1
|
Mutlu S, Fytianos K, Ferrié C, Scalise M, Mykoniati S, Gazdhar A, Blank F. Adoptive Transfer of T Cells as a Potential Therapeutic Approach in the Bleomycin-Injured Mouse Lung. J Gene Med 2025; 27:e70018. [PMID: 40159455 PMCID: PMC11955259 DOI: 10.1002/jgm.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/21/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a lethal disease with an unknown etiology and complex pathophysiology that are not fully understood. The disease involves intricate cellular interplay, particularly among various immune cells. Currently, there is no treatment capable of reversing the fibrotic process or aiding lung regeneration. Hepatocyte growth factor (HGF) has demonstrated antifibrotic properties, whereas the adoptive transfer of modified T cells is a well-established treatment for various malignancies. We aimed to understand the dynamics of T cells in the progression of lung fibrosis and to study the therapeutic benefit of adoptive T cell transfer in a bleomycin-injured mouse lung (BLM) model. METHODS T cells were isolated from the spleen of naïve mice and transfected in vitro with mouse HGF plasmid and were administered intratracheally to the mice lungs 7 days post-bleomycin injury to the lung. Lung tissue and bronchoalveolar lavage were collected and analyzed using flow cytometry, histology, qRT-PCR, ELISA, and hydroxyproline assay. RESULTS Our findings demonstrate the successful T cell therapy of bleomycin-induced lung injury through the adoptive transfer of HGF-transfected T cells in mice. This treatment resulted in decreased collagen deposition and a balancing of immune cell exhaustion and cytokine homeostasis compared with untreated controls. In vitro testing showed enhanced apoptosis in myofibroblasts induced by HGF-overexpressing T cells. CONCLUSIONS Taken together, our data highlight the great potential of adoptive T cell transfer as an emerging therapy to counteract lung fibrosis.
Collapse
Affiliation(s)
- Seyran Mutlu
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Kleanthis Fytianos
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | - Céline Ferrié
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | - Melanie Scalise
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | | | - Amiq Gazdhar
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | - Fabian Blank
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| |
Collapse
|
2
|
Schulte MC, Boll AC, Barcellona AT, Lopez EA, Schrum AG, Ulery BD. Peptide Antigen Modifications Influence the On-Target and Off-Target Antibody Response for an Influenza Subunit Vaccine. Vaccines (Basel) 2025; 13:51. [PMID: 39852830 PMCID: PMC11768957 DOI: 10.3390/vaccines13010051] [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: 10/08/2024] [Revised: 11/13/2024] [Accepted: 01/05/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Peptide amphiphile micelles (PAMs) are an exciting nanotechnology currently being studied for a variety of biomedical applications, especially for drug delivery. Specifically, PAMs can enhance in vivo trafficking, cell-targeting, and cell interactions/internalization. However, modifying peptides, as is commonly performed to induce micellization, can influence their bioactivity. In our previous work, murine antibody responses to PAMs containing the influenza antigen M22-16 were slightly incongruous with prior PAM vaccine studies using other antigens. In this current work, the effect of native protein linkages and non-native micellizing moieties on M2 immunogenicity was studied. METHODS PAMs were synthesized using an elongated M2 antigen (i.e., Palm2K-M21-24-(KE)4). The PAMs were characterized, then their immunogenicity was evaluated with bone marrow-derived dendritic cells and in mice. RESULTS Although the modification scheme yielded immunogenic PAMs, these PAMs induced a substantial amount of off-target antibody production compared to unmodified peptidyl micelles (PMs, M21-24 peptide). CONCLUSIONS While the impact PAM-induced off-target antibodies had on vaccine efficacy remains to be elucidated, on-target antibodies from both PAM- and PM-vaccinated mice were excitingly able to recognize the M2 antigen within the context of the full M2 protein. This provides preliminary evidence that the PAM-induced on-target antibodies will at minimum be able to recognize the influenza virus upon exposure.
Collapse
Affiliation(s)
- Megan C. Schulte
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
| | - Adam C. Boll
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
| | - Agustin T. Barcellona
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
| | - Elida A. Lopez
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA;
| | - Adam G. Schrum
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA;
- Department of Surgery, University of Missouri, Columbia, MO 65211, USA
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
| | - Bret D. Ulery
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
- Materials Science & Engineering Institute, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
3
|
Guo Y, Sheng X, Tang X, Xing J, Chi H, Zhan W. Immunoglobulin M-based local production in skin-associated lymphoid tissue of flounder (Paralichthys olivaceus) initiated by immersion with inactivated Edwardsiella tarda. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109982. [PMID: 39461396 DOI: 10.1016/j.fsi.2024.109982] [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: 08/16/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
Fish skin, the mucosal site most exposed to external antigens, requires protection by an efficient local mucosal immune system. The mucosal reserve of IgM is recognized as an immune strategy that blocks pathogen invasion to maintain homeostasis, whereas the mechanism of skin-associated local IgM production induced by mucosal antigens is not well know. In this study, we found that the skin of flounder (Paralichthys olivaceus) was equipped with the immune cellular and molecular basis for processing mucosal antigens and triggering local specific responses, i.e., CD4+ Zap-70+ T cells, CD4- Zap-70+ T/NK cells, IgM+ MHCII+ B cells, PNA+ MHCII+ antigen-presenting cells, UEA-1+ WGA+ and UEA-1+ WGA- antigen-sampling cells, as well as secreted IgM and pIgR, as demonstrated by indirect immunofluorescence assay using different antibodies and lectins. After immersion immunization with inactivated Edwardsiella tarda, qPCR assay displayed up-regulation of immune-related genes in flounder skin. Flow cytometry analysis and EdU labeling demonstrated that the mucosal inactivated vaccine induced local proliferation and increased amounts of cutaneous IgM+ B cells. Skin explant culture proved the local production of specific IgM in the skin, which could bind to the surface of E. tarda. ELISA, laser scanning confocal microscopy, and Western blot revealed that, in addition to the elevated IgM levels, pIgR protein level was significantly up-regulated in skin tissue and surface mucus containing the pIgR (secretory component, SC)-tetrameric IgM complex, indicating that mucosal vaccine stimulated up-regulation of IgM and pIgR, which were secreted as a complex into skin mucus to exert the protective effects as secretory IgM. These findings deepen the understanding of IgM-based local responses in the mucosal immunity of teleosts, which will be critical for subsequent investigation into the protective mechanism of mucosal vaccines for fish health.
