1
|
Park J, Pho T, Bhatnagar N, Mai LD, Rodriguez-Otero MR, Pal SS, Le CTT, Jenison SE, Li C, May GA, Arioka M, Kang SM, Champion JA. Multilayer Adjuvanted Influenza Protein Nanoparticles Improve Intranasal Delivery and Antigen-Specific Immunity. ACS NANO 2025; 19:7005-7025. [PMID: 39954231 PMCID: PMC11867023 DOI: 10.1021/acsnano.4c14735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Intranasal vaccination is a desired route for protection against influenza viruses by mucosal and systemic immunity. However, the nasal mucosa impedes the intranasal delivery of vaccines. Here, we formulated layer-by-layer (LBL) influenza vaccine nanoparticles for effective intranasal delivery by coating them with alternating mucoadhesive cationic chitosan and muco-inert anionic CpG adjuvants. The nanoparticle cores were formed by desolvating influenza M2e antigen and coating it with hemagglutinin (HA) antigen via biotin-streptavidin conjugation. LBL modification promoted nasal delivery and interaction with the resident immune cells. Intranasal administration with LBL nanoparticles significantly improved cellular and humoral immune responses against HA and M2e including high IgA titers, a hallmark of potent mucosal immunity and persistence of immune responses. Distinct trends for antigen-specific immune responses were observed for different routes of vaccination. The enhanced immune responses conferred mice protection against the influenza challenge and prominently reduced viral titers, demonstrating the effectiveness of intranasal LBL vaccine nanoparticles.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Thomas Pho
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Bioengineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Noopur Bhatnagar
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Linh D. Mai
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Mariela R. Rodriguez-Otero
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Bioengineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Surya Sekhar Pal
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Chau Thuy Tien Le
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Sarah E. Jenison
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Chenyu Li
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Grace A. May
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Marisa Arioka
- Department
of Chemistry, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Sang-Moo Kang
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Julie A. Champion
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Bioengineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
2
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [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: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
3
|
Bazjert J, Jawor P, Pisarek M, Baran R, Jachymek W, Stefaniak T. Local and systemic humoral immune responses to Histophilus somni recombinant antigens administered intranasally and subcutaneously to dairy calves. Sci Rep 2024; 14:27567. [PMID: 39528575 PMCID: PMC11555116 DOI: 10.1038/s41598-024-78605-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
Bovine respiratory disease (BRD) causes significant economic losses in dairy calves. Induction of an early immune response via parenteral vaccination is complicated by the interference of colostral immunity. In this study, we investigated early immunization against selected conserved bacterial antigens. Calves were vaccinated twice intranasally and then subcutaneously with Histophilus somni recombinant proteins (rOMP40 or rHsp60) mixed with one of two adjuvants: CpG ODN2007 or MPLA. The control group (Con) was treated with PBS. The first immunization was done between 24 and 48 h of life and then twice in two weeks intervals. Blood, nasal, and saliva secretion samples were collected directly before vaccination (S1-S3) and then on 42-44 (S4) and 59-61 (S5) day of life. Antibodies (IgG1/IgG2/IgM/IgA in serum; IgG1/IgA in secretions) against both vaccine antigens were quantified in all samples. Intranasal and subcutaneous vaccinations using the described formulas did not increase antibody reactivity against the tested proteins. The reactivity of serum IgG1, IgM, and IgA anti-rOMP40 antibodies was significantly higher in S1 in all groups than that in the other samplings (p˂0.01). Significant differences in the reactivity of serum anti-rOMP40 antibodies between groups were identified in S1 (IgA reactivity was higher in the CpG vs. MPLA group; p < 0.05), S4 (IgM reactivity was higher in Con vs. CpG group; p < 0.05), and S5 (IgG1 reactivity was higher in MPLA vs. Con group; p < 0.05). The lack of consistent changes in antibodies after immunization (S4 and S5) hinders the drawing of conclusions regarding the effect of immunization on antibody reactivity. In the future, establishing a proper immunization window and adjuvants for nasal vaccines against bacterial pathogens causing BRD in calves remains to be determined.
Collapse
Affiliation(s)
- Joanna Bazjert
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wrocław University of Environmental and Life Sciences, C.K. Norwida 31 Str, Wrocław, 50-375, Poland.
| | - Paulina Jawor
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wrocław University of Environmental and Life Sciences, C.K. Norwida 31 Str, Wrocław, 50-375, Poland
| | - Maciej Pisarek
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wrocław University of Environmental and Life Sciences, C.K. Norwida 31 Str, Wrocław, 50-375, Poland
| | - Rafał Baran
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wrocław University of Environmental and Life Sciences, C.K. Norwida 31 Str, Wrocław, 50-375, Poland
| | - Wojciech Jachymek
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Science, Rudolfa Weigla 12 Str, Wrocław, 53-114, Poland
| | - Tadeusz Stefaniak
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wrocław University of Environmental and Life Sciences, C.K. Norwida 31 Str, Wrocław, 50-375, Poland
| |
Collapse
|
4
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
5
|
Seefeld ML, Templeton EL, Lehtinen JM, Sinclair N, Yadav D, Hartwell BL. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front Immunol 2024; 15:1419527. [PMID: 39286244 PMCID: PMC11403286 DOI: 10.3389/fimmu.2024.1419527] [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: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.
Collapse
Affiliation(s)
- Madison L Seefeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Erin L Templeton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Justin M Lehtinen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Noah Sinclair
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Daman Yadav
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Brittany L Hartwell
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Miller A, Lombardo GP, Guerrera MC, Messina E, Marino S, Pellicanò F, Kotanska M, Pergolizzi S, Alesci A, Lauriano ER. Immunohistochemistry of the nasal cavity-associated lymphoid tissue in the dolphin (Stenella coeruleoalba, Meyen 1833). Microsc Res Tech 2024; 87:2103-2112. [PMID: 38683022 DOI: 10.1002/jemt.24592] [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: 02/17/2024] [Revised: 03/25/2024] [Accepted: 04/20/2024] [Indexed: 05/01/2024]
Abstract
The striped dolphin (Stenella coeruleoalba) is a medium-sized pelagic dolphin with a single external nasal opening (blowhole) located in the rostral and dorsal regions of the skull. The nasal cavity is divided into three sections: the olfactory, respiratory, and vestibular areas. The surface epithelium lining the regio vestibularis is the first tissue in the nose to be directly affected by environmental antigens. Cetaceans have a significant amount of mucosa-associated lymphoid tissue (MALT) located throughout their bodies. The lymphoid tissue found in the nasal mucosa is known as nose- or nasopharynx-associated lymphoid tissue (NALT). NALT has not yet been studied in dolphins, but it has been identified and documented in humans and laboratory rodents. This study utilized toll-like receptor 2 (TLR2), CD4, Langerin/CD207, and inducible nitric oxide synthase to characterize, for the first time, immune cells in the mucosal regio vestibularis of the S. coeruleoalba nasal cavity using confocal microscopy immunofluorescence techniques. The findings revealed scattered immune cells immunoreactive to the tested antibodies, present in both the epithelial tissue lining the nasal cavity vestibulum and the underlying connective tissue. This study enhances our comprehension of the immune system of cetaceans. RESEARCH HIGHLIGHTS: This study provides new insights into NALT in S. coeruleoalba. This research deepens the knowledge of the skin of cetaceans.
Collapse
Affiliation(s)
- Anthea Miller
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell''Annunziata, Messina, Italy
| | - Giorgia Pia Lombardo
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Maria Cristina Guerrera
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell''Annunziata, Messina, Italy
| | - Emmanuele Messina
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Sebastian Marino
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Filippo Pellicanò
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Magdalena Kotanska
- Department of Pharmacological Screening, Jagiellonian University Medical College, Krakow, Poland
| | - Simona Pergolizzi
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessio Alesci
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Eugenia Rita Lauriano
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
7
|
Komatsuzaki K, Kageshima H, Sekino Y, Suzuki Y, Ugajin T, Tamaoka M, Hanazawa R, Hirakawa A, Miyazaki Y. Local nasal immunotherapy with birch pollen-galactomannan conjugate-containing ointment in mice and humans. Allergol Int 2024; 73:290-301. [PMID: 37981502 DOI: 10.1016/j.alit.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Allergen immunotherapy (AIT) is the only disease-modifying treatment for immunoglobulin (Ig) E-mediated allergy. Owing to the high prevalence and early onset of hay fever and pollen-food allergy syndrome (PFAS), a safer and simpler treatment method than conventional AIT is needed. To develop a local nasal immunotherapy using an ointment containing hypoallergenic pollen and assess its efficacy in mice and healthy humans. METHODS Hypoallergenicity was achieved by combining pollen and galactomannan through the Maillard reaction to create birch pollen-galactomannan conjugate (BP-GMC). The binding of galactomannan to Bet v 1 was confirmed using electrophoresis and Western blotting (WB). Binding of specific IgE antibodies to BP-GMC was verified using enzyme-linked immunosorbent assay (ELISA) and basophil activation test (BAT). The localization of BP-GMC absorption was confirmed using a BALB/c mouse model. BP-GMC mixed with white petrolatum was intranasally administered to 10 healthy individuals (active drugs, 8; placebo, 2) for 14 days. RESULTS In electrophoresis and WB, no 17-kDa band was observed. In ELISA and BAT, BP-GMC did not react to specific IgE but was bound to IgA and IgG. In the mouse model, BP-GMC was detected in nasopharyngeal-associated lymphoid tissues. In the active drug group, the salivary-specific IgA level significantly increased on day 15 (p = 0.0299), while the serum-specific IgG level significantly increased on day 85 (p = 0.0006). CONCLUSIONS The BP-GMC ointment rapidly produced antagonistic antibodies against IgE; it is safe and easy to use and might serve as a therapeutic antigen for hay fever and PFAS.
Collapse
Affiliation(s)
- Keiko Komatsuzaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Hiroki Kageshima
- Bio & Healthcare Business Division, Wako Filter Technology Co., Ltd., Ibaraki, Japan
| | - Yuki Sekino
- Bio & Healthcare Business Division, Wako Filter Technology Co., Ltd., Ibaraki, Japan
| | - Yasuhiro Suzuki
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsukasa Ugajin
- Department of Dermatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Meiyo Tamaoka
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryoichi Hanazawa
- Department of Clinical Biostatistics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
8
|
Schülke S, Gilles S, Jirmo AC, Mayer JU. Tissue-specific antigen-presenting cells contribute to distinct phenotypes of allergy. Eur J Immunol 2023; 53:e2249980. [PMID: 36938688 DOI: 10.1002/eji.202249980] [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: 10/08/2022] [Revised: 01/19/2023] [Accepted: 03/13/2023] [Indexed: 03/21/2023]
Abstract
Antigen-presenting cells (APCs) are critical cells bridging innate and adaptive immune responses by taking up, processing, and presenting antigens to naïve T cells. At steady state, APCs thus control both tissue homeostasis and the induction of tolerance. In allergies however, APCs drive a Th2-biased immune response that is directed against otherwise harmless antigens from the environment. The main types of APCs involved in the induction of allergy are dendritic cells, monocytes, and macrophages. However, these cell types can be further divided into local, tissue-specific populations that differ in their phenotype, migratory capacity, T-cell activating potential, and production of effector molecules. Understanding if distinct populations of APCs contribute to either tissue-specific immune tolerance, allergen sensitization, or allergic inflammation will allow us to better understand disease pathology and develop targeted treatment options for different stages of allergic disease. Therefore, this review describes the main characteristics, phenotypes, and effector molecules of the APCs involved in the induction of allergen-specific Th2 responses in affected barrier sites, such as the skin, nose, lung, and gastrointestinal tract. Furthermore, we highlight open questions that remain to be addressed to fully understand the contribution of different APCs to allergic disease.
