1
|
Wantuch PL, Knoot CJ, Marino EC, Harding CM, Rosen DA. Klebsiella pneumoniae bioconjugate vaccine functional durability in mice. Vaccine 2025; 43:126536. [PMID: 39571358 DOI: 10.1016/j.vaccine.2024.126536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024]
Abstract
Klebsiella pneumoniae is a leading cause of hospital-acquired infections as well as the leading cause of neonatal sepsis worldwide. Further, increasing antibiotic resistance in this pathogen makes K. pneumoniae troublesome to treat. Despite its clinical importance, there is not yet an approved K. pneumoniae vaccine available. Here we tested antibody durability and long-term functionality of two previously reported bioconjugate vaccines targeting the K. pneumoniae capsular type K2 and O-antigen type O1v1. We demonstrate that both antibodies are durable in mice for up to six months with significant IgG titers. However, only the K2 antibodies exhibit functionality out to six months as evidenced by serum bactericidal activity and survival in a murine bacteremia challenge model. These results are another promising step towards demonstrating the clinical capacity of bioconjugate vaccines and their induction of durable antibody responses.
Collapse
Affiliation(s)
- Paeton L Wantuch
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Emily C Marino
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - David A Rosen
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
2
|
Zaleski MH, Chase LS, Hood ED, Wang Z, Nong J, Espy CL, Zamora ME, Wu J, Morrell LJ, Muzykantov VR, Myerson JW, Brenner JS. Conjugation Chemistry Markedly Impacts Toxicity and Biodistribution of Targeted Nanoparticles, Mediated by Complement Activation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2409945. [PMID: 39663706 DOI: 10.1002/adma.202409945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/21/2024] [Indexed: 12/13/2024]
Abstract
Conjugation chemistries are a major enabling technology for the development of drug delivery systems, from antibody-drug conjugates to antibody-targeted lipid nanoparticles inspired by the success of the COVID-19 vaccine. However, here it is shown that for antibody-targeted nanoparticles, the most popular conjugation chemistries directly participate in the activation of the complement cascade of plasma proteins. Their activation of complement leads to large changes in the biodistribution of nanoparticles (up to 140-fold increased uptake into phagocytes of the lungs) and multiple toxicities, including a 50% drop in platelet count. It is founded that the mechanism of complement activation varies dramatically between different conjugation chemistries. Dibenzocyclooctyne, a commonly used click-chemistry, caused aggregation of conjugated antibodies, but only on the surface of nanoparticles (not in bulk solution). By contrast, thiol-maleimide chemistry do not activate complement via its effects on antibodies, but rather because free maleimide bonded to albumin in plasma, and clustered albumin is then attacked by complement. Using these mechanistic insights, solutions are engineered that reduced the activation of complement for each class of conjugation chemistry. These results highlight that while conjugation chemistry is essential for the future of nanomedicine, it is not innocuous and must be designed with opsonins like complement in mind.
Collapse
Affiliation(s)
- Michael H Zaleski
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Liam S Chase
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Zhicheng Wang
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Carolann L Espy
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Marco E Zamora
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Jichuan Wu
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Lianne J Morrell
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., 354 BRB II/III, Philadelphia, PA, 19104, USA
- Department of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| |
Collapse
|
3
|
Chen H, Luo Y, Li X, Zhang Y, Zheng S, Chen J, Sun Y, Xie Y. The differences of characteristics and allergenicity between natural and recombinant tropomyosin of Macrobrachium nipponense. Food Chem 2024; 460:140610. [PMID: 39068796 DOI: 10.1016/j.foodchem.2024.140610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/06/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Tropomyosin (TM) is the main allergen of Macrobrachium nipponense. Recombinant allergens have great prospects in the detection, diagnosis, and treatment of food allergens. The purpose of this study was to compare the differences in structure and allergenicity between natural TM and recombinant TM. Recombinant TM of M. nipponense with a molecular weight of 38 kDa was successfully expressed in the Escherichia coli system. The amino acid sequence as well as secondary structure between natural and recombinant TM were similar, which were verified by mass and CD spectrometry, respectively. Studies showed that both natural TM and recombinant TM had strong allergenicity, and recombinant TM was more allergenic, which could be used as a substitute for natural TM in the diagnosis and treatment of shrimp allergy. This study provided stable and reliable allergen components for the detection of crustacean allergens and the diagnosis and treatment of food allergies caused by crustacean allergens.
Collapse
Affiliation(s)
- Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China
| | - Yeqing Luo
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Yingxue Zhang
- Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Shuangyan Zheng
- Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China
| | - Jiao Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Yaobin Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Yanhai Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China.
| |
Collapse
|
4
|
Archambault MJ, Tshibwabwa LM, Côté-Cyr M, Moffet S, Shiao TC, Bourgault S. Nanoparticles as Delivery Systems for Antigenic Saccharides: From Conjugation Chemistry to Vaccine Design. Vaccines (Basel) 2024; 12:1290. [PMID: 39591192 PMCID: PMC11598982 DOI: 10.3390/vaccines12111290] [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: 09/11/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Glycoconjugate vaccines have been effective in preventing numerous bacterial infectious diseases and have shown recent potential to treat cancers through active immunotherapy. Soluble polysaccharides elicit short-lasting immune responses and are usually covalently linked to immunogenic carrier proteins to enhance the antigen-specific immune response by stimulating T-cell-dependent mechanisms. Nonetheless, the conjugation of purified polysaccharides to carrier proteins complexifies vaccine production, and immunization with protein glycoconjugates can lead to the undesirable immunogenic interference of the carrier. Recently, the use of nanoparticles and nanoassemblies for the delivery of antigenic saccharides has gathered attention from the scientific community. Nanoparticles can be easily functionalized with a diversity of functionalities, including T-cell epitope, immunomodulator and synthetic saccharides, allowing for the modulation and polarization of the glycoantigen-specific immune response. Notably, the conjugation of glycan to nanoparticles protects the antigens from degradation and enhances their uptake by immune cells. Different types of nanoparticles, such as liposomes assembled from lipids, inorganic nanoparticles, virus-like particles and dendrimers, have been explored for glycovaccine design. The versatility of nanoparticles and their ability to induce robust immune responses make them attractive delivery platforms for antigenic saccharides. The present review aims at summarizing recent advancements in the use of nano-scaled systems for the delivery of synthetic glycoantigens. After briefly presenting the immunological mechanisms required to promote a robust immune response against antigenic saccharides, this review will offer an overview of the current trends in the nanoparticle-based delivery of glycoantigens.
Collapse
Affiliation(s)
- Marie-Jeanne Archambault
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| | - Laetitia Mwadi Tshibwabwa
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| | - Mélanie Côté-Cyr
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| | - Serge Moffet
- Glycovax Pharma Inc., Laval, QC H7V 5B7, Canada; (S.M.); (T.C.S.)
| | - Tze Chieh Shiao
- Glycovax Pharma Inc., Laval, QC H7V 5B7, Canada; (S.M.); (T.C.S.)
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| |
Collapse
|
5
|
Aymerich N, Schlotheuber LJ, Bucheli OTM, Portmann K, Baudry J, Eyer K. Antibody density on bacteria regulates C1q recruitment by monoclonal IgG but not IgM. Eur J Immunol 2024; 54:e2451228. [PMID: 39233515 DOI: 10.1002/eji.202451228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/06/2024]
Abstract
Antibodies that trigger the complement system play a pivotal role in the immune defense against pathogenic bacteria and offer potential therapeutic avenues for combating antibiotic-resistant bacterial infections, a rising global concern. To gain a deeper understanding of the key parameters regulating complement activation by monoclonal antibodies, we developed a novel bioassay for quantifying classical complement activation at the monoclonal antibody level, and employed this assay to characterize rare complement-activating antibacterial antibodies on the single-antibody level in postimmunization murine antibody repertoires. We characterized monoclonal antibodies from various antibody isotypes against specific pathogenic bacteria (Bordetella pertussis and Neisseria meningitidis) to broaden the scope of our findings. We demonstrated activation of the classical pathway by individual IgM- and IgG-secreting cells, that is, monoclonal IgM and IgG2a/2b/3 subclasses. Additionally, we could observe different epitope density requirements for efficient C1q binding depending on antibody isotype, which is in agreement with previously proposed molecular mechanisms. In short, we found that antibody density most crucially regulated C1q recruitment by monoclonal IgG isotypes, but not IgM isotypes. This study provides additional insights into important parameters for classical complement initiation by monoclonal antibodies, a knowledge that might inform antibody screening and vaccination efforts.
Collapse
Affiliation(s)
- Nathan Aymerich
- Laboratoire Colloïdes et Matériaux Divisés (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, Paris, France
| | - Luca J Schlotheuber
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Olivia T M Bucheli
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Kevin Portmann
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Jean Baudry
- Laboratoire Colloïdes et Matériaux Divisés (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, Paris, France
| | - Klaus Eyer
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
6
|
Euliano EM, Pogostin BH, Agrawal A, Yu MH, Baryakova TH, Graf TP, Kunkel AA, Cahue KA, Hartgerink JD, McHugh KJ. A TLR7 Agonist Conjugated to a Nanofibrous Peptide Hydrogel as a Potent Vaccine Adjuvant. Adv Healthc Mater 2024:e2402958. [PMID: 39460390 DOI: 10.1002/adhm.202402958] [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: 08/08/2024] [Indexed: 10/28/2024]
Abstract
Toll-like receptors (TLRs) recognize pathogen- and damage-associated molecular patterns and, in turn, trigger the release of cytokines and other immunostimulatory molecules. As a result, TLR agonists are increasingly being investigated as vaccine adjuvants. Many of these agonists are small molecules that quickly diffuse away from the vaccination site, limiting their co-localization with antigens and, thus, their effect. Here, the small-molecule TLR7 agonist 1V209 is conjugated to a positively-charged multidomain peptide (MDP) hydrogel, K2, which was previously shown to act as an adjuvant promoting humoral immunity. Mixing the 1V209-conjugated K2 50:50 with the unfunctionalized K2 produces hydrogels that retain the shear-thinning and self-healing physical properties of the original MDP while improving the solubility of 1V209 more than 200-fold compared to the unconjugated molecule. When co-delivered with ovalbumin as a model antigen, 1V209-functionalized K2 produces a robust Th2 immune response and an antigen-specific Th1 immune response superior to alum, a widely used vaccine adjuvant. Together, these results suggest that K2 MDP hydrogels functionalized with 1V209 are a promising adjuvant for vaccines against infectious diseases, especially those benefiting from a combined Th1 and Th2 immune response.
Collapse
Affiliation(s)
- Erin M Euliano
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Brett H Pogostin
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Anushka Agrawal
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Marina H Yu
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | | | - Tyler P Graf
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Alyssa A Kunkel
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Kiana A Cahue
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| |
Collapse
|
7
|
Nguyen GT, Le TT, Vu SDT, Nguyen TT, Le MTT, Pham VT, Nguyen HTT, Ho TT, Hoang HTT, Tran HX, Chu HH, Pham NB. A plant-based oligomeric CD2v extracellular domain antigen exhibits equivalent immunogenicity to the live attenuated vaccine ASFV-G-∆I177L. Med Microbiol Immunol 2024; 213:22. [PMID: 39412651 DOI: 10.1007/s00430-024-00804-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/07/2024] [Indexed: 12/28/2024]
Abstract
African swine fever (ASF), caused by the African swine fever virus (ASFV), is a deadly, highly contagious disease in both domestic pigs and wild boar. With mortality up to 100%, the disease has been making a serious impact on the swine industry worldwide. Because no effective antiviral treatment has been observed, proactive prevention such as vaccination remains the key to controlling the outbreak. In the pursuit of expediting vaccine development, our current work has made the first report for heterologous production of the viral outer envelope glycoprotein CD2v extracellular domain (CD2v ED), a proposed promising vaccine antigen candidate in the "green" synthetic host Nicotiana benthamiana. Protein oligomerization strategies were implemented to increase the immunogenicity of the target antigen. Herein, the protein was expressed in oligomeric forms based on the C-terminally fused GCN4pII trimerization motif and GCN4pII_TP oligomerization motif. Quantitative western blot analysis showed significantly higher expression of trimeric CD2v ED_GCN4pII with a yield of about 12 mg/100 g of fresh weight, in comparison to oligomeric CD2v ED_GCN4pII_TP, revealing the former is the better choice for further studies. The results of purification and size determination by size exclusion chromatography (SEC) illustrated that CD2v ED_GCN4pII was successfully produced in stable oligomeric forms throughout the extraction, purification, and analysis process. Most importantly, purified CD2v ED_GCN4pII was demonstrated to induce both humoral and cellular immunity responses in mice to extents equivalent to those of the live attenuated vaccine ASFV-G-∆I177L, suggesting it as the potential subunit vaccine candidate for preventing ASFV.
Collapse
Affiliation(s)
- Giang Thu Nguyen
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Thanh Thi Le
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Son Duy Thai Vu
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Tra Thi Nguyen
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - My Thi Tra Le
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Van Thi Pham
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Hien Thi Thu Nguyen
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Thuong Thi Ho
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Hang Thi Thu Hoang
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Hanh Xuan Tran
- National Veterinary Joint Stock Company - NAVETCO, Ho Chi Minh City, Viet Nam
| | - Ha Hoang Chu
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| | - Ngoc Bich Pham
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam.