Collapse
Affiliation(s)
- Yuan Guo
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| |
Collapse
|
4
|
Liu EG, Yin X, Siniscalco ER, Eisenbarth SC. Dendritic cells in food allergy, treatment, and tolerance. J Allergy Clin Immunol 2024; 154:511-522. [PMID: 38971539 PMCID: PMC11414995 DOI: 10.1016/j.jaci.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 07/08/2024]
Abstract
Food allergy is a growing problem with limited treatment options. It is important to understand the mechanisms of food tolerance and allergy to promote the development of directed therapies. Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) that prime adaptive immune responses, such as those involved in the development of oral tolerance and food allergies. The DC subsets in the gut and skin are defined by their surface markers and function. The default response to an ingested innocuous antigen is oral tolerance, which requires either gut DCs or a subset of newly identified RORγt+ APCs to induce the development of gut peripheral regulatory T cells. However, DCs in the skin, gut, and lung can also promote allergic sensitization when they are activated under certain inflammatory conditions, such as with alarmin release or gut dysbiosis. DCs also play a role in the responses to the various modalities of food immunotherapy. Langerhans cells in the skin appear to be necessary for the response to epicutaneous immunotherapy. It will be important to determine which real-world stimuli activate the DCs that prime allergic sensitization and discover methods to selectively initiate a tolerogenic program in APCs.
Collapse
Affiliation(s)
- Elise G Liu
- Section of Rheumatology, Allergy and Immunology, Department of Medicine, Yale University School of Medicine, New Haven, Conn
| | - Xiangyun Yin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Emily R Siniscalco
- Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Stephanie C Eisenbarth
- Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
5
|
Kim SH, Cho BH, Kim KS, Jang YS. Complement C5a promotes antigen cross-presentation by Peyer's patch monocyte-derived dendritic cells and drives a protective CD8 + T cell response. Cell Rep 2021; 35:108995. [PMID: 33852847 DOI: 10.1016/j.celrep.2021.108995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 01/25/2021] [Accepted: 03/24/2021] [Indexed: 01/05/2023] Open
Abstract
The complement fragment C5a is closely associated with adaptive immune induction in the mucosa. However, the mechanisms that control CD8+ T cell responses by C5a have not been extensively explored. This study reveals that C5/C5a in the Peyer's patch (PP) subepithelial dome increases upon oral Listeria infection. We hypothesize that C5aR+ PP cells play an important role in the induction of antigen-specific T cell immunity. Using single-cell RNA sequencing, we identify C5aR- and lysozyme-expressing dendritic cells (C5aR+ LysoDCs) in PP and examine their role in CD8+ T cell immune induction. Stimulation of C5aR+ LysoDCs by C5a increases reactive oxygen species levels, leading to efficient antigen cross-presentation, which elicits an antigen-specific CD8+ T cell response. In C5-deficient mice, oral co-administration of C5a and Listeria enhances Listeria-specific cytotoxic T cell levels. Collectively, these findings suggest a role of the complement system in intestinal T cell immunity.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology and The Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Korea
| | - Byeol-Hee Cho
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, 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
|
6
|
Zhang B, Liu E, Gertie JA, Joseph J, Xu L, Pinker EY, Waizman DA, Catanzaro J, Hamza KH, Lahl K, Gowthaman U, Eisenbarth SC. Divergent T follicular helper cell requirement for IgA and IgE production to peanut during allergic sensitization. Sci Immunol 2020; 5:5/47/eaay2754. [PMID: 32385053 DOI: 10.1126/sciimmunol.aay2754] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
Immunoglobulin A (IgA) is the dominant antibody isotype in the gut and has been shown to regulate microbiota. Mucosal IgA is also widely believed to prevent food allergens from penetrating the gut lining. Even though recent work has elucidated how bacteria-reactive IgA is induced, little is known about how IgA to food antigens is regulated. Although IgA is presumed to be induced in a healthy gut at steady state via dietary exposure, our data do not support this premise. We found that daily food exposure only induced low-level, cross-reactive IgA in a minority of mice. In contrast, induction of significant levels of peanut-specific IgA strictly required a mucosal adjuvant. Although induction of peanut-specific IgA required T cells and CD40L, it was T follicular helper (TFH) cell, germinal center, and T follicular regulatory (TFR) cell-independent. In contrast, IgG1 and IgE production to peanut required TFH cells. These data suggest an alternative paradigm in which the cellular mechanism of IgA production to food antigens is distinct from IgE and IgG1. We developed an equivalent assay to study this process in stool samples from healthy, nonallergic humans, which revealed substantial levels of peanut-specific IgA that were stable over time. Similar to mice, patients with loss of CD40L function had impaired titers of gut peanut-specific IgA. This work challenges two widely believed but untested paradigms about antibody production to dietary antigens: (i) the steady state/tolerogenic response to food antigens includes IgA production and (ii) TFH cells drive food-specific gut IgA.
