1
|
Sunagar R, Singh A, Kumar S. SARS-CoV-2: Immunity, Challenges with Current Vaccines, and a Novel Perspective on Mucosal Vaccines. Vaccines (Basel) 2023; 11:vaccines11040849. [PMID: 37112761 PMCID: PMC10143972 DOI: 10.3390/vaccines11040849] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The global rollout of COVID-19 vaccines has played a critical role in reducing pandemic spread, disease severity, hospitalizations, and deaths. However, the first-generation vaccines failed to block severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission, partially due to the limited induction of mucosal immunity, leading to the continuous emergence of variants of concern (VOC) and breakthrough infections. To meet the challenges from VOC, limited durability, and lack of mucosal immune response of first-generation vaccines, novel approaches are being investigated. Herein, we have discussed the current knowledge pertaining to natural and vaccine-induced immunity, and the role of the mucosal immune response in controlling SARS-CoV2 infection. We have also presented the current status of the novel approaches aimed at eliciting both mucosal and systemic immunity. Finally, we have presented a novel adjuvant-free approach to elicit effective mucosal immunity against SARS-CoV-2, which lacks the safety concerns associated with live-attenuated vaccine platforms.
Collapse
Affiliation(s)
| | - Amit Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
2
|
Bahramali G, Mashhadi Abolghasem Shirazi M, Hannan M, Aghasadeghi MR, Khosravy MS, Arjmand S, Sadat SM. Immunogenicity evaluation of a novel virus-like particle vaccine candidate against SARS-CoV-2 in BALB/c. Pathog Dis 2023; 81:ftad021. [PMID: 37667486 DOI: 10.1093/femspd/ftad021] [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: 07/19/2023] [Revised: 08/12/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023] Open
Abstract
The coronavirus disease (COVID-19) pandemic has imposed deployment of an effective vaccine as a worldwide health priority. The new variants of SARS-CoV-2 have also brought serious concerns due to virus eradiation hesitancy. In this study, we evaluated the protective immune system activity of a recombinant viral vector-based vaccine candidate encoding a fusion spike, membrane and nucleocapsid proteins, Spike (528-1273aa)-M-N, in BALB/c via two different routes of delivery, intranasal and subcutaneous. The immune responses were then assessed through specific SARS-CoV-2 antibodies, interleukin and granzyme B secretion. The outcomes showed that the IgG titer and IgA secretion was higher in intranasal route in comparison with the subcutaneous, and what is more, a higher titer of IL-4 was detected through the intranasal route, whereas IFN-γ was highly induced via the subcutaneous route. The cytotoxic cell activities were mostly achieved via subcutaneous route immunization. Vaccination with the target antigen is immunogenic and led to induction of specific antibodies. Both humoral and cellular immunity arms were well activated in immunized mice, especially through intranasal route with detectable IgA and IgG. Therefore, implication of the platform as a potential vaccine candidate has potential as a future prophylactic vaccine that guarantees further investigations for the assessment of its immunogenicity in humans.
Collapse
Affiliation(s)
- Golnaz Bahramali
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | | | - Mina Hannan
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Mohammad Reza Aghasadeghi
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Mohammad Sadeq Khosravy
- Department of Rabies Research (B), WHO Collaborating Center for Reference and Research on Rabies, Tehran 1316946551, Iran
| | - Sina Arjmand
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| |
Collapse
|
3
|
Dual-Targeting Polymer Nanoparticles Efficiently Deliver DNA Vaccine and Induce Robust Prophylactic Immunity against Spring Viremia of Carp Virus Infection. Microbiol Spectr 2022; 10:e0308522. [PMID: 36073822 PMCID: PMC9603200 DOI: 10.1128/spectrum.03085-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Spring viremia of carp virus (SVCV) is highly contagious and lethal to most cyprinid fish, causing serious economic losses to the carp aquaculture industry. Although DNA vaccines can generate long-term humoral and cellular immune responses, which provide protective immunity against SVCV, the major drawback of DNA vaccines is their low immunogenicity in clinical tests. Here, we construct a dual-targeted polymer DNA vaccine delivery platform (MCS-PCHG) by using mannosylated chitosan to encapsulate the poly(d,l-lactide-co-glycolide)-loaded DNA vaccine containing the heavy-chain CH3 region (CH3) of common carp IgM and the antigenic domain (G131c). The developed nanovaccine delivery platform showed good biocompatibility in vivo and in vitro. With the modification of the mannose moiety and the modification of CH3, the constructed MCS-PCHG could efficiently activate the maturation of antigen-presenting cells. Moreover, we observe significantly high level of immune-related genes expression, serum antigen-specific IgM, SVCV-neutralizing antibody titers in fish vaccinated with MCS-PCHG. Next, the protective efficacy of MCS-PCHG was further evaluated by challenge test. The highest survival rate (ca. 84%) was observed in fish vaccinated with MCS-PCHG after challenging with SVCV. This study presents a novel design for smart, dual-targeted polymer nanoparticles, which are inherently biocompatible, promising for targeted vaccine delivery. IMPORTANCE Spring viremia of carp virus (SVCV) affects global cyprinid fish farming industry, with no available commercial vaccine. Herein, we developed a dual-targeting polymer nanovaccine (MCS-PCHG) by using mannose and common carp IgM heavy chain CH3 region (CH3) as antigen presenting cell (APCs) recognition moiety, attaining the effective delivery of antigen. This dual-targeting polymer vaccine can efficiently activate the APCs, and further induce robust and durable adaptive immune response with good protection against SVCV infection. Our study provides valuable theoretical basis for developing efficient vaccine against infectious diseases in aquaculture.