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Viet Nam.
| |
Collapse
|
8
|
Vuitika L, Côrtes N, Malaquias VB, Silva JDQ, Lira A, Prates-Syed WA, Schimke LF, Luz D, Durães-Carvalho R, Balan A, Câmara NOS, Cabral-Marques O, Krieger JE, Hirata MH, Cabral-Miranda G. A self-adjuvanted VLPs-based Covid-19 vaccine proven versatile, safe, and highly protective. Sci Rep 2024; 14:24228. [PMID: 39414952 PMCID: PMC11484777 DOI: 10.1038/s41598-024-76163-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024] Open
Abstract
Vaccination has played a critical role in mitigating COVID-19. Despite the availability of licensed vaccines, there remains a pressing need for improved vaccine platforms that provide high protection, safety, and versatility, while also reducing vaccine costs. In response to these challenges, our aim is to create a self-adjuvanted vaccine against SARS-CoV-2, utilizing Virus-Like Particles (VLPs) as the foundation. To achieve this, we produced bacteriophage (Qβ) VLPs in a prokaryotic system and purified them using a rapid and cost-effective strategy involving organic solvents. This method aims to solubilize lipids and components of the cell membrane to eliminate endotoxins present in bacterial samples. For vaccine formulation, Receptor Binding Domain (RBD) antigens were conjugated using chemical crosslinkers, a process compatible with Good Manufacturing Practice (GMP) standards. Transmission Electron Microscopy (TEM) confirmed the expected folding and spatial configuration of the QβVLPs vaccine. Additionally, vaccine formulation assessment involved SDS-PAGE stained with Coomassie Brilliant Blue, Western blotting, and stereomicroscopic experiments. In vitro and in vivo evaluations of the vaccine formulation were conducted to assess its capacity to induce a protective immune response without causing side effects. Vaccine doses of 20 µg and 50 µg stimulated the production of neutralizing antibodies. In in vivo testing, the group of animals vaccinated with 50 µg of vaccine formulation provided complete protection against virus infection, maintaining stable body weight without showing signs of disease. In conclusion, the QβVLPs-RBD vaccine has proven to be effective and safe, eliminating the necessity for supplementary adjuvants and offering a financially feasible approach. Moreover, this vaccine platform demonstrates flexibility in targeting Variants of Concern (VOCs) via established conjugation protocols with VLPs.
Collapse
Affiliation(s)
- Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nelson Côrtes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vanessa B Malaquias
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jaqueline D Q Silva
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Aline Lira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Wasim A Prates-Syed
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29., University of São Paulo School of Medicine, São Paulo, Brazil
| | - Daniela Luz
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | - Ricardo Durães-Carvalho
- São Paulo School of Medicine, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
- Interunit Bioinformatics Graduate Program, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Andrea Balan
- Applied Structural Biology Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Niels O S Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29., University of São Paulo School of Medicine, São Paulo, Brazil
- DO'R Institute for research, São Paulo, Brazil, IDOR, São Paulo, Brazil
| | - José E Krieger
- Heart Institute, Clinical Hospital, Faculty of Medicine, Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mario H Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil.
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
9
|
Dvorscek AR, McKenzie CI, Stäheli VC, Ding Z, White J, Fabb SA, Lim L, O'Donnell K, Pitt C, Christ D, Hill DL, Pouton CW, Burnett DL, Brink R, Robinson MJ, Tarlinton DM, Quast I. Conversion of vaccines from low to high immunogenicity by antibodies with epitope complementarity. Immunity 2024; 57:2433-2452.e7. [PMID: 39305904 DOI: 10.1016/j.immuni.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/06/2024] [Accepted: 08/27/2024] [Indexed: 10/11/2024]
Abstract
Existing antibodies (Abs) have varied effects on humoral immunity during subsequent infections. Here, we leveraged in vivo systems that allow precise control of antigen-specific Abs and B cells to examine the impact of Ab dose, affinity, and specificity in directing B cell activation and differentiation. Abs competing with the B cell receptor (BCR) epitope showed affinity-dependent suppression. By contrast, Abs targeting a complementary epitope, not overlapping with the BCR, shifted B cell differentiation toward Ab-secreting cells. Such Abs allowed for potent germinal center (GC) responses to otherwise poorly immunogenic sites by promoting antigen capture and presentation by low-affinity B cells. These mechanisms jointly diversified the B cell repertoire by facilitating the recruitment of high- and low-affinity B cells into Ab-secreting cell, GC, and memory B cell fates. Incorporation of small amounts of monoclonal Abs into protein- or mRNA-based vaccines enhanced immunogenicity and facilitated sustained immune responses, with implications for vaccine design and our understanding of protective immunity.
Collapse
Affiliation(s)
- Alexandra R Dvorscek
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Craig I McKenzie
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Vera C Stäheli
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Zhoujie Ding
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Jacqueline White
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Kristy O'Donnell
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Catherine Pitt
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Danika L Hill
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Deborah L Burnett
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2010, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Marcus J Robinson
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - David M Tarlinton
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Isaak Quast
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia.
| |
Collapse
|
10
|
Colombatti Olivieri MA, Cuerda MX, Moyano RD, Gravisaco MJ, Pinedo MFA, Delgado FO, Calamante G, Mundo S, de la Paz Santangelo M, Romano MI, Alonso MN, Del Medico Zajac MP. Superior protection against paratuberculosis by a heterologous prime-boost immunization in a murine model. Vaccine 2024; 42:126055. [PMID: 38880691 DOI: 10.1016/j.vaccine.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Vaccination is the best strategy to control Paratuberculosis (PTB), which is a significant disease in cattle and sheep. Previously we showed the humoral and cellular immune response induced by a novel vaccine candidate against PTB based on the Argentinian Mycobacterium avium subspecies paratuberculosis (Map) 6611 strain. To improve 6611 immunogenicity and efficacy, we evaluated this vaccine candidate in mice with two different adjuvants and a heterologous boost with a recombinant modified vaccinia Ankara virus (MVA) expressing the antigen 85A (MVA85A). We observed that boosting with MVA85A did not improve total IgG or specific isotypes in serum induced by one or two doses of 6611 formulated with incomplete Freund's adjuvant (IFA). However, when 6611 was formulated with ISA201 adjuvant, MVA85A boost enhanced the production of IFNγ, Th1/Th17 cytokines (IL-2, TNF, IL-17A) and IL-6, IL-4 and IL-10. Also, this group showed the highest levels of IgG2b and IgG3 isotypes, both important for better protection against Map infection in the murine model. Finally, the heterologous scheme elicited the highest levels of protection after Map challenge (lowest CFU count and liver lesion score). In conclusion, our results encourage further evaluation of 6611 strain + ISA201 prime and MVA85A boost in bovines.
Collapse
MESH Headings
- Animals
- Mycobacterium avium subsp. paratuberculosis/immunology
- Immunization, Secondary/methods
- Mice
- Paratuberculosis/prevention & control
- Paratuberculosis/immunology
- Immunoglobulin G/blood
- Cytokines/metabolism
- Female
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Adjuvants, Immunologic/administration & dosage
- Disease Models, Animal
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Mice, Inbred BALB C
- Vaccinia virus/immunology
- Vaccinia virus/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Immunity, Cellular/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Freund's Adjuvant/administration & dosage
- Freund's Adjuvant/immunology
Collapse
Affiliation(s)
| | - María Ximena Cuerda
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Roberto Damián Moyano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María José Gravisaco
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Fiorella Alvarado Pinedo
- Centro de Diagnóstico e Investigaciones Veterinarias (CEDIVE) de la Facultad de Ciencias Veterinarias - Universidad de La Plata, Chascomús, Buenos Aires 7130, Argentina
| | - Fernando Oscar Delgado
- Instituto de Patobiologia Veterinaria (IPV), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Gabriela Calamante
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Silvia Mundo
- Cátedra de Inmunología de la Facultad de Ciencias Veterinarias - Universidad de Buenos Aires, Ciudad de Buenos Aires 1427, Argentina
| | - María de la Paz Santangelo
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Isabel Romano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Natalia Alonso
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina.
| | - María Paula Del Medico Zajac
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| |
Collapse
|
11
|
Acúrcio RC, Kleiner R, Vaskovich‐Koubi D, Carreira B, Liubomirski Y, Palma C, Yeheskel A, Yeini E, Viana AS, Ferreira V, Araújo C, Mor M, Freund NT, Bacharach E, Gonçalves J, Toister‐Achituv M, Fabregue M, Matthieu S, Guerry C, Zarubica A, Aviel‐Ronen S, Florindo HF, Satchi‐Fainaro R. Intranasal Multiepitope PD-L1-siRNA-Based Nanovaccine: The Next-Gen COVID-19 Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404159. [PMID: 39116324 PMCID: PMC11515909 DOI: 10.1002/advs.202404159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/28/2024] [Indexed: 08/10/2024]
Abstract
The first approved vaccines for human use against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are nanotechnology-based. Although they are modular, rapidly produced, and can reduce disease severity, the currently available vaccines are restricted in preventing infection, stressing the global demand for novel preventive vaccine technologies. Bearing this in mind, we set out to develop a flexible nanovaccine platform for nasal administration to induce mucosal immunity, which is fundamental for optimal protection against respiratory virus infection. The next-generation multiepitope nanovaccines co-deliver immunogenic peptides, selected by an immunoinformatic workflow, along with adjuvants and regulators of the PD-L1 expression. As a case study, we focused on SARS-CoV-2 peptides as relevant antigens to validate the approach. This platform can evoke both local and systemic cellular- and humoral-specific responses against SARS-CoV-2. This led to the secretion of immunoglobulin A (IgA), capable of neutralizing SARS-CoV-2, including variants of concern, following a heterologous immunization strategy. Considering the limitations of the required cold chain distribution for current nanotechnology-based vaccines, it is shown that the lyophilized nanovaccine is stable for long-term at room temperature and retains its in vivo efficacy upon reconstitution. This makes it particularly relevant for developing countries and offers a modular system adaptable to future viral threats.
Collapse
Affiliation(s)
- Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Ron Kleiner
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Daniella Vaskovich‐Koubi
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Bárbara Carreira
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Yulia Liubomirski
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Carolina Palma
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Adva Yeheskel
- The Blavatnik Center for Drug DiscoveryTel Aviv UniversityTel Aviv6997801Israel
| | - Eilam Yeini
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Ana S. Viana
- Center of Chemistry and BiochemistryFaculty of SciencesUniversity of LisbonLisbon1749‐016Portugal
| | - Vera Ferreira
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Carlos Araújo
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Michael Mor
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Natalia T. Freund
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Eran Bacharach
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - João Gonçalves
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | | | - Manon Fabregue
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Solene Matthieu
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Capucine Guerry
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Ana Zarubica
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | | | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Ronit Satchi‐Fainaro
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv6997801Israel
| |
Collapse
|
12
|
Wei P, Romanò C, Li C, Clergeaud G, Andresen TL, Henriksen JR, Hansen AE, Clausen MH. An intranasal cationic liposomal polysaccharide vaccine elicits humoral immune responses against Streptococcus pneumoniae. Commun Biol 2024; 7:1158. [PMID: 39284859 PMCID: PMC11405767 DOI: 10.1038/s42003-024-06806-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
Diseases caused by S. pneumoniae are the leading cause of child mortality. As antibiotic resistance of S. pneumoniae is rising, vaccination remains the most recommended solution. However, the existing pneumococcal polysaccharides vaccine (Pneumovax® 23) proved only to induce T-independent immunity, and strict cold chain dependence of the protein conjugate vaccine impedes its promotion in developing countries, where infections are most problematic. Affordable and efficient vaccines against pneumococcus are therefore in high demand. Here, we present an intranasal vaccine Lipo+CPS12F&αGC, containing the capsular polysaccharides of S. pneumoniae 12F and the iNKT agonist α-galactosylceramide in cationic liposomes. In BALB/cJRj mice, the vaccine effectively activates iNKT cells and promotes B cells maturation, stimulates affinity-matured IgA and IgG production in both the respiratory tract and systemic blood, and displays sufficient protection both in vivo and in vitro. The designed vaccine is a promising, cost-effective solution against pneumococcus, which can be expanded to cover more serotypes and pathogens.
Collapse
Affiliation(s)
- Peng Wei
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Cecilia Romanò
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Chengxin Li
- Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Gael Clergeaud
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Thomas L Andresen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Jonas R Henriksen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Anders E Hansen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Mads H Clausen
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark.
| |
Collapse
|
13
|
Hendy DA, Ma Y, Dixon TA, Murphy CT, Pena ES, Carlock MA, Ross TM, Bachelder EM, Ainslie KM, Fenton OS. Polymeric cGAMP microparticles affect the immunogenicity of a broadly active influenza mRNA lipid nanoparticle vaccine. J Control Release 2024; 372:168-175. [PMID: 38844178 PMCID: PMC11283345 DOI: 10.1016/j.jconrel.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/23/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024]
Abstract
Influenza outbreaks are a major burden worldwide annually. While seasonal vaccines do provide protection against infection, they are limited in that they need to be updated every year to account for the constantly mutating virus. Recently, lipid nanoparticles (LNPs) encapsulating mRNA have seen major success as a vaccine platform for SARS-CoV-2. Herein, we applied LNPs to deliver an mRNA encoding a computationally optimized broadly active (COBRA) influenza immunogen. These COBRA mRNA LNPs induced a broadly active neutralizing antibody response and protection after lethal influenza challenge. To further increase the immunogenicity of the COBRA mRNA LNPs, we combined them with acetalated dextran microparticles encapsulating a STING agonist. Contrary to recent findings, the STING agonist decreased the immunogenicity of the COBRA mRNA LNPs which was likely due to a decrease in mRNA translation as shown in vitro. Overall, this work aids in future selection of adjuvants to use with mRNA LNP vaccines.