Collapse
Affiliation(s)
- Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elise Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jake A Gertie
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Julie Joseph
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elisha Y Pinker
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Columbia University, New York, NY 10027, USA
| | - Daniel A Waizman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jason Catanzaro
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Pulmonology, Allergy, Immunology and Sleep Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Kedir Hussen Hamza
- Department for Experimental Medicine, Immunology Section, Lund University, Lund 221 84, Sweden
| | - Katharina Lahl
- Department for Experimental Medicine, Immunology Section, Lund University, Lund 221 84, Sweden.,Division of Biopharma, Institute for Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA. .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
7
|
Bruton K, Koenig JFE, Phelps A, Jordana M. Perturbations to Homeostasis in Experimental Models Revealed Innate Pathways Driving Food Allergy. Front Immunol 2020; 11:603272. [PMID: 33362786 PMCID: PMC7758527 DOI: 10.3389/fimmu.2020.603272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
While type 2 immunity has been conventionally viewed as beneficial against helminths, venoms, and poisons, and harmful in allergy, contemporary research has uncovered its critical role in the maintenance of homeostasis. The initiation of a type 2 immune response involves an intricate crosstalk between structural and immune cells. Structural cells react to physical and chemical tissue perturbations by secreting alarmins, which signal the innate immune system to restore homeostasis. This pathway acts autonomously in the context of sterile injury and in the presence of foreign antigen initiates an adaptive Th2 response that is beneficial in the context of venoms, toxins, and helminths, but not food allergens. The investigation of the triggers and mechanisms underlying food allergic sensitization in humans is elusive because sensitization is a silent process. Therefore, the central construct driving food allergy modeling is based on introducing perturbations of tissue homeostasis along with an allergen which will result in an immunological and clinical phenotype that is consistent with that observed in humans. The collective evidence from multiple models has revealed the pre-eminent role of innate cells and molecules in the elicitation of allergic sensitization. We posit that, with the expanding use of technologies capable of producing formidable datasets, models of food allergy will continue to have an indispensable role to delineate mechanisms and establish causal relationships.
Collapse
Affiliation(s)
| | | | | | - Manel Jordana
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton ON, Canada
| |
Collapse
|
8
|
Park J, Frizzell H, Zhang H, Cao S, Hughes SM, Hladik F, Koelle DM, Woodrow KA. Flt3-L enhances trans-epithelial migration and antigen presentation of dendritic cells adoptively transferred to genital mucosa. J Control Release 2020; 329:782-793. [PMID: 33035616 DOI: 10.1016/j.jconrel.2020.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
Dendritic cells (DCs) play a critical role in shaping adaptive immunity. Systemic transfer of DCs by intravenous injection has been widely investigated to inform the development of immunogenic DCs for use as cellular therapies. Adoptive transfer of DCs to mucosal sites has been limited but serves as a valuable tool to understand the role of the microenvironment on mucosal DC activation, maturation and antigen presentation. Here, we show that chitosan facilitates transmigration of DCs across the vaginal epithelium in the mouse female reproductive tract (FRT). In addition, ex vivo programming of DCs with fms-related tyrosine kinase 3 ligand (Flt3-L) was found to enhance translocation of intravaginally administered DCs to draining lymph nodes (dLNs) and stimulate in vivo proliferation of both antigen-specific CD4+ and CD8+ T cells (cross-presentation). Mucosal priming with chitosan and DC programming may hold great promise to enhance efficacy of DC-based vaccination to the female genital mucosa.
Collapse
Affiliation(s)
- Jaehyung Park
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Hannah Frizzell
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Hangyu Zhang
- Department of Bioengineering, University of Washington, Seattle, WA, USA; School of Biomedical Engineering, Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Shijie Cao
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Sean M Hughes
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Florian Hladik
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA; Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David M Koelle
- Departments of Laboratory Medicine, and Global Health, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA; Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
9
|
LTA1 and dmLT enterotoxin-based proteins activate antigen-presenting cells independent of PKA and despite distinct cell entry mechanisms. PLoS One 2020; 15:e0227047. [PMID: 31929548 PMCID: PMC6957164 DOI: 10.1371/journal.pone.0227047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 12/11/2019] [Indexed: 01/19/2023] Open
Abstract
Enterotoxin-based proteins are powerful manipulators of mucosal immunity. The A1 domain of heat-labile enterotoxin from E. coli, or LTA1, is a newer adjuvant from this family under investigation for intranasal vaccines. Although LTA1 has been examined in mouse vaccination studies, its ability to directly stimulate immune cells compared to related adjuvant proteins has not been well explored. Here, we perform the first rigorous examination of LTA1’s effect on antigen presenting cells (APC) using a human monocyte cell line THP-1. To better understand LTA1’s stimulatory effects, we compared it to dmLT, or LT-R192G/L211A, a related AB5 adjuvant in clinical trials for oral or parenteral vaccines. LTA1 and dmLT both activated APCs to upregulate MHC-II (HLA-DR), CD86, cytokine secretion (e.g., IL-1β) and inflammasome activation. The effect of LTA1 on surface marker changes (e.g., MHC-II) was highly dose-dependent whereas dmLT exhibited high MHC-II expression regardless of dose. In contrast, cytokine secretion profiles were similar and dose-dependent after both LTA1 and dmLT treatment. Cellular activation by LTA1 was independent of ganglioside binding, as pre-treatment with purified GM1 blocked the effect of dmLT but not LTA1. Unexpectedly, while activation of the inflammasome and cytokine secretion by LTA1 or dmLT was blocked by the protein kinase A inhibitor H89 (similar to previous reports), these responses were not inhibited by a more specific PKA peptide inhibitor or antagonist; thus Indicating that a novel and unknown mechanism is responsible for inflammasome activation and cytokine secretion by LT proteins. Lastly, LTA1 stimulated a similar cytokine profile in primary human monocytes as it did in THP1 cells, including IL-1β, IL-6, IL-8, MIP-1α, MIP-1β, and TNFα. Thus, we report that LTA1 protein programs a dendritic cell-like phenotype in APCs similar to dmLT in a mechanism that is independent of PKA activation and GM1 binding and entry.