Collapse
Affiliation(s)
- Stefan Schülke
- Vice President´s Research Group: Molecular Allergology, Paul-Ehrlich-Institut, Langen (Hesse), Germany
| | - Stefanie Gilles
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Adan C Jirmo
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Johannes U Mayer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
9
|
Petrone V, Fanelli M, Giudice M, Toschi N, Conti A, Maracchioni C, Iannetta M, Resta C, Cipriani C, Miele MT, Amati F, Andreoni M, Sarmati L, Rogliani P, Novelli G, Garaci E, Rasi G, Sinibaldi-Vallebona P, Minutolo A, Matteucci C, Balestrieri E, Grelli S. Expression profile of HERVs and inflammatory mediators detected in nasal mucosa as a predictive biomarker of COVID-19 severity. Front Microbiol 2023; 14:1155624. [PMID: 37283924 PMCID: PMC10239953 DOI: 10.3389/fmicb.2023.1155624] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction Our research group and others demonstrated the implication of the human endogenous retroviruses (HERVs) in SARS-CoV-2 infection and their association with disease progression, suggesting HERVs as contributing factors in COVID-19 immunopathology. To identify early predictive biomarkers of the COVID-19 severity, we analyzed the expression of HERVs and inflammatory mediators in SARS-CoV-2-positive and -negative nasopharyngeal/oropharyngeal swabs with respect to biochemical parameters and clinical outcome. Methods Residuals of swab samples (20 SARS-CoV-2-negative and 43 SARS-CoV-2-positive) were collected during the first wave of the pandemic and expression levels of HERVs and inflammatory mediators were analyzed by qRT-Real time PCR. Results The results obtained show that infection with SARS-CoV-2 resulted in a general increase in the expression of HERVs and mediators of the immune response. In particular, SARS-CoV-2 infection is associated with increased expression of HERV-K and HERV-W, IL-1β, IL-6, IL-17, TNF-α, MCP-1, INF-γ, TLR-3, and TLR-7, while lower levels of IL-10, IFN-α, IFN-β, and TLR-4 were found in individuals who underwent hospitalization. Moreover, higher expression of HERV-W, IL-1β, IL-6, IFN-α, and IFN-β reflected the respiratory outcome of patients during hospitalization. Interestingly, a machine learning model was able to classify hospitalized vs not hospitalized patients with good accuracy based on the expression levels of HERV-K, HERV-W, IL-6, TNF-a, TLR-3, TLR-7, and the N gene of SARS-CoV-2. These latest biomarkers also correlated with parameters of coagulation and inflammation. Discussion Overall, the present results suggest HERVs as contributing elements in COVID-19 and early genomic biomarkers to predict COVID-19 severity and disease outcome.
Collapse
Affiliation(s)
- Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Martina Giudice
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, United States
| | - Allegra Conti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Marco Iannetta
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Resta
- Respiratory Medicine Unit, Policlinic of Tor Vergata, Rome, Italy
| | - Chiara Cipriani
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Amati
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Andreoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Loredana Sarmati
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Respiratory Medicine Unit, Policlinic of Tor Vergata, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuromed IRCCS Institute, Pozzilli, IS, Italy
- University of Nevada, Department of Pharmacology, Reno, NV, United States
| | | | - Guido Rasi
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- Virology Unit, Policlinic of Tor Vergata, Rome, Italy
| |
Collapse
|
10
|
Zheng J, Lin J, Yang C, Ma Y, Liu P, Li Y, Yang Q. Characteristics of nasal mucosal barrier in lambs at different developmental stages. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 139:104587. [PMID: 36370908 DOI: 10.1016/j.dci.2022.104587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/04/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
The mucosal barriers of a lamb's nasal cavity are composed of a multi-layer barrier designed to protect against the invasion of harmful microorganisms. However, despite the protective measures, respiratory pathogens still infect the sheep from the nasal cavity. Therefore, our study aimed to investigate the characteristics of lamb's nasal cavity barrier at different developmental stages. For nasal histological characteristics, our study revealed that the conchoidal curvature of the inferior nasal conch and the number of glands significantly increased with lamb development. For nasal mucosal barrier characteristics, physical and immune barriers were carefully explored. Initially, we observed that the thickness and proliferative capacity of nasal epithelial significantly increased from fetal to 21 days, which then decreased at 60 days. Then, our study showed that the number of goblet cells (GCs) of 21 days old lamb was significantly higher than in other stages of development. Besides, we found that the number of nasal immune cells, such as dendritic cells, CD3+ T cells, IgA+ B cells, and nasal-associated lymphoid tissue (NALT), were all significantly increased not only from the proximal to distal side in the nasal cavity but also with their age. Totally, our study revealed various characteristics of the mucosal barriers of a lamb's nasal cavity, which provide a reference for explaining the susceptibility of respiratory tract infection in lambs.
Collapse
Affiliation(s)
- Jian Zheng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Jian Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Chengjie Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Yichao Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Peng Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Yucheng Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| |
Collapse
|
11
|
García-Silva I, Govea-Alonso DO, Rosales-Mendoza S. Current status of mucosal vaccines against SARS-CoV2: a hope for protective immunity. Expert Opin Biol Ther 2023; 23:207-222. [PMID: 36594264 DOI: 10.1080/14712598.2022.2156284] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The current vaccines used to fight against COVID-19 are effective, however the induction of protective immunity is a pending goal required to prevent viral transmission, prevent the generation of new variants, and ultimately eradicate SARS-CoV-2. Mucosal immunization stands as a promising approach to achieve protective immunity against SARS-CoV-2; therefore, it is imperative to innovate the current vaccines by developing mucosal candidates, focusing not only on their ability to prevent severe COVID-19 but to neutralize the virus before invasion of the respiratory system and other mucosal compartments. AREAS COVERED This review covers the current advances on the development of anti-COVID-19 mucosal vaccines. Biomedical literature, including PubMed and clinicaltrials.gov website, was analyzed to identify the state of the art for this field. The achievements in preclinical and clinical evaluations are presented and critically analyzed. EXPERT OPINION There is a significant advance on the development of mucosal vaccines against SARSCoV-2, which is a promise to increase the efficacy of immunization against this pathogen. Both preclinical and clinical evaluation for several candidates have been performed. The challenges in this road (e.g. low immunogenicity, a reduced number of adjuvants available, and inaccurate dosage) are identified and also critical perspectives for the field are provided.
Collapse
Affiliation(s)
- Ileana García-Silva
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| | - Dania O Govea-Alonso
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| |
Collapse
|
12
|
Lee J, Khang D. Mucosal delivery of nanovaccine strategy against COVID-19 and its variants. Acta Pharm Sin B 2022; 13:S2211-3835(22)00489-0. [PMID: 36438851 PMCID: PMC9676163 DOI: 10.1016/j.apsb.2022.11.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Despite the global administration of approved COVID-19 vaccines (e.g., ChAdOx1 nCoV-19®, mRNA-1273®, BNT162b2®), the number of infections and fatalities continue to rise at an alarming rate because of the new variants such as Omicron and its subvariants. Including COVID-19 vaccines that are licensed for human use, most of the vaccines that are currently in clinical trials are administered via parenteral route. However, it has been proven that the parenteral vaccines do not induce localized immunity in the upper respiratory mucosal surface, and administration of the currently approved vaccines does not necessarily lead to sterilizing immunity. This further supports the necessity of a mucosal vaccine that blocks the main entrance route of COVID-19: nasal and oral mucosal surfaces. Understanding the mechanism of immune regulation of M cells and dendritic cells and targeting them can be another promising approach for the successful stimulation of the mucosal immune system. This paper reviews the basic mechanisms of the mucosal immunity elicited by mucosal vaccines and summarizes the practical aspects and challenges of nanotechnology-based vaccine platform development, as well as ligand hybrid nanoparticles as potentially effective target delivery agents for mucosal vaccines.
Collapse
Affiliation(s)
- Junwoo Lee
- College of Medicine, Gachon University, Incheon 21999, South Korea
| | - Dongwoo Khang
- College of Medicine, Gachon University, Incheon 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
- Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon 21999, South Korea
- Department of Physiology, College of Medicine, Gachon University, Incheon 21999, South Korea
| |
Collapse
|
13
|
Ramos-Benitez MJ, Strich JR, Alehashemi S, Stein S, Rastegar A, de Jesus AA, Bhuyan F, Ramelli S, Babyak A, Perez-Valencia L, Vannella KM, Grubbs G, Khurana S, Gross R, Hadley K, Liang J, Mazur S, Postnikova E, Warner S, Holbrook MR, Busch LM, Warner B, Applefeld W, Warner S, Kadri SS, Davey RT, Goldbach-Mansky R, Chertow DS. Antiviral innate immunity is diminished in the upper respiratory tract of severe COVID-19 patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.11.08.22281846. [PMID: 36415460 PMCID: PMC9681051 DOI: 10.1101/2022.11.08.22281846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Understanding early innate immune responses to coronavirus disease 2019 (COVID-19) is crucial to developing targeted therapies to mitigate disease severity. Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 infection elicits interferon expression leading to transcription of IFN-stimulated genes (ISGs) to control viral replication and spread. SARS-CoV-2 infection also elicits NF-κB signaling which regulates inflammatory cytokine expression contributing to viral control and likely disease severity. Few studies have simultaneously characterized these two components of innate immunity to COVID-19. We designed a study to characterize the expression of interferon alpha-2 (IFNA2) and interferon beta-1 (IFNB1), both type-1 interferons (IFN-1), interferon-gamma (IFNG), a type-2 interferon (IFN-2), ISGs, and NF-κB response genes in the upper respiratory tract (URT) of patients with mild (outpatient) versus severe (hospitalized) COVID-19. Further, we characterized the weekly dynamics of these responses in the upper and lower respiratory tracts (LRTs) and blood of severe patients to evaluate for compartmental differences. We observed significantly increased ISG and NF-κB responses in the URT of mild compared with severe patients early during illness. This pattern was associated with increased IFNA2 and IFNG expression in the URT of mild patients, a trend toward increased IFNB1-expression and significantly increased STING/IRF3/cGAS expression in the URT of severe patients. Our by-week across-compartment analysis in severe patients revealed significantly higher ISG responses in the blood compared with the URT and LRT of these patients during the first week of illness, despite significantly lower expression of IFNA2, IFNB1, and IFNG in blood. NF-κB responses, however, were significantly elevated in the LRT compared with the URT and blood of severe patients during peak illness (week 2). Our data support that severe COVID-19 is associated with impaired interferon signaling in the URT during early illness and robust pro-inflammatory responses in the LRT during peak illness.
Collapse
Affiliation(s)
- Marcos J. Ramos-Benitez
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, Bethesda, MD 20892
- Ponce Health Science University and Ponce Research Institute, Department of Basic Sciences, School of Medicine, Ponce, Puerto Rico, USA
| | - Jeffrey R. Strich
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
- The United States Public Health Service Commissioned Corps, Rockville, MD, USA
| | - Sara Alehashemi
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy, and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Sydney Stein
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Andre Rastegar
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy, and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Adriana Almeida de Jesus
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy, and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Farzana Bhuyan
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy, and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Sabrina Ramelli
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Ashley Babyak
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Luis Perez-Valencia
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Kevin M. Vannella
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD
| | - Robin Gross
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD
| | - Kyra Hadley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD
| | - Janie Liang
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD
| | - Steven Mazur
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD
| | - Elena Postnikova
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD
| | - Seth Warner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Michael R. Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD
| | - Lindsay M. Busch
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Blake Warner
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Willard Applefeld
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Sarah Warner
- The United States Public Health Service Commissioned Corps, Rockville, MD, USA
| | - Sameer S Kadri
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Richard T Davey
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy, and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Daniel S. Chertow
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
- The United States Public Health Service Commissioned Corps, Rockville, MD, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
14
|
Qin T, Chen Y, Huangfu D, Yin Y, Miao X, Yin Y, Chen S, Peng D, Liu X. PA-X protein assists H9N2 subtype avian influenza virus in escaping immune response of mucosal dendritic cells. Transbound Emerg Dis 2022; 69:e3088-e3100. [PMID: 35855630 DOI: 10.1111/tbed.14665] [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: 04/15/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 11/27/2022]
Abstract
H9N2 subtype low pathogenicity avian influenza virus (AIV) poses a potential zoonotic risk. PA-X, a novel protein generated by PA gene ribosomal frameshift, is considered to be the virulence factor of H9N2 subtype AIVs. Our study found that rTX possessing PA-X protein enhanced the mammalian pathogenicity of H9N2 subtype AIVs compared with PA-X-deficient virus (rTX-FS). Furthermore, PA-X protein inhibited H9N2 subtype AIVs to infect dendritic cells (DCs), but not nonimmune cells (MDCK cells). Meanwhile, PA-X protein suppressed the phenotypic expression (CD80, CD86, CD40 and MHCII), early activation marker (CD69) and pro-inflammatory cytokines (IL-6 and TNF-α), whereas increased anti-inflammatory cytokine (IL-10) in DCs. After intranasally viral infection in mice, we found that PA-X protein of H9N2 subtype AIVs reduced CD11b+ and CD103+ subtype mucosal DCs recruitment to the nasal submucosa by inhibiting CCL20 expression. Moreover, PA-X protein abolished the migratory ability of CD11b+ and CD103+ DCs into draining cervical lymph nodes by down-regulating CCR7 expression. The rTX-infected DCs significantly impaired the allogeneic CD4+ T cell proliferation, suggesting PA-X protein suppressed the immune functions of DCs for hindering the downstream immune activation. These findings indicated that PA-X protein assisted H9N2 subtype AIVs in escaping immune response of mucosal DCs for enhancing the pathogenicity.