Collapse
Affiliation(s)
- Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Timothy A Dixon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Connor T Murphy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Erik S Pena
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Michael A Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, USA
| | - Ted M Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, USA; Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA.
| |
Collapse
|
14
|
Mandviwala AS, Huckriede ALW, Arankalle VA, Patil HP. Mucosal delivery of a prefusogenic-F, glycoprotein, and matrix proteins-based virus-like particle respiratory syncytial virus vaccine induces protective immunity as evidenced by challenge studies in mice. Virology 2024; 598:110194. [PMID: 39096774 DOI: 10.1016/j.virol.2024.110194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/18/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
RSV infection remains a serious threat to the children all over the world, especially, in the low-middle income countries. Vaccine delivery via the mucosa holds great potential for inducing local immune responses in the respiratory tract. Previously, we reported the development of highly immunogenic RSV virus-like-particles (RSV-VLPs) based on the conformationally stable prefusogenic-F protein (preFg), glycoprotein and matrix protein. Here, to explore whether mucosal delivery of RSV-VLPs is an effective strategy to induce RSV-specific mucosal and systemic immunity, RSV-VLPs were administered via the nasal, sublingual and pulmonary routes to BALB/c mice. The results demonstrate that immunization with the VLPs via the mucosal routes induced minimal mucosal response and yet facilitated modest levels of serum IgG antibodies, enhanced T cell responses and the expression of the lung-homing marker CXCR3 on splenocytes. Immunization with VLPs via all three mucosal routes provided protection against RSV challenge with no signs of RSV induced pathology.
Collapse
Affiliation(s)
- Ahmedali S Mandviwala
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Anke L W Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vidya A Arankalle
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Harshad P Patil
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India.
| |
Collapse
|
15
|
Cyster JG, Wilson PC. Antibody modulation of B cell responses-Incorporating positive and negative feedback. Immunity 2024; 57:1466-1481. [PMID: 38986442 PMCID: PMC11257158 DOI: 10.1016/j.immuni.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
Antibodies are powerful modulators of ongoing and future B cell responses. While the concept of antibody feedback has been appreciated for over a century, the topic has seen a surge in interest due to the evidence that the broadening of antibody responses to SARS-CoV-2 after a third mRNA vaccination is a consequence of antibody feedback. Moreover, the discovery that slow antigen delivery can lead to more robust humoral immunity has put a spotlight on the capacity for early antibodies to augment B cell responses. Here, we review the mechanisms whereby antibody feedback shapes B cell responses, integrating findings in humans and in mouse models. We consider the major influence of epitope masking and the diverse actions of complement and Fc receptors and provide a framework for conceptualizing the ways antigen-specific antibodies may influence B cell responses to any form of antigen, in conditions as diverse as infectious disease, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Patrick C Wilson
- Drukier Institute for Children's Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
16
|
Daley AD, Bénézech C. Fat-associated lymphoid clusters: Supporting visceral adipose tissue B cell function in immunity and metabolism. Immunol Rev 2024; 324:78-94. [PMID: 38717136 DOI: 10.1111/imr.13339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/09/2024] [Indexed: 07/23/2024]
Abstract
It is now widely understood that visceral adipose tissue (VAT) is a highly active and dynamic organ, with many functions beyond lipid accumulation and storage. In this review, we discuss the immunological role of this tissue, underpinned by the presence of fat-associated lymphoid clusters (FALCs). FALC's distinctive structure and stromal cell composition support a very different immune cell mix to that found in classical secondary lymphoid organs, which underlies their unique functions of filtration, surveillance, innate-like immune responses, and adaptive immunity within the serous cavities. FALCs are important B cell hubs providing B1 cell-mediated frontline protection against infection and supporting B2 cell-adaptative immune responses. Beyond these beneficial immune responses orchestrated by FALCs, immune cells within VAT play important homeostatic role. Dysregulation of immune cells during obesity and aging leads to chronic pathological "metabolic inflammation", which contributes to the development of cardiometabolic diseases. Here, we examine the emerging and complex functions of B cells in VAT homeostasis and the metabolic complications of obesity, highlighting the potential role that FALCs play and emphasize the areas where further research is needed.
Collapse
Affiliation(s)
- Alexander D Daley
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
17
|
Bissett C, Belij-Rammerstorfer S, Ulaszewska M, Smith H, Kailath R, Morris S, Powers C, Sebastian S, Sharpe HR, Allen ER, Wang Z, Cunliffe RF, Sallah HJ, Spencer AJ, Gilbert S, Tregoning JS, Lambe T. Systemic prime mucosal boost significantly increases protective efficacy of bivalent RSV influenza viral vectored vaccine. NPJ Vaccines 2024; 9:118. [PMID: 38926455 PMCID: PMC11208422 DOI: 10.1038/s41541-024-00912-1] [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: 01/25/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Although licensed vaccines against influenza virus have been successful in reducing pathogen-mediated disease, they have been less effective at preventing viral infection of the airways and current seasonal updates to influenza vaccines do not always successfully accommodate viral drift. Most licensed influenza and recently licensed RSV vaccines are administered via the intramuscular route. Alternative immunisation strategies, such as intranasal vaccinations, and "prime-pull" regimens, may deliver a more sterilising form of protection against respiratory viruses. A bivalent ChAdOx1-based vaccine (ChAdOx1-NP + M1-RSVF) encoding conserved nucleoprotein and matrix 1 proteins from influenza A virus and a modified pre-fusion stabilised RSV A F protein, was designed, developed and tested in preclinical animal models. The aim was to induce broad, cross-protective tissue-resident T cells against heterotypic influenza viruses and neutralising antibodies against RSV in the respiratory mucosa and systemically. When administered via an intramuscular prime-intranasal boost (IM-IN) regimen in mice, superior protection was generated against challenge with either RSV A, Influenza A H3N2 or H1N1. These results support further clinical development of a pan influenza & RSV vaccine administered in a prime-pull regimen.
Collapse
Affiliation(s)
- Cameron Bissett
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | | | - Marta Ulaszewska
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Holly Smith
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Reshma Kailath
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Claire Powers
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Sebastian
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah R Sharpe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Elizabeth R Allen
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, London, UK
| | - Robert F Cunliffe
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Alexandra J Spencer
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Sarah Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Hjálmsdóttir Á, Hasler F, Waeckerle-Men Y, Duda A, López-Deber MP, Pihlgren M, Vukicevic M, Kündig TM, Johansen P. T cell independent antibody responses with class switch and memory using peptides anchored on liposomes. NPJ Vaccines 2024; 9:115. [PMID: 38909055 PMCID: PMC11193769 DOI: 10.1038/s41541-024-00902-3] [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: 06/05/2023] [Accepted: 05/23/2024] [Indexed: 06/24/2024] Open
Abstract
Vaccines generally require T lymphocytes for B-cell activation and immunoglobulin class switching in response to peptide or protein antigens. In the absence of T cells, limited IgG class switch takes place, germinal centers are short-lived, and the B cells lack memory. Here, immunization of mice with liposomes containing 15mer peptides and monophosphoryl lipid A (MPLA) as adjuvant, induced T-cell independent (TI) IgG class switch within three days, as well as germinal center formation. The antibody responses were long-lived, strictly dependent on Toll-like receptor 4 (TLR4) signaling, partly dependent on Bruton's tyrosine kinase (BTK) signal transmission, and independent of signaling through T-cell receptors, MHC class II and inflammasome. The antibody response showed characteristics of both TI type 1 and TI type 2. All IgG subclasses could be boosted months after primary immunization, and the biological function of the secreted antibodies was demonstrated in murine models of allergic anaphylaxis and of bacterial infection. Moreover, antibody responses after immunization with peptide- and MPLA-loaded liposomes could be triggered in neonatal mice and in mice receiving immune-suppressants. This study demonstrates T-cell independent endogenous B-cell memory and recall responses in vivo using a peptide antigen. The stimulation of these antibody responses required a correct and dense assembly and administration of peptide and adjuvant on the surface of liposomes. In the future, TI vaccines may prove beneficial in pathological conditions in which T-cell immunity is compromised through disease or medicines or when rapid, antibody-mediated immune protection is needed.
Collapse
Affiliation(s)
| | - Fabio Hasler
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | | | - Agathe Duda
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Maria Pihlgren
- AC Immune SA, EPFL Innovation Park EPFL, Lausanne, Switzerland
| | | | - Thomas M Kündig
- Department of Dermatology, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Pål Johansen
- Department of Dermatology, University of Zurich, Zurich, Switzerland.
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Furiness KN, El Ansari YS, Oettgen HC, Kanagaratham C. Allergen-specific IgA and IgG antibodies as inhibitors of mast cell function in food allergy. FRONTIERS IN ALLERGY 2024; 5:1389669. [PMID: 38919913 PMCID: PMC11196826 DOI: 10.3389/falgy.2024.1389669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Food allergy, a group of adverse immune responses to normally innocuous food protein antigens, is an increasingly prevalent public health issue. The most common form is IgE-mediated food allergy in which food antigen-induced crosslinking of the high-affinity IgE-receptor, FcεRI, on the surface of mast cells triggers the release of inflammatory mediators that contribute to a wide range of clinical manifestations, including systemic anaphylaxis. Mast cells also play a critical function in adaptive immunity to foods, acting as adjuvants for food-antigen driven Th2 cell responses. While the diagnosis and treatment of food allergy has improved in recent years, no curative treatments are currently available. However, there is emerging evidence to suggest that both allergen-specific IgA and IgG antibodies can counter the activating effects of IgE antibodies on mast cells. Most notably, both antigen-specific IgA and IgG antibodies are induced in the course of oral immunotherapy. In this review, we highlight the role of mast cells in food allergy, both as inducers of immediate hypersensitivity reactions and as adjuvants for type 2 adaptive immune responses. Furthermore, we summarize current understanding of the immunomodulatory effects of antigen-specific IgA and IgG antibodies on IgE-induced mast cell activation and effector function. A more comprehensive understanding of the regulatory role of IgA and IgG in food allergy may provide insights into physiologic regulation of immune responses to ingested antigens and could seed novel strategies to treat allergic disease.
Collapse
Affiliation(s)
- Kameryn N. Furiness
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Yasmeen S. El Ansari
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Hans C. Oettgen
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Cynthia Kanagaratham
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
Tsareva A, Shelyakin PV, Shagina IA, Myshkin MY, Merzlyak EM, Kriukova VV, Apt AS, Linge IA, Chudakov DM, Britanova OV. Aberrant adaptive immune response underlies genetic susceptibility to tuberculosis. Front Immunol 2024; 15:1380971. [PMID: 38799462 PMCID: PMC11116662 DOI: 10.3389/fimmu.2024.1380971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/11/2024] [Indexed: 05/29/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) remains a major threat worldwide, although only a fraction of infected individuals develops tuberculosis (TB). TB susceptibility is shaped by multiple genetic factors, and we performed comparative immunological analysis of two mouse strains to uncover relevant mechanisms underlying susceptibility and resistance. C57BL/6 mice are relatively TB-resistant, whereas I/St mice are prone to develop severe TB, partly due to the MHC-II allelic variant that shapes suboptimal CD4+ T cell receptor repertoire. We investigated the repertoires of lung-infiltrating helper T cells and B cells at the progressed stage in both strains. We found that lung CD4+ T cell repertoires of infected C57BL/6 but not I/St mice contained convergent TCR clusters with functionally confirmed Mtb specificity. Transcriptomic analysis revealed a more prominent Th1 signature in C57BL/6, and expression of pro-inflammatory IL-16 in I/St lung-infiltrating helper T cells. The two strains also showed distinct Th2 signatures. Furthermore, the humoral response of I/St mice was delayed, less focused, and dominated by IgG/IgM isotypes, whereas C57BL/6 mice generated more Mtb antigen-focused IgA response. We conclude that the inability of I/St mice to produce a timely and efficient anti-Mtb adaptive immune responses arises from a suboptimal helper T cell landscape that also impacts the humoral response, leading to diffuse inflammation and severe disease.