Collapse
|
10
|
Valli E, Harriett AJ, Nowakowska MK, Baudier RL, Provosty WB, McSween Z, Lawson LB, Nakanishi Y, Norton EB. LTA1 is a safe, intranasal enterotoxin-based adjuvant that improves vaccine protection against influenza in young, old and B-cell-depleted (μMT) mice. Sci Rep 2019; 9:15128. [PMID: 31641151 PMCID: PMC6805908 DOI: 10.1038/s41598-019-51356-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 09/19/2019] [Indexed: 01/17/2023] Open
Abstract
Enterotoxin-based adjuvants including cholera toxin and heat-labile toxin (LT) are powerful manipulators of mucosal immunity; however, past clinical trials identified unacceptable neurological toxicity when LT or mutant AB5 adjuvant proteins were added to intranasal vaccines. Here, we examined the isolated enzymatic A1 domain of LT (LTA1) for intranasal safety and efficacy in combination with influenza (flu) vaccination. LTA1-treated mice exhibited no neurotoxicity, as measured by olfactory system testing and H&E staining of nasal tissue in contrast with cholera toxin. In vaccination studies, intranasal LTA1 enhanced immune responses to inactivated virus antigen and subsequent protection against H1N1 flu challenge in mice (8-week or 24-months). In addition, lung H1N1 viral titers post-challenge correlated to serum antibody responses; however, enhanced protection was also observed in μMT mice lacking B-cells while activation and recruitment of CD4 T-cells into the lung was apparent. Thus, we report that LTA1 protein is a novel, safe and effective enterotoxin adjuvant that improves protection of an intranasal flu vaccination by a mechanism that does not appear to require B-cells.
Collapse
Affiliation(s)
- E Valli
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - A J Harriett
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - M K Nowakowska
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - R L Baudier
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - W B Provosty
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - Z McSween
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - L B Lawson
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - Y Nakanishi
- Department of Pathology & Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - E B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA.
| |
Collapse
|
11
|
Abstract
Perhaps the best-studied mucosal adjuvants are the bacterially derived ADP-ribosylating enterotoxins. This adjuvant family includes heat-labile enterotoxin of Escherichia coli (LT), cholera toxin (CT), and mutants or subunits of LT and CT. These proteins promote a multifaceted antigen-specific response, including inflammatory Th1, Th2, Th17, cytotoxic T lymphocytes (CTLs), and antibodies. However, more uniquely among adjuvant classes, they induce antigen-specific IgA antibodies and long-lasting memory to coadministered antigens when delivered mucosally or even parenterally. The purpose of this minireview is to describe the general properties, history and creation, preclinical studies, clinical studies, mechanisms of action, and considerations for use of the most promising enterotoxin-based adjuvant to date, LT(R192G/L211A) or dmLT. This review is timely due to completed, ongoing, and planned clinical investigations of dmLT in multiple vaccine formulations by government, nonprofit, and industry groups in the United States and abroad.
Collapse
Affiliation(s)
- John D Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elizabeth B Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
12
|
Copland A, Diogo GR, Hart P, Harris S, Tran AC, Paul MJ, Singh M, Cutting SM, Reljic R. Mucosal Delivery of Fusion Proteins with Bacillus subtilis Spores Enhances Protection against Tuberculosis by Bacillus Calmette-Guérin. Front Immunol 2018; 9:346. [PMID: 29593708 PMCID: PMC5857916 DOI: 10.3389/fimmu.2018.00346] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/07/2018] [Indexed: 01/16/2023] Open
Abstract
Tuberculosis (TB) is the most deadly infectious disease in existence, and the only available vaccine, Bacillus Calmette-Guérin (BCG), is almost a century old and poorly protective. The immunological complexity of TB, coupled with rising resistance to antimicrobial therapies, necessitates a pipeline of diverse novel vaccines. Here, we show that Bacillus subtilis spores can be coated with a fusion protein 1 (“FP1”) consisting of Mycobacterium tuberculosis (Mtb) antigens Ag85B, ACR, and HBHA. The resultant vaccine, Spore-FP1, was tested in a murine low-dose Mtb aerosol challenge model. Mice were primed with subcutaneous BCG, followed by mucosal booster immunizations with Spore-FP1. We show that Spore-FP1 enhanced pulmonary control of Mtb, as evidenced by reduced bacterial burdens in the lungs. This was associated with elevated antigen-specific IgG and IgA titers in the serum and lung mucosal surface, respectively. Spore-FP1 immunization generated superior antigen-specific memory T-cell proliferation in both CD4+ and CD8+ compartments, alongside bolstered Th1-, Th17-, and Treg-type cytokine production, compared to BCG immunization alone. CD69+CD103+ tissue resident memory T-cells (Trm) were found within the lung parenchyma after mucosal immunization with Spore-FP1, confirming the advantages of mucosal delivery. Our data show that Spore-FP1 is a promising new TB vaccine that can successfully augment protection and immunogenicity in BCG-primed animals.
