1
|
Murphy CT, Bachelder EM, Ainslie KM. Mast cell activators as adjuvants for intranasal mucosal vaccines. Int J Pharm 2025; 672:125300. [PMID: 39914508 DOI: 10.1016/j.ijpharm.2025.125300] [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: 12/18/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
Mast cells have roles in immune regulation, allergy, and host response to pathogens. Compounds that activate mast cells (MCAs) can serve as vaccine adjuvants, potentially outperforming current FDA-approved options, especially for mucosal vaccines. While most vaccines are administered intramuscularly, intranasal and needle-free formulations offer benefits like improved compliance and accessibility. However, the lack of effective adjuvants limits mucosal vaccine development. This review explores MCAs as promising alternatives to traditional adjuvants, aiming to enhance mucosal vaccine efficacy. We summarize the nascent work of formulating MCAs like compound 48/80 into nanoparticles, with excipients such as chitosan and chitosan/alginate. Other MCAs like the peptide mastoparan 7 complexed with CpG have formed nanoparticle complexes that illustrate protective mucosal immunity in a model of influenza. The small molecule MCA ST101036, when encapsulated in acetalated dextran particles, has demonstrated enhanced immune responses and protection in a West Nile Virus model of infection. This review highlights the potential of MCAs as potent vaccine adjuvants, particularly for mucosal vaccines, and summarizes, recent advancements in formulating these activators into nanoparticles to enhance immune responses and protection.
Collapse
Affiliation(s)
- Connor T Murphy
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC Chapel Hill NC USA
| | - Eric M Bachelder
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC Chapel Hill NC USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC Chapel Hill NC USA; Department of Biomedical Engineering NC State/UNC Chapel Hill NC USA; Department of Microbiology and Immunology, School of Medicine, UNC Chapel Hill NC USA.
| |
Collapse
|
2
|
Bawazir M, Roy S, Ali H. The development of murine bone marrow-derived mast cells expressing functional human MRGPRX2 for ex vivo and in vivo studies. Front Immunol 2024; 15:1523393. [PMID: 39749337 PMCID: PMC11693745 DOI: 10.3389/fimmu.2024.1523393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction A subtype of human mast cells (MCs) found in the skin and to a lesser extent in the lung and gut express a novel G protein-coupled receptor (GPCR) known as Mas-related GPCR-X2 (MRGPRX2, mouse counterpart MrgprB2). In addition to drug-induced pseudoallergy and cutaneous disorders, MrgprB2 contributes to ulcerative colitis, IgE-mediated lung inflammation and systemic anaphylaxis. Interestingly, most agonists activate MRGPRX2 with higher potency than MrgprB2. In this study, we sought to replace mouse MrgprB2 with human MRGPRX2 and to study receptor function ex vivo and in vivo. Methods MrgprB2-/- bone marrow (BM) cells were transduced with retrovirus encoding MRGPRX2 and differentiated into BMMCs (MRGRPX2-BMMCs) ex vivo. Cell surface MRGPRX2 expression was determined by flow cytometry. Effects of substance P (SP) and LL-37 on Ca2+ mobilization, degranulation and TNF-α generation were determined. MRGPRX2-BMMCs were engrafted intraperitoneally into MC-deficient Wsh/Wsh mice. After 6-8 weeks, immunofluorescence staining was performed on peritoneal lavage cells (PLCs), and sections of small intestine and colon with anti c-Kit and anti-MRGPRX2 antibodies. SP-induced degranulation in PLCs obtained from engrafted mice was determined. Results MRGPRX2-BMMCs expressed cell surface MRGPRX2 and responded to both SP and LL-37 for Ca2+ mobilization, degranulation and TNF-α generation. Furthermore, Wsh/Wsh mice engrafted with MRGPRX2-BMMCs expressed the receptor in peritoneal, intestinal and colonic MCs. In addition, PLCs from engrafted mice responded to SP for degranulation. Conclusion Replacing mouse MrgprB2 with functional human MRGPRX2 in primary BMMCs and their engraftment in MC-deficient mice demonstrated the expression of this receptor in different tissues, which provides unique opportunities to study receptor signaling ex vivo and disease phenotype in vivo.
Collapse
Affiliation(s)
- Maram Bawazir
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral Diagnostic Sciences, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saptarshi Roy
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hydar Ali
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
3
|
Plum T, Feyerabend TB, Rodewald HR. Beyond classical immunity: Mast cells as signal converters between tissues and neurons. Immunity 2024; 57:2723-2736. [PMID: 39662090 DOI: 10.1016/j.immuni.2024.11.016] [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: 07/08/2024] [Revised: 10/15/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
Mast cells are regarded as effectors in immune defense against parasites and venoms and play an essential role in the pathology of allergic diseases. More recently, mast cells have been shown to receive stimuli derived from type 2 immunity, tissue damage, stress, and inflammation. Mast cells then rapidly convert these diverse signals into appropriate, organ-specific protective reflexes that can limit inflammation or reduce tissue damage. In this review, we consider functions of mast cells in sensations-such as pain, itch, and nausea-arising from tissue insults and inflammation and the ensuing protective responses. In light of emerging data highlighting the involvement of mast cells in neuroimmune communication, we also propose that mast cells are "signal converters" linking immunological and tissue states with nervous system responses.
Collapse
Affiliation(s)
- Thomas Plum
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| | - Thorsten B Feyerabend
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
4
|
Zelante T, Paolicelli G, Fallarino F, Gargaro M, Vascelli G, De Zuani M, Fric J, Laznickova P, Kohoutkova MH, Macchiarulo A, Dolciami D, Pieraccini G, Gaetani L, Scalisi G, Trevisan C, Frossi B, Pucillo C, De Luca A, Nunzi E, Spaccapelo R, Pariano M, Borghi M, Boscaro F, Romoli R, Mancini A, Gentili L, Renga G, Costantini C, Puccetti M, Giovagnoli S, Ricci M, Antonini M, Calabresi P, Puccetti P, Di Filippo M, Romani L. A microbially produced AhR ligand promotes a Tph1-driven tolerogenic program in multiple sclerosis. Sci Rep 2024; 14:6651. [PMID: 38509264 PMCID: PMC10954611 DOI: 10.1038/s41598-024-57400-8] [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: 11/15/2023] [Accepted: 03/18/2024] [Indexed: 03/22/2024] Open
Abstract
Multiple sclerosis is a debilitating autoimmune disease, characterized by chronic inflammation of the central nervous system. While the significance of the gut microbiome on multiple sclerosis pathogenesis is established, the underlining mechanisms are unknown. We found that serum levels of the microbial postbiotic tryptophan metabolite indole-3-carboxaldehyde (3-IAld) inversely correlated with disease duration in multiple sclerosis patients. Much like the host-derived tryptophan derivative L-Kynurenine, 3-IAld would bind and activate the Aryl hydrocarbon Receptor (AhR), which, in turn, controls endogenous tryptophan catabolic pathways. As a result, in peripheral lymph nodes, microbial 3-IAld, affected mast-cell tryptophan metabolism, forcing mast cells to produce serotonin via Tph1. We thus propose a protective role for AhR-mast-cell activation driven by the microbiome, whereby natural metabolites or postbiotics will have a physiological role in immune homeostasis and may act as therapeutic targets in autoimmune diseases.
Collapse
Affiliation(s)
- Teresa Zelante
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy.
- Interuniversity Consortium for Biotechnology, (CIB), 34149, Trieste, Italy.
| | - Giuseppe Paolicelli
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Francesca Fallarino
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Gianluca Vascelli
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Marco De Zuani
- International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jan Fric
- International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czech Republic
- Institute of Hematology and Blood Transfusion, U Nemocnice 2094/1, 128 20, Prague, Czech Republic
- International Clinical Research Centre, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petra Laznickova
- International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czech Republic
- International Clinical Research Centre, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Marcela Hortova Kohoutkova
- International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czech Republic
- International Clinical Research Centre, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Antonio Macchiarulo
- Department of Pharmaceutical Science, University of Perugia, 06132, Perugia, Italy
| | - Daniela Dolciami
- Department of Pharmaceutical Science, University of Perugia, 06132, Perugia, Italy
| | - Giuseppe Pieraccini
- Mass Spectrometry Center (CISM), University of Florence, 50139, Florence, Italy
| | - Lorenzo Gaetani
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Giulia Scalisi
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Caterina Trevisan
- Department of Medical and Biological Science, University of Udine, 33100, Udine, Italy
| | - Barbara Frossi
- Department of Medical and Biological Science, University of Udine, 33100, Udine, Italy
| | - Carlo Pucillo
- Department of Medical and Biological Science, University of Udine, 33100, Udine, Italy
| | - Antonella De Luca
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
- Center of Functional Genomics, C.U.R.Ge.F, University of Perugia, 06132, Perugia, Italy
| | - Roberta Spaccapelo
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
- Center of Functional Genomics, C.U.R.Ge.F, University of Perugia, 06132, Perugia, Italy
- Interuniversity Consortium for Biotechnology, (CIB), 34149, Trieste, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Monica Borghi
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Francesca Boscaro
- Mass Spectrometry Center (CISM), University of Florence, 50139, Florence, Italy
| | - Riccardo Romoli
- Mass Spectrometry Center (CISM), University of Florence, 50139, Florence, Italy
| | - Andrea Mancini
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Lucia Gentili
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, 06132, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, 06132, Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Science, University of Perugia, 06132, Perugia, Italy
| | - Martina Antonini
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Paolo Calabresi
- Unità di Neurologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
- Center of Functional Genomics, C.U.R.Ge.F, University of Perugia, 06132, Perugia, Italy
| | - Massimiliano Di Filippo
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi, 1, 06132, Perugia, Italy
- Center of Functional Genomics, C.U.R.Ge.F, University of Perugia, 06132, Perugia, Italy
| |
Collapse
|
5
|
Jiang Y, Lu L. New insight into the agonism of protease-activated receptors as an immunotherapeutic strategy. J Biol Chem 2024; 300:105614. [PMID: 38159863 PMCID: PMC10810747 DOI: 10.1016/j.jbc.2023.105614] [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: 05/26/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
The activation and mobilization of immune cells play a crucial role in immunotherapy. Existing therapeutic interventions, such as cytokines administration, aim to enhance immune cell activity. However, these approaches usually result in modest effectiveness and toxic side effects, thereby restricting their clinical application. Protease-activated receptors (PARs), a subfamily of G protein-coupled receptors, actively participate in the immune system by directly activating immune cells. The activation of PARs by proteases or synthetic ligands can modulate immune cell behavior, signaling, and responses to treat immune-related diseases, suggesting the significance of PARs agonism in immunotherapy. However, the agonism of PARs in therapeutical applications remains rarely discussed, since it has been traditionally considered that PARs activation facilitates disease progressions. This review aims to comprehensively summarize the activation, rather than inhibition, of PARs in immune-related physiological responses and diseases. Additionally, we will discuss the emerging immunotherapeutic potential of PARs agonism, providing a new strategic direction for PARs-mediated immunotherapy.