Collapse
Affiliation(s)
- Anastasiia Tsareva
- Precision Oncology Division, Boston Gene Laboratory, Waltham, MA, United States
| | - Pavel V. Shelyakin
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Irina A. Shagina
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Mikhail Yu. Myshkin
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Ekaterina M. Merzlyak
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Valeriia V. Kriukova
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Alexander S. Apt
- Laboratory for Immunogenetics, Central Tuberculosis Research Institute, Moscow, Russia
| | - Irina A. Linge
- Laboratory for Immunogenetics, Central Tuberculosis Research Institute, Moscow, Russia
| | - Dmitriy M. Chudakov
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Olga V. Britanova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
21
|
Chung YH, Zhao Z, Jung E, Omole AO, Wang H, Sutorus L, Steinmetz NF. Systemic Administration of Cowpea Mosaic Virus Demonstrates Broad Protection Against Metastatic Cancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308237. [PMID: 38430536 PMCID: PMC11095214 DOI: 10.1002/advs.202308237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/21/2023] [Indexed: 03/04/2024]
Abstract
The key challenge in cancer treatment is prevention of metastatic disease which is therapeutically resistant and carries poor prognoses necessitating efficacious prophylactic approaches that prevent metastasis and recurrence. It is previously demonstrated that cowpea mosaic virus (CPMV) induces durable antitumor responses when used in situ, i.e., intratumoral injection. As a new direction, it is showed that CPMV demonstrates widespread effectiveness as an immunoprophylactic agent - potent efficacy is demonstrated in four metastatic models of colon, ovarian, melanoma, and breast cancer. Systemic administration of CPMV stimulates the innate immune system, enabling attack of cancer cells; processing of the cancer cells and associated antigens leads to systemic, durable, and adaptive antitumor immunity. Overall, CPMV demonstrated broad efficacy as an immunoprophylactic agent in the rejection of metastatic cancer.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Moores Cancer CenterUniversity of California, San DiegoLa JollaCA92093USA
| | - Zhongchao Zhao
- Moores Cancer CenterUniversity of California, San DiegoLa JollaCA92093USA
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
- Center for Nano‐ImmunoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Eunkyeong Jung
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Anthony O. Omole
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Hanyang Wang
- Department of BiologyUniversity of California, San DiegoLa JollaCA92093USA
| | - Lucas Sutorus
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Nicole F. Steinmetz
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Moores Cancer CenterUniversity of California, San DiegoLa JollaCA92093USA
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
- Center for Nano‐ImmunoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
- Department of RadiologyUniversity of California, San DiegoLa JollaCA92093USA
- Institute for Materials Discovery and DesignUniversity of California, San DiegoLa JollaCA92093USA
- Center for Engineering in CancerUniversity of California, San DiegoLa JollaCA92093USA
- Shu and K.C. Chien and Peter Farrell CollaboratoryUniversity of California, San DiegoLa JollaCA92093USA
| |
Collapse
|
22
|
Gupta P, Sághy T, Bollmann M, Jin T, Ohlsson C, Carlsten H, Corciulo C, Engdahl C. Local Immune Activation and Age Impact on Humoral Immunity in Mice, with a Focus on IgG Sialylation. Vaccines (Basel) 2024; 12:479. [PMID: 38793730 PMCID: PMC11125885 DOI: 10.3390/vaccines12050479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/22/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Age alters the host's susceptibility to immune induction. Humoral immunity with circulating antibodies, particularly immunoglobulin G (IgG), plays an essential role in immune response. IgG glycosylation in the fragment crystallizable (Fc) region, including sialylation, is important in regulating the effector function by interacting with Fc gamma receptors (FcγRs). Glycosylation is fundamentally changed with age and inflammatory responses. We aimed to explore the regulation of humoral immunity by comparing responses to antigen-induced immune challenges in young and adult mice using a local antigen-induced arthritis mouse model. This study examines the differences in immune response between healthy and immune-challenged states across these groups. Our initial assessment of the arthritis model indicated that adult mice presented more severe knee swelling than their younger counterparts. In contrast, we found that neither histological assessment, bone mineral density, nor the number of osteoclasts differs. Our data revealed an age-associated but not immune challenge increase in total IgG; the only subtype affected by immune challenge was IgG1 and partially IgG3. Interestingly, the sialylation of IgG2b and IgG3 is affected by age and immune challenges but not stimulated further by immune challenges in adult mice. This suggests a shift in IgG towards a pro-inflammatory and potentially pathogenic state with age and inflammation.
Collapse
Affiliation(s)
- Priti Gupta
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (P.G.); (T.S.); (M.B.); (T.J.); (H.C.)
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
- SciLifeLab, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Tibor Sághy
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (P.G.); (T.S.); (M.B.); (T.J.); (H.C.)
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
- SciLifeLab, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Miriam Bollmann
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (P.G.); (T.S.); (M.B.); (T.J.); (H.C.)
- SciLifeLab, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (P.G.); (T.S.); (M.B.); (T.J.); (H.C.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| | - Hans Carlsten
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (P.G.); (T.S.); (M.B.); (T.J.); (H.C.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| | - Carmen Corciulo
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden;
| | - Cecilia Engdahl
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (P.G.); (T.S.); (M.B.); (T.J.); (H.C.)
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
- SciLifeLab, University of Gothenburg, 413 90 Gothenburg, Sweden
| |
Collapse
|
23
|
Hils M, Hoffard N, Iuliano C, Kreft L, Chakrapani N, Swiontek K, Fischer K, Eberlein B, Köberle M, Fischer J, Hilger C, Ohnmacht C, Kaesler S, Wölbing F, Biedermann T. IgE and anaphylaxis specific to the carbohydrate alpha-gal depend on IL-4. J Allergy Clin Immunol 2024; 153:1050-1062.e6. [PMID: 38135009 PMCID: PMC10997276 DOI: 10.1016/j.jaci.2023.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/23/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Alpha-gal (Galα1-3Galβ1-4GlcNAc) is a carbohydrate with the potential to elicit fatal allergic reactions to mammalian meat and drugs of mammalian origin. This type of allergy is induced by tick bites, and therapeutic options for this skin-driven food allergy are limited to the avoidance of the allergen and treatment of symptoms. Thus, a better understanding of the immune mechanisms resulting in sensitization through the skin is crucial, especially in the case of a carbohydrate allergen for which underlying immune responses are poorly understood. OBJECTIVE We aimed to establish a mouse model of alpha-gal allergy for in-depth immunologic analyses. METHODS Alpha-galactosyltransferase 1-deficient mice devoid of alpha-gal glycosylations were sensitized with the alpha-gal-carrying self-protein mouse serum albumin by repetitive intracutaneous injections in combination with the adjuvant aluminum hydroxide. The role of basophils and IL-4 in sensitization was investigated by antibody-mediated depletion. RESULTS Alpha-gal-sensitized mice displayed increased levels of alpha-gal-specific IgE and IgG1 and developed systemic anaphylaxis on challenge with both alpha-gal-containing glycoproteins and glycolipids. In accordance with alpha-gal-allergic patients, we detected elevated numbers of basophils at the site of sensitization as well as increased numbers of alpha-gal-specific B cells, germinal center B cells, and B cells of IgE and IgG1 isotypes in skin-draining lymph nodes. By depleting IL-4 during sensitization, we demonstrated for the first time that sensitization and elicitation of allergy to alpha-gal and correspondingly to a carbohydrate allergen is dependent on IL-4. CONCLUSION These findings establish IL-4 as a potential target to interfere with alpha-gal allergy elicited by tick bites.
Collapse
Affiliation(s)
- Miriam Hils
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nils Hoffard
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Caterina Iuliano
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Luisa Kreft
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Neera Chakrapani
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kyra Swiontek
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Konrad Fischer
- Department of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Bernadette Eberlein
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jörg Fischer
- Department of Dermatology, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany; Department of Dermatology and Allergology, University Hospital Augsburg, Augsburg, Germany
| | - Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Susanne Kaesler
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Florian Wölbing
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
24
|
Euliano EM, Pogostin BH, Agrawal A, Yu MH, Baryakova TH, Graf TP, Hartgerink JD, McHugh KJ. A TLR7 Agonist Conjugated to a Nanofibrous Peptide Hydrogel as a Potent Vaccine Adjuvant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583938. [PMID: 38496534 PMCID: PMC10942436 DOI: 10.1101/2024.03.07.583938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Toll-like receptors (TLRs) recognize pathogen- and damage-associated molecular patterns and, in turn, trigger the release of cytokines and other immunostimulatory molecules. As a result, TLR agonists are increasingly being investigated as vaccine adjuvants, though many of these agonists are small molecules that quickly diffuse away from the vaccination site, limiting their co-localization with antigens and, thus, their effect. Here, the small-molecule TLR7 agonist 1V209 is conjugated to a positively-charged multidomain peptide (MDP) hydrogel, K 2 , which was previously shown to act as an adjuvant promoting humoral immunity. Mixing the 1V209-conjugated K 2 50:50 with the unfunctionalized K 2 produces hydrogels that retain the shear-thinning and self-healing physical properties of the original MDP, while improving the solubility of 1V209 more than 200-fold compared to the unconjugated molecule. When co-delivered with ovalbumin as a model antigen, 1V209-functionalized K 2 produces antigen-specific IgG titers that were statistically similar to alum, the gold standard adjuvant, and a significantly lower ratio of Th2-associated IgG1 to Th1-associated IgG2a than alum, suggesting a more balanced Th1 and Th2 response. Together, these results suggest that K 2 MDP hydrogels functionalized with 1V209 are a promising adjuvant for vaccines against infectious diseases, especially those benefiting from a combined Th1 and Th2 immune response. Table of Contents Activation of toll-like receptors (TLRs) stimulates a signaling cascade to induce an immune response. A TLR7 agonist was conjugated to an injectable peptide hydrogel, which was then used to deliver a model vaccine antigen. This platform produced antibody titers similar to the gold standard adjuvant alum and demonstrated an improved balance between Th1- and Th2-mediated immunity over alum.
Collapse
|
25
|
Chen Q, Liu L, Guo S, Li L, Yu Y, Liu Z, Tan C, Chen H, Wang X. Characterization of the monoclonal antibody and the immunodominant B-cell epitope of African swine fever virus pA104R by using mouse model. Microbiol Spectr 2024; 12:e0140123. [PMID: 38305163 PMCID: PMC10913377 DOI: 10.1128/spectrum.01401-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 12/20/2023] [Indexed: 02/03/2024] Open
Abstract
The African swine fever virus (ASFV) structural protein pA104R is the only histone-like protein encoded by eukaryotic viruses. pA104R is an essential DNA-binding protein required for DNA replication and genome packaging of ASFV, which are vital for pathogen survival and proliferation. pA104R is an important target molecule for diagnosing, treating, and immune prevention of ASFV. This study characterized monoclonal antibodies (mAbs) against pA104R and found them to recognize natural pA104R in ASFV strains with different genotypes, showing high conservation. Confirmation analyses of pA104R epitopes using mAbs indicated the presence of immunodominant B-cell epitopes, and further characterization showed the high antigenic index and surface accessibility coefficients of the identified epitope. Furthermore, the pA104R protein functions through the polar interactions between the binding amino acid sites; however, these interactions may be blocked by the recognition of generated mAbs. Characterizing the immunodominant B-cell epitope of the ASFV critical proteins, such as pA104R, may contribute to developing sensitive diagnostic tools and vaccine candidate targets.IMPORTANCEAfrican swine fever (ASF) is a highly pathogenic, lethal, and contagious viral disease affecting domestic pigs and wild boars. As no effective vaccine or other treatments have been developed, the control of African swine fever virus (ASFV) relies heavily on virus detection and diagnosis. A potential serological target is the structural protein pA104R. However, the molecular basis of pA104R antigenicity remains unclear, and a specific monoclonal antibody (mAb) against this protein is still unavailable. In this study, mAbs against pA104R were characterized and found to recognize natural pA104R in ASFV strains with different genotypes. In addition, confirmation analyses of pA104R epitopes using mAbs indicated the presence of immunodominant B-cell epitopes, and further characterization showed the high antigenic index and surface accessibility coefficients of the identified epitope. Characteristics of the immunodominant B-cell epitope of ASFV proteins, such as pA104R, may contribute to developing sensitive diagnostic tools and identifying vaccine candidate targets.
Collapse
Affiliation(s)
- Qichao Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Lixinjie Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Shibang Guo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Liang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Yifeng Yu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Zhankui Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan, China
| |
Collapse
|
26
|
Freitas R, Miranda A, Ferreira D, Relvas-Santos M, Castro F, Ferreira E, Gaiteiro C, Soares J, Cotton S, Gonçalves M, Eiras M, Santos B, Palmeira C, Correia MP, Oliveira MJ, Sarmento B, Peixoto A, Santos LL, Silva AMN, Ferreira JA. A multivalent CD44 glycoconjugate vaccine candidate for cancer immunotherapy. J Control Release 2024; 367:540-556. [PMID: 38301927 DOI: 10.1016/j.jconrel.2024.01.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Cancer presents a high mortality rate due to ineffective treatments and tumour relapse with progression. Cancer vaccines hold tremendous potential due to their capability to eradicate tumour and prevent relapse. In this study, we present a novel glycovaccine for precise targeting and immunotherapy of aggressive solid tumours that overexpress CD44 standard isoform (CD44s) carrying immature Tn and sialyl-Tn (sTn) O-glycans. We describe an enzymatic method and an enrichment strategy to generate libraries of well-characterized cancer-specific CD44s-Tn and/or sTn glycoproteoforms, which mimic the heterogeneity found in tumours. We conjugated CD44-Tn-derived glycopeptides with carrier proteins making them more immunogenic, with further demonstration of the importance of this conjugation to overcome the glycopeptides' intrinsic toxicity. We have optimized the glycopeptide-protein maleimide-thiol conjugation chemistry to avoid undesirable cross-linking between carrier proteins and CD44s glycopeptides. The resulting glycovaccines candidates were well-tolerated in vivo, inducing both humoral and cellular immunity, including immunological memory. The generated antibodies exhibited specific reactivity against synthetic CD44s-Tn glycopeptides, CD44s-Tn glycoengineered cells, and human tumours. In summary, we present a promising prototype of a cancer glycovaccine for future therapeutical pre-clinical efficacy validation.