Collapse
Affiliation(s)
| | - Gil R Diogo
- St George's Medical School, London, United Kingdom
| | - Peter Hart
- St George's Medical School, London, United Kingdom
| | - Shane Harris
- St George's Medical School, London, United Kingdom
| | - Andy C Tran
- St George's Medical School, London, United Kingdom
| | | | | | - Simon M Cutting
- School of Biological Sciences, Royal Holloway University of London, Egham, United Kingdom
| | - Rajko Reljic
- St George's Medical School, London, United Kingdom
| |
Collapse
|
13
|
Da Silva C, Wagner C, Bonnardel J, Gorvel JP, Lelouard H. The Peyer's Patch Mononuclear Phagocyte System at Steady State and during Infection. Front Immunol 2017; 8:1254. [PMID: 29038658 PMCID: PMC5630697 DOI: 10.3389/fimmu.2017.01254] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022] Open
Abstract
The gut represents a potential entry site for a wide range of pathogens including protozoa, bacteria, viruses, or fungi. Consequently, it is protected by one of the largest and most diversified population of immune cells of the body. Its surveillance requires the constant sampling of its encounters by dedicated sentinels composed of follicles and their associated epithelium located in specialized area. In the small intestine, Peyer’s patches (PPs) are the most important of these mucosal immune response inductive sites. Through several mechanisms including transcytosis by specialized epithelial cells called M-cells, access to the gut lumen is facilitated in PPs. Although antigen sampling is critical to the initiation of the mucosal immune response, pathogens have evolved strategies to take advantage of this permissive gateway to enter the host and disseminate. It is, therefore, critical to decipher the mechanisms that underlie both host defense and pathogen subversive strategies in order to develop new mucosal-based therapeutic approaches. Whereas penetration of pathogens through M cells has been well described, their fate once they have reached the subepithelial dome (SED) remains less well understood. Nevertheless, it is clear that the mononuclear phagocyte system (MPS) plays a critical role in handling these pathogens. MPS members, including both dendritic cells and macrophages, are indeed strongly enriched in the SED, interact with M cells, and are necessary for antigen presentation to immune effector cells. This review focuses on recent advances, which have allowed distinguishing the different PP mononuclear phagocyte subsets. It gives an overview of their diversity, specificity, location, and functions. Interaction of PP phagocytes with the microbiota and the follicle-associated epithelium as well as PP infection studies are described in the light of these new criteria of PP phagocyte identification. Finally, known alterations affecting the different phagocyte subsets during PP stimulation or infection are discussed.
Collapse
Affiliation(s)
| | - Camille Wagner
- Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Johnny Bonnardel
- Laboratory of Myeloid Cell Ontogeny and Functional Specialisation, VIB Inflammation Research Center, Ghent, Belgium
| | | | - Hugues Lelouard
- Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
14
|
Pan Y, Lei Z, Wei X, Hao W. TAK1 deficiency in dendritic cells inhibits adaptive immunity in SRBC-immunized C57BL/6 mice. FEBS Open Bio 2016; 6:548-57. [PMID: 27419057 PMCID: PMC4887970 DOI: 10.1002/2211-5463.12062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 11/24/2022] Open
Abstract
Dendritic cells (DCs) are important in the initiation of primary T‐cell responses, while transforming growth factor‐β (TGF‐β)‐activated kinase 1 (TAK1) is a critical regulator of DC survival and homeostasis. This study evaluated the T‐cell dependent antibody response (TDAR) to sheep red blood cells (SRBC) on a DC‐specific TAK1‐deficient mice model. The results showed that TAK1 deficiency in DCs significantly suppressed the humoral and cellular immune response in mice. DC‐specific TAK1 deletion impaired splenic T‐cell population and conventional DCs, abolished the cytokine production of splenic T cells and down‐regulated some functional gene expression in the spleen. Collectively, this study suggests that TAK1 plays an essential role in the development of the humoral immune response.
Collapse
Affiliation(s)
- Yao Pan
- Department of ToxicologySchool of Public HealthPeking UniversityBeijingChina; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food SafetyBeijingChina
| | - Zhiming Lei
- Department of ToxicologySchool of Public HealthPeking UniversityBeijingChina; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food SafetyBeijingChina
| | - Xuetao Wei
- Department of ToxicologySchool of Public HealthPeking UniversityBeijingChina; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food SafetyBeijingChina
| | - Weidong Hao
- Department of ToxicologySchool of Public HealthPeking UniversityBeijingChina; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food SafetyBeijingChina
| |
Collapse
|
15
|
Lebens M, Terrinoni M, Karlsson SL, Larena M, Gustafsson-Hedberg T, Källgård S, Nygren E, Holmgren J. Construction and preclinical evaluation of mmCT, a novel mutant cholera toxin adjuvant that can be efficiently produced in genetically manipulated Vibrio cholerae. Vaccine 2016; 34:2121-8. [PMID: 26973069 DOI: 10.1016/j.vaccine.2016.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/12/2016] [Accepted: 03/02/2016] [Indexed: 10/22/2022]
Abstract
There is an urgent need for new adjuvants that are effective with mucosally administered vaccines. Cholera toxin (CT) is the most powerful known mucosal adjuvant but is much too toxic for human use. In an effort to develop a useful mucosal adjuvant we have generated a novel non-toxic mutant CT molecule that retains much of the adjuvant activity of native CT. This was achieved by making the enzymatically active A subunit (CTA) recalcitrant to the site-specific proteolytic cleavage ("nicking") required for toxicity, which was found to require mutations not only in the two residues rendering the molecule resistant to trypsin but also in neighboring sites protecting against cleavage by Vibrio cholerae proteases. This multiple-mutated CT (mmCT) adjuvant protein could be efficiently produced in and purified from the extracellular medium of CT-deleted V. cholerae. The mmCT completely lacked detectable enterotoxicity in an infant mouse model and had >1000-fold reduced cAMP inducing activity compared to native CT in a sensitive mammalian target cell system. It nonetheless proved to have potent adjuvant activity on mucosal and systemic antibody as well as cellular immune responses to mucosally co-administered antigens including oral cholera and intranasal influenza vaccines. We conclude that mmCT is an attractive novel non-toxic mucosal adjuvant for enhancing immune responses to co-administered mucosal vaccines.