Collapse
Affiliation(s)
- Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China.
| | - Lei Lu
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
6
|
O'Neill A, Mantri CK, Tan CW, Saron WAA, Nagaraj SK, Kala MP, Joy CM, Rathore APS, Tripathi S, Wang LF, St John AL. Mucosal SARS-CoV-2 vaccination of rodents elicits superior systemic T central memory function and cross-neutralising antibodies against variants of concern. EBioMedicine 2024; 99:104924. [PMID: 38113758 PMCID: PMC10772395 DOI: 10.1016/j.ebiom.2023.104924] [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: 02/08/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND COVID-19 vaccines used in humans are highly effective in limiting disease and death caused by the SARS-CoV-2 virus, yet improved vaccines that provide greater protection at mucosal surfaces, which could reduce break-through infections and subsequent transmission, are still needed. METHODS Here we tested an intranasal (I.N.) vaccination with the receptor binding domain of Spike antigen of SARS-CoV-2 (S-RBD) in combination with the mucosal adjuvant mastoparan-7 compared with the sub-cutaneous (S.C.) route, adjuvanted by either M7 or the gold-standard adjuvant, alum, in mice, for immunological read-outs. The same formulation delivered I.N. or S.C. was tested in hamsters to assess efficacy. FINDINGS I.N. vaccination improved systemic T cell responses compared to an equivalent dose of antigen delivered S.C. and T cell phenotypes induced by I.N. vaccine administration included enhanced polyfunctionality (combined IFN-γ and TNF expression) and greater numbers of T central memory (TCM) cells. These phenotypes were T cell-intrinsic and could be recalled in the lungs and/or brachial LNs upon antigen challenge after adoptive T cell transfer to naïve recipients. Furthermore, mucosal vaccination induced antibody responses that were similarly effective in neutralising the binding of the parental strain of S-RBD to its ACE2 receptor, but showed greater cross-neutralising capacity against multiple variants of concern (VOC), compared to S.C. vaccination. I.N. vaccination provided significant protection from lung pathology compared to unvaccinated animals upon challenge with homologous and heterologous SARS-CoV-2 strains in a hamster model. INTERPRETATION These results highlight the role of nasal vaccine administration in imprinting an immune profile associated with long-term T cell retention and diversified neutralising antibody responses, which could be applied to improve vaccines for COVID-19 and other infectious diseases. FUNDING This study was funded by Duke-NUS Medical School, the Singapore Ministry of Education, the National Medical Research Council of Singapore and a DBT-BIRAC Grant.
Collapse
Affiliation(s)
- Aled O'Neill
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Chinmay Kumar Mantri
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Chee Wah Tan
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| | - Wilfried A A Saron
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Santhosh Kambaiah Nagaraj
- Centre for Infectious Disease Research, Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, 560012, India
| | - Monica Palanichamy Kala
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Christy Margarat Joy
- Centre for Infectious Disease Research, Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, 560012, India
| | - Abhay P S Rathore
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; Department of Pathology, Duke University Medical Centre, Durham, North Carolina, 27705, USA
| | - Shashank Tripathi
- Centre for Infectious Disease Research, Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, 560012, India
| | - Lin-Fa Wang
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; SingHealth Duke-NUS Global Health Institute, Singapore
| | - Ashley L St John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; Department of Pathology, Duke University Medical Centre, Durham, North Carolina, 27705, USA; SingHealth Duke-NUS Global Health Institute, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
7
|
Tung CC, Rathore APS, St. John AL. Conventional and non-conventional antigen presentation by mast cells. DISCOVERY IMMUNOLOGY 2023; 2:kyad016. [PMID: 38567067 PMCID: PMC10917180 DOI: 10.1093/discim/kyad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/23/2023] [Accepted: 09/16/2023] [Indexed: 04/04/2024]
Abstract
Mast cells (MCs) are multifunctional immune cells that express a diverse repertoire of surface receptors and pre-stored bioactive mediators. They are traditionally recognized for their involvement in allergic and inflammatory responses, yet there is a growing body of literature highlighting their contributions to mounting adaptive immune responses. In particular, there is growing evidence that MCs can serve as antigen-presenting cells, owing to their often close proximity to T cells in both lymphoid organs and peripheral tissues. Recent studies have provided compelling support for this concept, by demonstrating the presence of antigen processing and presentation machinery in MCs and their ability to engage in classical and non-classical pathways of antigen presentation. However, there remain discrepancies and unresolved questions regarding the extent of the MC's capabilities with respect to antigen presentation. In this review, we discuss our current understanding of the antigen presentation by MCs and its influence on adaptive immunity.
Collapse
Affiliation(s)
- Chi-Ching Tung
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Ashley L St. John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore, Singapore
| |
Collapse
|
8
|
Raj S, Unsworth LD. Targeting active sites of inflammation using inherent properties of tissue-resident mast cells. Acta Biomater 2023; 159:21-37. [PMID: 36657696 DOI: 10.1016/j.actbio.2023.01.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/12/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
Mast cells play a pivotal role in initiating and directing host's immune response. They reside in tissues that primarily interface with the external environment. Activated mast cells respond to environmental cues throughout acute and chronic inflammation through releasing immune mediators via rapid degranulation, or long-term de novo expression. Mast cell activation results in the rapid release of a variety of unique enzymes and reactive oxygen species. Furthermore, the increased density of mast cell unique receptors like mas related G protein-coupled receptor X2 also characterizes the inflamed tissues. The presence of these molecules (either released mediators or surface receptors) are particular to the sites of active inflammation, and are a result of mast cell activation. Herein, the molecular design principles for capitalizing on these novel mast cell properties is discussed with the goal of manipulating localized inflammation. STATEMENT OF SIGNIFICANCE: Mast cells are immune regulating cells that play a crucial role in both innate and adaptive immune responses. The activation of mast cells causes the release of multiple unique profiles of biomolecules, which are specific to both tissue and disease. These unique characteristics are tightly regulated and afford a localized stimulus for targeting inflammatory diseases. Herein, these important mast cell attributes are discussed in the frame of highlighting strategies for the design of bioresponsive functional materials to target regions of inflammations.
Collapse
Affiliation(s)
- Shammy Raj
- Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering, 9211-116 Street NW, University of Alberta, Edmonton, AB, T6G1H9, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, Donadeo Innovation Centre for Engineering, 9211-116 Street NW, University of Alberta, Edmonton, AB, T6G1H9, Canada.
| |
Collapse
|
9
|
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
|
10
|
Ontiveros-Padilla L, Batty CJ, Hendy DA, Pena ES, Roque JA, Stiepel RT, Carlock MA, Simpson SR, Ross TM, Abraham SN, Staats HF, Bachelder EM, Ainslie KM. Development of a broadly active influenza intranasal vaccine adjuvanted with self-assembled particles composed of mastoparan-7 and CpG. Front Immunol 2023; 14:1103765. [PMID: 37033992 PMCID: PMC10081679 DOI: 10.3389/fimmu.2023.1103765] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Currently licensed vaccine adjuvants offer limited mucosal immunity, which is needed to better combat respiratory infections such as influenza. Mast cells (MCs) are emerging as a target for a new class of mucosal vaccine adjuvants. Here, we developed and characterized a nanoparticulate adjuvant composed of an MC activator [mastoparan-7 (M7)] and a TLR ligand (CpG). This novel nanoparticle (NP) adjuvant was co-formulated with a computationally optimized broadly reactive antigen (COBRA) for hemagglutinin (HA), which is broadly reactive against influenza strains. M7 was combined at different ratios with CpG and tested for in vitro immune responses and cytotoxicity. We observed significantly higher cytokine production in dendritic cells and MCs with the lowest cytotoxicity at a charge-neutralizing ratio of nitrogen/phosphate = 1 for M7 and CpG. This combination formed spherical NPs approximately 200 nm in diameter with self-assembling capacity. Mice were vaccinated intranasally with COBRA HA and M7-CpG NPs in a prime-boost-boost schedule. Vaccinated mice had significantly higher antigen-specific antibody responses (IgG and IgA) in serum and mucosa compared with controls. Splenocytes from vaccinated mice had significantly increased cytokine production upon antigen recall and the presence of central and effector memory T cells in draining lymph nodes. Finally, co-immunization with NPs and COBRA HA induced influenza H3N2-specific HA inhibition antibody titers across multiple strains and partially protected mice from a challenge against an H3N2 virus. These results illustrate that the M7-CpG NP adjuvant combination can induce a protective immune response with a broadly reactive influenza antigen via mucosal vaccination.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Erik S. Pena
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
| | - John A. Roque
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rebeca T. Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael A. Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Sean R. Simpson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ted M. Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Soman N. Abraham
- Departments of Pathology, Molecular Genetics and Microbiology and Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Herman F. Staats
- Department of Pathology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccines Institute, School of Medicine, Duke University, Durham, NC, United States
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Kristy M. Ainslie,
| |
Collapse
|
11
|
Mayavannan A, Shantz E, Haidl ID, Wang J, Marshall JS. Mast cells selectively produce inflammatory mediators and impact the early response to Chlamydia reproductive tract infection. Front Immunol 2023; 14:1166068. [PMID: 37138882 PMCID: PMC10150091 DOI: 10.3389/fimmu.2023.1166068] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Chlamydia trachomatis (C. trachomatis) is a Gram-negative obligate intracellular bacterium that causes reproductive tract complications in women, including ectopic pregnancies and tubal factor infertility. We hypothesized that mast cells, which are common at mucosal barriers, may contribute to responses to Chlamydia infection and aimed to define human mast cell responses to C. trachomatis. Methods Human cord blood-derived mast cells (CBMCs) were exposed to C. trachomatis to assess bacterial uptake, mast cell degranulation, gene expression, and production of inflammatory mediators. The role of formyl peptide receptors and Toll-like receptor 2 (TLR2) were investigated using pharmacological inhibitors and soluble TLR2. Mast cell-deficient mice and littermate controls were used to examine the in vivo role of mast cells in influencing the immune response to Chlamydia infection in the female reproductive tract. Results C. trachomatis bacteria were taken up by human mast cells but did not replicate efficiently inside CBMCs. C. trachomatis-activated mast cells did not degranulate but maintained viability and exhibited cellular activation with homotypic aggregation and upregulation of ICAM-1. However, they significantly enhanced the gene expression of IL1B, CCL3, NFKB1, CXCL8, and IL6. Inflammatory mediators were produced, including TNF, IL-1β, IL-1RA, IL-6, GM-CSF, IL-23, CCL3, CCL5, and CXCL8. Endocytic blockade resulted in reduced gene expression of IL6, IL1B, and CCL3, suggesting C. trachomatis induced mast cell activation in both extracellular and intracellular locations. The IL-6 response to C. trachomatis was reduced when CBMCs were treated with C. trachomatis coated with soluble TLR2. Mast cells derived from TLR2-deficient mice also demonstrated a reduced IL-6 response to C. muridarum. Five days following C. muridarum infection, mast cell-deficient mice showed attenuated CXCL2 production and significantly reduced numbers of neutrophils, eosinophils, and B cells in the reproductive tract when compared with mast cell-containing littermates. Discussion Taken together, these data demonstrate that mast cells are reactive to Chlamydia spp. through multiple mechanisms that include TLR2-dependent pathways. Mast cells also play an important role in shaping in vivo immune responses in Chlamydia reproductive tract infection through both effector cell recruitment and modification of the chemokine microenvironment.