Collapse
Affiliation(s)
- Rui Freitas
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal / INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Andreia Miranda
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal / INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Dylan Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal / INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal / INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal; REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Flávia Castro
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal / INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Eduardo Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
| | - Cristiana Gaiteiro
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
| | - Janine Soares
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Sofia Cotton
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Martina Gonçalves
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Mariana Eiras
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Beatriz Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Carlos Palmeira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; Immunology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; Health School of University Fernando Pessoa, 4249-004 Porto, Portugal
| | - Margareta P Correia
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP), 4200-072 Porto, Portugal
| | - Maria José Oliveira
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal / INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal / INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal; IUCS-CESPU, 4585-116 Gandra, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; Health School of University Fernando Pessoa, 4249-004 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal; Department of Surgical Oncology, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - André M N Silva
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal.
| |
Collapse
|
27
|
Collins AM, Ohlin M, Corcoran M, Heather JM, Ralph D, Law M, Martínez-Barnetche J, Ye J, Richardson E, Gibson WS, Rodriguez OL, Peres A, Yaari G, Watson CT, Lees WD. AIRR-C IG Reference Sets: curated sets of immunoglobulin heavy and light chain germline genes. Front Immunol 2024; 14:1330153. [PMID: 38406579 PMCID: PMC10884231 DOI: 10.3389/fimmu.2023.1330153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/27/2023] [Indexed: 02/27/2024] Open
Abstract
Introduction Analysis of an individual's immunoglobulin (IG) gene repertoire requires the use of high-quality germline gene reference sets. When sets only contain alleles supported by strong evidence, AIRR sequencing (AIRR-seq) data analysis is more accurate and studies of the evolution of IG genes, their allelic variants and the expressed immune repertoire is therefore facilitated. Methods The Adaptive Immune Receptor Repertoire Community (AIRR-C) IG Reference Sets have been developed by including only human IG heavy and light chain alleles that have been confirmed by evidence from multiple high-quality sources. To further improve AIRR-seq analysis, some alleles have been extended to deal with short 3' or 5' truncations that can lead them to be overlooked by alignment utilities. To avoid other challenges for analysis programs, exact paralogs (e.g. IGHV1-69*01 and IGHV1-69D*01) are only represented once in each set, though alternative sequence names are noted in accompanying metadata. Results and discussion The Reference Sets include less than half the previously recognised IG alleles (e.g. just 198 IGHV sequences), and also include a number of novel alleles: 8 IGHV alleles, 2 IGKV alleles and 5 IGLV alleles. Despite their smaller sizes, erroneous calls were eliminated, and excellent coverage was achieved when a set of repertoires comprising over 4 million V(D)J rearrangements from 99 individuals were analyzed using the Sets. The version-tracked AIRR-C IG Reference Sets are freely available at the OGRDB website (https://ogrdb.airr-community.org/germline_sets/Human) and will be regularly updated to include newly observed and previously reported sequences that can be confirmed by new high-quality data.
Collapse
Affiliation(s)
- Andrew M. Collins
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Mats Ohlin
- Department of Immunotechnology, and SciLifeLab, Lund University, Lund, Sweden
| | - Martin Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - James M. Heather
- Mass General Cancer Center, Massachusetts General Hospital, Charlestown, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Duncan Ralph
- Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Mansun Law
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Jesus Martínez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | - Jian Ye
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States
| | - Eve Richardson
- La Jolla Institute for Immunology, San Diego, CA, United States
| | - William S. Gibson
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Oscar L. Rodriguez
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Ayelet Peres
- Bioengineering Program, Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel
| | - Gur Yaari
- Bioengineering Program, Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel
| | - Corey T. Watson
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, KY, United States
| | - William D. Lees
- Institute of Structural and Molecular Biology, Birkbeck College, London, United Kingdom
- Human-Centered Computing and Information Science, Institute for Systems and Computer Engineering, Technology and Science, Porto, Portugal
| |
Collapse
|
28
|
Zhou Y, Li X, Guo Y, Wu Y, Yin L, Tu L, Hong S, Cai H, Ding F. Synthetic self-adjuvanted multivalent Mucin 1 (MUC1) glycopeptide vaccines with improved in vivo antitumor efficacy. MedComm (Beijing) 2024; 5:e484. [PMID: 38344400 PMCID: PMC10857776 DOI: 10.1002/mco2.484] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 10/28/2024] Open
Abstract
The tumor-associated glycoprotein Mucin 1 (MUC1) is aberrantly glycosylated on cancer cells and is considered a promising target for antitumor vaccines. The weak immunogenicity and low sequence homology of mouse mucins and human MUC1 are the main obstacles for the development of vaccines. Herein, a self-adjuvanted strategy combining toll-like receptor 2 lipopeptide ligands and T-cell epitopes and the multivalent effect were used to amplify the immune response and evade the unpredictable immunogenicity, generating two self-adjuvanted three-component MUC1 vaccines (mono- and trivalent MUC1 vaccines). To simulate the aberrantly glycosylated MUC1 glycoprotein, the MUC1 tandem repeat peptide was bounded with Tn antigens at T9, S15, and T16, and served as B-cell epitopes. Results showed that both vaccines elicited a robust antibody response in wild-type mice compared with a weaker response in MUC1 transgenic mice. The trivalent vaccine did not elevate the antibody response level compared with the monovalent vaccine; however, a more delayed tumor growth and prolonged survival time was realized in wild-type and transgenic mouse models treated with the trivalent vaccine. These results indicate that the self-adjuvanted three-component MUC1 vaccines, especially the trivalent vaccine, can trigger robust antitumor effects regardless of sequence homology, and, therefore, show promise for clinical translation.
Collapse
Affiliation(s)
- Yang Zhou
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Xinru Li
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Yajing Guo
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Ye Wu
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Lixin Yin
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Luyun Tu
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| |
Collapse
|
29
|
Yang YS, Chen HJ, Chen XC, Tang HJ, Chang FJ, Huang YL, Pan YL, Kesavan DK, Chen HY, Shang HS, Kuo SC, Chen TL, Chiang MH. Elizabethkingia anophelis outer membrane vesicles as a novel vaccine candidate against infection: insights into immune response and potential for passive immunity. mSphere 2023; 8:e0040023. [PMID: 38014949 PMCID: PMC10732079 DOI: 10.1128/msphere.00400-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE Elizabethkingia anophelis, a Gram-negative pathogen, causes infections such as bacteraemia, pneumonia, and neonatal meningitis. The pathogen resists most antimicrobial classes, making novel approaches urgently needed. In natural settings, Gram-negative bacteria secrete outer membrane vesicles (OMVs) that carry important molecules in the bacterial life cycle. These OMVs are enriched with proteins involved in virulence, survival, and carbohydrate metabolism, making them a promising source for vaccine development against the pathogen. This study investigated the efficacy of imipenem-induced OMVs (iOMVs) as a vaccine candidate against E. anophelis infection in a mouse pneumonia model. Mice immunized with iOMVs were completely protected during lethal-dose challenges. Passive immunization with hyperimmune sera and splenocytes conferred protection against lethal pneumonia. Further investigation is needed to understand the mechanisms underlying the protective effects of iOMV-induced passive immunity, such as the action on specific antibody subclasses or T cell subsets.
Collapse
Grants
- 109-2320-B-016-002-MY2, 110-2320-B-016-014, 111-2320-B-016-015, 112-2314-B-016-023, 112-2314-B-016-039, 112-2314-B-016-024-MY2 Ministry of Science and Technology, Taiwan (MOST)
- TSGH-E-111244, TSGH-E-112253 Tri-Service General Hospital (TSGH)
- CMNDMC11108, CMNDMC11206 Chi Mei Medical Center
- MND-MAB-110-049, MND-MAB-D-111072, MND-MAB-D-112115, MND-MAB-D-113078 MOD | Medical Affairs Bureau (MAB)
Collapse
Affiliation(s)
- Ya-Sung Yang
- Department of Internal Medicine, Division of Infectious Diseases and Tropical Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jui Chen
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Xiao-Chun Chen
- Department and Graduate institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jen Tang
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Fang-Ju Chang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yun-Ling Huang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ling Pan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Dinesh Kumar Kesavan
- School of Material Science, Nanyang Technological University, Singapore, Singapore
| | - Huan-Yuan Chen
- Inflammation Core Facility, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Sheng Shang
- Department of Pathology, Division of Clinical Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Te-Li Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hsien Chiang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
30
|
Seymour BJ, Trent B, Allen BE, Berlinberg AJ, Tangchittsumran J, Jubair WK, Chriswell ME, Liu S, Ornelas A, Stahly A, Alexeev EE, Dowdell AS, Sneed SL, Fechtner S, Kofonow JM, Robertson CE, Dillon SM, Wilson CC, Anthony RM, Frank DN, Colgan SP, Kuhn KA. Microbiota-dependent indole production stimulates the development of collagen-induced arthritis in mice. J Clin Invest 2023; 134:e167671. [PMID: 38113112 PMCID: PMC10866668 DOI: 10.1172/jci167671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Altered tryptophan catabolism has been identified in inflammatory diseases like rheumatoid arthritis (RA) and spondyloarthritis (SpA), but the causal mechanisms linking tryptophan metabolites to disease are unknown. Using the collagen-induced arthritis (CIA) model, we identified alterations in tryptophan metabolism, and specifically indole, that correlated with disease. We demonstrated that both bacteria and dietary tryptophan were required for disease and that indole supplementation was sufficient to induce disease in their absence. When mice with CIA on a low-tryptophan diet were supplemented with indole, we observed significant increases in serum IL-6, TNF, and IL-1β; splenic RORγt+CD4+ T cells and ex vivo collagen-stimulated IL-17 production; and a pattern of anti-collagen antibody isotype switching and glycosylation that corresponded with increased complement fixation. IL-23 neutralization reduced disease severity in indole-induced CIA. Finally, exposure of human colonic lymphocytes to indole increased the expression of genes involved in IL-17 signaling and plasma cell activation. Altogether, we propose a mechanism by which intestinal dysbiosis during inflammatory arthritis results in altered tryptophan catabolism, leading to indole stimulation of arthritis development. Blockade of indole generation may present a unique therapeutic pathway for RA and SpA.
Collapse
Affiliation(s)
| | - Brandon Trent
- Division of Rheumatology, Department of Medicine, and
| | | | | | | | | | | | - Sucai Liu
- Division of Rheumatology, Department of Medicine, and
| | - Alfredo Ornelas
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew Stahly
- Division of Rheumatology, Department of Medicine, and
| | - Erica E. Alexeev
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Alexander S. Dowdell
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sunny L. Sneed
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Jennifer M. Kofonow
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Charles E. Robertson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Stephanie M. Dillon
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cara C. Wilson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Robert M. Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sean P. Colgan
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | |
Collapse
|
31
|
Tran JQ, Muench MO, Gaillard B, Darst O, Tomayko MM, Jackman RP. Polyinosinic: polycytidylic acid induced inflammation enhances while lipopolysaccharide diminishes alloimmunity to platelet transfusion in mice. Front Immunol 2023; 14:1281130. [PMID: 38146372 PMCID: PMC10749330 DOI: 10.3389/fimmu.2023.1281130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Introduction Alloimmune responses against platelet antigens, which dominantly target the major histocompatibility complex (MHC), can cause adverse reactions to subsequent platelet transfusions, platelet refractoriness, or rejection of future transplants. Platelet transfusion recipients include individuals experiencing severe bacterial or viral infections, and how their underlying health modulates platelet alloimmunity is not well understood. Methods This study investigated the effect of underlying inflammation on platelet alloimmunization by modelling viral-like inflammation with polyinosinic-polycytidylic acid (poly(I:C)) or gram-negative bacterial infection with lipopolysaccharide (LPS), hypothesizing that underlying inflammation enhances alloimmunization. Mice were pretreated with poly(I:C), LPS, or nothing, then transfused with non-leukoreduced or leukoreduced platelets. Alloantibodies and allogeneic MHC-specific B cell (allo-B cell) responses were evaluated two weeks later. Rare populations of allo-B cells were identified using MHC tetramers. Results Relative to platelet transfusion alone, prior exposure to poly(I:C) increased the alloantibody response to allogeneic platelet transfusion whereas prior exposure to LPS diminished responses. Prior exposure to poly(I:C) had equivalent, if not moderately diminished, allo-B cell responses relative to platelet transfusion alone and exhibited more robust allo-B cell memory development. Conversely, prior exposure to LPS resulted in diminished allo-B cell frequency, activation, antigen experience, and germinal center formation and altered memory B cell responses. Discussion In conclusion, not all inflammatory environments enhance bystander responses and prior inflammation mediated by LPS on gram-negative bacteria may in fact curtail platelet alloimmunization.
Collapse
Affiliation(s)
- Johnson Q. Tran
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Marcus O. Muench
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Betty Gaillard
- Vitalant Research Institute, San Francisco, CA, United States
| | - Orsolya Darst
- Vitalant Research Institute, San Francisco, CA, United States
| | - Mary M. Tomayko
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, United States
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Rachael P. Jackman
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Janakiraman M, Leliavski A, Varadarajulu J, Jenne D, Krishnamoorthy G. An engineered Fc fusion protein that targets antigen-specific T cells and autoantibodies mitigates autoimmune disease. J Neuroinflammation 2023; 20:291. [PMID: 38057803 DOI: 10.1186/s12974-023-02974-9] [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: 05/04/2022] [Accepted: 11/28/2023] [Indexed: 12/08/2023] Open
Abstract
Current effective therapies for autoimmune diseases rely on systemic immunomodulation that broadly affects all T and/or B cell responses. An ideal therapeutic approach would combine autoantigen-specific targeting of both T and B cell effector functions, including efficient removal of pathogenic autoantibodies. Albeit multiple strategies to induce T cell tolerance in an autoantigen-specific manner have been proposed, therapeutic removal of autoantibodies remains a significant challenge. Here, we devised an approach to target both autoantigen-specific T cells and autoantibodies by producing a central nervous system (CNS) autoantigen myelin oligodendrocyte glycoprotein (MOG)-Fc fusion protein. We demonstrate that MOG-Fc fusion protein has significantly higher bioavailability than monomeric MOG and is efficient in clearing anti-MOG autoantibodies from circulation. We also show that MOG-Fc promotes T cell tolerance and protects mice from MOG-induced autoimmune encephalomyelitis. This multipronged targeting approach may be therapeutically advantageous in the treatment of autoimmunity.