Collapse
Affiliation(s)
- Michael Lebens
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden.
| | - Manuela Terrinoni
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden
| | - Stefan L Karlsson
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden
| | - Maximilian Larena
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden
| | - Tobias Gustafsson-Hedberg
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden
| | - Susanne Källgård
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden
| | - Erik Nygren
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden
| | - Jan Holmgren
- Department of Microbiology and Immunology and Gothenburg University Vaccine Research Institute (GUVAX), Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Box 435, SE-405 30 Göteborg, Sweden
| |
Collapse
|
16
|
Slütter B, Jiskoot W. Sizing the optimal dimensions of a vaccine delivery system: a particulate matter. Expert Opin Drug Deliv 2015; 13:167-70. [DOI: 10.1517/17425247.2016.1121989] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
17
|
Moro-Sibilot L, This S, Blanc P, Sanlaville A, Sisirak V, Bardel E, Boschetti G, Bendriss-Vermare N, Defrance T, Dubois B, Kaiserlian D. Plasmacytoid dendritic cells are dispensable for noninfectious intestinal IgA responses in vivo. Eur J Immunol 2015; 46:354-9. [PMID: 26518732 DOI: 10.1002/eji.201545977] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/06/2015] [Accepted: 10/27/2015] [Indexed: 11/10/2022]
Abstract
Intestinal DCs orchestrate gut immune homeostasis by dampening proinflammatory T-cell responses and inducing anti-inflammatory IgA responses. Although no specific DC subset has been strictly assigned so far to govern IgA response, some candidate subsets emerge. In particular, plasmacytoid DCs (pDCs), which notoriously promote anti-viral immunity and T-cell tolerance to innocuous antigens (Ags), contribute to IgA induction in response to intestinal viral infection and promote T-cell-independent IgA responses in vitro. Here, using two transgenic mouse models, we show that neither short-term nor long-term pDC depletion alters IgA class switch recombination in Peyer's patches and frequency of IgA plasma cells in intestinal mucosa at steady state, even in the absence of T-cell help. In addition, pDCs are dispensable for induction of intestinal IgA plasma cells in response to oral immunization with T-cell-dependent or T-cell-independent Ags, and are not required for proliferation and IgA switch of Ag-specific B cells in GALT. These results show that pDCs are dispensable for noninfectious IgA responses, and suggest that various DC subsets may play redundant roles in the control of intestinal IgA responses.
Collapse
Affiliation(s)
- Ludovic Moro-Sibilot
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France
| | - Sebastien This
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France
| | - Pascal Blanc
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France
| | - Amelien Sanlaville
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France.,Cancerology Research Center of Lyon, Inserm U1052 - CNRS UMR5286, Lyon, France
| | - Vanja Sisirak
- Departments of Pathology and Medicine, NYU Langone Medical Center, NY, USA
| | - Emilie Bardel
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France
| | - Gilles Boschetti
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France
| | | | - Thierry Defrance
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France
| | - Bertrand Dubois
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France.,Cancerology Research Center of Lyon, Inserm U1052 - CNRS UMR5286, Lyon, France
| | - Dominique Kaiserlian
- International Center for Infectiology Research (CIRI), Lyon, France.,Inserm U1111, Lyon, France.,CNRS UMR5308, Lyon, France.,Ecole Normale Superieure, Lyon, France.,Universite Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
18
|
Kau AL, Planer JD, Liu J, Rao S, Yatsunenko T, Trehan I, Manary MJ, Liu TC, Stappenbeck TS, Maleta KM, Ashorn P, Dewey KG, Houpt ER, Hsieh CS, Gordon JI. Functional characterization of IgA-targeted bacterial taxa from undernourished Malawian children that produce diet-dependent enteropathy. Sci Transl Med 2015; 7:276ra24. [PMID: 25717097 DOI: 10.1126/scitranslmed.aaa4877] [Citation(s) in RCA: 272] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To gain insights into the interrelationships among childhood undernutrition, the gut microbiota, and gut mucosal immune/barrier function, we purified bacterial strains targeted by immunoglobulin A (IgA) from the fecal microbiota of two cohorts of Malawian infants and children. IgA responses to several bacterial taxa, including Enterobacteriaceae, correlated with anthropometric measurements of nutritional status in longitudinal studies. The relationship between IgA responses and growth was further explained by enteropathogen burden. Gnotobiotic mouse recipients of an IgA(+) bacterial consortium purified from the gut microbiota of undernourished children exhibited a diet-dependent enteropathy characterized by rapid disruption of the small intestinal and colonic epithelial barrier, weight loss, and sepsis that could be prevented by administering two IgA-targeted bacterial species from a healthy microbiota. Dissection of a culture collection of 11 IgA-targeted strains from an undernourished donor, sufficient to transmit these phenotypes, disclosed that Enterobacteriaceae interacted with other consortium members to produce enteropathy. These findings indicate that bacterial targets of IgA responses have etiologic, diagnostic, and therapeutic implications for childhood undernutrition.
Collapse
Affiliation(s)
- Andrew L Kau
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA. Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph D Planer
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jie Liu
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Sindhuja Rao
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tanya Yatsunenko
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Indi Trehan
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Paediatrics and Child Health, College of Medicine, University of Malawi, Chichiri, Blantyre 3, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Community Health, College of Medicine, University of Malawi, Chichiri, Blantyre 3, Malawi
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kenneth M Maleta
- Department of Community Health, College of Medicine, University of Malawi, Chichiri, Blantyre 3, Malawi
| | - Per Ashorn
- Department for International Health, University of Tampere School of Medicine, Tampere 33014, Finland
| | - Kathryn G Dewey
- Department of Nutrition, and Program in International and Community Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Eric R Houpt
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Chyi-Song Hsieh
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey I Gordon
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
19
|
Wang Y, Xu B, Hu WW, Chen LJ, Wu CP, Lu BF, Shen YP, Jiang JT. High expression of CD11c indicates favorable prognosis in patients with gastric cancer. World J Gastroenterol 2015; 21:9403-9412. [PMID: 26309367 PMCID: PMC4541393 DOI: 10.3748/wjg.v21.i31.9403] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/13/2015] [Accepted: 06/10/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the relationship between CD11c expression level and prognosis in patients with gastric cancer (GC).