Collapse
Affiliation(s)
- Tao Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu, China
| | - Yulian Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Dandan Huangfu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yinyan Yin
- School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xinyu Miao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuncong Yin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu, China
| | - Sujuan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu, China
| | - Daxin Peng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu, China
| | - Xiufan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
15
|
Kanjanawasee D, Tantilipikorn P. LNIT-Local nasal immunotherapy in allergic rhinitis: revisited evidence and perspectives. Curr Opin Allergy Clin Immunol 2022; 22:259-267. [PMID: 35779069 DOI: 10.1097/aci.0000000000000830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Allergen immunotherapy (AIT) is a personalized treatment approach for the allergic airway disease. The most common routes of administration are subcutaneous and sublingual. Local nasal immunotherapy (LNIT) presents another alternative route for allergen desensitization. Nasal mucosa is the first entry site of pathogens and numerous lymphoid organs are located in this area, making LNIT a favorable method for triggering immune tolerance. LNIT has shown promising results in reducing symptoms and medication use in allergic rhinitis patients. Over time, difficulties in dosing adjustments have made this method less popular. Recent advances in intranasal drug delivery systems warrant re-examination of LNIT as a viable option for the treatment of the allergic airway disease. RECENT FINDINGS The scope of the review includes evidences of LNIT in human trials including comparison with placebo and conventional method of immunotherapy. Recent articles regarding the mechanism of LNIT and the challenges of intranasal drug delivery are reviewed. Advances in the LNIT delivery system which have overcome previous limitations demonstrate promising effects. SUMMARY LNIT presents a judicious alternative for noninjection AIT. The evidences from previous clinical trials and the novel improvement of drug delivery system will lead into the future allergen vaccine production.
Collapse
Affiliation(s)
- Dichapong Kanjanawasee
- Center of Research Excellence in Allergy and Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University
- Biodesign Innovation Center, Department of Parasitology
| | - Pongsakorn Tantilipikorn
- Center of Research Excellence in Allergy and Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University
- Division of Rhinology and Allergy, Department of Otorhinolaryngology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
16
|
Cai L, Xu H, Cui Z. Factors Limiting the Translatability of Rodent Model-Based Intranasal Vaccine Research to Humans. AAPS PharmSciTech 2022; 23:191. [PMID: 35819736 PMCID: PMC9274968 DOI: 10.1208/s12249-022-02330-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022] Open
Abstract
The intranasal route of vaccination presents an attractive alternative to parenteral routes and offers numerous advantages, such as the induction of both mucosal and systemic immunity, needle-free delivery, and increased patient compliance. Despite demonstrating promising results in preclinical studies, however, few intranasal vaccine candidates progress beyond early clinical trials. This discrepancy likely stems in part from the limited predictive value of rodent models, which are used frequently in intranasal vaccine research. In this review, we explored the factors that limit the translatability of rodent-based intranasal vaccine research to humans, focusing on the differences in anatomy, immunology, and disease pathology between rodents and humans. We also discussed approaches that minimize these differences and examined alternative animal models that would produce more clinically relevant research.
Collapse
Affiliation(s)
- Lucy Cai
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, USA
| | - Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, 2409 University Ave., A1900, Austin, Texas, 78712, USA
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, 2409 University Ave., A1900, Austin, Texas, 78712, USA.
| |
Collapse
|
17
|
Kasemsuk N, Ngaotepprutaram P, Kanjanawasee D, Suwanwech T, Durham SR, Canonica GW, Tantilipikorn P. Local nasal immunotherapy for allergic rhinitis: A systematic review and meta-analysis. Int Forum Allergy Rhinol 2022; 12:1503-1516. [PMID: 35543418 DOI: 10.1002/alr.23011] [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: 01/19/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Local nasal immunotherapy (LNIT), an alternative noninjection immunotherapy method, is theoretically an efficient method for inducing immunotolerance directly in the affected organ. LNIT is more convenient and less invasive than injection immunotherapy, with fewer systemic reactions. The development of adjuvants to overcome LNIT's limitations raises the possibility of it being an alternative allergen immunotherapy. OBJECTIVES To evaluate the clinical and immunological efficacy and safety of LNIT for patients with allergic rhinitis. METHODS A systematic search for randomized controlled trials comparing LNIT and placebo was performed using OVID Medline and Embase. Outcomes were total nasal symptom score (TNSS), symptom-medication score (SMS), medication score, immunological assessment, and nasal provocation threshold. Data were pooled for meta-analysis. RESULTS A total of 20 studies with 698 participants were included. The LNIT group had greater posttreatment improvement in TNSS, SMS, and medication score than control (TNSS: standardized mean difference [SMD], -1.37 [95% confidence interval [CI], -2.04 to -0.69]; SMS: SMD, -1.55 [95% CI, -2.83 to -0.28]; and medication score: SMD, -1.09 [95% CI, -1.35 to -0.83]). Immunological assessments showed no significant differences in serum-specific IgE (mean difference [MD], 6.35; 95% CI, -4.62 to 17.31), nasal IgE (MD, -0.59; 95% CI, -1.99 to 0.81), or nasal eosinophil cationic protein (MD, 7.63; 95% CI, -18.65 to 33.91). Only serum IgG significantly increased with LNIT (MD, 0.45; 95% CI, 0.20, 0.70). Posttreatment, nasal provocation threshold was higher with LNIT (MD, 27.30; 95% CI, 10.13-44.46). No significant adverse events were reported. CONCLUSIONS LNIT is a safe alternative allergen immunotherapy route without significant adverse events. It improves clinical symptoms, reduces medication usage, and increases the nasal provocation threshold.
Collapse
Affiliation(s)
- Navarat Kasemsuk
- Faculty of Medicine Siriraj Hospital, Division of Rhinology and Allergy, Department of Otorhinolaryngology, Mahidol University, Bangkok, Thailand.,Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Allergy and Immunology, Mahidol University, Bangkok, Thailand
| | - Premyot Ngaotepprutaram
- Faculty of Medicine Siriraj Hospital, Division of Rhinology and Allergy, Department of Otorhinolaryngology, Mahidol University, Bangkok, Thailand.,Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Allergy and Immunology, Mahidol University, Bangkok, Thailand
| | - Dichapong Kanjanawasee
- Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Allergy and Immunology, Mahidol University, Bangkok, Thailand.,Faculty of Medicine Siriraj Hospital, Biodesign Innovation Center, Department of Parasitology, Mahidol University, Bangkok, Thailand
| | - Triphoom Suwanwech
- Faculty of Medicine Siriraj Hospital, Division of Rhinology and Allergy, Department of Otorhinolaryngology, Mahidol University, Bangkok, Thailand.,Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Allergy and Immunology, Mahidol University, Bangkok, Thailand
| | - Stephen R Durham
- Allergy and Clinical Immunology, Imperial College London, London, UK
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Asthma & Allergy Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Pongsakorn Tantilipikorn
- Faculty of Medicine Siriraj Hospital, Division of Rhinology and Allergy, Department of Otorhinolaryngology, Mahidol University, Bangkok, Thailand.,Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Allergy and Immunology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
18
|
Xu H, Cai L, Hufnagel S, Cui Z. Intranasal vaccine: Factors to consider in research and development. Int J Pharm 2021; 609:121180. [PMID: 34637935 DOI: 10.1016/j.ijpharm.2021.121180] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
Most existing vaccines for human use are administered by needle-based injection. Administering vaccines needle-free intranasally has numerous advantages over by needle-based injection, but there are only a few intranasal vaccines that are currently approved for human use, and all of them are live attenuated influenza virus vaccines. Clearly, there are immunological as well as non-immunological challenges that prevent vaccine developers from choosing the intranasal route of administration. We reviewed current approved intranasal vaccines and pipelines and described the target of intranasal vaccines, i.e. nose and lymphoid tissues in the nasal cavity. We then analyzed factors unique to intranasal vaccines that need to be considered when researching and developing new intranasal vaccines. We concluded that while the choice of vaccine formulations, mucoadhesives, mucosal and epithelial permeation enhancers, and ligands that target M-cells are important, safe and effective intranasal mucosal vaccine adjuvants are needed to successfully develop an intranasal vaccine that is not based on live-attenuated viruses or bacteria. Moreover, more effective intranasal vaccine application devices that can efficiently target a vaccine to lymphoid tissues in the nasal cavity as well as preclinical animal models that can better predict intranasal vaccine performance in clinical trials are needed to increase the success rate of intranasal vaccines in clinical trials.
Collapse
Affiliation(s)
- Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Lucy Cai
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephanie Hufnagel
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States.
| |
Collapse
|
19
|
Hsieh MS, Hsu CW, Tu LL, Chai KM, Yu LL, Wu CC, Chen MY, Chiang CY, Liu SJ, Liao CL, Chen HW. Intranasal Vaccination With Recombinant Antigen-FLIPr Fusion Protein Alone Induces Long-Lasting Systemic Antibody Responses and Broad T Cell Responses. Front Immunol 2021; 12:751883. [PMID: 34707615 PMCID: PMC8543008 DOI: 10.3389/fimmu.2021.751883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022] Open
Abstract
A simple formulation is urgently needed for mucosal vaccine development. We employed formyl peptide receptor-like 1 inhibitory protein (FLIPr), an FcγR antagonist secreted by Staphylococcus aureus, as a vector to target ovalbumin (OVA) to dendritic cells (DCs) via intranasal administration. Our results demonstrate that intranasal administration of recombinant OVA-FLIPr fusion protein (rOVA-FLIPr) alone efficiently delivers OVA to DCs in nasal lymphoid tissue. Subsequently, OVA-specific IgG and IgA antibodies in the circulatory system and IgA antibodies in mucosal tissue were detected. Importantly, activation of OVA-specific CD4+ and CD8+ T cells and induction of a broad-spectrum cytokine secretion profile were detected after intranasal administration of rOVA-FLIPr alone in immunocompetent C57BL/6 mice. Furthermore, we employed immunodeficient AG129 mice as a Zika virus infection model and demonstrated that intranasal administration of recombinant Zika virus envelope protein domain III-FLIPr fusion protein induced protective immune responses against the Zika virus. These results suggest that antigen-FLIPr fusion protein alone via intranasal administration can be applied to mucosal vaccine development.
Collapse
Affiliation(s)
- Ming-Shu Hsieh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Wei Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ling-Ling Tu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Lu Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chiao-Chieh Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Mei-Yu Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
20
|
McGrath JJC, Thayaparan D, Cass SP, Mapletoft JP, Zeng PYF, Koenig JFE, Fantauzzi MF, Bagri P, Ly B, Heo R, Schenck LP, Shen P, Miller MS, Stämpfli MR. Cigarette smoke exposure attenuates the induction of antigen-specific IgA in the murine upper respiratory tract. Mucosal Immunol 2021; 14:1067-1076. [PMID: 34108594 DOI: 10.1038/s41385-021-00411-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 03/15/2021] [Accepted: 04/27/2021] [Indexed: 02/04/2023]
Abstract
The upper respiratory tract is highly exposed to airborne pathogens and serves as an important inductive site for protective antibody responses, including mucosal IgA and systemic IgG. However, it is currently unknown to what extent inhaled environmental toxins, such as a cigarette smoke, affect the ability to induce antibody-mediated immunity at this site. Using a murine model of intranasal lipopolysaccharide and ovalbumin (LPS/OVA) immunization, we show that cigarette smoke exposure compromises the induction of antigen-specific IgA in the upper airways and systemic circulation. Deficits in OVA-IgA were observed in conjunction with a reduced accumulation of OVA-specific IgA antibody-secreting cells (ASCs) in the nasal mucosa, inductive tissues (NALT, cervical lymph nodes, spleen) and the blood. Nasal OVA-IgA from smoke-exposed mice also demonstrated reduced avidity during the acute post-immunization period in association with an enhanced mutational burden in the cognate nasal Igha repertoire. Mechanistically, smoke exposure attenuated the ability of the nasal mucosa to upregulate VCAM-1 and pIgR, suggesting that cigarette smoke may inhibit both nasal ASC homing and IgA transepithelial transport. Overall, these findings demonstrate the immunosuppressive nature of tobacco smoke and illustrate the diversity of mechanisms through which this noxious stimulus can interfere with IgA-mediated immunity in the upper airways.