Collapse
|
4
|
Rothen DA, Krenger PS, Nonic A, Balke I, Vogt AS, Chang X, Manenti A, Vedovi F, Resevica G, Walton SM, Zeltins A, Montomoli E, Vogel M, Bachmann MF, Mohsen MO. Intranasal administration of a virus like particles-based vaccine induces neutralizing antibodies against SARS-CoV-2 and variants of concern. Allergy 2022; 77:2446-2458. [PMID: 35403221 PMCID: PMC9111403 DOI: 10.1111/all.15311] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND The highly contagious SARS-CoV-2 is mainly transmitted by respiratory droplets and aerosols. Consequently, people are required to wear masks and maintain a social distance to avoid spreading of the virus. Despite the success of the commercially available vaccines, the virus is still uncontained globally. Given the tropism of SARS-CoV-2, a mucosal immune reaction would help to reduce viral shedding and transmission locally. Only seven out of hundreds of ongoing clinical trials are testing the intranasal delivery of a vaccine against COVID-19. METHODS In the current study, we evaluated the immunogenicity of a traditional vaccine platform based on virus-like particles (VLPs) displaying RBD of SARS-CoV-2 for intranasal administration in a murine model. The candidate vaccine platform, CuMVTT -RBD, has been optimized to incorporate a universal T helper cell epitope derived from tetanus-toxin and is self-adjuvanted with TLR7/8 ligands. RESULTS CuMVTT -RBD vaccine elicited a strong systemic RBD- and spike-IgG and IgA antibodies of high avidity. Local immune response was assessed, and our results demonstrate a strong mucosal antibody and plasma cell production in lung tissue. Furthermore, the induced systemic antibodies could efficiently recognize and neutralize different variants of concern (VOCs). CONCLUSION Our data demonstrate that intranasal administration of CuMVTT -RBD induces a protective systemic and local specific antibody response against SARS-CoV-2 and its VOCs.
Collapse
Affiliation(s)
- Dominik A. Rothen
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Pascal S. Krenger
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Aleksandra Nonic
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Ina Balke
- Latvian Biomedical Research & Study CentreRigaLatvia
| | - Anne‐Cathrine S. Vogt
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Xinyue Chang
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | | | | | | | | | | | - Emanuele Montomoli
- VisMederi S.r.l.SienaItaly
- Department of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| | - Monique Vogel
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Martin F. Bachmann
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
- Nuffield Department of MedicineThe Jenner InstituteUniversity of OxfordOxfordUK
| | - Mona O. Mohsen
- Department of Rheumatology and ImmunologyUniversity HospitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
- Saiba AGPfaeffikonSwitzerland
| |
Collapse
|
5
|
Hsieh MS, Hsu CW, Tu LL, Chai KM, Yu LL, Wu CC, Chen MY, Chiang CY, Liu SJ, Liao CL, Chen HW. Intranasal Vaccination With Recombinant Antigen-FLIPr Fusion Protein Alone Induces Long-Lasting Systemic Antibody Responses and Broad T Cell Responses. Front Immunol 2021; 12:751883. [PMID: 34707615 PMCID: PMC8543008 DOI: 10.3389/fimmu.2021.751883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022] Open
Abstract
A simple formulation is urgently needed for mucosal vaccine development. We employed formyl peptide receptor-like 1 inhibitory protein (FLIPr), an FcγR antagonist secreted by Staphylococcus aureus, as a vector to target ovalbumin (OVA) to dendritic cells (DCs) via intranasal administration. Our results demonstrate that intranasal administration of recombinant OVA-FLIPr fusion protein (rOVA-FLIPr) alone efficiently delivers OVA to DCs in nasal lymphoid tissue. Subsequently, OVA-specific IgG and IgA antibodies in the circulatory system and IgA antibodies in mucosal tissue were detected. Importantly, activation of OVA-specific CD4+ and CD8+ T cells and induction of a broad-spectrum cytokine secretion profile were detected after intranasal administration of rOVA-FLIPr alone in immunocompetent C57BL/6 mice. Furthermore, we employed immunodeficient AG129 mice as a Zika virus infection model and demonstrated that intranasal administration of recombinant Zika virus envelope protein domain III-FLIPr fusion protein induced protective immune responses against the Zika virus. These results suggest that antigen-FLIPr fusion protein alone via intranasal administration can be applied to mucosal vaccine development.
Collapse
Affiliation(s)
- Ming-Shu Hsieh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Wei Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ling-Ling Tu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Lu Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chiao-Chieh Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Mei-Yu Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
van Beek LF, Welzen PLW, Teufel LU, Joosten I, Diavatopoulos DA, van Hest J, de Jonge MI. Bimodal Targeting of Human Leukocytes by Fc- and CpG-Decorated Polymersomes to Tune Immune Induction. Biomacromolecules 2021; 22:4422-4433. [PMID: 34554732 PMCID: PMC8512671 DOI: 10.1021/acs.biomac.1c00985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The use of well-defined
nanovesicles composed of amphiphilic block copolymers (polymersomes) for delivery
of adjuvants and antigens is a promising strategy for vaccine development.
However, the potency of nanoparticle vaccines depends on efficient
interaction with and activation of cells involved in antigen presentation,
which can be achieved by targeting cellular receptors. Here, we showed
that the Fc fragment display on the polymersome surface resulted in
markedly improved interactions with granulocytes, monocytes, and NK
cells, while for “naked” polymersomes, virtually no
binding to leukocytes was observed. Moreover, CpG-decorated polymersomes
were found to also interact with T and/or B cells. Interestingly,
whole blood stimulations with Fc fragment and CpG-decorated polymersomes
induced interleukin (IL)-6, IL-8, and TNF-α production, while
naked polymersomes did not induce any cytokine production. In conclusion,
specific immune induction by polymersomes can be controlled using
bimodal targeting of different immune receptors, which is an essential
feature for targeted vaccine delivery.
