548460 DOI: 10.1016/j.jbiotec.2014.01.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/30/2013] [Accepted: 01/29/2014] [Indexed: 01/04/2023]
Abstract
In concern with frequent recurrence of anthrax in endemic areas and inadvertent use of its spores as biological weapon, the development of an effective anthrax vaccine suitable for both human and veterinary needs is highly desirable. A simple oral delivery through expression in plant system could offer promising alternative to the current methods that rely on injectable vaccines extracted from bacterial sources. In the present study, we have expressed protective antigen (PA) gene in Indian mustard by Agrobacterium-mediated transformation and in tobacco by plastid transformation. Putative transgenic lines were verified for the presence of transgene and its expression by molecular analysis. PA expressed in transgenic lines was biologically active as evidenced by macrophage lysis assay. Intraperitoneal (i.p.) and oral immunization with plant PA in murine model indicated high serum PA specific IgG and IgA antibody titers. PA specific mucosal immune response was noted in orally immunized groups. Further, antibodies indicated lethal toxin neutralizing potential in-vitro and conferred protection against in-vivo toxin challenge. Oral immunization experiments demonstrated generation of immunoprotective response in mice. Thus, our study examines the feasibility of oral PA vaccine expressed in an edible plant system against anthrax.
Collapse
Affiliation(s)
- Jyotsna Gorantala
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sonam Grover
- Molecular Technology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Amit Rahi
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Prerna Chaudhary
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ravi Rajwanshi
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Neera Bhalla Sarin
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Rubio-Infante N, Govea-Alonso DO, Alpuche-Solís ÁG, García-Hernández AL, Soria-Guerra RE, Paz-Maldonado LMT, Ilhuicatzi-Alvarado D, Varona-Santos JT, Verdín-Terán L, Korban SS, Moreno-Fierros L, Rosales-Mendoza S. A chloroplast-derived C4V3 polypeptide from the human immunodeficiency virus (HIV) is orally immunogenic in mice. PLANT MOLECULAR BIOLOGY 2012; 78:337-49. [PMID: 22228408 DOI: 10.1007/s11103-011-9870-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 12/10/2011] [Indexed: 05/23/2023]
Abstract
Although the human immunodeficiency virus (HIV) causes one of the most important infectious diseases worldwide, attempts to develop an effective vaccine remain elusive. Designing recombinant proteins capable of eliciting significant and protective mammalian immune responses remain a priority. Moreover, large-scale production of proteins of interest at affordable cost remains a challenge for modern biotechnology. In this study, a synthetic gene encoding a C4V3 recombinant protein, known to induce systemic and mucosal immune responses in mammalian systems, has been introduced into tobacco chloroplasts to yield high levels of expression. Integration of the transgene into the tobacco plastome has been verified by Southern blot hybridization. The recombinant C4V3 protein is also detected in tobacco chloroplasts by confocal microscopy. Reactivity of the heterologous protein with both an anti-C4V3 rabbit serum as well as sera from HIV positive patients have been assayed using Western blots. When administered by the oral route in a four-weekly dose immunization scheme, the plant-derived C4V3 has elicited both systemic and mucosal antibody responses in BALB/c mice, as well as CD4+ T cell proliferation responses. These findings support the viability of using plant chloroplasts as biofactories for HIV candidate vaccines, and could serve as important vehicles for the development of a plant-based candidate vaccine against HIV.
Collapse
Affiliation(s)
- Néstor Rubio-Infante
- Laboratorio de Biofarmacéuticos Recombinantes, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, 78210 San Luis Potosí, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Rosales-Mendoza S, Rubio-Infante N, Govea-Alonso DO, Moreno-Fierros L. Current status and perspectives of plant-based candidate vaccines against the human immunodeficiency virus (HIV). PLANT CELL REPORTS 2012; 31:495-511. [PMID: 22159962 DOI: 10.1007/s00299-011-1194-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/03/2011] [Accepted: 11/18/2011] [Indexed: 05/31/2023]
Abstract
Genetically engineered plants are economical platforms for the large-scale production of recombinant proteins and have been used over the last 21 years as models for oral vaccines against a wide variety of human infectious and autoimmune diseases with promising results. The main inherent advantages of this approach consist in the absence of purification needs and easy production and administration. One relevant infectious agent is the human immunodeficiency virus (HIV), since AIDS evolved as an alarming public health problem implicating very high costs for government agencies in most African and developing countries. The design of an effective and inexpensive vaccine able to limit viral spread and neutralizing the viral entry is urgently needed. Due to the limited efficacy of the vaccines assessed in clinical trials, new HIV vaccines able to generate broad immune profiles are a priority in the field. This review discusses the current advances on the topic of using plants as alternative expression systems to produce functional vaccine components against HIV, including antigens from Env, Gag and early proteins such as Tat and Nef. Ongoing projects of our group based on the expression of chimeric proteins comprising C4 and V3 domains from gp120, as an approach to elicit broadly neutralizing antibodies are mentioned. The perspectives of the revised approaches, such as the great need of assessing the oral immunogenicity and a detailed immunological characterization of the elicited immune responses, are also discussed.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos recombinantes, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico.