METHODS: This retrospective survival study was performed from July 31, 2008 to June 30, 2014. Our study inclusion criteria included all the patients with GC who underwent surgical resection between January 1998 and December 2009 in the Third Affiliated Hospital of Soochow University. CD11c expression levels in 140 patients with GC at different UICC stages were evaluated using immunohistochemistry, and GC tissues from 16 cases were further verified by qRT-PCR. The χ2 test was used to compare the patient- and disease-related factors between the low CD11c expression group and the high expression group. Univariate probabilities of overall survival (OS) and disease-free survival (DFS) were assessed using the Kaplan-Meier method. The log rank test was used to compare survival curves. Different multivariate COX models were used to estimate the association between CD11c expression and both death and recurrence risk in GC patients.
RESULTS: The average CD11c expression level was 5.1 ± 1.8/high power field (HPF) in 10 gastritis samples, 4.5 ± 2.3/HPF in 10 gastric polyp samples and 9.7 ± 6.3/HPF in 140 gastric cancer samples, respectively. The CD11c expression level was significantly decreased from UICC stage I to stage IV (stage I: 16.0 ± 7.4, stage II: 10.4 ± 5.5, stage III: 9.4 ± 6.1, stage IV: 5.3 ± 3.2, P < 0.001). Patients in the high CD11c expression group had a greater 3- and 5-year OS probability and longer median survival time compared with the low CD11c expression group, (67.7% vs 39.2%; 51.4% vs 29.0%; 67.0 mo vs 28.0 mo; χ2 = 6.80, P = 0.009), and had a greater 3- and 5-year DFS probability and longer median DFS time (63.7% vs 24.0%; 49.1% vs 11.9%; 64.0 mo vs 18.0 mo; χ2 = 15.39, P < 0.001). Patients with high CD11c high expression had a reduced risk of death (HR = 0.56, 95%CI: 0.33-0.98, P < 0.05) and relapse (HR = 0.39, 95%CI: 0.23-0.67, P < 0.01) compared with patients with low CD11c expression after adjustment of potential confounders, with the exception of tumor size. However, the protective effect related to death (HR = 0.90, 95%CI: 0.49-1.67, P = 0.749) and relapse (HR = 0.65, 95%CI: 0.36-1.19, P = 0.160) disappeared when tumor size was incorporated into the model.
CONCLUSION: High expression of CD11c decreased the risk of death and relapse, and may be regarded as an alternative indicator of favorable prognosis in patients with GC.
Collapse
|
20
|
Cholera toxin adjuvant promotes a balanced Th1/Th2/Th17 response independently of IL-12 and IL-17 by acting on Gsα in CD11b⁺ DCs. Mucosal Immunol 2015; 8:815-27. [PMID: 25425266 DOI: 10.1038/mi.2014.111] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 10/06/2014] [Indexed: 02/04/2023]
Abstract
Despite an extensive literature on the mechanism of action of cholera toxin (CT), we still lack critical information about how the toxin acts as an adjuvant and, especially, which dendritic cells (DCs) are the target cells. Although a T helper type 2 (Th2)-skewing effect of CT is most commonly reported, effective priming of Th17 cells as well as suppression of Th1 responses are well documented. However, the ability of CT to block interferon regulatory factor 8 (IRF8) function and interleukin (IL)-12 production in DCs, which blocks CD8α DC and Th1 cell development, is inconsistent with priming of Th1 and CD8 T cells in many other reports. This prompted us to investigate the adjuvant effect of CT in wild-type, IL-12p40-/-, Batf3-/-, and IL-17A-/- mice and in mice that selectively lack the Gsα target protein for CT adenosine diphosphate (ADP)-ribosylation in DCs. We found that CT promoted Th1 priming independently of IL-12, and whereas Th2 and also Th17 responses were augmented, the gut IgA responses did not require IL-17A. Adjuvanticity was intact in Batf3-/- mice, lacking CD8α(+) DCs, but completely lost in mice with Gsα-deficient CD11c cells. Thus, our data demonstrate that the adjuvant effect requires Gsα expression in CD11b(+) DCs, and that priming of mucosal IgA and CD4 T cells appears unbiased and is independent of IL-12 and IL-17A.
Collapse
|
21
|
Fougeron D, Van Maele L, Songhet P, Cayet D, Hot D, Van Rooijen N, Mollenkopf HJ, Hardt WD, Benecke AG, Sirard JC. Indirect Toll-like receptor 5-mediated activation of conventional dendritic cells promotes the mucosal adjuvant activity of flagellin in the respiratory tract. Vaccine 2015; 33:3331-41. [PMID: 26003491 DOI: 10.1016/j.vaccine.2015.05.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 04/17/2015] [Accepted: 05/11/2015] [Indexed: 01/17/2023]
Abstract
The Toll-like receptor 5 (TLR5) agonist flagellin is an effective adjuvant for vaccination. Recently, we demonstrated that the adaptive responses stimulated by intranasal administration of flagellin and antigen were linked to TLR5 signaling in the lung epithelium. The present study sought to identify the antigen presenting cells involved in this adjuvant activity. We first found that the lung dendritic cells captured antigen very efficiently in a process independent of TLR5. However, TLR5-mediated signaling specifically enhanced the maturation of lung dendritic cells. Afterward, the number of antigen-bound and activated conventional dendritic cells (both CD11b(+) and CD103(+)) increased in the mediastinal lymph nodes in contrast to monocyte-derived dendritic cells. These data suggested that flagellin-activated lung conventional dendritic cells migrate to the draining lymph nodes. The lymph node dendritic cells, in particular CD11b(+) cells, were essential for induction of CD4 T-cell response. Lastly, neutrophils and monocytes were recruited into the lungs by flagellin administration but did not contribute to the adjuvant activity. The functional activation of conventional dendritic cells was independent of direct TLR5 signaling, thereby supporting the contribution of maturation signals produced by flagellin-stimulated airway epithelium. In conclusion, our results demonstrated that indirect TLR5-dependent stimulation of airway conventional dendritic cells is essential to flagellin's mucosal adjuvant activity.