Collapse
Affiliation(s)
- Animamalar Mayavannan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Emily Shantz
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Ian D. Haidl
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jun Wang
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Canadian Center for Vaccinology, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
| | - Jean S. Marshall
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- *Correspondence: Jean S. Marshall,
| |
Collapse
|
12
|
St John AL, Rathore APS, Ginhoux F. New perspectives on the origins and heterogeneity of mast cells. Nat Rev Immunol 2023; 23:55-68. [PMID: 35610312 DOI: 10.1038/s41577-022-00731-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 01/06/2023]
Abstract
Mast cells are immune cells of the haematopoietic lineage that are now thought to have multifaceted functions during homeostasis and in various disease states. Furthermore, while mast cells have been known for a long time to contribute to allergic disease in adults, recent studies, mainly in mice, have highlighted their early origins during fetal development and potential for immune functions, including allergic responses, in early life. Our understanding of the imprinting of mast cells by particular tissues of residence and their potential for regulatory interactions with organ systems such as the peripheral immune, nervous and vascular systems is also rapidly evolving. Here, we discuss the origins of mast cells and their diverse and plastic phenotypes that are influenced by tissue residence. We explore how divergent phenotypes and functions might result from both their hard-wired 'nature' defined by their ontogeny and the 'nurture' they receive within specialized tissue microenvironments.
Collapse
Affiliation(s)
- Ashley L St John
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| | - Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Florent Ginhoux
- Singapore Immunology Network, A*STAR, Singapore, Singapore.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
13
|
Maki Y, Kushibiki T, Sano T, Ogawa T, Komai E, Takahashi S, Kitagami E, Serizawa Y, Nagaoka R, Yokomizo S, Ono T, Ishihara M, Miyahira Y, Kashiwagi S, Kawana A, Kimizuka Y. 1270 nm near-infrared light as a novel vaccine adjuvant acts on mitochondrial photoreception in intradermal vaccines. Front Immunol 2022; 13:1028733. [PMID: 36439134 PMCID: PMC9684730 DOI: 10.3389/fimmu.2022.1028733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/20/2022] [Indexed: 04/13/2024] Open
Abstract
With the development of laser technology in the 1960s, a technique was developed to inject intradermal vaccines immediately after irradiating the skin with laser light to elicit an adjuvant effect, referred to as "laser adjuvant." We have been investigating the mechanism of laser adjuvant in influenza mouse models using noninvasive continuous-wave (CW) near-infrared (NIR) light mainly at a wavelength of 1064 nm, and have shown that the production of reactive-oxygen-species (ROS) in the skin and mast cells in the skin tissue plays an important role in the laser adjuvant effect. The new wavelength of 1270 nm NIR light is characterized by its ability to elicit the same vaccine adjuvant effect as other wavelengths at a lower energy, and may be suitable for clinical applications. In this study, we investigated the physiological activity of CW1270 nm NIR light in mast cells, its biological activity on mouse skin, and the durability of the vaccine adjuvant effect in influenza vaccine mouse models. We show that irradiation of mast cells with 1270 nm NIR light produced ROS and ATP, and irradiation of isolated mitochondria also produced ATP. In mouse skin, the relative expression levels of chemokine mRNAs, such as Ccl2 and Ccl20, were increased by irradiation with 1270 and 1064 nm NIR light at minimum safe irradiance. However, the relative expression of Nfkb1 was increased at 1064 nm, but not at 1270 nm. Serum anti-influenza IgG antibody titers increased early after immunization with 1064 nm, whereas with 1270 nm, there was not only an early response of antibody production but also persistence of antibody titers over the medium- to long-term. Thus, to our knowledge, we show for the first time that 1270 nm NIR light induces ROS and ATP production in mitochondria as photoreceptors, initiating a cascade of laser adjuvant effects for intradermal vaccines. Additionally, we demonstrate that there are wavelength-specific variations in the mechanisms and effects of laser adjuvants. In conclusion, CW1270 nm NIR light is expected to be clinically applicable as a novel laser adjuvant that is equivalent or superior to 1064 nm NIR light, because it can be operated at low energy and has a wavelength-specific adjuvant effect with medium- to long-lasting antibody titer.
Collapse
Affiliation(s)
- Yohei Maki
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Toshihiro Kushibiki
- Department of Medical Engineering, National Defense Medical College, Tokorozawa, Japan
| | - Tomoya Sano
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Takunori Ogawa
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Eri Komai
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Shusaku Takahashi
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Etsuko Kitagami
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yusuke Serizawa
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Ryosuke Nagaoka
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Shinya Yokomizo
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Takeshi Ono
- Department of Global Infectious Diseases and Tropical Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Miya Ishihara
- Department of Medical Engineering, National Defense Medical College, Tokorozawa, Japan
| | - Yasushi Miyahira
- Department of Global Infectious Diseases and Tropical Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Akihiko Kawana
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yoshifumi Kimizuka
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
14
|
Garcia-Rodriguez KM, Goenka A, Thomson DD, Bahri R, Tontini C, Salcman B, Hernandez-Pando R, Bulfone-Paus S. Bacillus Calmette-Guérin-Induced Human Mast Cell Activation Relies on IL-33 Priming. Int J Mol Sci 2022; 23:7549. [PMID: 35886897 PMCID: PMC9320129 DOI: 10.3390/ijms23147549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Bacillus Calmette-Guérin (BCG) vaccine is an attenuated strain of Mycobacterium bovis that provides weak protection against tuberculosis (TB). Mast cells (MCs) are tissue-resident immune cells strategically that serve as the first line of defence against pathogenic threats. In this study, we investigated the response of human MCs (hMCs) to BCG. We found that naïve hMCs exposed to BCG did not secrete cytokines, degranulate, or support the uptake and intracellular growth of bacteria. Since we could show that in hMCs IL-33 promotes the transcription of host-pathogen interaction, cell adhesion and activation genes, we used IL-33 for cell priming. The treatment of hMCs with IL-33, but not IFN-γ, before BCG stimulation increased IL-8, MCP-1 and IL-13 secretion, and induced an enhanced expression of the mycobacteria-binding receptor CD48. These effects were comparable to those caused by the recombinant Mycobacterium tuberculosis (Mtb) 19-KDa lipoprotein. Finally, stimulation of hMCs with IL-33 incremented MC-BCG interactions. Thus, we propose that IL-33 may improve the immunogenicity of BCG vaccine by sensitising hMCs.
Collapse
Affiliation(s)
- Karen M. Garcia-Rodriguez
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.M.G.-R.); (D.D.T.); (R.B.); (C.T.); (B.S.)
- School of Materials, Faculty of Science and Engineering, University of Manchester, Manchester M13 9PL, UK
| | - Anu Goenka
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TH, UK;
| | - Darren D. Thomson
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.M.G.-R.); (D.D.T.); (R.B.); (C.T.); (B.S.)
- MRC Centre for Medical Mycology, University of Exeter, Exeter EX4 4PY, UK
| | - Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.M.G.-R.); (D.D.T.); (R.B.); (C.T.); (B.S.)
| | - Chiara Tontini
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.M.G.-R.); (D.D.T.); (R.B.); (C.T.); (B.S.)
| | - Barbora Salcman
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.M.G.-R.); (D.D.T.); (R.B.); (C.T.); (B.S.)
| | - Rogelio Hernandez-Pando
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14080, Mexico;
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (K.M.G.-R.); (D.D.T.); (R.B.); (C.T.); (B.S.)
| |
Collapse
|
15
|
de Santana CJC, Pires Júnior OR, Fontes W, Palma MS, Castro MS. Mastoparans: A Group of Multifunctional α-Helical Peptides With Promising Therapeutic Properties. Front Mol Biosci 2022; 9:824989. [PMID: 35813822 PMCID: PMC9263278 DOI: 10.3389/fmolb.2022.824989] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Biologically active peptides have been attracting increasing attention, whether to improve the understanding of their mechanisms of action or in the search for new therapeutic drugs. Wasp venoms have been explored as a remarkable source for these molecules. In this review, the main findings on the group of wasp linear cationic α-helical peptides called mastoparans were discussed. These compounds have a wide variety of biological effects, including mast cell degranulation, activation of protein G, phospholipase A2, C, and D activation, serotonin and insulin release, and antimicrobial, hemolytic, and anticancer activities, which could lead to the development of new therapeutic agents.
Collapse
Affiliation(s)
- Carlos José Correia de Santana
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Osmindo Rodrigues Pires Júnior
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Mário Sérgio Palma
- Department of Basic and Applied Biology, Institute of Biosciences of Rio Claro, São Paulo State University, UNESP, Rio Claro, Brazil
| | - Mariana S. Castro
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
- *Correspondence: Mariana S. Castro,
| |
Collapse
|
16
|
Johnson-Weaver BT, Choi HW, Yang H, Granek JA, Chan C, Abraham SN, Staats HF. Nasal Immunization With Small Molecule Mast Cell Activators Enhance Immunity to Co-Administered Subunit Immunogens. Front Immunol 2021; 12:730346. [PMID: 34566991 PMCID: PMC8461742 DOI: 10.3389/fimmu.2021.730346] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/23/2021] [Indexed: 01/02/2023] Open
Abstract
Mast cell activators are a novel class of mucosal vaccine adjuvants. The polymeric compound, Compound 48/80 (C48/80), and cationic peptide, Mastoparan 7 (M7) are mast cell activators that provide adjuvant activity when administered by the nasal route. However, small molecule mast cell activators may be a more cost-efficient adjuvant alternative that is easily synthesized with high purity compared to M7 or C48/80. To identify novel mast cell activating compounds that could be evaluated for mucosal vaccine adjuvant activity, we employed high-throughput screening to assess over 55,000 small molecules for mast cell degranulation activity. Fifteen mast cell activating compounds were down-selected to five compounds based on in vitro immune activation activities including cytokine production and cellular cytotoxicity, synthesis feasibility, and selection for functional diversity. These small molecule mast cell activators were evaluated for in vivo adjuvant activity and induction of protective immunity against West Nile Virus infection in BALB/c mice when combined with West Nile Virus envelope domain III (EDIII) protein in a nasal vaccine. We found that three of the five mast cell activators, ST101036, ST048871, and R529877, evoked high levels of EDIII-specific antibody and conferred comparable levels of protection against WNV challenge. The level of protection provided by these small molecule mast cell activators was comparable to the protection evoked by M7 (67%) but markedly higher than the levels seen with mice immunized with EDIII alone (no adjuvant 33%). Thus, novel small molecule mast cell activators identified by high throughput screening are as efficacious as previously described mast cell activators when used as nasal vaccine adjuvants and represent next-generation mast cell activators for evaluation in mucosal vaccine studies.