| | | | | | | |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Abstract
Recombinant protein pharmaceuticals are now widely used in treatment of chronic diseases, and several recombinant protein subunit vaccines are approved for human and veterinary use. With growing demand for complex protein pharmaceuticals, such as monoclonal antibodies, manufacturing capacity is becoming limited. There is increasing need for safe, scalable, and economical alternatives to mammalian cell culture-based manufacturing systems, which require substantial capital investment for new manufacturing facilities. Since a seminal paper reporting immunoglobulin expression in transgenic plants was published in 1989, there have been many technological advances in plant expression systems to the present time where production of proteins in leaf tissues of nonfood crops such as Nicotiana species is considered a viable alternative. In particular, transient expression systems derived from recombinant plant viral vectors offer opportunities for rapid expression screening, construct optimization, and expression scale-up. Extraction of recombinant proteins from Nicotiana leaf tissues can be achieved by collection of secreted protein fractions, or from a total protein extract after grinding the leaves with buffer. After separation from solids, the major purification challenge is contamination with elements of the photosynthetic complex, which can be solved by application of a variety of facile and proven strategies. In conclusion, the technologies required for safe, efficient, scalable manufacture of recombinant proteins in Nicotiana leaf tissues have matured to the point where several products have already been tested in phase I clinical trials and will soon be followed by a rich pipeline of recombinant vaccines, microbicides, and therapeutic proteins.
Collapse
|
32
|
Granell A, Fernández del-Carmen A, Orzáez D. In planta production of plant-derived and non-plant-derived adjuvants. Expert Rev Vaccines 2010; 9:843-58. [PMID: 20673009 DOI: 10.1586/erv.10.80] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recombinant antigen production in plants is a safe and economically sound strategy for vaccine development, particularly for oral/mucosal vaccination, but subunit vaccines usually suffer from weak immunogenicity and require adjuvants that escort the antigens, target them to relevant sites and/or activate antigen-presenting cells for elicitation of protective immunity. Genetic fusions of antigens with bacterial adjuvants as the B subunit of the cholera toxin have been successful in inducing protective immunity of plant-made vaccines. In addition, several plant compounds, mainly plant defensive molecules as lectins and saponins, have shown strong adjuvant activities. The molecular diversity of the plant kingdom offers a vast source of non-bacterial compounds with adjuvant activity, which can be assayed in emerging plant manufacturing systems for the design of new plant vaccine formulations.
Collapse
Affiliation(s)
- Antonio Granell
- Instituto de Biología Molecular y Celular de Plantas, CSIC-Universidad Politécnica de Valencia, Spain
| | | | | |
Collapse
|
33
|
Salyaev RK, Rigano MM, Rekoslavskaya NI. Development of plant-based mucosal vaccines against widespread infectious diseases. Expert Rev Vaccines 2010; 9:937-46. [PMID: 20673015 DOI: 10.1586/erv.10.81] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mucosal vaccination is a perspective for the control of infectious diseases, since it is capable of inducing humoral and cell-mediated responses. In addition, the delivery of vaccines to mucosal surfaces makes immunization practice safe and acceptable, and eliminates needle-associated risks. Transgenic plants can be used as bioreactors for the production of mucosally delivered protective antigens. This technology shows great promise to simplify and decrease the cost of vaccine delivery. Herein, we review the development of mucosally administered vaccines expressed in transgenic plants. In particular, we evaluate the advantages and disadvantages of using plants for the production of mucosal vaccines against widespread infectious diseases such as HIV, hepatitis B and TB.