Collapse
Affiliation(s)
- Joshua J C McGrath
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Danya Thayaparan
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Steven P Cass
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Jonathan P Mapletoft
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Peter Y F Zeng
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Joshua F E Koenig
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Matthew F Fantauzzi
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Puja Bagri
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Bruce Ly
- Biomedical Discovery & Commercialization Program, McMaster University, Hamilton, ON, Canada
| | - Rachel Heo
- Health Sciences Undergraduate Program, McMaster University, Hamilton, ON, Canada
| | - L Patrick Schenck
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Biochemistry Graduate Program, McMaster University, Hamilton, ON, Canada.,Weston Family Foundation, Toronto, ON, Canada
| | - Pamela Shen
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Merck & Co., Inc., West Point, PA, USA
| | - Matthew S Miller
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Martin R Stämpfli
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada. .,Department of Medicine, McMaster University, Hamilton, ON, Canada. .,Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada. .,State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, China. .,CSL Biologics Research Center, Bern, Switzerland.
| |
Collapse
|
21
|
de Souza JNR, de Castro FDOF, de Souza CL, El Cheikh MR, Ramos HVL, da Fonseca SG, Costa CC. Is There a Difference between the Preoperative and Postoperative Serum Levels of Interleukin-6 and Tumor Necrosis Factor-α in Children Submitted to Adenotonsillectomy? Int Arch Otorhinolaryngol 2021; 26:e208-e212. [PMID: 35602273 PMCID: PMC9122772 DOI: 10.1055/s-0041-1730301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/20/2020] [Indexed: 11/04/2022] Open
Abstract
Introduction
Palatine and pharyngeal tonsils are the first line of defense against pathogens. Clinically, two alterations may require surgical removal of the tonsils: hypertrophy and recurrent tonsillitis. The two conditions probably result from a dysfunction of the immune system.
Objective
To evaluate possible differences in the plasma levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-10 (IL-10) in patients submitted to adenotonsillectomy.
Methods
Prospective, longitudinal study with 25 children undergoing adenotonsillectomy separated into 3 different groups: recurrent tonsillitis (RT), composed of 7 patients; recurrent hypertrophy tonsillitis (RTTH), with 8 patients; and the tonsillar hypertrophy (TH) group, with 10 patients. Ten healthy control children (SD) were also included in the study. Peripheral blood was collected, and plasma was separated to measure the levels of TNF-α, IL-6, and IL-10. The Mann-Whitney test was used for statistical analysis.
Results
The plasma level of IL-6 was higher in the RT (
p
= 0.0394) and TH (
p
= 0.0009) groups, compared with the control group. The TH group also had higher levels of IL-6 than the RT group (
p
= 0.039). The IL-6/IL-10 ratio was higher in the RT (
p
= 0.029) and TH (
p
= 0.0005) groups compared with the control group. Between the RT and RTTH groups, the IL-6/IL-10 ratio was higher in the RT group, with a statistically significant difference (
p
= 0.0091).
Conclusion
Patients with a history of chronic tonsillitis had higher levels of IL-6, compared with the control group.
Collapse
Affiliation(s)
| | | | - Camila Lemes de Souza
- Institute of Tropical Pathology and Public Health, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Hugo Valter Lisboa Ramos
- Otorhinolaryngology, Centro de Reabilitação e Readaptação Dr. Henrique Santilo (CRER), Goiânia, GO, Brazil
| | | | - Claudiney Candido Costa
- Otorhinolaryngology, Centro de Reabilitação e Readaptação Dr. Henrique Santilo (CRER), Goiânia, GO, Brazil
| |
Collapse
|
22
|
Zhao K, Sun B, Shi C, Sun Y, Jin Z, Hu G. Intranasal immunization with O-2'-Hydroxypropyl trimethyl ammonium chloride chitosan nanoparticles loaded with Newcastle disease virus DNA vaccine enhances mucosal immune response in chickens. J Nanobiotechnology 2021; 19:240. [PMID: 34380522 PMCID: PMC8359106 DOI: 10.1186/s12951-021-00983-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/01/2021] [Indexed: 11/16/2022] Open
Abstract
Background There has been a great interest in developing strategies for enhancing antigen delivery to the mucosal immune system as well as identifying mucosal active immunostimulating agents. To elevate the potential of O-2ʹ-Hydroxypropyl trimethyl ammonium chloride chitosan (O-2ʹ-HACC) as an adjuvant and mucosal immune delivery carrier for DNA vaccine, we prepared the O-2ʹ-HACC loaded with Newcastle disease virus (NDV) F gene plasmid DNA and C3d6 molecular adjuvant (O-2ʹ-HACC/pFDNA microparticles). Results The O-2ʹ-HACC/pFDNA exhibited a regular spherical morphology with a particle size of 202.3 ± 0.52 nm, a zeta potential of 50.8 ± 8.21 mV, encapsulation efficiency of 90.74 ± 1.10%, and a loading capacity of 49.84 ± 1.20%. The plasmid DNA could be sustainably released from the O-2ʹ-HACC/pFDNA after an initial burst release. Intranasal vaccination of chickens immunized with O-2ʹ-HACC/pFDNA not only induced higher anti-NDV IgG and sIgA antibody titers but also significantly promoted lymphocyte proliferation and produced higher levels of IL-2, IL-4, IFN-γ, CD4+, and CD8 + T lymphocytes compared with the NDV commercial live attenuated vaccine. Intranasal delivery of the O-2ʹ-HACC/pFDNA enhanced humoral, cellular, and mucosal immune responses and protected chickens from the infection of highly virulent NDV compared with the intramuscular delivery. Conclusions Collectively, our findings indicated that the O-2ʹ-HACC could be used as a vaccine adjuvant and delivery system for mucosal immunity and have an immense application promise. Graphic Abstract ![]()
Collapse
Affiliation(s)
- Kai Zhao
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, 318000, China. .,Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China.
| | - Beini Sun
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China
| | - Ci Shi
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China
| | - Yanwei Sun
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China
| | - Zheng Jin
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, 318000, China.,Key Laboratory of Chemical Engineering Process and Technology for High-Efficiency Conversion, College of Chemistry and Material Sciences, Heilongjiang University, Harbin, 150080, China
| | - Gaowei Hu
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, 318000, China
| |
Collapse
|
23
|
Raineri EJM, Altulea D, van Dijl JM. Staphylococcal trafficking and infection - from 'nose to gut' and back. FEMS Microbiol Rev 2021; 46:6321165. [PMID: 34259843 PMCID: PMC8767451 DOI: 10.1093/femsre/fuab041] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/11/2021] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus is an opportunistic human pathogen, which is a leading cause of infections worldwide. The challenge in treating S. aureus infection is linked to the development of multidrug-resistant strains and the mechanisms employed by this pathogen to evade the human immune defenses. In addition, S. aureus can hide asymptomatically in particular ‘protective’ niches of the human body for prolonged periods of time. In the present review, we highlight recently gained insights in the role of the human gut as an endogenous S. aureus reservoir next to the nasopharynx and oral cavity. In addition, we address the contribution of these ecological niches to staphylococcal transmission, including the roles of particular triggers as modulators of the bacterial dissemination. In this context, we present recent advances concerning the interactions between S. aureus and immune cells to understand their possible roles as vehicles of dissemination from the gut to other body sites. Lastly, we discuss the factors that contribute to the switch from colonization to infection. Altogether, we conclude that an important key to uncovering the pathogenesis of S. aureus infection lies hidden in the endogenous staphylococcal reservoirs, the trafficking of this bacterium through the human body and the subsequent immune responses.
Collapse
Affiliation(s)
- Elisa J M Raineri
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dania Altulea
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
24
|
Galletti JG, de Paiva CS. Age-related changes in ocular mucosal tolerance: Lessons learned from gut and respiratory tract immunity. Immunology 2021; 164:43-56. [PMID: 33837534 DOI: 10.1111/imm.13338] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/28/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
The ocular surface is the part of the visual system directly exposed to the environment, and it comprises the cornea, the first refractive tissue layer and its surrounding structures. The ocular surface has evolved to keep the cornea smooth and wet, a prerequisite for proper sight, and also protected. To this aim, the ocular surface is a bona fide mucosal niche with an immune system capable of fighting against dangerous pathogens. However, due to the potential harmful effects of uncontrolled inflammation, the ocular surface has several mechanisms to keep the immune response in check. Specifically, the ocular surface is maintained inflammation-free and functional by a particular form of peripheral tolerance known as mucosal tolerance, markedly different from the immune privilege of intraocular structures. Remarkably, conjunctival tolerance is akin to the oral and respiratory tolerance mechanisms found in the gut and airways, respectively. And also similarly, this form of immunoregulation in the eye is affected by ageing just as it is in the digestive and respiratory tracts. With ageing comes an increased prevalence of immune-based ocular surface disorders, which could be related to an age-related impairment of conjunctival tolerance. The purpose of this review was to summarize the present knowledge of ocular mucosal tolerance and how it is affected by the ageing process in the light of the current literature on mucosal immunoregulation of the gut and airways.
Collapse
Affiliation(s)
- Jeremias G Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (IMEX), National Academy of Medicine/CONICET, Buenos Aires, Argentina
| | - Cintia S de Paiva
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
25
|
Kim B, Lee YE, Yeon JW, Go GY, Byun J, Lee K, Lee HK, Hur JK, Jang M, Kim TH. A novel therapeutic modality using CRISPR-engineered dendritic cells to treat allergies. Biomaterials 2021; 273:120798. [PMID: 33895493 DOI: 10.1016/j.biomaterials.2021.120798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/22/2022]
Abstract
Despite the important roles of dendritic cells (DCs) in airway allergies, current therapeutic strategies such as drugs, allergen immunotherapy and biologics haven't been targeted at them. In this study, we established a promising DC-based therapeutic approach for the alleviation of allergic rhinitis (AR)-associated allergic reactions, using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated targeted gene disruption. RNA sequencing analysis revealed upregulation of vacuolar protein sorting 37 B (VPS37B) in AR-derived DCs, indicating a novel molecular target. Following antigen presentation, VPS37A and VPS37B enabled endocytosis of the mannose receptor, which recognizes the house dust mite (HDM) allergen Der p 1. DCs with targeted disruption of VPS37A/B alleviated Th2 cytokine production when co-cultured in vitro with allogeneic naïve CD4+ T cell from patients with AR. Furthermore, nasal administration of Vps37a/b-disrupted bone marrow DCs to a mouse model of AR resulted in strongly reduced AR-related symptoms. Thus, this novel modality using genetically engineered DCs can provide an effective therapeutic and preventative strategy for allergic diseases.