Collapse
Affiliation(s)
| | - Hae Woong Choi
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
| | - Hang Yang
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Josh A. Granek
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Cliburn Chan
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Soman N. Abraham
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
- Department of Immunology, School of Medicine, Duke University, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Herman F. Staats
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
- Department of Immunology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| |
Collapse
|
17
|
Oliveira MP, Prates J, Gimenes AD, Correa SG, Oliani SM. Annexin A1 Mimetic Peptide Ac 2-26 Modulates the Function of Murine Colonic and Human Mast Cells. Front Immunol 2021; 12:689484. [PMID: 34557187 PMCID: PMC8452975 DOI: 10.3389/fimmu.2021.689484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022] Open
Abstract
Mast cells (MCs) are main effector cells in allergic inflammation and after activation, they release stored (histamine, heparin, proteases) and newly synthesized (lipid mediators and cytokines) substances. In the gastrointestinal tract the largest MC population is located in the lamina propria and submucosa whereas several signals such as the cytokine IL-4, seem to increase the granule content and to stimulate a remarkable expansion of intestinal MCs. The broad range of MC-derived bioactive molecules may explain their involvement in many different allergic disorders of the gastrointestinal tract. Annexin A1 (AnxA1) is a 37 KDa glucocorticoid induced monomeric protein selectively distributed in certain tissues. Its activity can be reproduced by mimetic peptides of the N-terminal portion, such as Ac2-26, that share the same receptor FPR-L1. Although previous reports demonstrated that AnxA1 inhibits MC degranulation in murine models, the effects of exogenous peptide Ac2-26 on intestinal MCs or the biological functions of the Ac2-26/FPR2 system in human MCs have been poorly studied. To determine the effects of Ac2-26 on the function of MCs toward the possibility of AnxA1-based therapeutics, we treated WT and IL-4 knockout mice with peptide Ac2-26, and we examined the spontaneous and compound 48/80 stimulated colonic MC degranulation and cytokine production. Moreover, in vitro, using human mast cell line HMC-1 we demonstrated that exogenous AnxA1 peptide is capable of interfering with the HMC-1 degranulation in a direct pathway through formyl peptide receptors (FPRs). We envisage that our results can provide therapeutic strategies to reduce the release of MC mediators in inflammatory allergic processes.
Collapse
Affiliation(s)
- Marcia Pereira Oliveira
- Laboratory of Interdisciplinary Medical Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Janesly Prates
- Department of Biology, Institute of Bioscience, Humanities and Exact Science, São Paulo State University (Unesp), São José do Rio Preto, Brazil
| | | | - Silvia Graciela Correa
- Departamento de Bioquímica Clinica-Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI - CONICET) - Facultad de Ciencias Quimicas- Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Sonia Maria Oliani
- Department of Biology, Institute of Bioscience, Humanities and Exact Science, São Paulo State University (Unesp), São José do Rio Preto, Brazil
- Advanced Research Center in Medicine, CEPAM –Unilago, São José do Rio Preto, Brazil
- Federal University of São Paulo, Post Graduate Program in Structural and Functional Biology, Escola Paulista de Medicina (Unifesp-EPM), São Paulo, Brazil
| |
Collapse
|
18
|
Roy S, Chompunud Na Ayudhya C, Thapaliya M, Deepak V, Ali H. Multifaceted MRGPRX2: New insight into the role of mast cells in health and disease. J Allergy Clin Immunol 2021; 148:293-308. [PMID: 33957166 PMCID: PMC8355064 DOI: 10.1016/j.jaci.2021.03.049] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/03/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Cutaneous mast cells (MCs) express Mas-related G protein-coupled receptor-X2 (MRGPRX2; mouse ortholog MrgprB2), which is activated by an ever-increasing number of cationic ligands. Antimicrobial host defense peptides (HDPs) generated by keratinocytes contribute to host defense likely by 2 mechanisms, one involving direct killing of microbes and the other via MC activation through MRGPRX2. However, its inappropriate activation may cause pseudoallergy and likely contribute to the pathogenesis of rosacea, atopic dermatitis, allergic contact dermatitis, urticaria, and mastocytosis. Gain- and loss-of-function missense single nucleotide polymorphisms in MRGPRX2 have been identified. The ability of certain ligands to serve as balanced or G protein-biased agonists has been defined. Small-molecule HDP mimetics that display both direct antimicrobial activity and activate MCs via MRGPRX2 have been developed. In addition, antibodies and reagents that modulate MRGPRX2 expression and signaling have been generated. In this article, we provide a comprehensive update on MrgprB2 and MRGPRX2 biology. We propose that harnessing MRGPRX2's host defense function by small-molecule HDP mimetics may provide a novel approach for the treatment of antibiotic-resistant cutaneous infections. In contrast, MRGPRX2-specific antibodies and inhibitors could be used for the modulation of allergic and inflammatory diseases that are mediated via this receptor.
Collapse
Affiliation(s)
- Saptarshi Roy
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pa
| | - Chalatip Chompunud Na Ayudhya
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pa
| | - Monica Thapaliya
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pa
| | - Vishwa Deepak
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pa
| | - Hydar Ali
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pa.
| |
Collapse
|
19
|
Hafezi B, Chan L, Knapp JP, Karimi N, Alizadeh K, Mehrani Y, Bridle BW, Karimi K. Cytokine Storm Syndrome in SARS-CoV-2 Infections: A Functional Role of Mast Cells. Cells 2021; 10:1761. [PMID: 34359931 PMCID: PMC8308097 DOI: 10.3390/cells10071761] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/27/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Cytokine storm syndrome is a cascade of escalated immune responses disposing the immune system to exhaustion, which might ultimately result in organ failure and fatal respiratory distress. Infection with severe acute respiratory syndrome-coronavirus-2 can result in uncontrolled production of cytokines and eventually the development of cytokine storm syndrome. Mast cells may react to viruses in collaboration with other cells and lung autopsy findings from patients that died from the coronavirus disease that emerged in 2019 (COVID-19) showed accumulation of mast cells in the lungs that was thought to be the cause of pulmonary edema, inflammation, and thrombosis. In this review, we present evidence that a cytokine response by mast cells may initiate inappropriate antiviral immune responses and cause the development of cytokine storm syndrome. We also explore the potential of mast cell activators as adjuvants for COVID-19 vaccines and discuss the medications that target the functions of mast cells and could be of value in the treatment of COVID-19. Recognition of the cytokine storm is crucial for proper treatment of patients and preventing the release of mast cell mediators, as impeding the impacts imposed by these mediators could reduce the severity of COVID-19.
Collapse
Affiliation(s)
- Bahareh Hafezi
- Department of Clinical Science, School of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad 9177948974, Iran; (B.H.); (N.K.)
| | - Lily Chan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Jason P. Knapp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Negar Karimi
- Department of Clinical Science, School of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad 9177948974, Iran; (B.H.); (N.K.)
| | - Kimia Alizadeh
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA;
| | - Yeganeh Mehrani
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| |
Collapse
|
20
|
Noel JC, Berin MC. Role of innate immunity and myeloid cells in susceptibility to allergic disease. Ann N Y Acad Sci 2021; 1499:42-53. [PMID: 34159612 DOI: 10.1111/nyas.14654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Allergic diseases, including asthma, food allergy, eczema, and allergic rhinitis, are common diseases increasing in prevalence. Allergy, a failure of immune tolerance to innocuous environmental allergens, is characterized by allergen-specific immune responses, including IgE antibodies and T helper and T follicular helper cells producing type 2 cytokines. Despite the central role of adaptive immunity in pathophysiology of allergy, there is a growing body of evidence indicating an important role for the innate immune system in allergic disease. In this review, we focus on epithelial-mononuclear phagocyte communication in the control of allergy and tolerance. We discuss studies on early life environmental exposures and allergy susceptibility, and the evidence for innate training of mononuclear phagocytes as the mechanistic link between exposure and health or disease.
Collapse
Affiliation(s)
- Justine C Noel
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - M Cecilia Berin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
21
|
Lichterman JN, Reddy SM. Mast Cells: A New Frontier for Cancer Immunotherapy. Cells 2021; 10:cells10061270. [PMID: 34063789 PMCID: PMC8223777 DOI: 10.3390/cells10061270] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 12/24/2022] Open
Abstract
Mast cells are unique tissue-resident immune cells of the myeloid lineage that have long been implicated in the pathogenesis of allergic and autoimmune disorders. More recently, mast cells have been recognized as key orchestrators of anti-tumor immunity, modulators of the cancer stroma, and have also been implicated in cancer cell intrinsic properties. As such, mast cells are an underrecognized but very promising target for cancer immunotherapy. In this review, we discuss the role of mast cells in shaping cancer and its microenvironment, the interaction between mast cells and cancer therapies, and strategies to target mast cells to improve cancer outcomes. Specifically, we address (1) decreasing cell numbers through c-KIT inhibition, (2) modulating mast cell activation and phenotype (through mast cell stabilizers, FcεR1 signaling pathway activators/inhibitors, antibodies targeting inhibitory receptors and ligands, toll like receptor agonists), and (3) altering secreted mast cell mediators and their downstream effects. Finally, we discuss the importance of translational research using patient samples to advance the field of mast cell targeting to optimally improve patient outcomes. As we aim to expand the successes of existing cancer immunotherapies, focused clinical and translational studies targeting mast cells in different cancer contexts are now warranted.
Collapse
Affiliation(s)
- Jake N. Lichterman
- Division of Hematology/Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Sangeetha M. Reddy
- Division of Hematology/Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: ; Tel.: +1-214-648-4180
| |
Collapse
|
22
|
Natarajan S, Govender K, Shobo A, Baijnath S, Arvidsson PI, Govender T, Lin J, Maguire GE, Naicker T, Kruger HG. Potential of brain mast cells for therapeutic application in the immune response to bacterial and viral infections. Brain Res 2021; 1767:147524. [PMID: 34015358 DOI: 10.1016/j.brainres.2021.147524] [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: 02/15/2021] [Revised: 04/21/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
A wide range of microorganisms can infect the central nervous system (CNS). The immune response of the CNS provides limited protection against microbes penetrating the blood-brain barrier. This results in a neurological deficit and sometimes leads to high morbidity and mortality rates despite advanced therapies. For the last two decades, different studies have expanded our understanding of the molecular basis of human neuroinfectious diseases, especially concerning the contributions of mast cell interactions with other central nervous system compartments. Brain mast cells are multifunctional cells derived from the bone marrow and reside in the brain. Their proximity to blood vessels, their role as "first responders" their unique receptors systems and their ability to rapidly release pathogen responsive mediators enable them to exert a crucial defensive role in the host-defense system. This review describes key biological and physiological functions of mast cells, concerning their ability to recognize pathogens via various receptor systems, followed by a coordinated and selective mediator release upon specific interactions with pathogenic stimulating factors. The goal of this review is to direct attention to the possibilities for therapeutic applications of mast cells against bacterial and viral related infections. We also focus on opportunities for future research activating mast cells via adjuvants.