Collapse
Affiliation(s)
- Mathangi Janakiraman
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Alexei Leliavski
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jeeva Varadarajulu
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Dieter Jenne
- Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Gurumoorthy Krishnamoorthy
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
33
|
Raman SNT, Zetner A, Hashem AM, Patel D, Wu J, Gravel C, Gao J, Zhang W, Pfeifle A, Tamming L, Parikh K, Cao J, Tam R, Safronetz D, Chen W, Johnston MJ, Wang L, Sauve S, Rosu-Myles M, Domselaar GV, Li X. Bivalent vaccines effectively protect mice against influenza A and respiratory syncytial viruses. Emerg Microbes Infect 2023; 12:2192821. [PMID: 36927227 PMCID: PMC10171128 DOI: 10.1080/22221751.2023.2192821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Influenza and Respiratory Syncytial virus (RSV) infections together contribute significantly to the burden of acute lower respiratory tract infections. Despite the disease burden, no approved RSV vaccine is available. While approved vaccines are available for influenza, seasonal vaccination is required to maintain protection. In addition to both being respiratory viruses, they follow a common seasonality, which warrants the necessity for a concerted vaccination approach. Here, we designed bivalent vaccines by utilizing highly conserved sequences, targeting both influenza A and RSV, as either a chimeric antigen or individual antigens separated by a ribosome skipping sequence. These vaccines were found to be effective in protecting the animals from challenge by either virus, with mechanisms of protection being substantially interrogated in this communication.
Collapse
Affiliation(s)
- Sathya N. Thulasi Raman
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Adrian Zetner
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Devina Patel
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Jianguo Wu
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Caroline Gravel
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Jun Gao
- Centre for Vaccines Clinical Trials and Biostatistics, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, Canada
| | - Wanyue Zhang
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Annabelle Pfeifle
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Levi Tamming
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Karan Parikh
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Jingxin Cao
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Roger Tam
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - David Safronetz
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council of Canada, Ottawa, Canada
| | - Michael J.W. Johnston
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Chemistry, Carleton University, Ottawa, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Simon Sauve
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Michael Rosu-Myles
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Gary Van Domselaar
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Xuguang Li
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
34
|
Tomas NM, Schnarre A, Dehde S, Lucas R, Hermans-Borgmeyer I, Kretz O, Koellner SMS, Wiech T, Koch-Nolte F, Seifert L, Huber TB, Zahner G. Introduction of a novel chimeric active immunization mouse model of PLA2R1-associated membranous nephropathy. Kidney Int 2023; 104:916-928. [PMID: 37598854 DOI: 10.1016/j.kint.2023.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/22/2023]
Abstract
The phospholipase A2 receptor 1 (PLA2R1) is the major target antigen in patients with membranous nephropathy (MN), an antibody-mediated autoimmune glomerular disease. Investigation of MN pathogenesis has been hampered by the lack of reliable animal models. Here, we overcome this issue by generating a transgenic mouse line expressing a chimeric PLA2R1 (chPLA2R1) consisting of three human PLA2R1 domains (cysteine-rich, fibronectin type-II and CTLD1) and seven murine PLA2R1 domains (CTLD2-8) specifically in podocytes. Mice expressing the chPLA2R1 were healthy at birth and showed no major glomerular alterations when compared to mice with a wild-type PLA2R1 status. Upon active immunization with human PLA2R1 (hPLA2R1), chPLA2R1-positive mice developed anti-hPLA2R1 antibodies, a nephrotic syndrome, and all major histological features of MN, including granular deposition of mouse IgG and complement components in immunofluorescence and subepithelial electron-dense deposits and podocyte foot process effacement in electron microscopy. In order to investigate the role of the complement system in this model, we further crossed chPLA2R1-positive mice with mice lacking the central complement component C3 (C3-/- mice). Upon immunization with hPLA2R1, chPLA2R1-positive C3-/- mice had substantially less severe albuminuria and nephrotic syndrome when compared to chPLA2R1-positive mice with a wild-type C3 status. In conclusion, we introduce a novel active immunization model of PLA2R1-associated MN and demonstrate a pathogenic role of the complement system in this model.
Collapse
Affiliation(s)
- Nicola M Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Annabel Schnarre
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Silke Dehde
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renke Lucas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah M S Koellner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Nephropathology Section, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Larissa Seifert
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gunther Zahner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
35
|
Seymour BJ, Trent B, Allen B, Berlinberg AJ, Tangchittsumran J, Jubair WK, Chriswell ME, Liu S, Ornelas A, Stahly A, Alexeev EE, Dowdell AS, Sneed SL, Fechtner S, Kofonow JM, Robertson CE, Dillon SM, Wilson CC, Anthony RM, Frank DN, Colgan SP, Kuhn KA. Microbiota-dependent indole production is required for the development of collagen-induced arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.561693. [PMID: 37873395 PMCID: PMC10592798 DOI: 10.1101/2023.10.13.561693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Altered tryptophan catabolism has been identified in inflammatory diseases like rheumatoid arthritis (RA) and spondyloarthritis (SpA), but the causal mechanisms linking tryptophan metabolites to disease are unknown. Using the collagen-induced arthritis (CIA) model we identify alterations in tryptophan metabolism, and specifically indole, that correlate with disease. We demonstrate that both bacteria and dietary tryptophan are required for disease, and indole supplementation is sufficient to induce disease in their absence. When mice with CIA on a low-tryptophan diet were supplemented with indole, we observed significant increases in serum IL-6, TNF, and IL-1β; splenic RORγt+CD4+ T cells and ex vivo collagen-stimulated IL-17 production; and a pattern of anti-collagen antibody isotype switching and glycosylation that corresponded with increased complement fixation. IL-23 neutralization reduced disease severity in indole-induced CIA. Finally, exposure of human colon lymphocytes to indole increased expression of genes involved in IL-17 signaling and plasma cell activation. Altogether, we propose a mechanism by which intestinal dysbiosis during inflammatory arthritis results in altered tryptophan catabolism, leading to indole stimulation of arthritis development. Blockade of indole generation may present a novel therapeutic pathway for RA and SpA.
Collapse
Affiliation(s)
- Brenda J. Seymour
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brandon Trent
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brendan Allen
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Adam J. Berlinberg
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jimmy Tangchittsumran
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Widian K. Jubair
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alfredo Ornelas
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Stahly
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erica E. Alexeev
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexander S. Dowdell
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sunny L. Sneed
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sabrina Fechtner
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer M. Kofonow
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charles E. Robertson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie M. Dillon
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cara C. Wilson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert M. Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sean P. Colgan
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
36
|
da Silva BB, da Silva Junior AB, Araújo LDS, Santos ENFN, da Silva ACM, Florean EOPT, van Tilburg MF, Guedes MIF. Subcutaneous, Oral, and Intranasal Immunization of BALB/c Mice with Leishmania infantum K39 Antigen Induces Non-Protective Humoral Immune Response. Trop Med Infect Dis 2023; 8:444. [PMID: 37755905 PMCID: PMC10534909 DOI: 10.3390/tropicalmed8090444] [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: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Visceral leishmaniasis is a high-burden disease caused by parasites of the Leishmania genus. The K39 kinesin is a highly antigenic protein of Leishmania infantum, but little is known about the immune response elicited by this antigen. We evaluated the humoral immune response of female BALB/c mice (n = 6) immunized with the rK39-HFBI construct, formed by the fusion of the K39 antigen to a hydrophobin partner. The rK39-HFBI construct was administered through subcutaneous, oral, and intranasal routes using saponin as an adjuvant. We analyzed the kinetics of IgG, IgG1, and IgG2a production. The groups were then challenged by an intravenous infection with L. infantum promastigote cells. The rK39-HFBI antigen-induced high levels of total IgG (p < 0.05) in all groups, but only the subcutaneous route was associated with increased production of IgG1 and IgG2a 42 days after immunization (p < 0.05), suggesting a potential secondary immune response following the booster dose. There was no reduction in the splenic parasite load; thus, the rK39-HFBI failed to protect the mice against infection under the tested conditions. The results presented here demonstrate that the high antigenicity of the K39 antigen does not contribute to a protective immune response against visceral leishmaniasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Maria Izabel Florindo Guedes
- Laboratory of Biotechnology and Molecular Biology, Northeast Biotechnology Network (RENORBIO), State University of Ceará, Fortaleza 60714903, Brazil
| |
Collapse
|
37
|
Gupta P, Sághy T, Nordqvist J, Nilsson J, Carlsten H, Horkeby K, Henning P, Engdahl C. Impact of estrogen on IgG glycosylation and serum protein glycosylation in a murine model of healthy postmenopause. Front Endocrinol (Lausanne) 2023; 14:1243942. [PMID: 37766692 PMCID: PMC10519799 DOI: 10.3389/fendo.2023.1243942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/07/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction The glycosylation of immunoglobulin (Ig) G regulates IgG interaction capability with Fc gamma receptors found in all immune cells. In pathogenic conditions, estrogen can impact IgG levels and glycosylation. Following menopause, when estrogen levels decline affecting the immune system and potentially leading to a heightened susceptibility of immune activation. Purpose In this study, we aim to determine if estrogen levels can regulate IgG glycosylation in postmenopausal healthy situations. Methods Mice were ovariectomized to simulate an estrogen-deficient postmenopausal status and then treated with 17-beta-estradiol (E2) at different doses and different administration strategies. Results Using a highly sensitive liquid chromatography-tandem mass spectrometry (MS/MS) glycoproteomic method, we demonstrated that E2 treatment increased the degree of glycosylation on IgG-Fc with both galactosylation and sialylation in the position required for interaction with Fc gamma receptors. We also observed that only long-term estrogen deficiency reduces IgG levels and that estrogen status had no impact on total IgG sialylation on both Fab and Fc domains or general glycoprotein sialylation evaluated by ELISA. Furthermore, E2 status did not affect the total sialic acid content of total cells in lymphoid organs and neither B cells nor plasma cells. Conclusion The study concluded that E2 treatment does not affect total serum glycoprotein sialylation but alters IgG glycosylation, including IgG sialylation, implying that estrogen functions as an intrinsic modulator of IgG sialylation and could thereby be one pathway by which estrogen modulates immunity.
Collapse
Affiliation(s)
- Priti Gupta
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Tibor Sághy
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Jauquline Nordqvist
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jonas Nilsson
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hans Carlsten
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin Horkeby
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Engdahl
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
38
|
Hendy DA, Lifshits LM, Batty CJ, Carlock MA, Ross TM, Mousa JJ, Bachelder EM, Ainslie KM. Zinc Carnosine Metal-Organic Coordination Polymer as a Potent Broadly Active Influenza Vaccine Platform with In Vitro Shelf-Stability. Mol Pharm 2023; 20:4687-4697. [PMID: 37603310 DOI: 10.1021/acs.molpharmaceut.3c00424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Current seasonal influenza vaccines are limited in that they need to be reformulated every year in order to account for the constant mutation of the virus. Hemagglutinin (HA) immunogens have been developed using a computationally optimized broadly reactive antigen (COBRA) methodology, which are able to elicit an antibody response that neutralizes antigenically distinct influenza strains; however, subunit proteins are not immunogenic enough on their own to generate a substantial immune response. Due to this, different delivery strategies and adjuvants can be used to improve immunogenicity. Recently, we reported a new coordination polymer composed of the dipeptide carnosine and zinc (ZnCar) that is able to deliver protein antigens along with CpG to generate a potent immune response. In the present work, ZnCar was used to deliver the COBRA HA immunogen Y2 and the adjuvant CpG. We incorporated Y2 into ZnCar using two different methods to assess which would be the most immunogenic. Mice vaccinated with Y2 and CpG complexed with ZnCar showed an improved humoral and cellular response when compared to mice vaccinated with soluble Y2 and CpG. Further, we demonstrate in vitro that when Y2 and CpG are coordinated with ZnCar, they are protected from degradation at 40 °C for 3 months or 24 °C for 6 months. Overall, ZnCar shows promise as a delivery vehicle for subunit vaccines, given its superior immunogenicity and in vitro storage stability.