Collapse
Affiliation(s)
- Lucille F van Beek
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Pascal L W Welzen
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Lisa U Teufel
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Irma Joosten
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Dimitri A Diavatopoulos
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jan van Hest
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Marien I de Jonge
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
7
|
Hu Z, Liu X. "Antigen Camouflage and Decoy" Strategy to Overcome Interference From Maternally Derived Antibody With Newcastle Disease Virus-Vectored Vaccines: More Than a Simple Combination. Front Microbiol 2021; 12:735250. [PMID: 34512613 PMCID: PMC8432293 DOI: 10.3389/fmicb.2021.735250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/09/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Zenglei Hu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
8
|
Tummillo KM, Hazlett KR. Co-Opting Host Receptors for Targeted Delivery of Bioconjugates-From Drugs to Bugs. Molecules 2021; 26:molecules26051479. [PMID: 33803208 PMCID: PMC7963163 DOI: 10.3390/molecules26051479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 11/16/2022] Open
Abstract
Bioconjugation has allowed scientists to combine multiple functional elements into one biological or biochemical unit. This assembly can result in the production of constructs that are targeted to a specific site or cell type in order to enhance the response to, or activity of, the conjugated moiety. In the case of cancer treatments, selectively targeting chemotherapies to the cells of interest limit harmful side effects and enhance efficacy. Targeting through conjugation is also advantageous in delivering treatments to difficult-to-reach tissues, such as the brain or infections deep in the lung. Bacterial infections can be more selectively treated by conjugating antibiotics to microbe-specific entities; helping to avoid antibiotic resistance across commensal bacterial species. In the case of vaccine development, conjugation is used to enhance efficacy without compromising safety. In this work, we will review the previously mentioned areas in which bioconjugation has created new possibilities and advanced treatments.
Collapse
Affiliation(s)
- Kristen M. Tummillo
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
- Admera Health, South Plainfield, NJ 07080, USA
| | - Karsten R.O. Hazlett
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
- Correspondence: ; Tel.: +1-518-262-2338
| |
Collapse
|
9
|
Wiedinger K, McCauley J, Bitsaktsis C. Isotype-specific outcomes in Fc gamma receptor targeting of PspA using fusion proteins as a vaccination strategy against Streptococcus pneumoniae infection. Vaccine 2020; 38:5634-5646. [PMID: 32646816 DOI: 10.1016/j.vaccine.2020.06.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/22/2020] [Accepted: 06/22/2020] [Indexed: 02/08/2023]
Abstract
Streptococcus pneumoniae (Spn) remains a considerable threat to public health despite the availability of antibiotics and polysaccharide conjugate vaccines. The lack of mucosal immunity in addition to capsular polysaccharide diversity, has proved to be problematic in developing a universal vaccine against Spn. Targeting antigen to Fc receptors is an attractive way to augment both innate and adaptive immunity against mucosal pathogens, by promoting interactions with activating Fcγ receptors (FcγR) that mediate diverse immunomodulatory functions. The effect of targeting FcγR is highly influenced by the IgG subclass, which bares differential affinities for activating and inhibitory FcγR. In the current study we demonstrate targeting activating FcγR with fusion proteins consisting of PspA and IgG2a Fc enhance PspA-specific immune responses, and effectively protect against mucosal Spn challenge. Specifically, targeting PspA to FcγR polarized alveolar macrophage to the AM1 phenotype and increased conventional dendritic cell subsets in the lung in addition to augmenting Th1 cytokines and PspA-specific IgG and IgA. In contrast, fusion proteins consisting of PspA fused to the IgG1 Fc provided minimal benefit over administration of PspA alone, as a result of interaction with the inhibitory FcγRIIB. Protective efficacy of the IgG1 fusion protein was significantly enhanced in animals deficient for FcγRIIB accompanied by increased B cell maturation and proliferation levels in these animals. These studies demonstrate FcγR targeting is an effective strategy for inducing potent cellular and humoral responses via mucosal immunization with Fc fusion proteins, however, careful consideration of the Fc region utilized is required since Fc isotype subclass heavily influenced immunization induced effector functions and survival against lethal Spn challenge. Fc-engineering with specific attention to FcγRIIB engagement presents a valuable vaccine strategy for protecting against Spn infection.
Collapse
Affiliation(s)
- Kari Wiedinger
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA.
| | - James McCauley
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA.
| | | |
Collapse
|
10
|
Abstract
Antibody/antigen binding results in immune complexes (IC) that have a variety of regulatory functions. One important feature is the enhanced host immune activation against antigen contained in the complex. ICs play important roles at several critical steps that lead to B and T cell activation, including antigen targeting/retention, facilitated antigen uptake, antigen presenting cell activation and proper balancing of positive and negative stimulatory signals. In both poultry industry and clinical health care, ICs have been used as preventive and therapeutic vaccines. With our deepening understanding of antibody biology, particularly in light of new revelations of regulatory functions of Fc receptors, mechanistically more precise engineering has spearheaded tailored use of this tool for infection control and cancer therapy. IC-based treatment and prophylaxis have been tested to different extents in HBV, HIV and influenza viral infection control and are actively examined as an alternative treatment for several forms of tumor. As a part of this book series, this chapter aims to discuss the mechanistic aspects of IC signaling and their impact on immune cells. We give samples how this old technology has been used by practitioners over the last several decades and suggest potential paths for future development of IC-based immune therapy.
Collapse
Affiliation(s)
- Yu-Mei Wen
- Key Laboratory of Molecular Virology, Shanghai Medical College, School of Basic Medical Sciences, Fudan University, Shanghai, China. .,Shanghai Medical College, Fudan University, Rm 401, Fuxing Bldg, 131 Yi Xue Yuan Rd, Shanghai, 200032, China.