Collapse
Affiliation(s)
- Satheesh Natarajan
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Kamini Govender
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Adeola Shobo
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Sooraj Baijnath
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Per I Arvidsson
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa; Science for Life Laboratory, Drug Discovery and Development, Platform and Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Sweden
| | - Thavendran Govender
- Department of Chemistry, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa
| | - Johnson Lin
- School of Life Sciences, University of KwaZulu-Natal, Durban 4001, South Africa; School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Glenn Em Maguire
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Tricia Naicker
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa.
| |
Collapse
|
23
|
Mantri CK, Soundarajan G, Saron WAA, Rathore APS, Alonso S, St. John AL. Maternal Immunity and Vaccination Influence Disease Severity in Progeny in a Novel Mast Cell-Deficient Mouse Model of Severe Dengue. Viruses 2021; 13:v13050900. [PMID: 34066286 PMCID: PMC8152039 DOI: 10.3390/v13050900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
Sub-neutralizing concentrations of antibodies in dengue infected patients is a major risk factor for the development of dengue hemorrhagic fever and dengue shock syndrome. Here, we describe a mouse model with a deficiency in mast cells (MCs) in addition to a deficiency in Type-I and II IFN receptors for studying dengue virus (DENV) infection. We used this model to understand the influence of MCs in a maternal antibody-dependent model of severe dengue, where offspring born to DENV-immune mothers are challenged with a heterologous DENV serotype. Mice lacking both MCs and IFN receptors were found susceptible to primary DENV infection and showed morbidity and mortality. When these mice were immunized, pups born to DENV-immune mothers were found to be protected for a longer duration from a heterologous DENV challenge. In the absence of MCs and type-I interferon signaling, IFN-γ was found to protect pups born to naïve mothers but had the opposite effect on pups born to DENV-immune mothers. Our results highlight the complex interactions between MCs and IFN-signaling in influencing the role of maternal antibodies in DENV-induced disease severity.
Collapse
Affiliation(s)
- Chinmay Kumar Mantri
- Program in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore 169857, Singapore; (G.S.); (W.A.A.S.)
- Correspondence: (C.K.M.); (A.L.S.J.)
| | - Gayathri Soundarajan
- Program in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore 169857, Singapore; (G.S.); (W.A.A.S.)
| | - Wilfried A. A. Saron
- Program in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore 169857, Singapore; (G.S.); (W.A.A.S.)
| | - Abhay P. S. Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Sylvie Alonso
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Ashley L. St. John
- Program in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore 169857, Singapore; (G.S.); (W.A.A.S.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA;
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
- SingHealth Duke-National University of Singapore Global Health Institute, Singapore 168753, Singapore
- Correspondence: (C.K.M.); (A.L.S.J.)
| |
Collapse
|
24
|
IgE-activated mast cells enhance TLR4-mediated antigen-specific CD4 + T cell responses. Sci Rep 2021; 11:9686. [PMID: 33958642 PMCID: PMC8102524 DOI: 10.1038/s41598-021-88956-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/19/2021] [Indexed: 12/30/2022] Open
Abstract
Mast cells are potent mediators of allergy and asthma, yet their role in regulating adaptive immunity remains ambiguous. On the surface of mast cells, the crosslinking of IgE bound to FcεRI by a specific antigen recognized by that IgE triggers the release of immune mediators such as histamine and cytokines capable of activating other immune cells; however, little is known about the mast cell contribution to the induction of endogenous, antigen-specific CD4+ T cells. Here we examined the effects of specific mast cell activation in vivo on the initiation of an antigen-specific CD4+ T cell response. While CD4+ T cells were not enhanced by FcεRI stimulation alone, their activation was synergistically enhanced when FcεRI activation was combined with TLR4 stimulation. This enhanced activation was dependent on global TLR4 stimulation but appeared to be less dependent on mast cell expressed TLR4. This study provides important new evidence to support the role of mast cells as mediators of the antigen-specific adaptive immune response.
Collapse
|
25
|
Zhu W, Dong C, Wei L, Wang BZ. Promising Adjuvants and Platforms for Influenza Vaccine Development. Pharmaceutics 2021; 13:pharmaceutics13010068. [PMID: 33430259 PMCID: PMC7825707 DOI: 10.3390/pharmaceutics13010068] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Influenza is one of the major threats to public health. Current influenza vaccines cannot provide effective protection against drifted or shifted influenza strains. Researchers have considered two important strategies to develop novel influenza vaccines with improved immunogenicity and broader protective efficacy. One is applying fewer variable viral antigens, such as the haemagglutinin stalk domain. The other is including adjuvants in vaccine formulations. Adjuvants are promising and helpful boosters to promote more rapid and stronger immune responses with a dose-sparing effect. However, few adjuvants are currently licensed for human influenza vaccines, although many potential candidates are in different trials. While many advantages have been observed using adjuvants in influenza vaccine formulations, an improved understanding of the mechanisms underlying viral infection and vaccination-induced immune responses will help to develop new adjuvant candidates. In this review, we summarize the works related to adjuvants in influenza vaccine research that have been used in our studies and other laboratories. The review will provide perspectives for the utilization of adjuvants in developing next-generation and universal influenza vaccines.
Collapse
|
26
|
Duguay BA, Lu L, Arizmendi N, Unsworth LD, Kulka M. The Possible Uses and Challenges of Nanomaterials in Mast Cell Research. THE JOURNAL OF IMMUNOLOGY 2020; 204:2021-2032. [PMID: 32253270 DOI: 10.4049/jimmunol.1800658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 12/19/2019] [Indexed: 11/19/2022]
Abstract
Mast cells are tissue-resident immune cells that are involved in inflammation and fibrosis but also serve beneficial roles, including tissue maintenance, angiogenesis, pathogen clearance, and immunoregulation. Their multifaceted response and the ability of their mediators to target multiple organs and tissues means that mast cells play important roles in numerous conditions, including asthma, atopic dermatitis, drug sensitivities, ischemic heart disease, Alzheimer disease, arthritis, irritable bowel syndrome, infections (parasites, bacteria and viruses), and cancer. As a result, mast cells have become an important target for drug discovery and diagnostic research. Recent work has focused on applying novel nanotechnologies to explore cell biology. In this brief review, we will highlight the use of nanomaterials to modify mast cell functions and will discuss the potential of these technologies as research tools for understanding mast cell biology.
Collapse
Affiliation(s)
- Brett A Duguay
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada
| | - Lei Lu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Narcy Arizmendi
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada; and
| | - Marianna Kulka
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
27
|
Willows S, Kulka M. Harnessing the Power of Mast Cells in unconventional Immunotherapy Strategies and Vaccine Adjuvants. Cells 2020; 9:cells9122713. [PMID: 33352850 PMCID: PMC7766453 DOI: 10.3390/cells9122713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/03/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Mast cells are long-lived, granular, myeloid-derived leukocytes that have significant protective and repair functions in tissues. Mast cells sense disruptions in the local microenvironment and are first responders to physical, chemical and biological insults. When activated, mast cells release growth factors, proteases, chemotactic proteins and cytokines thereby mobilizing and amplifying the reactions of the innate and adaptive immune system. Mast cells are therefore significant regulators of homeostatic functions and may be essential in microenvironmental changes during pathogen invasion and disease. During infection by helminths, bacteria and viruses, mast cells release antimicrobial factors to facilitate pathogen expulsion and eradication. Mast cell-derived proteases and growth factors protect tissues from insect/snake bites and exposure to ultraviolet radiation. Finally, mast cells release mediators that promote wound healing in the inflammatory, proliferative and remodelling stages. Since mast cells have such a powerful repertoire of functions, targeting mast cells may be an effective new strategy for immunotherapy of disease and design of novel vaccine adjuvants. In this review, we will examine how certain strategies that specifically target and activate mast cells can be used to treat and resolve infections, augment vaccines and heal wounds. Although these strategies may be protective in certain circumstances, mast cells activation may be deleterious if not carefully controlled and any therapeutic strategy using mast cell activators must be carefully explored.
Collapse
Affiliation(s)
- Steven Willows
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Dr, Edmonton, AB T6G 2M9, Canada;
| | - Marianna Kulka
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Dr, Edmonton, AB T6G 2M9, Canada;
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Correspondence: ; Tel.: +1-780-641-1687
| |
Collapse
|
28
|
Cossette B, Kelly SH, Collier JH. Intranasal Subunit Vaccination Strategies Employing Nanomaterials and Biomaterials. ACS Biomater Sci Eng 2020; 7:1765-1779. [DOI: 10.1021/acsbiomaterials.0c01291] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Benjamin Cossette
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| | - Sean H. Kelly
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| | - Joel H. Collier
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| |
Collapse
|
29
|
El Ansari YS, Kanagaratham C, Oettgen HC. Mast Cells as Regulators of Adaptive Immune Responses in Food Allergy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:711-718. [PMID: 33380933 PMCID: PMC7757069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mast cells are a critical first line of defense against endogenous and environmental threats. Their participation in innate immunity is well characterized; activation of toll like receptors as well as receptors for complement, adenosine, and a host of other ligands leads to mast cell release of preformed mediators contained within granules along with newly synthesized arachidonic acid metabolites, cytokines, and chemokines. These confer protective effects including the induction of mucus secretion, smooth muscle contraction, and activation of common itch and pain sensations, all of which act to promote expulsion of noxious agents. While their innate immune role as sentinel cells is well established, recent research has brought into focus their separate but also critical function in adaptive immunity particularly in the setting of IgE mediated food allergies. Crosslinking of FcεR1, the high affinity receptor for IgE, when bound to IgE and antigen, triggers the release of the same factors and elicits the same physiologic responses that occur after activation by innate stimuli. Though IgE-activated mast cells are best known for their role in acute allergic reactions, including the most severe manifestation, anaphylaxis, accumulating evidence has suggested an immunoregulatory effect in T cell-mediated immunity, modulating the balance between type 2 immunity and tolerance. In this review, we outline how mast cells act as adjuvants for food antigen driven Th2 cell responses, while curtailing Treg function.
Collapse
Affiliation(s)
- Yasmeen S. El Ansari
- Boston Children’s Hospital and Harvard Medical School,
Boston, MA
- Institute of Laboratory Medicine, Philipps University
Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Boston Children’s Hospital and Harvard Medical School,
Boston, MA
- Department of Pediatrics, Harvard Medical School,
Boston, MA
| | - Hans C. Oettgen
- Boston Children’s Hospital and Harvard Medical School,
Boston, MA
- Department of Pediatrics, Harvard Medical School,
Boston, MA
| |
Collapse
|
30
|
Mast Cell Functions Linking Innate Sensing to Adaptive Immunity. Cells 2020; 9:cells9122538. [PMID: 33255519 PMCID: PMC7761480 DOI: 10.3390/cells9122538] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Although mast cells (MCs) are known as key drivers of type I allergic reactions, there is increasing evidence for their critical role in host defense. MCs not only play an important role in initiating innate immune responses, but also influence the onset, kinetics, and amplitude of the adaptive arm of immunity or fine-tune the mode of the adaptive reaction. Intriguingly, MCs have been shown to affect T-cell activation by direct interaction or indirectly, by modifying the properties of antigen-presenting cells, and can even modulate lymph node-borne adaptive responses remotely from the periphery. In this review, we provide a summary of recent findings that explain how MCs act as a link between the innate and adaptive immunity, all the way from sensing inflammatory insult to orchestrating the final outcome of the immune response.