Collapse
Affiliation(s)
- Rurick K Salyaev
- Siberian Institute of Plant Physiology and Biochemistry of The Siberian Branch of the RAS, Irkutsk, Russia.
| | | | | |
Collapse
|
34
|
Rybicki EP. Plant-made vaccines for humans and animals. PLANT BIOTECHNOLOGY JOURNAL 2010; 8:620-37. [PMID: 20233333 PMCID: PMC7167690 DOI: 10.1111/j.1467-7652.2010.00507.x] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 11/30/2009] [Accepted: 12/02/2009] [Indexed: 05/17/2023]
Abstract
The concept of using plants to produce high-value pharmaceuticals such as vaccines is 20 years old this year and is only now on the brink of realisation as an established technology. The original reliance on transgenic plants has largely given way to transient expression; proofs of concept for human and animal vaccines and of efficacy for animal vaccines have been established; several plant-produced vaccines have been through Phase I clinical trials in humans and more are scheduled; regulatory requirements are more clear than ever, and more facilities exist for manufacture of clinic-grade materials. The original concept of cheap edible vaccines has given way to a realisation that formulated products are required, which may well be injectable. The technology has proven its worth as a means of cheap, easily scalable production of materials: it now needs to find its niche in competition with established technologies. The realised achievements in the field as well as promising new developments will be reviewed, such as rapid-response vaccines for emerging viruses with pandemic potential and bioterror agents.
Collapse
Affiliation(s)
- Edward P Rybicki
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, South Africa. ed.rybicki@ uct.ac.za
| |
Collapse
|
35
|
Chia MY, Hsiao SH, Chan HT, Do YY, Huang PL, Chang HW, Tsai YC, Lin CM, Pang VF, Jeng CR. The immunogenicity of DNA constructs co-expressing GP5 and M proteins of porcine reproductive and respiratory syndrome virus conjugated by GPGP linker in pigs. Vet Microbiol 2010; 146:189-99. [PMID: 20570063 DOI: 10.1016/j.vetmic.2010.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 04/26/2010] [Accepted: 05/03/2010] [Indexed: 01/13/2023]
Abstract
The heterodimer of glycoprotein 5 (GP5) and non-glycosylated matrix protein (M) is the leading target for the development of new generation of vaccines against porcine reproductive and respiratory syndrome virus (PRRSV) infection. It has been demonstrated that DNA vaccine co-expressing GP5 and M proteins as a fusion protein aroused better immunogenicity than that expressing GP5 or M alone, but it was no better than the DNA vaccine co-expressing GP5 and M proteins with two different promoters. Altered natural conformation of the co-expressed GP5 and M fusion protein was considered as the major cause. Glycine-proline-glycine-proline (GPGP) linker can minimize the conformational changes in tertiary structure and provide flexibility of the peptide chain. The objective of this study was to evaluate whether the immunogenicity of DNA constructs co-expressing GP5 and M proteins linked by GPGP could be enhanced in pigs. Three recombinant DNA constructs expressing GP5/M fusion protein without GPGP linker (pcDNA-56), GP5/M fusion protein conjugated by GPGP linker (pcDNA-5L6), and M/GP5 fusion protein conjugated by GPGP linker (pcDNA-6L5) were established. Sixteen PRRSV-free pigs were randomly assigned to four groups and inoculated intramuscularly with 3 consecutive doses of 500 μg of empty vector pcDNA3.1, pcDNA-56, pcDNA-5L6 or pcDNA-6L5 each at a 2-week interval followed by challenge with 5 × 10(5) TCID(50) PRRSV at 3 weeks after the final inoculation. All pcDNA-56-, pcDNA-5L6-, and pcDNA-6L5- but not pcDNA-3.1-inoculated pigs developed neutralizing antibodies (NAs) 3 weeks after the final inoculation and a gradual increase in NA titers after PRRSV challenge, indicating that pigs inoculated with these DNA constructs could establish a sufficient immune memory. The pcDNA-5L6- and pcDNA-6L5-inoculated pigs displayed lower level and shorter period of viremia and lower tissue viral load following PRRSV challenge than did the pcDNA-56-inoculated pigs. The strategy of co-expressing GPGP-linked GP5 and M fusion protein may be a promising approach for future PRRSV vaccine development, possibly via the improvement of natural conformation of the target fusion protein.