| | - Yan Shi
- Department of Basic Medical Sciences, Center for Life Sciences, Institute of Immunology, Tsinghua University, Beijing, China.,Department of Microbiology, Immunology & Infectious Diseases and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.,D301 Medical Sciences Bldg, Tsinghua University, Beijing, 00084, China
| |
Collapse
|
11
|
Duffy EB, Periasamy S, Hunt D, Drake JR, Harton JA. FcγR mediates TLR2- and Syk-dependent NLRP3 inflammasome activation by inactivated Francisella tularensis LVS immune complexes. J Leukoc Biol 2016; 100:1335-1347. [PMID: 27365531 PMCID: PMC5110000 DOI: 10.1189/jlb.2a1215-555rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 06/09/2016] [Accepted: 06/15/2016] [Indexed: 01/08/2023] Open
Abstract
IgG (mAb)-opsonized, inactivated Francisella tularensis LVS (iFt-mAb) enhances TLR2-dependent IL-6 production by macrophages via Fcγ receptors (FcγR). In mice, vaccination with iFt-mAb provides IgA-dependent protection against lethal challenge with Ft LVS. Because inflammasome maturation of IL-1β is thought important for antibody-mediated immunity, we considered the possibility that iFt-mAb elicits an FcγR-dependent myeloid cell inflammasome response. Herein, we find that iFt-mAb enhances macrophage and dendritic cell IL-1β responses in a TLR2- and FcγR-dependent fashion. Although iFt-mAb complexes bind FcγR and are internalized, sensing of cytosolic DNA by absent in melanoma 2 (AIM2) is not required for the IL-1β response. In contrast, ASC, caspase-1, and NLR family pyrin domain-containing 3 (NLRP3) are indispensable. Further, FcγR-mediated spleen tyrosine kinase (Syk) signaling is required for this NLRP3-dependent IL-1β response, but the alternative IL-1β convertase caspase-8 is insufficient. Finally, iFt-mAb-vaccinated wild-type mice exhibit a significant delay in time to death, but IL-1R1- or Nlrp3-deficient mice vaccinated in this way are not protected and lack appreciable Francisella-specific antibodies. This study demonstrates that FcγR-mediated Syk activation leads to NLRP3 inflammasome-dependent IL-1β production in macrophages and suggests that an Nlrp3- and IL-1R-dependent process contributes to the IgA response important for protection against Ft LVS. These findings extend our understanding of cellular responses to inactivated pathogen-opsonized vaccine, establish FcγR-elicited Syk kinase-mediated NLRP3 inflammasome activation, and provide additional insight toward understanding crosstalk between TLR and FcγR signals.
Collapse
Affiliation(s)
- Ellen B Duffy
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sivakumar Periasamy
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Danielle Hunt
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - James R Drake
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Jonathan A Harton
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| |
Collapse
|
12
|
Soleimanpour S, Mohammadi A, Ghazvini K, Jamehdar SA, Sadeghian H, Taghiabadi M, Rezaee SAR. Construction of Mycobacterium tuberculosis ESAT-6 fused to human Fcγ of IgG1: To target FcγR as a delivery system for enhancement of immunogenicity. Gene 2016; 580:111-117. [PMID: 26778208 DOI: 10.1016/j.gene.2016.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 11/27/2015] [Accepted: 01/06/2016] [Indexed: 01/07/2023]
Abstract
In order to prevent spreading of Mycobacterium tuberculosis (Mtb), it is necessary to discover effective vaccines, fast and reliable diagnosis, and appropriate treatment schemes. In the present study, an Fc-tagged recombinant Mtb-ESAT-6 was produced to make a selective delivery system for promoting cellular immunity. To determine 3D structure of the recombinant protein, model building was performed in MODELLER9v13 program. After preparation of Mtb-DNA and Fcγ1 cDNA, they were amplified by specific primers to make ESAT-6 and Fcγ1 products to fuse them in frame using splicing by overlap extension (SOEing)-PCR. After TA cloning, the construct was sequenced to confirm no errors have been introduced. The recombinant DNA was then subcloned into PDR2EF1α eukaryotic expression vector. The plasmid sequenced over the sites at which two DNA fragments were cloned to ensure that the ligation had generated an in-frame fusion of the genes. The CHO cells were then stably transected by PDR2EF1α-ESAT-6:Fcγ1 vector using lipofectamin and the expression and its binding to the Fcγ receptor (FcγRI) on APCs were confirmed by immunofluorescence assay (IFA). The IFA results demonstrated that ESAT6:Fcγ1 was expressed in engineered CHO cells. Semi-scale protein production and purification using HiTrap-PA column showed a high secretion of the recombinant protein by Western blotting method. The molecular weight of the monomer in the SDS-PAGE was equal to a protein of 50kDa, which dimerizes by disulfide bond of Fcγ fragments. Since, ESAT6:Fcγ1 protein dimerizes and bind to FcγRs, therefore, Fc-tagged protein could target APCs for inducing appropriate immune response or using in interferon-based assays.
Collapse
Affiliation(s)
- Saman Soleimanpour
- Microbiology & Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Mohammadi
- Microbiology & Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kiarash Ghazvini
- Microbiology & Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Amel Jamehdar
- Microbiology & Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Sadeghian
- Microbiology & Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboubeh Taghiabadi
- Microbiology & Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - S A R Rezaee
- Inflammation and Inflammatory Research Center, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Mikulska JE. Analysis of Response Elements Involved in the Regulation of the Human Neonatal Fc Receptor Gene (FCGRT). PLoS One 2015; 10:e0135141. [PMID: 26252948 PMCID: PMC4529178 DOI: 10.1371/journal.pone.0135141] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/19/2015] [Indexed: 12/26/2022] Open
Abstract
Human epithelial, endothelial and PMA-differentiated THP-1 cell lines were used as model systems to study the transcriptional regulation of the human FCGRT gene encoding the alpha chain of hFcRn. The data obtained from site-directed mutagenesis in transient transfection experiments indicate that the Sp1 sites at positions -641, -635, and -313, CF1/YY1 elements at positions -586 and -357, and the AP-1 motif at -276 within the-660/-233 fragment of the human FCGRT promoter (hFCGRT) participate in the regulation of human FCGRT in all selected cell lines. However, their individual contribution to promoter activity is not equivalent. The Sp1 binding site at -313 and the AP-1 site at -276 are critical for the activity of the hFCGRT promoter in epithelial and endothelial cells. Moreover, the CF1/YY1 site at -586 in differentiated THP-1 cells, plays an essential role in the transcriptional activity of the promoter. In addition, the C/EBPbeta binding site at -497 of the hFCGRT promoter in epithelial and endothelial cells, and the C/EBPbeta motif located at -497 and -233 within the hFCGRT promoter in differentiated THP-1 cells may function as positive regulatory sequences in response to LPS or PMA stimulation. EMSA and supershift analyses showed that the functionally identified binding motifs in the hFCGRT promoter were able to specifically interact with their corresponding (Sp1, Sp2, Sp3, c-Fos, c-Jun, YY1, and C/EBPbeta or C/EBPdelta) transcription factors (TFs), suggesting their possible involvement in the regulation of the human FCGRT gene expression.