Collapse
|
31
|
St. John AL, Choi HW, Walker QD, Blough B, Kuhn CM, Abraham SN, Staats HF. Novel mucosal adjuvant, mastoparan-7, improves cocaine vaccine efficacy. NPJ Vaccines 2020; 5:12. [PMID: 32047657 PMCID: PMC7002721 DOI: 10.1038/s41541-020-0161-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/14/2020] [Indexed: 12/29/2022] Open
Abstract
Cocaine is one of the most potent and addictive psychostimulants known and there are no available pharmacotherapies to treat cocaine addiction. Here we describe a novel cocaine vaccine employing the mucosal adjuvant and mast cell-activating oligopeptide, mastoparan-7 (M7), to achieve optimal IgA antibody responses in mucosal secretions and effective induction of humoral immunity using a short immunization protocol. This formulation, using a hapten-carrier system to deliver cocaine as antigen, also reduced cocaine penetration of the blood brain barrier and protected mice from its psychoactive effects by reducing cocaine-induced locomotion. Surprisingly, the magnitude of cocaine-specific antibody titers induced by each adjuvant was not the major determinant of functional protection from cocaine challenge. A side-by-side comparison of the two haptens, cocaine and its analog GNC demonstrated that cocaine haptenation resulted in superior functional protection when used in combination with the novel mucosal adjuvant, M7. These results provide a new potential strategy for combatting cocaine addiction through mucosal vaccination.
Collapse
Affiliation(s)
- Ashley L. St. John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, 169857 Singapore
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228 Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore, 168753 Singapore
| | - Hae Woong Choi
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Present Address: Korea University, Division of Life Sciences, 108 Hana-Science Building, 145 Anam-ro, Seongbuk-gu, Seoul, South Korea
| | - Q. David Walker
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Bruce Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709 USA
| | - Cynthia M. Kuhn
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Soman N. Abraham
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, 169857 Singapore
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Immunology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710 USA
| | - Herman F. Staats
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
32
|
Olivera A, Rivera J. Paradigm Shifts in Mast Cell and Basophil Biology and Function: An Emerging View of Immune Regulation in Health and Disease. Methods Mol Biol 2020; 2163:3-31. [PMID: 32766962 DOI: 10.1007/978-1-0716-0696-4_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The physiological role of the mast cell and basophil has for many years remained enigmatic. In this chapter, we briefly summarize some of the more recent studies that shed new light on the role of mast cells and basophils in health and disease. What we gain from these studies is a new appreciation for mast cells and basophils as sentinels in host defense and a further understanding that dysregulation of mast cell and basophil function can be a component of various diseases other than allergies. Perhaps the most important insight reaped from this work is the increasing awareness that mast cells and basophils can function as immunoregulatory cells that modulate the immune response in health and disease. Collectively, the recent knowledge provides new challenges and opportunities toward the development of novel therapeutic strategies to augment host protection and modify disease through manipulation of mast cell and basophil function.
Collapse
Affiliation(s)
- Ana Olivera
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Juan Rivera
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
|
34
|
Imiquimod enhances DNFB mediated contact hypersensitivity in mice. Int Immunopharmacol 2019; 72:284-291. [DOI: 10.1016/j.intimp.2019.04.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/22/2019] [Accepted: 04/12/2019] [Indexed: 11/30/2022]
|
35
|
Optimized Mucosal Modified Vaccinia Virus Ankara Prime/Soluble gp120 Boost HIV Vaccination Regimen Induces Antibody Responses Similar to Those of an Intramuscular Regimen. J Virol 2019; 93:JVI.00475-19. [PMID: 31068425 DOI: 10.1128/jvi.00475-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/01/2019] [Indexed: 12/29/2022] Open
Abstract
The benefits of mucosal vaccines over injected vaccines are difficult to ascertain, since mucosally administered vaccines often induce serum antibody responses of lower magnitude than those induced by injected vaccines. This study aimed to determine if mucosal vaccination using a modified vaccinia virus Ankara expressing human immunodeficiency virus type 1 (HIV-1) gp120 (MVAgp120) prime and a HIV-1 gp120 protein boost could be optimized to induce serum antibody responses similar to those induced by an intramuscularly (i.m.) administered MVAgp120 prime/gp120 boost to allow comparison of an i.m. immunization regimen to a mucosal vaccination regimen for the ability to protect against a low-dose rectal simian-human immunodeficiency virus (SHIV) challenge. A 3-fold higher antigen dose was required for intranasal (i.n.) immunization with gp120 to induce serum anti-gp120 IgG responses not significantly different than those induced by i.m. immunization. gp120 fused to the adenovirus type 2 fiber binding domain (gp120-Ad2F), a mucosal targeting ligand, exhibited enhanced i.n. immunogenicity compared to gp120. MVAgp120 was more immunogenic after i.n. delivery than after gastric or rectal delivery. Using these optimized vaccines, an i.n. MVAgp120 prime/combined i.m. (gp120) and i.n. (gp120-Ad2F) boost regimen (i.n./i.m.-plus-i.n.) induced serum anti-gp120 antibody titers similar to those induced by the intramuscular prime/boost regimen (i.m./i.m.) in rabbits and nonhuman primates. Despite the induction of similar systemic anti-HIV-1 antibody responses, neither the i.m./i.m. nor the i.n./i.m.-plus-i.n. regimen protected against a repeated low-dose rectal SHIV challenge. These results demonstrate that immunization regimens utilizing the i.n. route are able to induce serum antigen-specific antibody responses similar to those induced by systemic immunization.IMPORTANCE Mucosal vaccination is proposed as a method of immunization able to induce protection against mucosal pathogens that is superior to protection provided by parenteral immunization. However, mucosal vaccination often induces serum antigen-specific immune responses of lower magnitude than those induced by parenteral immunization, making the comparison of mucosal and parenteral immunization difficult. We identified vaccine parameters that allowed an immunization regimen consisting of an i.n. prime followed by boosters administered by both i.n. and i.m. routes to induce serum antibody responses similar to those induced by i.m. prime/boost vaccination. Additional studies are needed to determine the potential benefit of mucosal immunization for HIV-1 and other mucosally transmitted pathogens.
Collapse
|
36
|
Choi HW, Chan C, Shterev ID, Lynch HE, Robinette TJ, Johnson-Weaver BT, Shi J, Sempowski GD, Kim SY, Dickson JK, Gooden DM, Abraham SN, Staats HF. Identification of Novel Mast Cell Activators Using Cell-Based High-Throughput Screening. SLAS DISCOVERY 2019; 24:628-640. [PMID: 30917061 DOI: 10.1177/2472555219834699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mast cells (MCs) are known to regulate innate and adaptive immunity. MC activators have recently been described as safe and effective vaccine adjuvants. Many currently known MC activators are inadequate for in vivo applications, however, and research on identifying novel MC activators is limited. In this study, we identified novel MC activators by using high-throughput screening (HTS) assays using approximately 55,000 small molecules. Data sets obtained by the primary HTS assays were statistically evaluated using quality control rules and the B-score calculation, and compounds with B-scores of >3.0 were chosen as mast cell activators (hits). These hits were re-evaluated with secondary and tertiary HTS assays, followed by further statistical analysis. From these hits, we selected 15 compounds that caused degranulation in murine and human MCs, with potential for flexible chemical modification for further study. Among these 15 compounds, ST101036, ST029248, and ST026567 exhibited higher degranulation potency than other hit compounds in both human and mouse MCs. In addition, the 15 compounds identified promote de novo synthesis of cytokines and induce the release of eicosanoids from human and mouse MCs. HTS enabled us to identify small-molecule MC activators with unique properties that may be useful as vaccine adjuvants.
Collapse
Affiliation(s)
- Hae Woong Choi
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Cliburn Chan
- 2 Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Ivo D Shterev
- 3 Duke Regional Biocontainment Laboratory, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Heather E Lynch
- 3 Duke Regional Biocontainment Laboratory, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,5 Departments of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Taylor J Robinette
- 5 Departments of Medicine, Duke University School of Medicine, Durham, NC, USA
| | | | - Jianling Shi
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Gregory D Sempowski
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA.,3 Duke Regional Biocontainment Laboratory, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,5 Departments of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - So Young Kim
- 6 Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | | | - David M Gooden
- 8 Department of Chemistry, Duke University, Durham, NC, USA
| | - Soman N Abraham
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA.,9 Department of Molecular Genetics & Microbiology, Duke University Medical Center, Durham, NC, USA.,10 Department of Immunology, Duke University Medical Center, Durham, NC, USA.,11 Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore
| | - Herman F Staats
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,10 Department of Immunology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
37
|
Bento D, Jesus S, Lebre F, Gonçalves T, Borges O. Chitosan Plus Compound 48/80: Formulation and Preliminary Evaluation as a Hepatitis B Vaccine Adjuvant. Pharmaceutics 2019; 11:pharmaceutics11020072. [PMID: 30744102 PMCID: PMC6409706 DOI: 10.3390/pharmaceutics11020072] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/22/2019] [Accepted: 02/04/2019] [Indexed: 11/16/2022] Open
Abstract
Current vaccine research is mostly based on subunit antigens. Despite the better toxicity profile of these antigens they are often poorly immunogenic, so adjuvant association has been explored as a strategy to obtain a potent vaccine formulation. Recently, mast cell activators were recognized as a new class of vaccine adjuvants capable of potentiating mucosal and systemic immune responses. In this study, a co-adjuvanted delivery system was developed and characterized, combining the mast cell activator C48/80 with chitosan nanoparticles (Chi-C48/80 NPs), and the results were compared with plain chitosan nanoparticles. The adsorption of model antigens onto the NP surface as well as the biocompatibility of the system was not affected by the incorporation of C48/80 in the formulation. The stability of the nanoparticles was demonstrated by studying the variation of size and zeta potential at different times, and the ability to be internalized by antigen presenting cells was confirmed by confocal microscopy. Vaccination studies with hepatitis B surface antigen loaded Chi-C48/80 NPs validated the adjuvanticity of the delivery system, demonstrating for the first time a successful association between a mast cell activator and chitosan nanoparticles as a vaccine adjuvant for hepatitis B virus, applied to a nasal vaccination strategy.
Collapse
Affiliation(s)
- Dulce Bento
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-0504 Coimbra, Portugal.
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - Sandra Jesus
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-0504 Coimbra, Portugal.
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - Filipa Lebre
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-0504 Coimbra, Portugal.
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - Teresa Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-0504 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Olga Borges
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-0504 Coimbra, Portugal.