Collapse
Affiliation(s)
- Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill 27599, North Carolina, United States
| | - Liubov M Lifshits
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill 27599, North Carolina, United States
| | - Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill 27599, North Carolina, United States
| | - Michael A Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie 33331-3609, Florida, United States
| | - Ted M Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie 33331-3609, Florida, United States
- Center for Vaccines and Immunology, University of Georgia, Athens 30602-0002, Georgia, United States
- Department of Infectious Diseases, University of Georgia, Athens 30602-0002, Georgia, United States
| | - Jarrod J Mousa
- Center for Vaccines and Immunology, University of Georgia, Athens 30602-0002, Georgia, United States
- Department of Infectious Diseases, University of Georgia, Athens 30602-0002, Georgia, United States
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill 27599, North Carolina, United States
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill 27599, North Carolina, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill 27599, North Carolina, United States
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill 27599, North Carolina, United States
| |
Collapse
|
39
|
Ferguson MR, Delgado KN, McBride S, Orbe IC, La Vake CJ, Caimano MJ, Mendez Q, Moraes TF, Schryvers AB, Moody MA, Radolf JD, Weiner MP, Hawley KL. Use of Epivolve phage display to generate a monoclonal antibody with opsonic activity directed against a subdominant epitope on extracellular loop 4 of Treponema pallidum BamA (TP0326). Front Immunol 2023; 14:1222267. [PMID: 37675118 PMCID: PMC10478084 DOI: 10.3389/fimmu.2023.1222267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/19/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction Syphilis, a sexually transmitted infection caused by the spirochete Treponema pallidum (Tp), is resurging globally. Tp's repertoire of outer membrane proteins (OMPs) includes BamA (β-barrel assembly machinery subunit A/TP0326), a bipartite protein consisting of a 16-stranded β-barrel with nine extracellular loops (ECLs) and five periplasmic POTRA (polypeptide transport-associated) domains. BamA ECL4 antisera promotes internalization of Tp by rabbit peritoneal macrophages. Methods Three overlapping BamA ECL4 peptides and a two-stage, phage display strategy, termed "Epivolve" (for epitope evolution) were employed to generate single-chain variable fragments (scFvs). Additionally, antisera generated by immunizing mice and rabbits with BamA ECL4 displayed by a Pyrococcus furiosus thioredoxin scaffold (PfTrxBamA/ECL4). MAbs and antisera reactivities were evaluated by immunoblotting and ELISA. A comparison of murine and rabbit opsonophagocytosis assays was conducted to evaluate the functional ability of the Abs (e.g., opsonization) and validate the mouse assay. Sera from Tp-infected mice (MSS) and rabbits (IRS) were evaluated for ECL4-specific Abs using PfTrxBamA/ECL4 and overlapping ECL4 peptides in immunoblotting and ELISA assays. Results Each of the five mAbs demonstrated reactivity by immunoblotting and ELISA to nanogram amounts of PfTrxBamA/ECL4. One mAb, containing a unique amino acid sequence in both the light and heavy chains, showed activity in the murine opsonophagocytosis assay. Mice and rabbits hyperimmunized with PfTrxBamA/ECL4 produced opsonic antisera that strongly recognized the ECL presented in a heterologous scaffold and overlapping ECL4 peptides, including S2. In contrast, Abs generated during Tp infection of mice and rabbits poorly recognized the peptides, indicating that S2 contains a subdominant epitope. Discussion Epivolve produced mAbs target subdominant opsonic epitopes in BamA ECL4, a top syphilis vaccine candidate. The murine opsonophagocytosis assay can serve as an alternative model to investigate the opsonic potential of vaccinogens. Detailed characterization of BamA ECL4-specific Abs provided a means to dissect Ab responses elicited by Tp infection.
Collapse
Affiliation(s)
- Mary R. Ferguson
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | | | | | - Isabel C. Orbe
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Carson J. La Vake
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Melissa J. Caimano
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States
| | - Qiana Mendez
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | - Trevor F. Moraes
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Anthony B. Schryvers
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - M. Anthony Moody
- Duke Human Vaccine Institute, Durham, NC, United States
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, United States
| | - Justin D. Radolf
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States
- Department of Immunology, UConn Health, Farmington, CT, United States
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - Michael P. Weiner
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | - Kelly L. Hawley
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Immunology, UConn Health, Farmington, CT, United States
- Division of Infectious Diseases and Immunology, Connecticut Children’s, Hartford, CT, United States
| |
Collapse
|
40
|
Scholte FEM, Karaaslan E, O’Neal TJ, Sorvillo TE, Genzer SC, Welch SR, Coleman-McCray JD, Spengler JR, Kainulainen MH, Montgomery JM, Pegan SD, Bergeron E, Spiropoulou CF. Vaccination with the Crimean-Congo hemorrhagic fever virus viral replicon vaccine induces NP-based T-cell activation and antibodies possessing Fc-mediated effector functions. Front Cell Infect Microbiol 2023; 13:1233148. [PMID: 37671145 PMCID: PMC10475602 DOI: 10.3389/fcimb.2023.1233148] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/10/2023] [Indexed: 09/07/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV; family Nairoviridae) is a tick-borne pathogen that frequently causes lethal disease in humans. CCHFV has a wide geographic distribution, and cases have been reported in Africa, Asia, the Middle East, and Europe. Availability of a safe and efficacious vaccine is critical for restricting outbreaks and preventing disease in endemic countries. We previously developed a virus-like replicon particle (VRP) vaccine that provides complete protection against homologous and heterologous lethal CCHFV challenge in mice after a single dose. However, the immune responses induced by this vaccine are not well characterized, and correlates of protection remain unknown. Here we comprehensively characterized the kinetics of cell-mediated and humoral immune responses in VRP-vaccinated mice, and demonstrate that they predominantly target the nucleoprotein (NP). NP antibodies are not associated with protection through neutralizing activity, but VRP vaccination results in NP antibodies possessing Fc-mediated antibody effector functions, such as complement activation (ADCD) and antibody-mediated cellular phagocytosis (ADCP). This suggests that Fc-mediated effector functions may contribute to this vaccine's efficacy.
Collapse
Affiliation(s)
- F. E. M. Scholte
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - E. Karaaslan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - T. J. O’Neal
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - T. E. Sorvillo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - S. C. Genzer
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - S. R. Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - J. D. Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - J. R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - M. H. Kainulainen
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - J. M. Montgomery
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - S. D. Pegan
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - E. Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - C. F. Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| |
Collapse
|
41
|
Emami S, Rojas Converso T, Persson JJ, Johansson-Lindbom B. Insertion of an immunodominant T helper cell epitope within the Group A Streptococcus M protein promotes an IFN-γ-dependent shift from a non-protective to a protective immune response. Front Immunol 2023; 14:1241485. [PMID: 37654501 PMCID: PMC10465795 DOI: 10.3389/fimmu.2023.1241485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/28/2023] [Indexed: 09/02/2023] Open
Abstract
The common pathogen Group A Streptococcus (GAS, Streptococcus pyogenes) is an extracellular bacterium that is associated with a multitude of infectious syndromes spanning a wide range of severity. The surface-exposed M protein is a major GAS virulence factor that is also target for protective antibody responses. In this study, we use a murine immunization model to investigate aspects of the cellular and molecular foundation for protective adaptive immune responses generated against GAS. We show that a wild type M1 GAS strain induces a non-protective antibody response, while an isogenic strain carrying the immunodominant 2W T helper cell epitope within the M protein elicits an immune response that is protective against the parental non-recombinant M1 GAS strain. Although the two strains induce total anti-GAS IgG levels of similar magnitude, only the 2W-carrying strain promotes elevated titers of the complement-fixing IgG2c subclass. Protection is dependent on IFN-γ, and IFN-γ-deficient mice show a specific reduction in IgG2c levels. Our findings suggest that inclusion of the 2W T cell epitope in the M protein confers essential qualitative alterations in the adaptive immune response against GAS, and that sparsity in IFN-γ-promoting Th cell epitopes in the M protein may constitute an immune evasion mechanism, evolved to allow the pathogen to avoid attack by complement-fixing antibodies.
Collapse
|
42
|
Grund M, Choi SJ, Powell L, Lukomski S. Intranasal immunization with a Bucl8-based vaccine ameliorates bacterial burden and pathological inflammation, and promotes an IgG2a/b dominant response in an outbred mouse model of Burkholderia infection. Front Immunol 2023; 14:1177650. [PMID: 37545515 PMCID: PMC10399622 DOI: 10.3389/fimmu.2023.1177650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Burkholderia pseudomallei is a gram-negative bacterium that is the etiological agent of the tropical disease melioidosis. Currently, there is no licensed vaccine for melioidosis, but numerous candidates are being tested for protective efficacy and characterization of the elicited immune response. Our lab has previously reported the immunogenicity of a Bucl8-protein-based peptide antigen, designated L1-CRM197 (Cross-reacting material 197). When given subcutaneously, this vaccine formulation promoted a strong Th2 (IgG1) antibody response, however immunization did not protect from death. In this study, we hypothesized that an intranasally administered L1-CRM197 vaccine would induce protective mucosal immunity. To evaluate vaccine efficacy, we developed a surrogate Burkholderia infection model that employs outbred CD-1 mice which imitates the immunogenetic diversity of humans. Mice were immunized with either L1-CRM197 adjuvanted with fluorinated cyclic diguanosine monophosphate (FCDG) or with FCDG-only control. These mice were then challenged intranasally with an infectious dose of a luminescent strain of B. thailandensis E264 two weeks post-immunization, and correlates of protection were assessed in euthanized mice on days 1, 2, 3, and 7 post-infection. Overall, intranasal vaccination, compared to subcutaneous administration, induced a stronger Th1 (IgG2a/2b) to Th2 (IgG1) antibody response and promoted anti-L1 nasal, pulmonary, and systemic IgA. Additionally, sera IgG from L1-CRM197-vaccinated mice recognized whole-cell B. thailandensis and B. pseudomallei, a select agent exempt strain Bp82. Vaccination ameliorated disease indicators, including luminescent signal and bacterial cell counts, weight and temperature loss, and organ weight, which negatively correlated with IgG2a antibody levels and mucosa-stimulating cytokines IL-13 and IL-9. L1-CRM197-vaccinated mice also had earlier resolution of inflammatory and tissue-damaging cytokines compared to the FCDG-only controls. These results suggest a balanced humoral and cell-mediated response, along with mucosa-based immunity are beneficial for protection. Future efforts should further assess mucosal cellular and humoral mechanisms of protection and test such protection, using aerosolized B. pseudomallei select agent strain(s).
Collapse
Affiliation(s)
| | | | | | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
43
|
Gronke K, Nguyen M, Santamaria N, Schumacher J, Yang Y, Sonnert N, Leopold S, Martin AL, Hallet R, Richter K, Schubert DA, Daniel GM, Dylus D, Forkel M, Vieira SM, Schwinge D, Schramm C, Lassen KG, Piali L, Palm NW, Bieniossek C, Kriegel MA. Human Th17- and IgG3-associated autoimmunity induced by a translocating gut pathobiont. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.546430. [PMID: 37425769 PMCID: PMC10327010 DOI: 10.1101/2023.06.29.546430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Extraintestinal autoimmune diseases are multifactorial with translocating gut pathobionts implicated as instigators and perpetuators in mice. However, the microbial contributions to autoimmunity in humans remain largely unclear, including whether specific pathological human adaptive immune responses are triggered by such pathobionts. We show here that the translocating pathobiont Enterococcus gallinarum induces human IFNγ + Th17 differentiation and IgG3 subclass switch of anti- E. gallinarum RNA and correlating anti-human RNA autoantibody responses in patients with systemic lupus erythematosus and autoimmune hepatitis. Human Th17 induction by E. gallinarum is cell-contact dependent and involves TLR8-mediated human monocyte activation. In murine gnotobiotic lupus models, E. gallinarum translocation triggers IgG3 anti-RNA autoantibody titers that correlate with renal autoimmune pathophysiology and with disease activity in patients. Overall, we define cellular mechanisms of how a translocating pathobiont induces human T- and B-cell-dependent autoimmune responses, providing a framework for developing host- and microbiota-derived biomarkers and targeted therapies in extraintestinal autoimmune diseases. One Sentence Summary Translocating pathobiont Enterococcus gallinarum promotes human Th17 and IgG3 autoantibody responses linked to disease activity in autoimmune patients.
Collapse
|
44
|
McNitt DH, Joosse BA, Thomas JW, Bonami RH. Productive Germinal Center Responses Depend on the Nature of Stimuli Received by Anti-Insulin B Cells in Type 1 Diabetes-Prone Mice. Immunohorizons 2023; 7:384-397. [PMID: 37261716 PMCID: PMC10448785 DOI: 10.4049/immunohorizons.2300036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Islet autoantibodies, including those directed at insulin, predict type 1 diabetes (T1D) in mice and humans and signal immune tolerance breach by B lymphocytes. High-affinity insulin autoantibodies and T follicular helper cell involvement implicate germinal centers (GCs) in T1D. The VH125SD BCR transgenic model, in which 1-2% of peripheral B lymphocytes recognize insulin, enables direct study of insulin-binding B cells. Our prior studies showed that anti-insulin B cell receptor transgene site-directed to H chain locus mice fail to generate insulin Ab following T-dependent immunization, but it was unclear whether anti-insulin B cells were blocked for GC initiation, survival, or differentiation into Ab-secreting cells. Here, we show that insulin-binding B cells in T1D-prone anti-insulin B cell receptor transgene site-directed to H chain locus mice can spontaneously adopt a GC phenotype and undergo class switching to the IgG1 isotype, with little if any switching to IgG2b. T-dependent immunizations with insulin SRBC or insulin CFA drove anti-insulin B lymphocytes to adopt a GC phenotype, despite blunted insulin Ab production. Dual immunization against self (insulin) and foreign (4-hydroxy-3-nitrophenylacetyl hapten conjugated to keyhole limpet hemocyanin) Ags showed an anti-insulin (but not anti-4-hydroxy-3-nitrophenylacetyl) Ab block that tracked with increased expression of the apoptosis marker, activated caspase 3, in self-reactive GC B cells. Finally, T-independent immunization with insulin conjugated to Brucella abortus ring test Ag released immune tolerance to allow robust expansion of anti-insulin GC B cells and IgG-switched insulin Ab production. Overall, these data pinpoint GC survival and Ab-secreting cell differentiation as immune tolerance blocks that limit T-dependent, but not T-independent, stimulation of anti-insulin B cell responses.