Collapse
Affiliation(s)
- Byoungjae Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea; Neuroscience Research Institute, Korea University, College of Medicine, Seoul, 02841, Republic of Korea
| | - Young Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul, 02792, Republic of Korea; Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Woo Yeon
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Ga-Yeon Go
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul, 02792, Republic of Korea
| | - Junhyoung Byun
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Kijeong Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Hyomin K Lee
- Department of Medicine, Major in Medical Genetics, Graduate School, Hanyang University, Seoul, 04763, Republic of Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea; Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Mihue Jang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul, 02792, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
26
|
Bernasconi V, Norling K, Gribonika I, Ong LC, Burazerovic S, Parveen N, Schön K, Stensson A, Bally M, Larson G, Höök F, Lycke N. A vaccine combination of lipid nanoparticles and a cholera toxin adjuvant derivative greatly improves lung protection against influenza virus infection. Mucosal Immunol 2021; 14:523-536. [PMID: 32807838 DOI: 10.1038/s41385-020-0334-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 02/04/2023]
Abstract
This is a proof-of-principle study demonstrating that the combination of a cholera toxin derived adjuvant, CTA1-DD, and lipid nanoparticles (LNP) can significantly improve the immunogenicity and protective capacity of an intranasal vaccine. We explored the self-adjuvanted universal influenza vaccine candidate, CTA1-3M2e-DD (FPM2e), linked to LNPs. We found that the combined vector greatly enhanced survival against a highly virulent PR8 strain of influenza virus as compared to when mice were immunized with FPM2e alone. The combined vaccine vector enhanced early endosomal processing and peptide presentation in dendritic cells and upregulated co-stimulation. The augmenting effect was CTA1-enzyme dependent. Whereas systemic anti-M2e antibody and CD4+ T-cell responses were comparable to those of the soluble protein, the local respiratory tract IgA and the specific Th1 and Th17 responses were strongly enhanced. Surprisingly, the lung tissue did not exhibit gross pathology upon recovery from infection and M2e-specific lung resident CD4+ T cells were threefold higher than in FPM2e-immunized mice. This study conveys optimism as to the protective ability of a combination vaccine based on LNPs and various forms of the CTA1-DD adjuvant platform, in general, and, more specifically, an important way forward to develop a universal vaccine against influenza.
Collapse
Affiliation(s)
- Valentina Bernasconi
- Department of Microbiology and Immunology, Institute of Biomedicine, Mucosal Immunobiology and Vaccine Center (MIVAC), University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Karin Norling
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Inta Gribonika
- Department of Microbiology and Immunology, Institute of Biomedicine, Mucosal Immunobiology and Vaccine Center (MIVAC), University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Li Ching Ong
- Department of Microbiology and Immunology, Institute of Biomedicine, Mucosal Immunobiology and Vaccine Center (MIVAC), University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Sabina Burazerovic
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Nagma Parveen
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Karin Schön
- Department of Microbiology and Immunology, Institute of Biomedicine, Mucosal Immunobiology and Vaccine Center (MIVAC), University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Anneli Stensson
- Department of Microbiology and Immunology, Institute of Biomedicine, Mucosal Immunobiology and Vaccine Center (MIVAC), University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Marta Bally
- Department of Clinical Microbiology and Wallenberg Centre for Molecular Medicine, Umeå University, 901 85, Umeå, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Fredrik Höök
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Nils Lycke
- Department of Microbiology and Immunology, Institute of Biomedicine, Mucosal Immunobiology and Vaccine Center (MIVAC), University of Gothenburg, 405 30, Gothenburg, Sweden.
| |
Collapse
|
27
|
Gallo O, Locatello LG, Mazzoni A, Novelli L, Annunziato F. The central role of the nasal microenvironment in the transmission, modulation, and clinical progression of SARS-CoV-2 infection. Mucosal Immunol 2021; 14:305-316. [PMID: 33244161 PMCID: PMC7690066 DOI: 10.1038/s41385-020-00359-2] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023]
Abstract
The novel coronavirus SARS-CoV-2 enters into the human body mainly through the ACE2 + TMPRSS2+ nasal epithelial cells. The initial host response to this pathogen occurs in a peculiar immune microenvironment that, starting from the Nasopharynx-Associated Lymphoid Tissue (NALT) system, is the product of a long evolutionary process that is aimed to first recognize exogenous airborne agents. In the present work, we want to critically review the latest molecular and cellular findings on the mucosal response to SARS-CoV-2 in the nasal cavity and in NALT, and to analyze its impact in the subsequent course of COVID-19. Finally, we want to explore the possibility that the regulation of the systemic inflammatory network against the virus can be modulated starting from the initial phases of the nasal and nasopharyngeal response and this may have several clinical and epidemiological implications starting from a mucosal vaccine development.
Collapse
Affiliation(s)
- Oreste Gallo
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| | - Luca Giovanni Locatello
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luca Novelli
- Department of Pathology, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy,Flow Cytometry and Immunotherapy Diagnostic Center, Careggi University Hospital, Largo Brambilla, 3, 50134, Florence, Italy
| |
Collapse
|
28
|
Zhao Y, Liu Y, Yi F, Zhang J, Xu Z, Liu Y, Tao Y. Type 2 diabetes mellitus impaired nasal immunity and increased the risk of hyposmia in COVID-19 mild pneumonia patients. Int Immunopharmacol 2021; 93:107406. [PMID: 33601246 PMCID: PMC7826056 DOI: 10.1016/j.intimp.2021.107406] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 01/05/2023]
Abstract
In patients with COVID-19, type 2 diabetes mellitus (T2DM) can impair the function of nasal-associated lymphoid tissue (NALT) and result in olfactory dysfunction. Exploring the causative alterations of T2DM within the nasal mucosa and NALT could provide insight into the pathogenic mechanisms and bridge the gap between innate immunity and adaptive immunity for virus clearance. Here, we designed a case-control study to compare the olfactory function (OF) among the groups of normal control (NC), COVID-19 mild pneumonia (MP), and MP patients with T2DM (MPT) after a 6–8 months’ recovery, in which MPT had a higher risk of hyposmia than MP and NC. No significant difference was found between the MP and NC. This elevated risk of hyposmia indicated that T2DM increased COVID-19 susceptibility in the nasal cavity with unknown causations. Therefore, we used the T2DM animal model (db/db mice) to evaluate how T2DM increased COVID-19 associated susceptibilities in the nasal mucosa and lymphoid tissues. Db/db mice demonstrated upregulated microvasculature ACE2 expression and significant alterations in lymphocytes component of NALT. Specifically, db/db mice NALT had increased immune-suppressive TCRγδ+ CD4−CD8− T and decreased immune-effective CD4+/CD8+ TCRβ+ T cells and decreased mucosa-protective CD19+ B cells. These results indicated that T2DM could dampen the first-line defense of nasal immunity, and further mechanic studies of metabolic damage and NALT restoration should be one of the highest importance for COVID-19 healing.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yujie Liu
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Fangzheng Yi
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jun Zhang
- Department of Internal Medicine, University of California at Davis, Davis, 95616, United States
| | - Zhaohui Xu
- Department of Disease Prevention and Control, Xijing 986 Hospital, Fourth Military Medical University, Xi'an 710000, China.
| | - Yehai Liu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Ye Tao
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
29
|
Kim SH, Jang YS. Recent Insights into Cellular Crosstalk in Respiratory and Gastrointestinal Mucosal Immune Systems. Immune Netw 2020; 20:e44. [PMID: 33425429 PMCID: PMC7779865 DOI: 10.4110/in.2020.20.e44] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 02/08/2023] Open
Abstract
The human body is continuously threatened by pathogens, and the immune system must maintain a balance between fighting infection and becoming over-activated. Mucosal surfaces cover several anatomically diverse organs throughout the body, such as the respiratory and gastrointestinal tracts, and are directly exposed to the external environment. Various pathogens invade the body through mucosal surfaces, making the mucosa the frontline of immune defense. The immune systems of various mucosal tissues display distinctive features that reflect the tissues' anatomical and functional characteristics. This review discusses the cellular components that constitute the respiratory and gastrointestinal tracts; in particular, it highlights the complex interactions between epithelial and immune cells to induce Ag-specific immune responses in the lung and gut. This information on mucosal immunity may facilitate understanding of the defense mechanisms against infectious agents that invade mucosal surfaces, such as severe acute respiratory syndrome coronavirus 2, and provide insight into effective vaccine development.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology and The Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Korea
| | - Yong-Suk Jang
- Department of Molecular Biology and The Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Korea.,Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
30
|
Bedford JG, Heinlein M, Garnham AL, Nguyen THO, Loudovaris T, Ge C, Mannering SI, Elliott M, Tangye SG, Kedzierska K, Gray DHD, Heath WR, Wakim LM. Unresponsiveness to inhaled antigen is governed by conventional dendritic cells and overridden during infection by monocytes. Sci Immunol 2020; 5:5/52/eabb5439. [DOI: 10.1126/sciimmunol.abb5439] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022]
Abstract
The nasal-associated lymphoid tissues (NALTs) are mucosal-associated lymphoid organs embedded in the submucosa of the nasal passage. NALTs represent a known site for the deposition of inhaled antigens, but little is known of the mechanisms involved in the induction of immunity within this lymphoid tissue. We find that during the steady state, conventional dendritic cells (cDCs) within the NALTs suppress T cell responses. These cDCs, which are also prevalent within human NALTs (tonsils/adenoids), express a unique transcriptional profile and inhibit T cell proliferation via contact-independent mechanisms that can be diminished by blocking the actions of reactive oxygen species and prostaglandin E2. Although the prevention of unrestrained immune activation to inhaled antigens appears to be the default function of NALT cDCs, inflammation after localized virus infection recruited monocyte-derived DCs (moDCs) to this region, which diluted out the suppressive DC pool, and permitted local T cell priming. Accommodating for inflammation-induced temporal changes in NALT DC composition and function, we developed an intranasal vaccine delivery system that coupled the recruitment of moDCs with the sustained release of antigen into the NALTs, and we were able to substantially improve T cell responses after intranasal immunization. Thus, homeostasis and immunity to inhaled antigens is tuned by inflammatory signals that regulate the balance between conventional and moDC populations within the NALTs.
Collapse
Affiliation(s)
- James G. Bedford
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Melanie Heinlein
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Alexandra L. Garnham
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Thi H. O. Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Tom Loudovaris
- Immunology and Diabetes Unit, St Vincent’s Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Chenghao Ge
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- School of Medicine, Tsinghua University, Beijing, China
| | - Stuart I. Mannering
- Immunology and Diabetes Unit, St Vincent’s Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Michael Elliott
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
- Chris O’Brien Lifehouse Cancer Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia
| | - Stuart G. Tangye
- Immunity & Inflammation Theme, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
- St Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Daniel H. D. Gray
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - William R. Heath
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Linda M. Wakim
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| |
Collapse
|
31
|
Das PK, Salinas I. Fish nasal immunity: From mucosal vaccines to neuroimmunology. FISH & SHELLFISH IMMUNOLOGY 2020; 104:165-171. [PMID: 32497724 DOI: 10.1016/j.fsi.2020.05.076] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 06/11/2023]
Abstract
Like terrestrial vertebrates, bony fishes have a nasopharynx-associated lymphoid tissue (NALT) that protects the host against invading pathogens. Despite nasal immunity being a relatively new field in fish immunology, the investigation of nasal immune systems has already illuminated fundamental aspects of teleost mucosal immune systems as well as neuroimmunology. In this review, we highlight the importance of nasal infections in bony fish and the progress that has been made towards understanding how fish respond locally and systemically to nasal infection or vaccination. We also want to highlight the complex interactions between neurons and immune cells that occur in the olfactory organ during the course of an immune response. We predict that similar neuroimmune interactions govern immune responses at all mucosal tissues in bony fish. Understanding the principles of mucosal immune responses in teleost NALT has therefore revealed important aspects of fish mucosal immunity that are critical for mucosal vaccination in aquaculture.
Collapse
Affiliation(s)
- Pankoj Kumar Das
- University of New Mexico, Department of Biology, Center for Evolutionary and Theoretical Immunology (CETI), Albuquerque, NM, USA
| | - Irene Salinas
- University of New Mexico, Department of Biology, Center for Evolutionary and Theoretical Immunology (CETI), Albuquerque, NM, USA.
| |
Collapse
|
32
|
Abstract
Mucosal surfaces represent important routes of entry into the human body for the majority of pathogens, and they constitute unique sites for targeted vaccine delivery. Nanoparticle-based drug delivery systems are emerging technologies for delivering and improving the efficacy of mucosal vaccines. Recent studies have provided new insights into formulation and delivery aspects of importance for the design of safe and efficacious mucosal subunit vaccines based on nanoparticles. These include novel nanomaterials, their physicochemical properties and formulation approaches, nanoparticle interaction with immune cells in the mucosa, and mucosal immunization and delivery strategies. Here, we present recent progress in the application of nanoparticle-based approaches for mucosal vaccine delivery and discuss future research challenges and opportunities in the field.