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| |
Collapse
|
38
|
Stassen M, Hartmann AK, Delgado SJ, Dehmel S, Braun A. Mast cells within cellular networks. J Allergy Clin Immunol 2019; 144:S46-S54. [PMID: 30731122 DOI: 10.1016/j.jaci.2019.01.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 11/27/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022]
Abstract
Mast cells are highly versatile in terms of their mode of activation by a host of stimuli and their ability to flexibly release a plethora of biologically highly active mediators. Within the immune system, mast cells can best be designated as an active nexus interlinking innate and adaptive immunity. Here we try to draw an arc from initiation of acute inflammatory reactions to microbial pathogens to development of adaptive immunity and allergies. This multifaceted nature of mast cells is made possible by interaction with multiple cell types of immunologic and nonimmunologic origin. Examples for the former include neutrophils, eosinophils, T cells, and professional antigen-presenting cells. These interactions allow mast cells to orchestrate inflammatory innate reactions and complex adaptive immunity, including the pathogenesis of allergies. Important partners of nonimmunologic origin include cells of the sensory neuronal system. The intimate association between mast cells and sensory nerve fibers allows bidirectional communication, leading to neurogenic inflammation. Evidence is accumulating that this mast cell/nerve crosstalk is of pathophysiologic relevance in patients with allergic diseases, such as asthma.
Collapse
Affiliation(s)
- Michael Stassen
- Institute for Immunology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Ann-Kathrin Hartmann
- Institute for Immunology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sharon Jiménez Delgado
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH) research network, Member of the Cluster of Excellence Regenerative Biology to Reconstructive Therapy (REBIRTH), Hannover, Germany; Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Susann Dehmel
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH) research network, Member of the Cluster of Excellence Regenerative Biology to Reconstructive Therapy (REBIRTH), Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH) research network, Member of the Cluster of Excellence Regenerative Biology to Reconstructive Therapy (REBIRTH), Hannover, Germany; Institute of Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
39
|
Roy S, Ganguly A, Haque M, Ali H. Angiogenic Host Defense Peptide AG-30/5C and Bradykinin B 2 Receptor Antagonist Icatibant Are G Protein Biased Agonists for MRGPRX2 in Mast Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:1229-1238. [PMID: 30651343 DOI: 10.4049/jimmunol.1801227] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/12/2018] [Indexed: 01/21/2023]
Abstract
AG-30/5C is an angiogenic host defense peptide that activates human mast cells (MC) via an unknown mechanism. Using short hairpin RNA-silenced human MC line LAD2 and stably transfected RBL-2H3 cells, we demonstrate that AG-30/5C induces MC degranulation via Mas-related G protein-coupled receptor X2 (MRGPRX2). Most G protein-coupled receptors signal via parallel and independent pathways mediated by G proteins and β-arrestins. AG-30/5C and compound 48/80 induced similar maximal MC degranulation via MRGPRX2, which was abolished by pertussis toxin. However, compound 48/80 induced a robust β-arrestin activation as determined by transcriptional activation following arrestin translocation (Tango), but AG-30/5C did not. Overnight culture of MC with compound 48/80 resulted in reduced cell surface MRGPRX2 expression, and this was associated with a significant decrease in subsequent MC degranulation in response to compound 48/80 or AG-30/5C. However, AG-30/5C pretreatment had no effect on cell surface MRGPRX2 expression or degranulation in response to compound 48/80 or AG-30/5C. Icatibant, a bradykinin B2 receptor antagonist, promotes MC degranulation via MRGPRX2 and causes pseudoallergic drug reaction. Icatibant caused MC degranulation via a pertussis toxin-sensitive G protein but did not activate β-arrestin. A screen of the National Institutes of Health Clinical Collection library led to the identification of resveratrol as an inhibitor of MRGPRX2. Resveratrol inhibited compound 48/80-induced Tango and MC degranulation in response to compound 48/80, AG-30/5C, and Icatibant. This study demonstrates the novel finding that AG-30/5C and Icatibant serve as G protein-biased agonists for MRGPRX2, but compound 48/80 signals via both G protein and β-arrestin with distinct differences in receptor regulation.
Collapse
Affiliation(s)
- Saptarshi Roy
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Anirban Ganguly
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Maureen Haque
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Hydar Ali
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
40
|
Arifuzzaman M, Mobley YR, Choi HW, Bist P, Salinas CA, Brown ZD, Chen SL, Staats HF, Abraham SN. MRGPR-mediated activation of local mast cells clears cutaneous bacterial infection and protects against reinfection. SCIENCE ADVANCES 2019; 5:eaav0216. [PMID: 30613778 PMCID: PMC6314830 DOI: 10.1126/sciadv.aav0216] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/27/2018] [Indexed: 05/22/2023]
Abstract
Mast cells (MCs) are strategically distributed at barrier sites and prestore various immunocyte-recruiting cytokines, making them ideal targets for selective activation to treat peripheral infections. Here, we report that topical treatment with mastoparan, a peptide MC activator (MCA), enhances clearance of Staphylococcus aureus from infected mouse skins and accelerates healing of dermonecrotic lesions. Mastoparan functions by activating connective tissue MCs (CTMCs) via the MRGPRX2 (Mas-related G protein-coupled receptor member X2) receptor. Peripheral CTMC activation, in turn, enhances recruitment of bacteria-clearing neutrophils and wound-healing CD301b+ dendritic cells. Consistent with MCs playing a master coordinating role, MC activation also augmented migration of various antigen-presenting dendritic cells to draining lymph nodes, leading to stronger protection against a second infection challenge. MCAs therefore orchestrate both the innate and adaptive immune arms, which could potentially be applied to combat peripheral infections by a broad range of pathogens.
Collapse
Affiliation(s)
- Mohammad Arifuzzaman
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Yuvon R. Mobley
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Hae Woong Choi
- Department of Pathology, Duke University, Durham, NC 27710, USA
| | - Pradeep Bist
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore
| | | | - Zachary D. Brown
- Undergraduate Program in Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Swaine L. Chen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Infectious Diseases Group, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Herman F. Staats
- Department of Pathology, Duke University, Durham, NC 27710, USA
- Department of Immunology, Duke University, Durham, NC 27710, USA
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Soman N. Abraham
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- Department of Pathology, Duke University, Durham, NC 27710, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore
- Department of Immunology, Duke University, Durham, NC 27710, USA
- Corresponding author.
| |
Collapse
|
41
|
Kimizuka Y, Katagiri W, Locascio JJ, Shigeta A, Sasaki Y, Shibata M, Morse K, Sîrbulescu RF, Miyatake M, Reeves P, Suematsu M, Gelfand J, Brauns T, Poznansky MC, Tsukada K, Kashiwagi S. Brief Exposure of Skin to Near-Infrared Laser Modulates Mast Cell Function and Augments the Immune Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:3587-3603. [PMID: 30420435 PMCID: PMC6289684 DOI: 10.4049/jimmunol.1701687] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
The treatment of skin with a low-power continuous-wave (CW) near-infrared (NIR) laser prior to vaccination is an emerging strategy to augment the immune response to intradermal vaccine, potentially substituting for chemical adjuvant, which has been linked to adverse effects of vaccines. This approach proved to be low cost, simple, small, and readily translatable compared with the previously explored pulsed-wave medical lasers. However, little is known on the mode of laser-tissue interaction eliciting the adjuvant effect. In this study, we sought to identify the pathways leading to the immunological events by examining the alteration of responses resulting from genetic ablation of innate subsets including mast cells and specific dendritic cell populations in an established model of intradermal vaccination and analyzing functional changes of skin microcirculation upon the CW NIR laser treatment in mice. We found that a CW NIR laser transiently stimulates mast cells via generation of reactive oxygen species, establishes an immunostimulatory milieu in the exposed tissue, and provides migration cues for dermal CD103+ dendritic cells without inducing prolonged inflammation, ultimately augmenting the adaptive immune response. These results indicate that use of an NIR laser with distinct wavelength and power is a safe and effective tool to reproducibly modulate innate programs in skin. These mechanistic findings would accelerate the clinical translation of this technology and warrant further explorations into the broader application of NIR lasers to the treatment of immune-related skin diseases.
Collapse
Affiliation(s)
- Yoshifumi Kimizuka
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Wataru Katagiri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129
- Graduate School of Fundamental Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology, 14152 Huddinge, Sweden
| | - Joseph J Locascio
- Alzheimer's Disease Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114
| | - Ayako Shigeta
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Yuri Sasaki
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Mai Shibata
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Kaitlyn Morse
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Mizuki Miyatake
- Faculty of Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan; and
| | - Patrick Reeves
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-0016, Japan
| | - Jeffrey Gelfand
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Timothy Brauns
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129
| | - Kosuke Tsukada
- Graduate School of Fundamental Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
- Faculty of Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan; and
| | - Satoshi Kashiwagi
- Vaccine and Immunotherapy Center, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129;
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129
| |
Collapse
|
42
|
Dudeck J, Froebel J, Kotrba J, Lehmann CHK, Dudziak D, Speier S, Nedospasov SA, Schraven B, Dudeck A. Engulfment of mast cell secretory granules on skin inflammation boosts dendritic cell migration and priming efficiency. J Allergy Clin Immunol 2018; 143:1849-1864.e4. [PMID: 30339853 DOI: 10.1016/j.jaci.2018.08.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 07/13/2018] [Accepted: 08/26/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Mast cells (MCs) are best known as key effector cells of allergic reactions, but they also play an important role in host defense against pathogens. Despite increasing evidence for a critical effect of MCs on adaptive immunity, the underlying mechanisms are poorly understood. OBJECTIVE Here we monitored MC intercellular communication with dendritic cells (DCs), MC activation, and degranulation and tracked the fate of exocytosed mast cell granules (MCGs) during skin inflammation. METHODS Using a strategy to stain intracellular MCGs in vivo, we tracked the MCG fate after skin inflammation-induced MC degranulation. Furthermore, exogenous MCGs were applied to MC-deficient mice by means of intradermal injection. MCG effects on DC functionality and adaptive immune responses in vivo were assessed by combining intravital multiphoton microscopy with flow cytometry and functional assays. RESULTS We demonstrate that dermal DCs engulf the intact granules exocytosed by MCs on skin inflammation. Subsequently, the engulfed MCGs are actively shuttled to skin-draining lymph nodes and finally degraded inside DCs within the lymphoid tissue. Most importantly, MCG uptake promotes DC maturation and migration to skin-draining lymph nodes, partially through MC-derived TNF, and boosts their T-cell priming efficiency. Surprisingly, exogenous MCGs alone are sufficient to induce a prominent DC activation and T-cell response. CONCLUSION Our study highlights a unique feature of peripheral MCs to affect lymphoid tissue-borne adaptive immunity over distance by modifying DC functionality through delivery of granule-stored mediators.