Collapse
Affiliation(s)
- Min-Yuan Chia
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, Taipei 106, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Azizi A, Ghunaim H, Diaz-Mitoma F, Mestecky J. Mucosal HIV vaccines: A holy grail or a dud? Vaccine 2010; 28:4015-26. [DOI: 10.1016/j.vaccine.2010.04.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 03/08/2010] [Accepted: 04/05/2010] [Indexed: 12/13/2022]
|
37
|
Shen R, Drelichman ER, Bimczok D, Ochsenbauer C, Kappes JC, Cannon JA, Tudor D, Bomsel M, Smythies LE, Smith PD. GP41-specific antibody blocks cell-free HIV type 1 transcytosis through human rectal mucosa and model colonic epithelium. THE JOURNAL OF IMMUNOLOGY 2010; 184:3648-55. [PMID: 20208001 DOI: 10.4049/jimmunol.0903346] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Monostratified epithelial cells translocate HIV type 1 (HIV-1) from the apical to the basolateral surface via vesicular transcytosis. Because acutely transmitted HIV-1 is almost exclusively CCR5-tropic and human intestinal epithelial cells preferentially transcytose CCR5-tropic virus, we established epithelial monolayers using polarized HT-29 cells transduced to express CCR5, and an explant system using normal human rectal mucosa, to characterize biological parameters of epithelial cell transcytosis of HIV-1 and assess antiviral Ab blockade of transcytosis. The amount of cell-free HIV-1 transcytosed through the epithelial monolayer increased linearly in relation to the amount of virus applied to the apical surface, indicating transcytosis efficiency was constant (r(2) = 0.9846; p < 0.0001). The efficiency of HIV-1 transcytosis ranged between 0.05 and 1.21%, depending on the virus strain, producer cell type and gp120 V1-V3 loop signature. Inoculation of HIV-1 neutralizing Abs to the immunodominant region (7B2) or the conserved membrane proximal external region (2F5) of gp41 or to cardiolipin (IS4) onto the apical surface of epithelial monolayers prior to inoculation of virus significantly reduced HIV-1 transcytosis. 2F5 was the most potent of these IgG1 Abs. Dimeric IgA and monomeric IgA, but not polymeric IgM, 2F5 Abs also blocked HIV-1 transcytosis across the epithelium and, importantly, across explanted normal human rectal mucosa, with monomeric IgA substantially more potent than dimeric IgA in effecting transcytosis blockade. These findings underscore the potential role of transcytosis blockade in the prevention of HIV-1 transmission across columnar epithelium such as that of the rectum.
Collapse
Affiliation(s)
- Ruizhong Shen
- Division of Gastroenterology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Mestecky J, Moldoveanu Z, Smith PD, Hel Z, Alexander RC. Mucosal immunology of the genital and gastrointestinal tracts and HIV-1 infection. J Reprod Immunol 2009; 83:196-200. [PMID: 19853927 DOI: 10.1016/j.jri.2009.07.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 04/16/2009] [Accepted: 07/06/2009] [Indexed: 01/07/2023]
Abstract
The male and female genital tracts are protected by a local immune system that displays features distinguishing them from other mucosal sites. In contrast to the intestinal tract, where locally produced IgA is the dominant Ig, secretions of the male and female genital tract contain predominantly IgG of both local and systemic origin. Genital tract tissues also lack mucosal lymphoepithelial inductive sites analogous to intestinal Peyer's patches; consequently, local immunization or infections with sexually transmitted pathogens induce low immune responses. Human immunodeficiency virus 1 (HIV-1) infection must be primarily considered as a mucosal disease with extensive involvement of the systemic immune compartment. Although the majority of infections is acquired through the genital mucosa, a high rate of virus replication and profound CD4(+) T cell depletion occurs in the intestinal mucosa and other mucosal tissues shortly after infection. Evaluation of HIV-specific antibodies in sera and external secretions, including vaginal washes and semen, unexpectedly revealed a selective lack of IgA responses. Moreover, specific antibody-secreting cells in peripheral blood were of the IgG isotype, even in mucosally infected individuals. Whether humoral responses to previously or newly encountered antigens are compromised in HIV-1-infected persons is under current investigation.