Collapse
Affiliation(s)
- Joanna E. Mikulska
- Department of Immunochemistry, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- * E-mail:
| |
Collapse
|
14
|
Wang S, Liu H, Zhang X, Qian F. Intranasal and oral vaccination with protein-based antigens: advantages, challenges and formulation strategies. Protein Cell 2015; 6:480-503. [PMID: 25944045 PMCID: PMC4491048 DOI: 10.1007/s13238-015-0164-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/10/2015] [Indexed: 02/06/2023] Open
Abstract
Most pathogens initiate their infections at the human mucosal surface. Therefore, mucosal vaccination, especially through oral or intranasal administration routes, is highly desired for infectious diseases. Meanwhile, protein-based antigens provide a safer alternative to the whole pathogen or DNA based ones in vaccine development. However, the unique biopharmaceutical hurdles that intranasally or orally delivered protein vaccines need to overcome before they reach the sites of targeting, the relatively low immunogenicity, as well as the low stability of the protein antigens, require thoughtful and fine-tuned mucosal vaccine formulations, including the selection of immunostimulants, the identification of the suitable vaccine delivery system, and the determination of the exact composition and manufacturing conditions. This review aims to provide an up-to-date survey of the protein antigen-based vaccine formulation development, including the usage of immunostimulants and the optimization of vaccine delivery systems for intranasal and oral administrations.
Collapse
Affiliation(s)
- Shujing Wang
- Department of Pharmacology and Pharmaceutical Sciences, School of Medicine and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | | | | | | |
Collapse
|
15
|
Downmodulation of vaccine-induced immunity and protection against the intracellular bacterium Francisella tularensis by the inhibitory receptor FcγRIIB. J Immunol Res 2015; 2015:840842. [PMID: 25961064 PMCID: PMC4417568 DOI: 10.1155/2015/840842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 02/08/2023] Open
Abstract
Fc gamma receptor IIB (FcγRIIB) is the only Fc gamma receptor (FcγR) which negatively regulates the immune response, when engaged by antigen- (Ag-) antibody (Ab) complexes. Thus, the generation of Ag-specific IgG in response to infection or immunization has the potential to downmodulate immune protection against infection. Therefore, we sought to determine the impact of FcγRIIB on immune protection against Francisella tularensis (Ft), a Category A biothreat agent. We utilized inactivated Ft (iFt) as an immunogen. Naïve and iFt-immunized FcγRIIB knockout (KO) or wildtype (WT) mice were challenged with Ft-live vaccine strain (LVS). While no significant difference in survival between naïve FcγRIIB KO versus WT mice was observed, iFt-immunized FcγRIIB KO mice were significantly better protected than iFt-immunized WT mice. Ft-specific IgA in serum and bronchial alveolar lavage, as well as IFN-γ, IL-10, and TNF-α production by splenocytes harvested from iFt-immunized FcγRIIB KO, were also significantly elevated. In addition, iFt-immunized FcγRIIB KO mice exhibited a reduction in proinflammatory cytokine levels in vivo at 5 days after challenge, which correlates with increased survival following Ft-LVS challenge in published studies. Thus, these studies demonstrate for the first time the ability of FcγRIIB to regulate vaccine-induced IgA production and downmodulate immunity and protection. The immune mechanisms behind the above observations and their potential impact on vaccine development are discussed.
Collapse
|
16
|
Girard A, Roques E, Massie B, Archambault D. Flagellin in fusion with human rotavirus structural proteins exerts an adjuvant effect when delivered with replicating but non-disseminating adenovectors through the intrarectal route. Mol Biotechnol 2014; 56:394-407. [PMID: 24271565 DOI: 10.1007/s12033-013-9723-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human rotavirus (HRV) is the worldwide leading cause of gastroenteritis in young children. Two live attenuated HRV vaccines have been approved since 2006. However, these live vaccines still have potential risks including reversion of virulence. Adenoviruses are suitable vectors for mucosal administration of subunit vaccines. In addition to the adjuvant effect of certain adenovirus components, the use of an adjuvant like flagellin is also another means to increase the immune response to the immunogen. The aim of this study was to determine whether flagellin in fusion with HRV structural proteins stimulates the innate immune response and enhances the HRV-specific immune response when delivered through the intrarectal route with replicating but non-disseminating adenovector (R-AdV). Salmonella typhimurium flagellin B (FljB) in fusion with HRV VP4Δ::VP7 protein induced IL-1β production in J774A.1 macrophages exposed to the R-AdV. Intrarectal administration of R-AdVs expressing either VP4Δ::VP7 or VP4Δ::VP7::FljB in BALB/c mice resulted in HRV-specific mixed Th1/Th2 immune responses. The HRV-specific antibody response elicited with the use of R-AdV expressing VP4Δ::VP7::FljB was higher than that with R-AdV expressing VP4Δ::VP7. The results also show that the replication capability of R-AdVs contributed to enhance the HRV-specific immune response as compared with that obtained with non-replicative AdVs. This work lays the foundation for using the R-AdV system and FljB-adjuvanted formulation to elicit a mucosal immune response specific to HRV.