Collapse
Affiliation(s)
- Jan Dudeck
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Julia Froebel
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Johanna Kotrba
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christian H K Lehmann
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital of Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital of Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, München-Neuherberg, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; DFG-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology and Lomonosov Moscow State University, Moscow, Russia
| | - Burkhart Schraven
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Anne Dudeck
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany.
| |
Collapse
|
43
|
Schubert N, Lisenko K, Auerbach C, Weitzmann A, Ghouse SM, Muhandes L, Haase C, Häring T, Schulze L, Voehringer D, Gunzer F, Müller W, Feyerabend TB, Rodewald HR, Dudeck A, Roers A. Unimpaired Responses to Vaccination With Protein Antigen Plus Adjuvant in Mice With Kit-Independent Mast Cell Deficiency. Front Immunol 2018; 9:1870. [PMID: 30210490 PMCID: PMC6123530 DOI: 10.3389/fimmu.2018.01870] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/30/2018] [Indexed: 12/23/2022] Open
Abstract
Innate inflammatory responses are crucial for induction and regulation of T cell and antibody responses. Mast cell (MC)-deficient Kit mutant mice showed impaired adaptive immunity, suggesting that MCs provide essential adjuvant activities, and pharmacological MC activation was proposed as a new adjuvant principle. However, the Kit mutations result in complex alterations of the immune system in addition to MC deficiency. We revisited the role of MCs in vaccination responses using Mcpt5-Cre R26DTA/DTA and Cpa3Cre/+ mice that lack connective tissue MCs or all MCs, respectively, but feature an otherwise normal immune system. These animals showed no impairment of T and B cell responses to intradermal vaccination with protein antigen plus complete Freund’s adjuvant. Moreover, we demonstrate that the adjuvant effects of the MC secretagogue c48/80 in intradermal or mucosal immunization are independent of the presence of MCs. We hence find no evidence for a regulation by MCs of adaptive immune responses to protein antigens. The finding that immunological MC functions differ from those suggested by experiments in Kit mutants, emphasizes the importance of rigorous tests in Kit-independent MC-deficiency models.
Collapse
Affiliation(s)
- Nadja Schubert
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Katharina Lisenko
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Christian Auerbach
- Medical Faculty Carl Gustav Carus, Institute of Medical Microbiology and Hygiene, University of Technology Dresden, Dresden, Germany
| | - Anke Weitzmann
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Shanawaz Mohammed Ghouse
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Lina Muhandes
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Christa Haase
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Tobias Häring
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Livia Schulze
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Florian Gunzer
- Medical Faculty Carl Gustav Carus, Institute of Medical Microbiology and Hygiene, University of Technology Dresden, Dresden, Germany
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Anne Dudeck
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany.,Medical Faculty, Institute for Molecular and Clinical Immunology, Otto von Guericke University, Magdeburg, Germany
| | - Axel Roers
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| |
Collapse
|
44
|
Ren S, Wang Q, Zhang Y, Song Y, Dong X, Zhang W, Qin X, Liu M, Yu T. Imiquimod enhances the potency of an exogenous BM-DC based vaccine against mouse melanoma. Int Immunopharmacol 2018; 64:69-77. [PMID: 30149266 DOI: 10.1016/j.intimp.2018.08.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
Abstract
Dendritic cell (DC) vaccine is a potent immunotherapeutic approach for cancer treatment, but the clinical efficacy needs to be improved. In this study, we evaluated the combinational effect of Toll-like receptor 7 (TLR7) agonist Imiquimod and BM-DC vaccine against mouse melanoma and explored the potential mechanisms. We found that topical application of Imiquimod cream caused skin inflammation and enhanced exogenous BM-DC homing to draining lymph nodes. Imiquimod treatment enhanced DC vaccine efficacy against B16-OVA melanoma. The combinational modality enhanced cytotoxicity of splenic lymphocyte to tumor cells and inhibited CD4+FOXP3+Treg cell production. TLR7 mRNA expression was confirmed in both MC/9 mast cells and DCs. MC/9 cells treated by R837 (soluble form of Imiquimod) enhanced CD80, CD86, MHC-II and CCR7 expression on DCs. R837 inhibited B16-OVA cell growth in vitro. Our findings suggest that Imiquimod can be used as a potent adjuvant in the formulation of a DC-based tumor fighting vaccine. The mechanisms underlying these effects of Imiquimod are related with enhanced DC homing to DLNs, inhibition of Treg's production, direct tumor cell toxicity and synergistic function with mast cell in enhancing DC activation.
Collapse
Affiliation(s)
- Shurong Ren
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China.
| | - Qiubo Wang
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Yanli Zhang
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Yancheng Song
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Xue Dong
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Wendi Zhang
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Xianfei Qin
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Mingyue Liu
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Ting Yu
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| |
Collapse
|
45
|
Attia Z, Rowe JC, Kim E, Varikuti S, Steiner HE, Zaghawa A, Hassan H, Cormet-Boyaka E, Satoskar AR, Boyaka PN. Inhibitors of elastase stimulate murine B lymphocyte differentiation into IgG- and IgA-producing cells. Eur J Immunol 2018; 48:1295-1301. [PMID: 29710424 DOI: 10.1002/eji.201747264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/17/2018] [Accepted: 04/25/2018] [Indexed: 01/11/2023]
Abstract
It is well established that dendritic cells and macrophages play a role in antigen presentation to B and T cells and in shaping B and T cell responses via cytokines they produce. We have previously reported that depletion of neutrophils improves the production of mucosal IgA after sublingual immunization with Bacillus anthracis edema toxin as adjuvant. These past studies also demonstrated that an inverse correlation exists between the number of neutrophils and production of IgA by B cells. Using specific inhibitors of elastase, we addressed whether the elastase activity of neutrophil could be the factor that interferes with production of IgA and possibly other immunoglobulin isotypes. We found that murine splenocytes and mesenteric lymph node cells cultured for 5 days in the presence of neutrophil elastase inhibitors secreted higher levels of IgG and IgA than cells cultured in the absence of inhibitors. The effect of the inhibitors was dose-dependent and was consistent with increased frequency of CD138+ cells expressing IgG or IgA. Finally, neutrophil elastase inhibitors increased transcription of mRNA for AID, IL-10, BAFF and APRIL, factors involved in B cell differentiation. These findings identify inhibitors of elastase as potential adjuvants for increasing production of antibodies.
Collapse
Affiliation(s)
- Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA.,Department of Medicine and Infectious Diseases, University of Sadat City, Sadat City, Egypt
| | - John C Rowe
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Haley E Steiner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Ahmad Zaghawa
- Department of Medicine and Infectious Diseases, University of Sadat City, Sadat City, Egypt
| | - Hany Hassan
- Department of Medicine and Infectious Diseases, University of Sadat City, Sadat City, Egypt
| | | | - Abhay R Satoskar
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
46
|
Mast cell activators as novel immune regulators. Curr Opin Pharmacol 2018; 41:89-95. [PMID: 29843056 DOI: 10.1016/j.coph.2018.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/14/2018] [Indexed: 11/22/2022]
Abstract
Mast cells are an important cell type of the innate immune system that when activated, play a crucial role in generating protective innate host responses after bacterial and viral infection. Additionally, activated mast cells influence lymph node composition to regulate the induction of adaptive immune responses. The recognition that mast cells play a beneficial role in host responses to microbial infection and induction of adaptive immunity has provided the rationale to evaluate mast cell activators for use as antimicrobials or vaccine adjuvants. This review summarizes the role of mast cell activators in antimicrobial responses while also discussing the use of different classes of mast cell activators as potent vaccine adjuvants that enhance the induction of protective immune responses.
Collapse
|
47
|
Li X, Wu Y, Huang S, Lu F. Disodium cromoglycate may act as a novel adjuvant for UV-attenuated Toxoplasma gondii vaccine in mouse model. Parasitol Int 2018; 67:351-356. [PMID: 29421521 DOI: 10.1016/j.parint.2018.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/09/2017] [Accepted: 02/02/2018] [Indexed: 02/09/2023]
Abstract
We have proven the beneficial effects during acute Toxoplasma gondii infection when mast cells were inhibited by disodium cromoglycate (DSCG). Here we investigated the adjuvant effect of DSCG on the protective efficacy of UV-attenuated T. gondii (UV-Tg) vaccine. Mice were infected with 102Tg alone or infected with 102Tg plus DSCG (Tg + DSCG), immunized with 105 UV-Tg and challenged with 102Tg (UV-Tg + Tg) or immunized with 105 UV-Tg plus DSCG and challenged with 102Tg (UV-Tg + DSCG + Tg). Compared to Tg group, Tg + DSCG, UV-Tg + Tg, and UV-Tg + DSCG + Tg showed significantly prolonged survival times, decreased parasite burdens, reduced liver histopathologies, and increased levels of Th1 and Th2 cytokines and IL-17 in the livers and spleens by using quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR). Compared to UV-Tg + Tg, UV-Tg + DSCG + Tg had significantly longer survival time, lower tissue parasite burden and histopathological score, and higher levels of Th1 and Th2 cytokines and IL-17 in the livers or spleens. Our data suggest that DSCG may play an adjuvant role in the immunization induced by UV-attenuated T. gondii in mice, by promoting cellular immune response against T. gondii challenge.
Collapse
Affiliation(s)
- Xi Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, Guangdong, China
| | - Yifan Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, Guangdong, China
| | - Shiguang Huang
- School of Stomatology, Jinan University, Guangzhou 510632, China.
| | - Fangli Lu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
48
|
|
49
|
Oral hepatitis B vaccine: chitosan or glucan based delivery systems for efficient HBsAg immunization following subcutaneous priming. Int J Pharm 2018; 535:261-271. [PMID: 29133207 DOI: 10.1016/j.ijpharm.2017.11.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 12/18/2022]
|
50
|
Jesus S, Soares E, Borchard G, Borges O. Adjuvant Activity of Poly-ε-caprolactone/Chitosan Nanoparticles Characterized by Mast Cell Activation and IFN-γ and IL-17 Production. Mol Pharm 2017; 15:72-82. [PMID: 29160080 DOI: 10.1021/acs.molpharmaceut.7b00730] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Polymeric nanoparticles (NPs) are extremely attractive vaccine adjuvants, able to promote antigen delivery and in some instances, exert intrinsic immunostimulatory properties that enhance antigen specific humoral and cellular immune responses. The poly-ε-caprolactone (PCL)/chitosan NPs were designed with the aim of being able to combine the properties of the 2 polymers in the preparation of an adjuvant for the hepatitis B surface antigen (HBsAg). This article reports important results of an in vitro mechanistic study and immunization studies with HBsAg associated with different concentrations of the nanoparticles. The results revealed that PCL/chitosan NPs promoted mast cell (MC) activation (β-hexosaminidase release) and that its adjuvant effect is not mediated by the TNF-α secretion. Moreover, we demonstrated that HBsAg loaded PCL/chitosan NPs, administered through the subcutaneous (SC) route, were able to induce higher specific antibody titers without increasing IgE when compared to a commercial vaccine, and that the IgG titers are nanoparticle-dose dependent. The results also revealed the NPs' capability to promote a cellular immune response against HBsAg, characterized by the production of IFN-γ and IL-17. These results demonstrated that PCL/chitosan NPs are a good hepatitis B antigen adjuvant, with direct influence on the intensity and type of the immune response generated.
Collapse
Affiliation(s)
- Sandra Jesus
- Faculty of Pharmacy, University of Coimbra , 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra , 3000-548 Coimbra, Portugal
| | - Edna Soares
- Faculty of Pharmacy, University of Coimbra , 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra , 3000-548 Coimbra, Portugal
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, Unssssiversity of Lausanne , 1211 Geneva, Switzerland
| | - Olga Borges
- Faculty of Pharmacy, University of Coimbra , 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra , 3000-548 Coimbra, Portugal
| |
Collapse
|