Collapse
|
33
|
Sohn M, Na HY, Ryu SH, Choi W, In H, Shin HS, Park JS, Shim D, Shin SJ, Park CG. Two Distinct Subsets Are Identified from the Peritoneal Myeloid Mononuclear Cells Expressing both CD11c and CD115. Immune Netw 2019; 19:e15. [PMID: 31281712 PMCID: PMC6597442 DOI: 10.4110/in.2019.19.e15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/21/2019] [Accepted: 05/06/2019] [Indexed: 12/23/2022] Open
Abstract
To this date, the criteria to distinguish peritoneal macrophages and dendritic cells (DCs) are not clear. Here we delineate the subsets of myeloid mononuclear cells in the mouse peritoneal cavity. Considering phenotypical, functional, and ontogenic features, peritoneal myeloid mononuclear cells are divided into 5 subsets: large peritoneal macrophages (LPMs), small peritoneal macrophages (SPMs), DCs, and 2 MHCII+CD11c+CD115+ subpopulations (i.e., MHCII+CD11c+CD115+CD14−CD206− and MHCII+CD11c+CD115+CD14+CD206+). Among them, 2 subsets of competent Ag presenting cells are demonstrated with distinct functional characteristics, one being DCs and the other being MHCII+CD11c+CD115+CD14−CD206− cells. DCs are able to promote fully activated T cells and superior in expanding cytokine producing inflammatory T cells, whereas MHCII+CD11c+CD115+CD14−CD206− cells generate partially activated T cells and possess a greater ability to induce Treg under TGF-β and retinoic acid conditions. While the development of DCs and MHCII+CD11c+CD115+CD14−CD206− cells are responsive to the treatment of FLT3 ligand and GM-CSF, the number of LPMs, SPMs, and MHCII+CD11c+CD115+CD14+CD206+ cells are only influenced by the injection of GM-CSF. In addition, the analysis of gene expression profiles among MHCII+ peritoneal myeloid mononuclear cells reveals that MHCII+CD11c+CD115+CD14+CD206+ cells share high similarity with SPMs, whereas MHCII+CD11c+CD115+CD14−CD206− cells are related to peritoneal DC2s. Collectively, our study identifies 2 distinct subpopulations of MHCII+CD11c+CD115+ cells, 1) MHCII+CD11c+CD115+CD14−CD206− cells closely related to peritoneal DC2s and 2) MHCII+CD11c+CD115+CD14+CD206+ cells to SPMs.
Collapse
Affiliation(s)
- Moah Sohn
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hye Young Na
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seul Hye Ryu
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Wanho Choi
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hyunju In
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hyun Soo Shin
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji Soo Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Dahee Shim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, Korea.,Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Sung Jae Shin
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea.,Department of Microbiology, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Chae Gyu Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
34
|
A recombinant bovine herpesvirus-4 vectored vaccine delivered via intranasal nebulization elicits viral neutralizing antibody titers in cattle. PLoS One 2019; 14:e0215605. [PMID: 31002724 PMCID: PMC6474629 DOI: 10.1371/journal.pone.0215605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/05/2019] [Indexed: 02/05/2023] Open
Abstract
Recombinant herpesvirus vaccine vectors offer distinct advantages in next-generation vaccine development, primarily due to the ability to establish persistent infections to provide sustainable antigen responses in the host. Recombinant bovine herpesvirus-4 (BoHV-4) has been previously shown to elicit protective immunity in model laboratory animal species against a variety of pathogens. For the first time, we describe the induction of antigen-specific immune responses to two delivered antigens in the host species after intranasal nebulization of recombinant BoHV-4 expressing the chimeric peptide containing the bovine viral diarrhea virus (BVDV) glycoprotein E2 and the bovine herpesvirus 1 (BoHV-1) glycoprotein D (BoHV-4-A-CMV-IgK-gE2gD-TM). In this study, four cattle were immunized via intranasal nebulization with the recombinant BoHV-4 construct. Two of the cattle were previously infected with wild-type BoHV-4, and both developed detectable serologic responses to BVDV and BoHV-1. All four immunized cattle developed detectable viral neutralizing antibody responses to BVDV, and one steer developed a transient viral neutralizing response to BoHV-1. Approximately one year after immunization, immunosuppressive doses of the glucocorticoid dexamethasone were administered intravenously to all four cattle. Within two weeks of immunosuppression, all animals developed viral neutralizing antibody responses to BoHV-1, and all animals maintained BVDV viral neutralizing capacity. Overall, nebulization of BoHV-4-A-CMV-IgK-gE2gD-TM persistently infects cattle, is capable of eliciting antigen-specific immunity following immunization, including in the presence of pre-existing BoHV-4 immunity, and recrudescence of the virus boosts the immune response to BoHV-4-vectored antigens. These results indicate that BoHV-4 is a viable and attractive vaccine delivery platform for use in cattle.
Collapse
|
35
|
Lima SF, Bicalho MLDS, Bicalho RC. The Bos taurus maternal microbiome: Role in determining the progeny early-life upper respiratory tract microbiome and health. PLoS One 2019; 14:e0208014. [PMID: 30840624 PMCID: PMC6402649 DOI: 10.1371/journal.pone.0208014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 11/10/2018] [Indexed: 12/28/2022] Open
Abstract
Natural transference of maternal microbes to the neonate, especially at birth via the vaginal canal, has recently been recognized in humans and cows; however, its microbial influence on calf health has not yet been documented. We compared the bacterial communities in vaginal and fecal samples from 81 pregnant dairy cows versus those in nasopharyngeal and fecal samples collected at 3, 14 and 35 days of life from their respective progeny. The microbiota of the calf upper respiratory tract (URT), regardless of calf age, was found to be highly similar to the maternal vaginal microbiota. Calf fecal microbiota clustered closely to the maternal fecal microbiota, progressing toward an adult-like state over the first 35 days when relative abundances of taxa were considered. Sixty-four, 65 and 87% of the detected OTUs were shared between cow and calf fecal microbiota at days 3, 14 and 35 respectively, whereas 73, 76 and 87% were shared between maternal vaginal microbiome and calf URT microbiota at days 3, 14 and 35, respectively. Bacteroidetes, Ruminococcus, Clostridium, and Blautia were the top four genera identified in maternal and calf fecal samples. Mannheimia, Moraxella, Bacteroides, Streptococcus and Pseudomonas were the top five genera identified in maternal vaginal and calf URT samples. Mannheimia was relatively more abundant in the vaginal microbiota of cows whose progeny were diagnosed with respiratory and middle ear disease. Our results indicate that maternal vaginal microbiota potentially influences the initial bacterial colonization of the calf URT, and that might have an important impact on the health of the calf respiratory tract and middle ear.
Collapse
Affiliation(s)
- Svetlana Ferreira Lima
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Marcela Lucas de Souza Bicalho
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Rodrigo Carvalho Bicalho
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
36
|
van Aalst S, Jansen MAA, Ludwig IS, van der Zee R, van Eden W, Broere F. Routing dependent immune responses after experimental R848-adjuvated vaccination. Vaccine 2018; 36:1405-1413. [PMID: 29409680 DOI: 10.1016/j.vaccine.2018.01.077] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/15/2017] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
Abstract
Most traditional vaccines are administered via the intramuscular route. Other routes of administration however, can induce equal or improved protective memory responses and might provide practical advantages such as needle-free immunization, dose sparing and induction of tissue-specific (mucosal) immunity. Here we explored the differences in immunological outcome after immunization with model antigens via two promising immunization routes (intradermal and intranasal) with or without the experimental adjuvant and TLR7/8-agonist R848. Because the adaptive immune response is largely determined by the local innate cells at the site of immunization, the effect of R848-adjuvation on local cellular recruitment, antigenic uptake by antigen-presenting cells and the initiation of the adaptive response were analyzed for the two routes of administration. We show a general immune-stimulating effect of R848 irrespective of the route of administration. This includes influx of neutrophils, macrophages and dendritic cells to the respective draining lymph nodes and an increase in antigen-positive antigen-presenting cells which leads for both intradermal and intranasal immunization to a mainly TH1 response. Furthermore, both intranasal and intradermal R848-adjuvated immunization induces a local shift in DC subsets; frequencies of CD11b+DC increase whereas CD103+DC decrease in relative abundance in the draining lymph node. In spite of these similarities, the outcome of immune responses differs for the respective immunization routes in both magnitude and cytokine profile. Via the intradermal route, the induced T-cell response is higher compared to that after intranasal immunization, which corresponds with the local higher uptake of antigen by antigen-presenting cells after intradermal immunization. Furthermore, R848-adjuvation enhances ex vivo IL-10 and IL-17 production after intranasal, but not intradermal, T-cell activation. Quite the opposite, intradermal immunization leads to a decrease in IL-10 production by the vaccine induced T-cells. This knowledge may lead to a more rational development of novel adjuvanted vaccines administered via non-traditional routes.
Collapse
Affiliation(s)
- Susan van Aalst
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.
| | - Manon A A Jansen
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.
| | - Irene S Ludwig
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.
| | - Ruurd van der Zee
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.
| | - Femke Broere
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
37
|
Pigni M, Ashok D, Stevanin M, Acha-Orbea H. Establishment and Characterization of a Functionally Competent Type 2 Conventional Dendritic Cell Line. Front Immunol 2018; 9:1912. [PMID: 30197645 PMCID: PMC6117413 DOI: 10.3389/fimmu.2018.01912] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/02/2018] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) are the most potent antigen presenting cells and possess an incomparable ability to activate and instruct T cells, which makes them one of the cornerstones in the regulation of the cross-talk between innate and adaptive immunity. Therefore, a deep understanding of DC biology lays the foundations to describe and to harness the mechanisms that regulate the development of the adaptive response, with clear implications in a vast array of fields such as the study of autoimmune diseases and the development of new vaccines. However, the great difficulty to obtain large quantities of viable non-activated DCs for experimentation have considerably hindered the progress of DC research. Several strategies have been proposed to overcome these limitations by promoting an increase of DC abundance in vivo, by inducing DC development from DC progenitors in vitro and by generating stable DC lines. In the past years, we have described a method to derive immortalized stable DC lines, named MutuDCs, from the spleens of Mushi1 mice, a transgenic mouse strain that express the simian virus 40 Large T-oncogene in the DCs. The comparison of these DC lines with the vast variety of DC subsets described in vivo has shown that all the MutuDC lines that we have generated so far have phenotypic and functional features of type 1 conventional DCs (cDC1s). With the purpose of deriving DC lines with characteristics of type 2 conventional DCs (cDC2s), we bred a new Batf3-/- Mushi1 murine line in which the development of the cDC1 subset is severely defective. The new MutuDC line that we generated from Batf3-/- Mushi1 mice was phenotypically and functionally characterized in this work. Our results demonstrated that all the tested characteristics of this new cell line, including the expression of subset-determining transcription factors, the profile of cytokine production and the ability to present antigens, are comparable with the features of splenic CD4- cDC2s. Therefore, we concluded that our new cell line, that we named CD4- MutuDC2 line, represents a valuable model for the CD4- cDC2 subset.
Collapse
Affiliation(s)
| | | | | | - Hans Acha-Orbea
- Department of Biochemistry CIIL, University of Lausanne, Épalinges, Switzerland
| |
Collapse
|
38
|
Vangeti S, Yu M, Smed-Sörensen A. Respiratory Mononuclear Phagocytes in Human Influenza A Virus Infection: Their Role in Immune Protection and As Targets of the Virus. Front Immunol 2018; 9:1521. [PMID: 30018617 PMCID: PMC6037688 DOI: 10.3389/fimmu.2018.01521] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
Emerging viruses have become increasingly important with recurrent epidemics. Influenza A virus (IAV), a respiratory virus displaying continuous re-emergence, contributes significantly to global morbidity and mortality, especially in young children, immunocompromised, and elderly people. IAV infection is typically confined to the airways and the virus replicates in respiratory epithelial cells but can also infect resident immune cells. Clearance of infection requires virus-specific adaptive immune responses that depend on early and efficient innate immune responses against IAV. Mononuclear phagocytes (MNPs), comprising monocytes, dendritic cells, and macrophages, have common but also unique features. In addition to being professional antigen-presenting cells, MNPs mediate leukocyte recruitment, sense and phagocytose pathogens, regulate inflammation, and shape immune responses. The immune protection mediated by MNPs can be compromised during IAV infection when the cells are also targeted by the virus, leading to impaired cytokine responses and altered interactions with other immune cells. Furthermore, it is becoming increasingly clear that immune cells differ depending on their anatomical location and that it is important to study them where they are expected to exert their function. Defining tissue-resident MNP distribution, phenotype, and function during acute and convalescent human IAV infection can offer valuable insights into understanding how MNPs maintain the fine balance required to protect against infections that the cells are themselves susceptible to. In this review, we delineate the role of MNPs in the human respiratory tract during IAV infection both in mediating immune protection and as targets of the virus.