Collapse
Affiliation(s)
- Aurélie Girard
- Department of Biological Sciences, University of Québec at Montréal, P.O. Box 8888, Succursale Centre-Ville, Montreal, QC, H3C 3P8, Canada
| | | | | | | |
Collapse
|
17
|
Baker K, Rath T, Pyzik M, Blumberg RS. The Role of FcRn in Antigen Presentation. Front Immunol 2014; 5:408. [PMID: 25221553 PMCID: PMC4145246 DOI: 10.3389/fimmu.2014.00408] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/12/2014] [Indexed: 01/06/2023] Open
Abstract
Immunoglobulins are unique molecules capable of simultaneously recognizing a diverse array of antigens and themselves being recognized by a broad array of receptors. The abundance specifically of the IgG subclass and the variety of signaling receptors to which it binds render this an important immunomodulatory molecule. In addition to the classical Fcγ receptors that bind IgG at the cell surface, the neonatal Fc receptor (FcRn) is a lifelong resident of the endolysosomal system of most hematopoietic cells where it determines the intracellular fate of both IgG and IgG-containing immune complexes (IgG IC). Cross-linking of FcRn by multivalent IgG IC within antigen presenting cells such as dendritic cells initiates specific mechanisms that result in trafficking of the antigen-bearing IgG IC into compartments from which the antigen can successfully be processed into peptide epitopes compatible with loading onto both major histocompatibility complex class I and II molecules. In turn, this enables the synchronous activation of both CD4(+) and CD8(+) T cell responses against the cognate antigen, thereby bridging the gap between the humoral and cellular branches of the adaptive immune response. Critically, FcRn-driven T cell priming is efficient at very low doses of antigen due to the exquisite sensitivity of the IgG-mediated antigen delivery system through which it operates. FcRn-mediated antigen presentation has important consequences in tissue compartments replete with IgG and serves not only to determine homeostatic immune activation at a variety of sites but also to induce inflammatory responses upon exposure to antigens perceived as foreign. Therapeutically targeting the pathway by which FcRn enables T cell activation in response to IgG IC is thus a highly attractive prospect not only for the treatment of diseases that are driven by immune complexes but also for manipulating local immune responses against defined antigens such as those present during infections and cancer.
Collapse
Affiliation(s)
- Kristi Baker
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Timo Rath
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nueremberg, Erlangen, Germany
| | - Michal Pyzik
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Digestive Diseases Center, Boston, MA, USA
| |
Collapse
|
18
|
Devriendt B, Goddeeris BM, Cox E. The Fcγ receptor expression profile on porcine dendritic cells depends on the nature of the stimulus. Vet Immunol Immunopathol 2013; 152:43-9. [DOI: 10.1016/j.vetimm.2012.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
19
|
Iglesias BV, Bitsaktsis C, Pham G, Drake JR, Hazlett KRO, Porter K, Gosselin EJ. Multiple mechanisms mediate enhanced immunity generated by mAb-inactivated F. tularensis immunogen. Immunol Cell Biol 2012; 91:139-48. [PMID: 23247654 PMCID: PMC3570671 DOI: 10.1038/icb.2012.66] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We have previously demonstrated that immunization with inactivated Francisella tularensis, a Category A intracellular mucosal pathogen, combined with IgG2a anti-F. tularensis monoclonal antibody, enhances protection against subsequent F. tularensis challenge. To understand the mechanism(s) involved, we examined the binding, internalization, presentation, and in vivo trafficking of inactivated F. tularensis in the presence and absence of opsonizing monoclonal antibody. We found that when inactivated F. tularensis is combined with anti-F. tularensis monoclonal antibody, presentation to F. tularensis-specific T cells is enhanced, this enhancement is Fc receptor-dependent, and requires a physical linkage between the monoclonal antibody and the inactivated F. tularensis immunogen. This enhanced presentation is due, in part, to enhanced binding and internalization of inactivated F. tularensis by antigen presenting cells, and involves interactions with multiple Fc receptor types. Furthermore, targeting inactivated F. tularensis to Fc receptors enhances dendritic cell maturation and extends the time period over which antigen presenting cells stimulate T cells. In vivo trafficking studies reveal enhanced transport of inactivated F. tularensis immunogen to the Nasal Associated Lymphoid Tissue in the presence of monoclonal antibody, which is FcRn-dependent. In summary, these are the first comprehensive studies using a single vaccine protection model/immunogen to establish the array of mechanisms involved in enhanced immunity/protection mediated by an Fc receptor-targeted mucosal immunogen. These results demonstrate that multiple cellular/immune mechanisms contribute to Fc receptor-enhanced immunity.
Collapse
Affiliation(s)
- Bibiana V Iglesias
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Mucosal immunization with an unadjuvanted vaccine that targets Streptococcus pneumoniae PspA to human Fcγ receptor type I protects against pneumococcal infection through complement- and lactoferrin-mediated bactericidal activity. Infect Immun 2011; 80:1166-80. [PMID: 22158740 DOI: 10.1128/iai.05511-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Targeting an antigen to Fc receptors (FcR) can enhance the immune response to the antigen in the absence of adjuvant. Furthermore, we recently demonstrated that intranasal immunization with an FcγR-targeted antigen enhances protection against a category A intracellular mucosal pathogen, Francisella tularensis. To determine if a similar strategy could be applied to the important pathogen Streptococcus pneumoniae, we used an improved mucosal FcR-targeting strategy that specifically targets human FcγR type I (hFcγRI). A humanized single-chain antibody component in which the variable domain binds to hFcγRI [anti-hFcγRI (H22)] was linked in a fusion protein with the pneumococcal surface protein A (PspA). PspA is known to elicit protection against pneumococcal sepsis, carriage, and pneumonia in mouse models when administered with adjuvants. Anti-hFcγRI-PspA or recombinant PspA (rPspA) alone was used to intranasally immunize wild-type (WT) and hFcγRI transgenic (Tg) mice in the absence of adjuvant. The hFcγRI Tg mice receiving anti-hFcγRI-PspA exhibited elevated S. pneumoniae-specific IgA, IgG2c, and IgG1 antibodies in serum and bronchoalveolar lavage fluid. Neither immunogen was effective in protecting WT mice in the absence of adjuvant, but when PspA was targeted to hFcγRI as the anti-hFcγRI-PspA fusion, enhanced protection against lethal S. pneumoniae challenge was observed in the hFcγRI Tg mice compared to mice given nontargeted rPspA alone. Immune sera from the anti-hFcγRI-PspA-immunized Tg mice showed enhanced complement C3 deposition on bacterial surfaces, and protection was dependent upon an active complement system. Immune serum also showed an enhanced bactericidal activity directed against S. pneumoniae that appears to be lactoferrin mediated.