Collapse
Affiliation(s)
- Jiri Mestecky
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | | | | | | |
Collapse
|
40
|
Tudor D, Derrien M, Diomede L, Drillet AS, Houimel M, Moog C, Reynes JM, Lopalco L, Bomsel M. HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG-seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4(+) cell infection: an IgA gene and functional analysis. Mucosal Immunol 2009; 2:412-26. [PMID: 19587640 DOI: 10.1038/mi.2009.89] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
AIDS is mainly a sexually transmitted disease, and accordingly, mucosal tissues are the primary sites of natural human immunodeficiency virus type-1 (HIV-1) transmission. Mucosal immunoglobulin A (IgA) antibody specific for HIV-1 envelope gp41 subunit is one correlate of protection in individuals who are highly sexually exposed to HIV-1 but remain persistently IgG seronegative (HEPS). Understanding these peculiar IgAs at the gene and functional level is possible only with monoclonal IgAs. We have constructed a mucosal Fab IgA library from HEPS and have characterized a series of HIV-1 IgAs specific for gp41 that, in vitro, are transcytosis-blocking and infection-neutralizing. Characterization of their IgA genes shows that Fab specific for the gp41 membrane-proximal region harbors a long heavy-chain CDR3 loop (CDRH3) similar to the two broadly neutralizing IgG monoclonal antibodies, 2F5 and 4E10. Furthermore, the selected Fab IgA shows extensive somatic mutations that cluster in the CDR regions, indicating that affinity maturation due to an antigen-driven process had occurred in HEPS individuals, presumably upon multiple exposures to HIV. This analysis of HEPS monoclonal IgA gives a unique opportunity to correlate an antibody function (resistance to a pathogen in vivo) with an antibody gene. Such neutralizing monoclonal IgAs could be used in microbicide formulation.
Collapse
Affiliation(s)
- D Tudor
- Entrée Muqueuse du VIH et Immunité Muqueuse, (Mucosal Entry of HIV-1 and Mucosal Immunity), Departement de Biologie Cellulaire, (Cell Biology Department), Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
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
|
42
|
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
|
43
|
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
|
44
|
Marusic C, Vitale A, Pedrazzini E, Donini M, Frigerio L, Bock R, Dix PJ, McCabe MS, Bellucci M, Benvenuto E. Plant-based strategies aimed at expressing HIV antigens and neutralizing antibodies at high levels. Nef as a case study. Transgenic Res 2009; 18:499-512. [PMID: 19169897 PMCID: PMC2758358 DOI: 10.1007/s11248-009-9244-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 01/06/2009] [Indexed: 12/31/2022]
Abstract
The first evidence that plants represent a valid, safe and cost-effective alternative to traditional expression systems for large-scale production of antigens and antibodies was described more than 10 years ago. Since then, considerable improvements have been made to increase the yield of plant-produced proteins. These include the use of signal sequences to target proteins to different cellular compartments, plastid transformation to achieve high transgene dosage, codon usage optimization to boost gene expression, and protein fusions to improve recombinant protein stability and accumulation. Thus, several HIV/SIV antigens and neutralizing anti-HIV antibodies have recently been successfully expressed in plants by stable nuclear or plastid transformation, and by transient expression systems based on plant virus vectors or Agrobacterium-mediated infection. The current article gives an overview of plant expressed HIV antigens and antibodies and provides an account of the use of different strategies aimed at increasing the expression of the accessory multifunctional HIV-1 Nef protein in transgenic plants.
Collapse
Affiliation(s)
- Carla Marusic
- Dipartimento BAS, Sezione Genetica e Genomica Vegetale, ENEA, C.R. Casaccia, via Anguillarese 301, 00123, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Plant-produced vaccines: promise and reality. Drug Discov Today 2008; 14:16-24. [PMID: 18983932 DOI: 10.1016/j.drudis.2008.10.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 09/30/2008] [Accepted: 10/06/2008] [Indexed: 11/21/2022]
Abstract
Plant-produced vaccines are a much-hyped development of the past two decades, whose time to embrace reality may have finally come. Vaccines have been developed against viral, bacterial, parasite and allergenic antigens, for humans and for animals; a wide variety of plants have been used for stable transgenic expression as well as for transient expression via Agrobacterium tumefaciens and plant viral vectors. A great many products have shown significant immunogenicity; several have shown efficacy in target animals or in animal models. The realised potential of plant-produced vaccines is discussed, together with future prospects for production and registration.