Collapse
Affiliation(s)
- Sindhu Vangeti
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Meng Yu
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
39
|
Abhyankar MM, Orr MT, Lin S, Suraju MO, Simpson A, Blust M, Pham T, Guderian JA, Tomai MA, Elvecrog J, Pedersen K, Petri WA, Fox CB. Adjuvant composition and delivery route shape immune response quality and protective efficacy of a recombinant vaccine for Entamoeba histolytica. NPJ Vaccines 2018; 3:22. [PMID: 29900011 PMCID: PMC5988657 DOI: 10.1038/s41541-018-0060-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
Amebiasis caused by Entamoeba histolytica is the third leading cause of parasitic mortality globally, with some 100,000 deaths annually, primarily among young children. Protective immunity to amebiasis is associated with fecal IgA and IFN-γ in humans; however, no vaccine exists. We have previously identified recombinant LecA as a potential protective vaccine antigen. Here we describe the development of a stable, manufacturable PEGylated liposomal adjuvant formulation containing two synthetic Toll-like receptor (TLR) ligands: GLA (TLR4) and 3M-052 (TLR7/8). The liposomes stimulated production of monocyte/macrophage chemoattractants MCP-1 and Mip-1β, and Th1-associated cytokines IL-12p70 and IFN-γ from human whole blood dependent on TLR ligand composition and dose. The liposomes also demonstrated acceptable physicochemical compatibility with the recombinant LecA antigen. Whereas mice immunized with LecA and GLA-liposomes demonstrated enhanced antigen-specific fecal IgA titers, mice immunized with LecA and 3M-052-liposomes showed a stronger Th1 immune profile. Liposomes containing GLA and 3M-052 together elicited both LecA-specific fecal IgA and Th1 immune responses. Furthermore, the quality of the immune response could be modulated with modifications to the liposomal formulation based on PEG length. Compared to subcutaneous administration, the optimized liposome adjuvant composition with LecA antigen administered intranasally resulted in significantly enhanced fecal IgA, serum IgG2a, as well as systemic IFN-γ and IL-17A levels in mice. The optimized intranasal regimen provided greater than 80% protection from disease as measured by parasite antigen in the colon. This work demonstrates the physicochemical and immunological characterization of an optimized mucosal adjuvant system containing a combination of TLR ligands with complementary activities and illustrates the importance of adjuvant composition and route of delivery to enhance a multifaceted and protective immune response to amebiasis.
Collapse
Affiliation(s)
- Mayuresh M. Abhyankar
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA USA
| | - Mark T. Orr
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
- Department of Global Health, University of Washington, Seattle, WA USA
| | - Susan Lin
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
| | - Mohammed O. Suraju
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA USA
| | | | | | - Tiep Pham
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
| | | | - Mark A. Tomai
- 3M Drug Delivery Systems, 3M Center, 275-3E-10, St. Paul, MN USA
| | - James Elvecrog
- 3M Drug Delivery Systems, 3M Center, 275-3E-10, St. Paul, MN USA
| | | | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA USA
| | - Christopher B. Fox
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
- Department of Global Health, University of Washington, Seattle, WA USA
| |
Collapse
|
40
|
Takaki H, Ichimiya S, Matsumoto M, Seya T. Mucosal Immune Response in Nasal-Associated Lymphoid Tissue upon Intranasal Administration by Adjuvants. J Innate Immun 2018; 10:515-521. [PMID: 29860261 DOI: 10.1159/000489405] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/12/2018] [Indexed: 12/23/2022] Open
Abstract
The nasal administration of vaccines directed against diseases caused by upper respiratory tract infections of pathogens, such as the influenza virus, mimics the natural infection of pathogens and induces immunoglobulin A (IgA) production in the nasal cavity to effectively protect viral entry. Therefore, the development of a nasally administered vaccine is a research objective. Because the antigenicity of influenza split vaccines is low, nasal inoculation with the vaccine alone does not induce strong IgA production in the nasal cavity. However, the addition of adjuvants activates the innate immune response, enhancing antigen-specific IgA production and the T-cell response. Although the development of suitable adjuvants for nasal vaccinations is in progress, the mechanism by which adjuvants promote the immune response is still unclear. In this review, we discuss the mucosal immune response, especially in the nasal-associated lymphoid tissue, induced in response to the intranasal inoculation of an influenza vaccine and adjuvants in animal models.
Collapse
Affiliation(s)
- Hiromi Takaki
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, .,Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo,
| | - Shingo Ichimiya
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Misako Matsumoto
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tsukasa Seya
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
41
|
Dommaschk A, Lang LF, Maus R, Stolper J, Welte T, Maus UA. Colonization-induced protection against invasive pneumococcal disease in mice is independent of CD103 driven adaptive immune responses. Eur J Immunol 2018; 48:965-974. [PMID: 29543979 DOI: 10.1002/eji.201747236] [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: 07/18/2017] [Revised: 02/22/2018] [Accepted: 03/08/2018] [Indexed: 11/09/2022]
Abstract
Nasopharyngeal colonization with Streptococcus pneumoniae (the pneumococcus) is known to mount protective adaptive immune responses in rodents and humans. However, the cellular response of the nasopharyngeal compartment to pneumococcal colonization and its importance for the ensuing adaptive immune response is only partially defined. Here we show that nasopharyngeal colonization with S. pneumoniae triggered substantial expansion of both integrin αE (CD103) positive dendritic cells (DC) and T lymphocytes in nasopharynx, nasal-associated lymphoid tissue (NALT) and cervical lymph nodes (CLN) of WT mice. However, nasopharyngeal de-colonization and pneumococcus-specific antibody responses were similar between WT and CD103 KO mice or Batf3 KO mice. Also, naïve WT mice passively immunized with antiserum from previously colonized WT and CD103 KO mice were similarly protected against invasive pneumococcal disease (IPD). In summary, the data show that CD103 is dispensable for pneumococcal colonization-induced adaptive immune responses in mice.
Collapse
Affiliation(s)
- Anne Dommaschk
- Department of Experimental Pneumology, Hannover School of Medicine, Hannover, Germany
| | - Lara F Lang
- Department of Experimental Pneumology, Hannover School of Medicine, Hannover, Germany
| | - Regina Maus
- Department of Experimental Pneumology, Hannover School of Medicine, Hannover, Germany
| | - Jennifer Stolper
- Department of Experimental Pneumology, Hannover School of Medicine, Hannover, Germany
| | - Tobias Welte
- Clinic for Pneumology, Hannover School of Medicine, Hannover, Germany.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Ulrich A Maus
- Department of Experimental Pneumology, Hannover School of Medicine, Hannover, Germany.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| |
Collapse
|
42
|
Nurjadi D, Heeg K, Weber ANR, Zanger P. Toll-like receptor 9 (TLR-9) promotor polymorphisms and gene expression are associated with persistent Staphylococcus aureus nasal carriage. Clin Microbiol Infect 2018; 24:1210.e7-1210.e12. [PMID: 29458158 DOI: 10.1016/j.cmi.2018.02.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/21/2017] [Accepted: 02/08/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Toll-like receptor (TLR) 9 could have importance in human Staphylococcus aureus immunity, but population-level evidence for this hypothesis is missing. METHODS We phenotyped S. aureus nasal carriage of 603 volunteers using four consecutive swabs, genotyped TLR9 promotor variants in 106 persistent carriers and 219 noncarriers, measured TLR9-mRNA expression in whole blood after stimulation with viable S. aureus and studied mutual associations of carriage, transcriptional activity and single nucleotide polymorphisms while accounting for sex and hormone contraceptive use (HCU). RESULTS The -1486 (rs187084) and -1237 (rs5743836) CT haplotype was more common in noncarriers (185/438, 42%) than in carriers (63/212, 30%), with the TT haplotype showing a reverse association (noncarriers, 180/438, 41%; carriers 117/212, 55%) (χ2 p 0.001). Mean TLR9 mRNA expression in whole blood was higher in noncarriers (ratiocarriers/noncarriers 0.63; 95% confidence interval, 0.43-0.92; p 0.017). A duplication of TLR9 transcriptional activity lowered the odds of persistent S. aureus carriage by 37% in the overall group (odds ratio = 0.63; 95% confidence interval, 0.42-0.94; p 0.022) and by 54% in women (odds ratio = 0.46; 95% confidence interval, 0.23-0.90; p 0.023). Promotor haplotype and HCU had a combined effect on TLR9 transcription (interaction model): women in the TT (risk) haplotype/HCU- stratum (baseline) had lower mRNA levels than women in the CT (protective) haplotype/HCU- (ratio 1.92; p 0.055), the CT haplotype/HCU+ (ratio 2.02; p 0.032) and the TT haplotype/HCU+ (ratio 2.59; p < 0.004) strata. No such associations were observed in men. CONCLUSIONS We provide evidence that TLR9 affects human S. aureus immunity and present potential explanations for differences according to sex in S. aureus colonization and infection.
Collapse
Affiliation(s)
- D Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospitals, Heidelberg, Germany; Institute of Tropical Medicine, University Hospitals, Tübingen, Germany
| | - K Heeg
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospitals, Heidelberg, Germany
| | - A N R Weber
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard-Karls-Universität, Tübingen, Germany
| | - P Zanger
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospitals, Heidelberg, Germany; Heidelberg Institute of Public Health, University Hospitals, Ruprecht-Karls-Universität, Heidelberg, Germany.
| |
Collapse
|
43
|
Abstract
Advances in CNS immunity and anatomy bridge the CNS and the peripheral immune system. Region-specific antiviral responses alter BBB permeability during viral invasion. CNS barriers have anatomical specializations with tailored defenses against pathogens. Immunocytotherapies for persistent CNS infections can promote non-cytopathic viral clearance.
The central nervous system (CNS) is an immunologically specialized organ where restrictive barrier structures protect the parenchyma from inflammation and infection. This protection is important in preventing damage to non-renewable resident cell populations, such as neurons, responsible for functions ranging from executive to autonomic. Despite these barriers, the CNS can be infected through several entry portals, giving rise to meningitis and encephalitis. Following infection, resident cells recruit peripherally derived immune cells to sites of viral infection. In this review, we discuss recent advances in immune recruitment and entry at barrier structures as well as current immunotherapeutic strategies for the treatment of persistent viral infections.
Collapse
|
44
|
Bankvall M, Jontell M, Wold A, Östman S. Tissue-specific Differences in Immune Cell Subsets Located in the Naso-oropharyngeal-associated Lymphoid Tissues. Scand J Immunol 2017; 87:15-27. [PMID: 29077981 DOI: 10.1111/sji.12625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/21/2017] [Indexed: 01/09/2023]
Abstract
Defining the immune cells within the naso-oropharyngeal-associated lymphoid tissues would promote the development of efficient orally and nasally delivered immunotherapies. The aim was to compare murine antigen-presenting cells (APCs) and T cell subsets in the nose-associated lymphoid tissues (NALT), cervical lymph nodes (CLN), mesenteric lymph nodes (MLN) and peripheral lymph nodes (PLN) using flow cytometry and in vitro proliferation assays. Overall, the NALT contained a higher proportion of APCs and a lower proportion of T cells compared to the CLN, MLN and PLN. The APCs of the NALT more often belonged to the CD11c+ CD11b+ and the CD11cneg CD11b+ subsets as compared to the other sites. Both of these APC populations showed little sign of activation, that is low expression of the markers CD40, CD86 and IAd. Instead, the APCs of the NALT more often co-expressed CX3CR1 and CD206, markers associated with a tolerogenic function. No increase in the proportion of regulatory T cells was observed in the NALT. Instead, the T cells frequently exhibited a memory/effector phenotype, expressing the homing markers α4β7, CCR4 and CCR9, but rarely the naïve phenotype cell surface marker CD45RB. In contrast, the T cells at the other sites were mostly of the naïve phenotype. In addition, cells from the NALT did not proliferate upon in vitro stimulation with Con A, whereas the cells from the other sites did. Taken together, these results suggest that the NALT is primarily an effector site rather than one for activation and differentiation, despite it being regarded as a site of induction.