Collapse
|
21
|
Optimized adenovirus-antibody complexes stimulate strong cellular and humoral immune responses against an encoded antigen in naive mice and those with preexisting immunity. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 19:84-95. [PMID: 22089246 DOI: 10.1128/cvi.05319-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The immune response to recombinant adenoviruses is the most significant impediment to their clinical use for immunization. We test the hypothesis that specific virus-antibody combinations dictate the type of immune response generated against the adenovirus and its transgene cassette under certain physiological conditions while minimizing vector-induced toxicity. In vitro and in vivo assays were used to characterize the transduction efficiency, the T and B cell responses to the encoded transgene, and the toxicity of 1 × 10(11) adenovirus particles mixed with different concentrations of neutralizing antibodies. Complexes formed at concentrations of 500 to 0.05 times the 50% neutralizing dose (ND(50)) elicited strong virus- and transgene-specific T cell responses. The 0.05-ND(50) formulation elicited measurable anti-transgene antibodies that were similar to those of virus alone (P = 0.07). This preparation also elicited very strong transgene-specific memory T cell responses (28.6 ± 5.2% proliferation versus 7.7 ± 1.4% for virus alone). Preexisting immunity significantly reduced all responses elicited by these formulations. Although lower concentrations (0.005 and 0.0005 ND(50)) of antibody did not improve cellular and humoral responses in naïve animals, they did promote strong cellular (0.005 ND(50)) and humoral (0.0005 ND(50)) responses in mice with preexisting immunity. Some virus-antibody complexes may improve the potency of adenovirus-based vaccines in naïve individuals, while others can sway the immune response in those with preexisting immunity. Additional studies with these and other virus-antibody ratios may be useful to predict and model the type of immune responses generated against a transgene in those with different levels of exposure to adenovirus.
Collapse
|
22
|
Pal I, Ramsey JD. The role of the lymphatic system in vaccine trafficking and immune response. Adv Drug Deliv Rev 2011; 63:909-22. [PMID: 21683103 DOI: 10.1016/j.addr.2011.05.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 01/26/2011] [Indexed: 01/13/2023]
Abstract
The development and improvement of vaccines has been a significant endeavor on the part of the medical community for more than the last two centuries, and the success of these efforts is obvious when one considers the millions of lives that have been saved. Recent work in the field of vaccines, however, indicates that vaccines may be developed for even more challenging diseases than those previously addressed. It will be important in achieving this feat to account for the physical and chemical processes related to vaccine trafficking, rather than solely relying on our knowledge of the pathogen and our empirical experience. A thorough understanding of the lymphatic system is essential considering the role it plays in antigen trafficking and all immunological activity. This review describes the results of recent work that provides insight into the physiological processes of the lymphatic system and its various components with an emphasis on vaccine antigen trafficking from the administration site to secondary lymphoid tissues and the ensuing immune response. The review also discusses current challenges in designing vaccines and presents modern strategies for designing vaccines to better interface with the lymphatic system.
Collapse
|
23
|
Girard A, Saron W, Bergeron-Sandoval LP, Sarhan F, Archambault D. Flagellin produced in plants is a potent adjuvant for oral immunization. Vaccine 2011; 29:6695-703. [PMID: 21745522 DOI: 10.1016/j.vaccine.2011.06.092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 06/14/2011] [Accepted: 06/24/2011] [Indexed: 11/24/2022]
Abstract
The aim of this study was to produce adjuvant with high biosafety, efficacy and low cost. Towards this goal, the plant Nicotiana benthamiana transient expression system was successfully used to express Salmonella typhimurium's flagellin (FljB). The yield of the expressed FljB was 280 mg per kg of fresh weight (FW) leaves. The lyophilized plant powder containing plant expressing FljB was mixed with ovalbumin (OVA) and used for oral immunization of BALB/c mice. The ELISA analysis showed higher and accelerated OVA-specific serum antibody responses in mice given the mixture when compared to animals receiving OVA alone. Furthermore, FljB elicited a mixed Th1/Th2 response as shown by the presence of specific anti-OVA IgG1, IgG2a and IgG2b isotypes. OVA-specific IgAs were also detected in mice given the mixture. Cell-mediated immune response to OVA was induced by FljB as determined by a spleen lymphocyte specific proliferation test. No immune response was generated against FljB. In conclusion, our results showed for the first time the production of FljB in plants and the efficient use of the crude lyophilized extract as an adjuvant for oral immunization.
Collapse
Affiliation(s)
- Aurélie Girard
- University of Québec at Montréal, Department of Biological Sciences, PO Box 8888, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3P8
| | | | | | | | | |
Collapse
|
24
|
Rapid immune responses to a botulinum neurotoxin Hc subunit vaccine through in vivo targeting to antigen-presenting cells. Infect Immun 2011; 79:3388-96. [PMID: 21576339 DOI: 10.1128/iai.00166-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The clostridial botulinum neurotoxins (BoNTs) are the most potent protein toxins known. The carboxyl-terminal fragment of the toxin heavy chain (Hc) has been intensively investigated as a BoNT vaccine immunogen. We sought to determine whether targeting Hc to antigen-presenting cells (APCs) could accelerate the immune responses to vaccination with BoNT serotype A (BoNT/A) Hc. To test this hypothesis, we targeted Hc to the Fc receptors for IgG (FcγRs) expressed by dendritic cells (DCs) and other APCs. Hc was expressed as a fusion protein with a recombinant ligand for human FcγRs (R4) to produce HcR4 or a similar ligand for murine FcγRs to produce HcmR4. HcR4, HcmR4, and Hc were produced as secreted proteins using baculovirus-mediated expression in SF9 insect cells. In vitro receptor binding assays showed that HcR4 effectively targets Hc to all classes of FcγRs. APCs loaded with HcR4 or HcmR4 are substantially more effective at stimulating Hc-reactive T cells than APCs loaded with nontargeted Hc. Mice immunized with a single dose of HcmR4 or HcR4 had earlier and markedly higher Hc-reactive antibody titers than mice immunized with nontargeted Hc. These results extend to BoNT neutralizing antibody titers, which are substantially higher in mice immunized with HcmR4 than in mice immunized with Hc. Our results demonstrate that targeting Hc to FcγRs augments the pace and magnitude of immune responses to Hc.