Collapse
|
46
|
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
|
47
|
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
|
48
|
Meyers A, Chakauya E, Shephard E, Tanzer FL, Maclean J, Lynch A, Williamson AL, Rybicki EP. Expression of HIV-1 antigens in plants as potential subunit vaccines. BMC Biotechnol 2008; 8:53. [PMID: 18573204 PMCID: PMC2443125 DOI: 10.1186/1472-6750-8-53] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 06/23/2008] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) has infected more than 40 million people worldwide, mainly in sub-Saharan Africa. The high prevalence of HIV-1 subtype C in southern Africa necessitates the development of cheap, effective vaccines. One means of production is the use of plants, for which a number of different techniques have been successfully developed. HIV-1 Pr55Gag is a promising HIV-1 vaccine candidate: we compared the expression of this and a truncated Gag (p17/p24) and the p24 capsid subunit in Nicotiana spp. using transgenic plants and transient expression via Agrobacterium tumefaciens and recombinant tobamovirus vectors. We also investigated the influence of subcellular localisation of recombinant protein to the chloroplast and the endoplasmic reticulum (ER) on protein yield. We partially purified a selected vaccine candidate and tested its stimulation of a humoral and cellular immune response in mice. RESULTS Both transient and transgenic expression of the HIV antigens were successful, although expression of Pr55Gag was low in all systems; however, the Agrobacterium-mediated transient expression of p24 and p17/p24 yielded best, to more than 1 mg p24/kg fresh weight. Chloroplast targeted protein levels were highest in transient and transgenic expression of p24 and p17/p24. The transiently-expressed p17/p24 was not immunogenic in mice as a homologous vaccine, but it significantly boosted a humoral and T cell immune response primed by a gag DNA vaccine, pTHGagC. CONCLUSION Transient agroinfiltration was best for expression of all of the recombinant proteins tested, and p24 and p17/p24 were expressed at much higher levels than Pr55Gag. Our results highlight the usefulness of plastid signal peptides in enhancing the production of recombinant proteins meant for use as vaccines. The p17/p24 protein effectively boosted T cell and humoral responses in mice primed by the DNA vaccine pTHGagC, showing that this plant-produced protein has potential for use as a vaccine.
Collapse
MESH Headings
- AIDS Vaccines/biosynthesis
- AIDS Vaccines/genetics
- Adjuvants, Immunologic/genetics
- Agrobacterium tumefaciens/genetics
- Animals
- Chloroplasts/genetics
- Chloroplasts/metabolism
- Endoplasmic Reticulum/genetics
- Endoplasmic Reticulum/metabolism
- Female
- Gene Expression
- Gene Expression Regulation, Plant
- Genes, gag
- Genetic Vectors
- HIV Antigens/biosynthesis
- HIV Antigens/genetics
- HIV Antigens/immunology
- HIV Infections/immunology
- HIV Seronegativity
- HIV-1/genetics
- Humans
- Mice
- Mice, Inbred BALB C
- Plants, Genetically Modified
- Nicotiana/genetics
- Tobamovirus/genetics
- Transformation, Genetic
- Vaccines, Subunit/biosynthesis
- Vaccines, Subunit/genetics
- gag Gene Products, Human Immunodeficiency Virus/biosynthesis
- gag Gene Products, Human Immunodeficiency Virus/genetics
Collapse
Affiliation(s)
- Ann Meyers
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Ereck Chakauya
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
- CSIR Biosciences, Pretoria 0001, South Africa
| | - Enid Shephard
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- MRC/UCT Liver Research Centre, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Fiona L Tanzer
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - James Maclean
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Alisson Lynch
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Observatory 7925, South Africa
| | - Edward P Rybicki
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| |
Collapse
|
49
|
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
|
50
|
Girard MP, Bansal GP, Pedroza-Martins L, Dodet B, Mehra V, Schito M, Mathieson B, Delfraissy JF, Bradac J. Mucosal immunity and HIV/AIDS vaccines. Report of an International Workshop, 28-30 October 2007. Vaccine 2008; 26:3969-77. [PMID: 18513838 PMCID: PMC7131112 DOI: 10.1016/j.vaccine.2008.04.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Accepted: 04/22/2008] [Indexed: 11/22/2022]
Abstract
In October 2007, a joint ANRS-NIH workshop was held on “Mucosal immunity and HIV/AIDS vaccines” in Veyrier-du-Lac, France. Goal of the meeting was to discuss recent developments in the understanding of viral entry and dissemination at mucosal surfaces, rationale for designing vaccines to elicit mucosal immune responses by various routes of immunization, and the types of immune responses elicited. Lessons were drawn from existing vaccines against viral mucosal infections, from the recent failure of the Merck Ad5/HIV vaccine and from attempts at mucosal immunization against SIV. This report summarizes the main concepts and conclusions that came out of the meeting.
Collapse
|