Collapse
Affiliation(s)
- Dudley H. McNitt
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Bryan A. Joosse
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - James W. Thomas
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
45
|
Gupta P, Hu Z, Kopparapu PK, Deshmukh M, Sághy T, Mohammad M, Jin T, Engdahl C. The impact of TLR2 and aging on the humoral immune response to Staphylococcus aureus bacteremia in mice. Sci Rep 2023; 13:8850. [PMID: 37258615 DOI: 10.1038/s41598-023-35970-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/26/2023] [Indexed: 06/02/2023] Open
Abstract
Aging alters immunoglobulin production, affecting the humoral immune response. Toll-like receptor 2 (TLR2) recognizes Staphylococcus aureus (S. aureus) which causes bacteremia with high mortality in the elderly. To understand how TLR2 and aging affect the humoral immune response in bacteremia, four groups of mice (wild type-young, wild type-old, TLR2-/--young, and TLR2-/--old) were used to analyze immunoglobulin levels in healthy conditions as well as 10 days after intravenous injection with S. aureus. We found that aging increased the levels of both IgM and IgG. Increased IgG in aged mice was controlled by TLR2. In bacteremia infection, aged mice failed to mount proper IgM response in both wild-type (WT) and TLR2-/- mice, whereas IgG response was impaired in both aged and TLR2-/- mice. Aged mice displayed reduced IgG1 and IgG2a response irrespective of TLR2 expression. However, impaired IgG2b response was only found in aged WT mice and not in TLR2-/- mice. Both aging and TLR2-/- increased the levels of anti-staphylococcal IgM in bacteremia. Aging increased sialylated IgG in WT mice but not in TLR2-/- mice. IgG sialylation was not affected by the infection in neither of the mice. In summary, aging increases all immunoglobulins except IgG1. However, aged mice fail to mount a proper antibody response to S. aureus bacteremia. TLR2 plays the regulatory role in IgG but not IgM response to infection.
Collapse
Affiliation(s)
- Priti Gupta
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden.
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Center, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- SciLifeLab, University of Gothenburg, Box 413, 405 30, Gothenburg, Sweden.
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden
- Centre for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden
| | - Meghshree Deshmukh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden
| | - Tibor Sághy
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Center, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Box 413, 405 30, Gothenburg, Sweden
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Cecilia Engdahl
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box- 480, 413 45, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Center, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Box 413, 405 30, Gothenburg, Sweden
| |
Collapse
|
46
|
Tarrés-Freixas F, Aguilar-Gurrieri C, Rodríguez de la Concepción ML, Urrea V, Trinité B, Ortiz R, Pradenas E, Blanco P, Marfil S, Molinos-Albert LM, Barajas A, Pons-Grífols A, Ávila-Nieto C, Varela I, Cervera L, Gutiérrez-Granados S, Segura MM, Gòdia F, Clotet B, Carrillo J, Blanco J. An engineered HIV-1 Gag-based VLP displaying high antigen density induces strong antibody-dependent functional immune responses. NPJ Vaccines 2023; 8:51. [PMID: 37024469 PMCID: PMC10077320 DOI: 10.1038/s41541-023-00648-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 03/17/2023] [Indexed: 04/08/2023] Open
Abstract
Antigen display on the surface of Virus-Like Particles (VLPs) improves immunogenicity compared to soluble proteins. We hypothesised that immune responses can be further improved by increasing the antigen density on the surface of VLPs. In this work, we report an HIV-1 Gag-based VLP platform engineered to maximise the presence of antigen on the VLP surface. An HIV-1 gp41-derived protein (Min), including the C-terminal part of gp41 and the transmembrane domain, was fused to HIV-1 Gag. This resulted in high-density MinGag-VLPs. These VLPs demonstrated to be highly immunogenic in animal models using either a homologous (VLP) or heterologous (DNA/VLP) vaccination regimen, with the latter yielding 10-fold higher anti-Gag and anti-Min antibody titres. Despite these strong humoral responses, immunisation with MinGag-VLPs did not induce neutralising antibodies. Nevertheless, antibodies were predominantly of an IgG2b/IgG2c profile and could efficiently bind CD16-2. Furthermore, we demonstrated that MinGag-VLP vaccination could mediate a functional effect and halt the progression of a Min-expressing tumour cell line in an in vivo mouse model.
Collapse
Affiliation(s)
- Ferran Tarrés-Freixas
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | | | | | - Victor Urrea
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Benjamin Trinité
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Raquel Ortiz
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Edwards Pradenas
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Pau Blanco
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Sílvia Marfil
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Luis Manuel Molinos-Albert
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Ana Barajas
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Anna Pons-Grífols
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Carlos Ávila-Nieto
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Ismael Varela
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Laura Cervera
- Grup d'Enginyeria Cel•lular i Bioprocessos, Department of Chemical, Biological and Environmental Engineering, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, 08913, Cerdanyola del Vallès, Catalonia, Spain
| | - Sònia Gutiérrez-Granados
- Grup d'Enginyeria Cel•lular i Bioprocessos, Department of Chemical, Biological and Environmental Engineering, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, 08913, Cerdanyola del Vallès, Catalonia, Spain
| | - María Mercedes Segura
- Grup d'Enginyeria Cel•lular i Bioprocessos, Department of Chemical, Biological and Environmental Engineering, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, 08913, Cerdanyola del Vallès, Catalonia, Spain
| | - Francesc Gòdia
- Grup d'Enginyeria Cel•lular i Bioprocessos, Department of Chemical, Biological and Environmental Engineering, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, 08913, Cerdanyola del Vallès, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Barcelona, Spain
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain.
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, 08916, Barcelona, Spain.
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916, Badalona, Catalonia, Spain.
- University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Barcelona, Spain.
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, 08916, Barcelona, Spain.
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
47
|
Prakash A, Medved J, Arneja A, Niebuhr C, Li AN, Tarrah S, Boscia AR, Burnett ED, Singh A, Salazar JE, Xu W, Santhanakrishnan M, Hendrickson JE, Luckey CJ. Class switching is differentially regulated in RBC alloimmunization and vaccination. Transfusion 2023; 63:826-838. [PMID: 36907655 PMCID: PMC10851675 DOI: 10.1111/trf.17301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 03/13/2023]
Abstract
BACKGROUND Studies of human patients have shown that most anti-RBC alloantibodies are IgG1 or IgG3 subclasses, although it is unclear why transfused RBCs preferentially drive these subclasses over others. Though mouse models allow for the mechanistic exploration of class-switching, previous studies of RBC alloimmunization in mice have focused more on the total IgG response than the relative distribution, abundance, or mechanism of IgG subclass generation. Given this major gap, we compared the IgG subclass distribution generated in response to transfused RBCs relative to protein in alum vaccination, and determined the role of STAT6 in their generation. STUDY DESIGN AND METHODS WT mice were either immunized with Alum/HEL-OVA or transfused with HOD RBCs and levels of anti-HEL IgG subtypes were measured using end-point dilution ELISAs. To study the role of STAT6 in IgG class-switching, we first generated and validated novel STAT6 KO mice using CRISPR/cas9 gene editing. STAT6 KO mice were then transfused with HOD RBCs or immunized with Alum/HEL-OVA, and IgG subclasses were quantified by ELISA. RESULTS When compared with antibody responses to Alum/HEL-OVA, transfusion of HOD RBCs induced lower levels of IgG1, IgG2b, and IgG2c but similar levels of IgG3. Class switching to most IgG subtypes remained largely unaffected in STAT6 deficient mice in response to HOD RBC transfusion, with the one exception being IgG2b. In contrast, STAT6 deficient mice showed altered levels of all IgG subtypes following Alum vaccination. DISCUSSION Our results show that anti-RBC class-switching occurs via alternate mechanisms when compared with the well-studied immunogen alum vaccination.
Collapse
Affiliation(s)
- Anupam Prakash
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Jelena Medved
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Abhinav Arneja
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Conrad Niebuhr
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Andria N. Li
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Soraya Tarrah
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Alexis R. Boscia
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Emily D. Burnett
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Aanika Singh
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Juan E. Salazar
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Wenhao Xu
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Manjula Santhanakrishnan
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jeanne E. Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
48
|
Rojas-Ortega DA, Rojas-Hernández S, Sánchez-Mendoza ME, Gómez-López M, Sánchez-Camacho JV, Rosales-Cruz E, Yépez MMC. Role of FcγRIII in the nasal cavity of BALB/c mice in the primary amebic meningoencephalitis protection model. Parasitol Res 2023; 122:1087-1105. [PMID: 36913025 PMCID: PMC10009362 DOI: 10.1007/s00436-023-07810-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/01/2023] [Indexed: 03/14/2023]
Abstract
Different mechanisms of the host immune response against the primary amebic meningoencephalitis (PAM) in the mouse protection model have been described. It has been proposed that antibodies opsonize Naegleria fowleri trophozoites; subsequently, the polymorphonuclear cells (PMNs) surround the trophozoites to avoid the infection. FcγRs activate signaling pathways of adapter proteins such as Syk and Hck on PMNs to promote different effector cell functions which are induced by the Fc portion of the antibody-antigen complexes. In this work, we analyzed the activation of PMNs, epithelial cells, and nasal passage cells via the expression of Syk and Hck genes. Our results showed an increment of the FcγRIII and IgG subclasses in the nasal cavity from immunized mice as well as Syk and Hck expression was increased, whereas in the in vitro assay, we observed that when the trophozoites of N. fowleri were opsonized with IgG anti-N. fowleri and interacted with PMN, the expression of Syk and Hck was also increased. We suggest that PMNs are activated via their FcγRIII, which leads to the elimination of the trophozoites in vitro, while in the nasal cavity, the adhesion and consequently infection are avoided.
Collapse
Affiliation(s)
- Diego Alexander Rojas-Ortega
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Saúl Rojas-Hernández
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - María Elena Sánchez-Mendoza
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Modesto Gómez-López
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Jennifer Viridiana Sánchez-Camacho
- Laboratorio de Inmunología Molecular, Instituto Politécnico Nacional, Escuela Superior de Medicina, Salvador Diaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de Mexico, México
| | - Erika Rosales-Cruz
- Laboratorio de Investigación en Hematopatología, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Ciudad de Mexico, México
| | | |
Collapse
|
49
|
Hendy DA, Johnson-Weaver BT, Batty CJ, Bachelder EM, Abraham SN, Staats HF, Ainslie KM. Delivery of small molecule mast cell activators for West Nile Virus vaccination using acetalated dextran microparticles. Int J Pharm 2023; 634:122658. [PMID: 36731641 PMCID: PMC9975031 DOI: 10.1016/j.ijpharm.2023.122658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
Recently, there has been increasing interest in the activation of mast cells to promote vaccine efficacy. Several mast cell activating (MCA) compounds have been reported such as M7 and Compound 48/80 (C48/80). While these MCAs have been proven to be efficacious vaccine adjuvants, their translatability is limited by batch-to-batch variability, challenging large-scale manufacturing, and poor in vivo stability for the M7 peptide. Due to this, high throughput screening was performed to identify small molecule MCAs. Several potent MCAs were identified via this screening, but the in vivo translatability of the compounds was limited due to their poor aqueous solubility. To enhance the delivery of these MCAs we encapsulated them in acetalated dextran (Ace-DEX) microparticles (MPs). We have previously utilized Ace-DEX MPs for vaccine delivery due to their passive targeting to phagocytic cells, acid sensitivity, and tunable degradation. Four different MCA loaded MPs were combined with West Nile Virus Envelope III protein (EDIII) and their vaccine adjuvant activities were compared in vivo. MPs containing the small molecule MCA ST101036 produced the highest anti-EDIII IgG titers of all the MCAs tested. Further, ST101036 MPs produced higher titers than ST101036 formulated with PEG as a cosolvent which highlights the benefit of Ace-DEX MPs over a conventional formulation technique. Finally, in a mouse model of West Nile Virus infection ST101036 MPs produced similar survival to soluble M7 (80-90%). Overall, these data show that ST101036 MPs produce a robust antibody response against EDIII and survival emphasizing the benefits of using Ace-DEX as a delivery platform for the poorly soluble ST101036.
Collapse
Affiliation(s)
- Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | | | - Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | | | - Herman F Staats
- Department of Pathology, Duke University, USA; Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
50
|
Wabnitz H, Cruz-Leal Y, Lazarus AH. Antigen-specific IgG subclass composition in recipient mice can indicate the degree of red blood cell alloimmunization as well as discern between primary and secondary immunization. Transfusion 2023; 63:619-628. [PMID: 36591986 DOI: 10.1111/trf.17232] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Despite the vast antigen disparity between donor and recipient red blood cells (RBCs), only 2%-6% of transfusion patients mount an alloantibody response. Recently, RBC antigen density has been proposed as one of the factors that can influence alloimmunization, however, there has been no characterization of the role of antigen density along with RBC dose in primary and secondary immunization. STUDY DESIGN AND METHODS To generate RBCs that express distinct antigen copy numbers, different quantities of hen egg lysozyme (HEL) were coupled to murine RBCs. The HEL-RBCs were subsequently transfused into recipient mice at different RBC doses and their HEL-specific IgM, IgG, and IgG subclass response was evaluated. RESULTS Productive immune responses could be generated through a high copy number antigen transfused at low RBC doses or a low copy number transfused at high RBC doses. Further, primary but submaximal humoral immunization predominantly induced the IgG2b and IgG3 subclasses. In contrast, a maximal primary immunization or a secondary immunization induced all four IgG subclasses. DISCUSSION Our results confirm the existence of an antigen threshold for productive immune responses but indicate that a high antigen copy number alone might not be enough to induce a response, but rather a combination of both antigen copy number and cell dosage may determine the outcome of immunization. Further, this study provides a proof of concept that the IgG subclass composition can be an indicator of the level of RBC alloimmunization as well as discern between primary and secondary immunization at least in this murine model.
Collapse
Affiliation(s)
- Hanna Wabnitz
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Yoelys Cruz-Leal
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Alan H Lazarus
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|