Collapse
|
25
|
Devriendt B, De Geest BG, Cox E. Designing oral vaccines targeting intestinal dendritic cells. Expert Opin Drug Deliv 2011; 8:467-83. [DOI: 10.1517/17425247.2011.561312] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
26
|
Gallo P, Gonçalves R, Mosser DM. The influence of IgG density and macrophage Fc (gamma) receptor cross-linking on phagocytosis and IL-10 production. Immunol Lett 2010; 133:70-7. [PMID: 20670655 DOI: 10.1016/j.imlet.2010.07.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 05/27/2010] [Accepted: 07/20/2010] [Indexed: 10/19/2022]
Abstract
We have previously demonstrated that the addition of immune complexes (IC) to stimulated macrophages could profoundly influence cytokine production. In the present work we sought to determine the density of IgG on immune complexes necessary to mediate phagocytosis, inhibit IL-12 production and induce IL-10 production from stimulated macrophages. We developed immune complexes with predictable average densities of surface-bound immunoglobulin. We show that a threshold amount of IgG was necessary to mediate attachment of IC to macrophages. At progressively higher densities of IgG, Fc receptor-mediated phagocytosis resulted in an inhibition of IL-12 production and then an induction of IL-10. The reciprocal alterations in these two cytokines occurred when as little as one optimally opsonized SRBC was bound per macrophage. Macrophage IL-10 induction by immune complexes was associated with the activation of the MAP kinase, ERK, which was progressively increased as a function of IgG density. We conclude that signal transduction through the macrophage Fcγ receptors vary as a function of signal strength. At moderate IgG densities, especially in the presence of complement, efficient phagocytosis occurs in the absence of cytokine alterations. At slightly higher IgG densities IL-12 production is shut off and eventually IL-10 induction occurs. Thus, the myriad events emanating from FcγR ligation depends on the density of immune complexes, allowing the Fc receptors to fine-tune cellular responses depending on the extent of receptor cross-linking.
Collapse
Affiliation(s)
- Paul Gallo
- Department of Cell Biology and Molecular Genetics and the Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA
| | | | | |
Collapse
|
27
|
Mohamed W, Sethi S, Darji A, Mraheil MA, Hain T, Chakraborty T. Antibody targeting the ferritin-like protein controls Listeria infection. Infect Immun 2010; 78:3306-14. [PMID: 20439472 PMCID: PMC2897390 DOI: 10.1128/iai.00210-10] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 04/02/2010] [Accepted: 04/25/2010] [Indexed: 11/20/2022] Open
Abstract
The acquisition of iron during the infection process is essential for the growth of pathogenic microorganisms (S. C. Andrews, Adv. Microb. Physiol. 40:281-351, 1998; H. M. Baker, B. F. Anderson, and E. N. Baker, Proc. Natl. Acad. Sci. U. S. A. 100:3579-3583, 2003). Since the solubility of iron is low and it is toxic at low concentrations, following uptake, iron is stored in subcellular microenvironments in the iron storage protein ferritin (C. Cheers and M. Ho, J. Reticuloendothel. Soc. 34:299-309, 1983). Here, we show that ferritin-like proteins (Frl) are highly conserved in the genus Listeria and demonstrate that these proteins are present in both the cytoplasm and cell wall fractions of these bacteria. Even though Frl is expressed under different growth conditions, transcriptional mapping revealed that its regulation is complex. When bacteria are grown in brain heart infusion medium, extracellular expression involves both sigma A (SigA)- and sigma B (SigB)-dependent promoters; however, during intracellular growth, initiation of transcription is additionally SigB dependent. The expression of Frl is greatly enhanced in bacteria grown in the presence of blood, and a mutant strain lacking the frl gene was defective for growth in this medium. Using the monoclonal antibody (MAb) specific for Frl, we demonstrate that administration of anti-Frl MAb prior to infection confers antilisterial resistance in vivo, evidenced in reduced bacterial load and increased survival rates, thereby demonstrating the in vivo significance of upregulated cell surface-associated Frl expression. In vitro studies revealed that the antilisterial resistance is due to increased listerial phagocytosis.
Collapse
Affiliation(s)
- Walid Mohamed
- Institute for Medical Microbiology, Justus-Liebig-University, Frankfurter Str. 107, D-35392 Giessen, Germany, Centre de Recerca en Sanitat Animal, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Shneh Sethi
- Institute for Medical Microbiology, Justus-Liebig-University, Frankfurter Str. 107, D-35392 Giessen, Germany, Centre de Recerca en Sanitat Animal, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Ayub Darji
- Institute for Medical Microbiology, Justus-Liebig-University, Frankfurter Str. 107, D-35392 Giessen, Germany, Centre de Recerca en Sanitat Animal, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Mobarak A. Mraheil
- Institute for Medical Microbiology, Justus-Liebig-University, Frankfurter Str. 107, D-35392 Giessen, Germany, Centre de Recerca en Sanitat Animal, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Torsten Hain
- Institute for Medical Microbiology, Justus-Liebig-University, Frankfurter Str. 107, D-35392 Giessen, Germany, Centre de Recerca en Sanitat Animal, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Trinad Chakraborty
- Institute for Medical Microbiology, Justus-Liebig-University, Frankfurter Str. 107, D-35392 Giessen, Germany, Centre de Recerca en Sanitat Animal, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| |
Collapse
|