Collapse
Affiliation(s)
- M Bankvall
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - M Jontell
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - A Wold
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S Östman
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
45
|
Shen Y, Hu W, Wei Y, Feng Z, Yang Q. The immune mechanism of Mycoplasma hyopneumoniae 168 vaccine strain through dendritic cells. BMC Vet Res 2017; 13:285. [PMID: 28915878 PMCID: PMC5603027 DOI: 10.1186/s12917-017-1194-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022] Open
Abstract
Background Mycoplasma hyopneumoniae (Mhp) causes porcine enzootic pneumonia, a disease that cause major economic losses in the pig industry. Dendritic cells (DCs), the most effective antigen-presenting cells, are widely distributed beneath respiratory epithelium, DCs uptake and present antigens to T cells, to initiate protective immune responses in different infections. In this study, we investigated the role of porcine DCs in vaccine Mhp-168 exposure. Results The antigen presenting ability of DCs were improved by vaccine Mhp-168 exposure. DCs could activate T-cell proliferation by up-regulating the antigen presenting molecule MHCII expression and co-stimulatory molecule CD80/86. However, the up-regulation of IL-10 and accompany with down-regulation of IFN-γ gene level may account for the limitation of attenuated Mhp-168 strain use as vaccine alone. Conclusion These findings are benefit for exploring the protection mechanisms and the possible limitations of this attenuated Mhp-168 vaccine.
Collapse
Affiliation(s)
- Yumeng Shen
- Veterinary College, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Weiwei Hu
- Veterinary College, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Yanna Wei
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, People's Republic of China
| | - Zhixin Feng
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, People's Republic of China
| | - Qian Yang
- Veterinary College, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China.
| |
Collapse
|
46
|
Smole U, Schabussova I, Pickl WF, Wiedermann U. Murine models for mucosal tolerance in allergy. Semin Immunol 2017; 30:12-27. [PMID: 28807539 DOI: 10.1016/j.smim.2017.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/21/2017] [Indexed: 02/07/2023]
Abstract
Immunity is established by a fine balance to discriminate between self and non-self. In addition, mucosal surfaces have the unique ability to establish and maintain a state of tolerance also against non-self constituents such as those represented by the large numbers of commensals populating mucosal surfaces and food-derived or air-borne antigens. Recent years have seen a dramatic expansion in our understanding of the basic mechanisms and the involved cellular and molecular players orchestrating mucosal tolerance. As a direct outgrowth, promising prophylactic and therapeutic models for mucosal tolerance induction against usually innocuous antigens (derived from food and aeroallergen sources) have been developed. A major theme in the past years was the introduction of improved formulations and novel adjuvants into such allergy vaccines. This review article describes basic mechanisms of mucosal tolerance induction and contrasts the peculiarities but also the interdependence of the gut and respiratory tract associated lymphoid tissues in that context. Particular emphasis is put on delineating the current prophylactic and therapeutic strategies to study and improve mucosal tolerance induction in allergy.
Collapse
Affiliation(s)
- Ursula Smole
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
47
|
Mulligan JK, O’Connell BP, Pasquini W, Mulligan RM, Smith S, Soler ZM, Atkinson C, Schlosser RJ. Impact of tobacco smoke on upper airway dendritic cell accumulation and regulation by sinonasal epithelial cells. Int Forum Allergy Rhinol 2017; 7:777-785. [PMID: 28574651 PMCID: PMC5544557 DOI: 10.1002/alr.21955] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND In these studies we examined the impact of environmental tobacco smoke (ETS) and active smoking on sinonasal dendritic cell (DC) subsets in controls or patients with chronic rhinosinusitis with nasal polyps (CRSwNP). In subsequent in-vitro investigations, we examined the influence of cigarette smoke extract (CSE) on human sinonasal epithelial cells' (HSNECs) ability to regulate DC functions. METHODS Sinonasal tissue, blood, and hair were collected from patients undergoing sinus surgery. Smoking status and ETS exposure were determined by hair nicotine. DC subsets were examined by flow cytometric analysis. Monocyte-derived dendritic cells (moDCs) were treated with conditioned medium from non-smoked-exposed HSNECs (NS-HSNECs) or cigarette-smoke-extract-exposed HSNECs (CSE-HSNECs) to assess the impact of CSE exposure on HSNEC regulation of moDC functions. RESULTS Control subjects who were active smokers displayed increased sinonasal moDC and myeloid dendritic 1 (mDC1) cells and reduced mDC2 cells, whereas, in CRSwNP patients, only moDC and mDC2 cells were altered. ETS was found to increase only moDCs in the CRSwNP patients. In vitro, CSE stimulated HSNEC secretion of the moDC regulatory products chemokine (C-C motif) ligand 20, prostaglandin E2 , and granulocyte-macrophage colony-stimulating factor. CSE exposure also promoted HSNECs to stimulate monocyte and moDC migration. moDCs treated with CSE-HSNEC media stimulated an increase in antigen uptake and expression of CD80 and CD86. Last, CSE-HSNEC-treated moDCs secreted increased levels of interleukin-10, interferon-γ, and thymic stromal lymphopoietin. CONCLUSION Active smoking, and to a lesser degree ETS, alters the sinonasal composition of DCs. A potential mechanism to account for this is that cigarette smoke stimulates HSNECs to induce moDC migration, maturation, and activation.
Collapse
Affiliation(s)
- Jennifer K. Mulligan
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Brendan P. O’Connell
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Whitney Pasquini
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Ryan M. Mulligan
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Sarah Smith
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Zachary M. Soler
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Carl Atkinson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Rodney J. Schlosser
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina
| |
Collapse
|
48
|
Sichien D, Lambrecht BN, Guilliams M, Scott CL. Development of conventional dendritic cells: from common bone marrow progenitors to multiple subsets in peripheral tissues. Mucosal Immunol 2017; 10:831-844. [PMID: 28198365 DOI: 10.1038/mi.2017.8] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/14/2017] [Indexed: 02/04/2023]
Abstract
Our understanding of conventional dendritic cell (cDC) development and the functional specializations of distinct subsets in the peripheral tissues has increased greatly in recent years. Here, we review cDC development from the distinct progenitors in the bone marrow through to the distinct cDC subsets found in barrier tissues, providing an overview of the different subsets described in each location. In addition, we detail the transcription factors and local signals that have been proposed to control this developmental process. Importantly, despite these significant advances, numerous questions remain to be answered regarding cDC development. For example, it remains unclear whether the different subsets described, such as the CD103+CD11b+ and CD103-CD11b+ cDCs in the intestines, truly represent different populations or rather distinct developmental or activation stages. Furthermore, whether distinct progenitors exist for these cDC subsets remains to be determined. Thus in the last part of this review we discuss what we believe will be the main questions facing the field for the coming years.
Collapse
Affiliation(s)
- D Sichien
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - B N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, The Netherlands
| | - M Guilliams
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - C L Scott
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
49
|
Cao Y, Zhang E, Yang J, Yang Y, Yu J, Xiao Y, Li W, Zhou D, Li Y, Zhao B, Yan H, Lu M, Zhong M, Yan H. Frontline Science: Nasal epithelial GM-CSF contributes to TLR5-mediated modulation of airway dendritic cells and subsequent IgA response. J Leukoc Biol 2017; 102:575-587. [PMID: 28522600 DOI: 10.1189/jlb.3hi0816-368rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 11/24/2022] Open
Abstract
Flagellin, as a TLR5 agonist, is an established mucosal adjuvant for enhancing mucosal IgA responses by i.n. immunization. Nasal epithelial cells (NECs) are the first sentinel cells to be exposed to antigen and adjuvant in i.n. immunization, and it is suggested that they play an important role in the mucosal adjuvant activity of flagellin. However, the molecular mechanism leading to modulation and the response by flagellin-activated NECs remain unknown. We aimed to identify the soluble mediator(s) from flagellin-activated NECs that modulate the functions of airway dendritic cells (DCs) and enhance subsequent IgA response. In vitro studies showed that compared with the TLR4 agonist LPS, flagellin directly triggered slight up-regulation of CD80 on airway DCs but was insufficient to affect CD86 expression and DC-mediated IgA response. With the use of an in vitro system for culturing mouse primary NECs (mNECs), we demonstrated that flagellin-activated mNECs could functionally modulate airway DCs, which manifested as significant up-regulation of CD80/CD86 and enhancement of IgA production. The functional modulation of airway DCs was dependent on TLR5 activation of mNECs rather than direct TLR5 activation of airway DCs. With the use of cytokine array and antibody-blocking assays, we further identified that GM-CSF, a cytokine secreted from TLR5-activated mNECs, contributes to the activation of mNECs to airway DCs and subsequent IgA enhancement. In vivo blocking experiments confirmed that GM-CSF is an important factor in recombinant flagellin derived from Salmonella typhi (FliC)-induced airway DC activation and antigen-specific IgA enhancement. Our data directly demonstrate that nasal epithelial GM-CSF contributes to TLR5-mediated modulation of airway DCs and a subsequent IgA response.
Collapse
Affiliation(s)
- Yuan Cao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Ejuan Zhang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Jingyi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Yi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Jie Yu
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Yang Xiao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Wei Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Dihan Zhou
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Yaoming Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Bali Zhao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Hu Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, Essen, Germany
| | - Maohua Zhong
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China; and
| |
Collapse
|
50
|
Baharom F, Rankin G, Blomberg A, Smed-Sörensen A. Human Lung Mononuclear Phagocytes in Health and Disease. Front Immunol 2017; 8:499. [PMID: 28507549 PMCID: PMC5410584 DOI: 10.3389/fimmu.2017.00499] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/11/2017] [Indexed: 12/17/2022] Open
Abstract
The lungs are vulnerable to attack by respiratory insults such as toxins, allergens, and pathogens, given their continuous exposure to the air we breathe. Our immune system has evolved to provide protection against an array of potential threats without causing collateral damage to the lung tissue. In order to swiftly detect invading pathogens, monocytes, macrophages, and dendritic cells (DCs)-together termed mononuclear phagocytes (MNPs)-line the respiratory tract with the key task of surveying the lung microenvironment in order to discriminate between harmless and harmful antigens and initiate immune responses when necessary. Each cell type excels at specific tasks: monocytes produce large amounts of cytokines, macrophages are highly phagocytic, whereas DCs excel at activating naïve T cells. Extensive studies in murine models have established a division of labor between the different populations of MNPs at steady state and during infection or inflammation. However, a translation of important findings in mice is only beginning to be explored in humans, given the challenge of working with rare cells in inaccessible human tissues. Important progress has been made in recent years on the phenotype and function of human lung MNPs. In addition to a substantial population of alveolar macrophages, three subsets of DCs have been identified in the human airways at steady state. More recently, monocyte-derived cells have also been described in healthy human lungs. Depending on the source of samples, such as lung tissue resections or bronchoalveolar lavage, the specific subsets of MNPs recovered may differ. This review provides an update on existing studies investigating human respiratory MNP populations during health and disease. Often, inflammatory MNPs are found to accumulate in the lungs of patients with pulmonary conditions. In respiratory infections or inflammatory diseases, this may contribute to disease severity, but in cancer patients this may improve clinical outcomes. By expanding on this knowledge, specific lung MNPs may be targeted or modulated in order to attain favorable responses that can improve preventive or treatment strategies against respiratory infections, lung cancer, or lung inflammatory diseases.
Collapse
Affiliation(s)
- Faezzah Baharom
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Gregory Rankin
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Anna Smed-Sörensen
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|