Collapse
Affiliation(s)
- Delphine Fougeron
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France; Centre National de la Recherche Scientifique, UMR 8204, F-59019 Lille, France; Université de Lille, F-59000 Lille, France
| | - Laurye Van Maele
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France; Centre National de la Recherche Scientifique, UMR 8204, F-59019 Lille, France; Université de Lille, F-59000 Lille, France
| | - Pascal Songhet
- Institute of Microbiology, Eidgenössische Technische Hochschule Zürich, CH-8093 Zürich, Switzerland
| | - Delphine Cayet
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France; Centre National de la Recherche Scientifique, UMR 8204, F-59019 Lille, France; Université de Lille, F-59000 Lille, France
| | - David Hot
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France; Centre National de la Recherche Scientifique, UMR 8204, F-59019 Lille, France; Université de Lille, F-59000 Lille, France
| | - Nico Van Rooijen
- Department of Molecular Cell Biology, VU Medical Center, NL-1007 Amsterdam, The Netherlands
| | | | - Wolf-Dietrich Hardt
- Institute of Microbiology, Eidgenössische Technische Hochschule Zürich, CH-8093 Zürich, Switzerland
| | - Arndt G Benecke
- Institut des Hautes Études Scientifiques and Centre National de la Recherche Scientifique, F-91440 Bures-sur-Yvette, France
| | - Jean-Claude Sirard
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France; Centre National de la Recherche Scientifique, UMR 8204, F-59019 Lille, France; Université de Lille, F-59000 Lille, France.
| |
Collapse
|
22
|
Dahlgren MW, Gustafsson-Hedberg T, Livingston M, Cucak H, Alsén S, Yrlid U, Johansson-Lindbom B. T follicular helper, but not Th1, cell differentiation in the absence of conventional dendritic cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:5187-99. [PMID: 25917099 DOI: 10.4049/jimmunol.1401938] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/18/2015] [Indexed: 11/19/2022]
Abstract
Development of long-lived humoral immunity is dependent on CXCR5-expressing T follicular helper (Tfh) cells, which develop concomitantly to effector Th cells that support cellular immunity. Conventional dendritic cells (cDCs) are critical APCs for initial priming of naive CD4(+) T cells but, importantly, also provide accessory signals that govern effector Th cell commitment. To define the accessory role of cDCs during the concurrent development of Tfh and effector Th1 cells, we performed high-dose Ag immunization in conjunction with the Th1-biased adjuvant polyinosinic:polycytidylic acid (pI:C). In the absence of cDCs, pI:C failed to induce Th1 cell commitment and IgG2c production. However, cDC depletion did not impair Tfh cell differentiation or germinal center formation, and long-lived IgG1 responses of unaltered affinity developed in mice lacking cDCs at the time point for immunization. Thus, cDCs are required for the pI:C-driven Th1 cell fate commitment but have no crucial accessory function in relation to Tfh cell differentiation.
Collapse
Affiliation(s)
| | - Tobias Gustafsson-Hedberg
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Megan Livingston
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Helena Cucak
- Immunology Section, Lund University, 221 84 Lund, Sweden; and
| | - Samuel Alsén
- Immunology Section, Lund University, 221 84 Lund, Sweden; and Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | | |
Collapse
|
23
|
Kim SH, Yang IY, Kim J, Lee KY, Jang YS. Antimicrobial peptide LL-37 promotes antigen-specific immune responses in mice by enhancing Th17-skewed mucosal and systemic immunities. Eur J Immunol 2015; 45:1402-13. [PMID: 25655317 DOI: 10.1002/eji.201444988] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 01/13/2015] [Accepted: 02/03/2015] [Indexed: 01/17/2023]
Abstract
The human antimicrobial peptide LL-37 is known to have chemotactic and modulatory activities on various cells including monocytes, T cells, and epithelial cells. Given that LL-37 enhances chemotactic attraction and modulates the activity of DCs, it is conceivable that it might play a role as an immune adjuvant by skewing the immune environment toward immunostimulatory conditions. In this study, we characterized the mucosal adjuvant activity of LL-37 using model and pathogenic Ags. When LL-37-conjugated Ag was administered orally to mice, a tolerogenic Peyer's patch environment was altered to cell populations containing IL-6-secreting CD11c(+), CD11c(+) CD70(+), and Th17 cells capable of evoking a subsequent LL-37-conjugated Ag-specific immune response in both systemic and mucosal immune compartments. In addition, we showed presentation of formyl peptide receptor, an LL-37 receptor, on M cells, which may aid the initiation of an LL-37-mediated enhanced immune response through targeting and transcytosis of the conjugated Ag. Based on our findings, we conclude that LL-37 has potential as an oral mucosal adjuvant, not only by enhancing the delivery of LL-37-conjugated Ag to M cells, but also by triggering T-cell-mediated Ag-specific immune responses through modulation of the mucosal immune environment.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology, Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea.,Research Center of Bioactive Materials, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Korea
| | - In-Young Yang
- Department of Molecular Biology, Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea
| | - Ju Kim
- Jeonju Biomaterials Institute, Chonbuk National University, Jeonju, Korea
| | - Kyung-Yeol Lee
- Department of Oral Microbiology, Institute of Oral Bioscience, Chonbuk National University, Jeonju, Korea
| | - Yong-Suk Jang
- Department of Molecular Biology, Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea.,Research Center of Bioactive Materials, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Korea
| |
Collapse
|
24
|
|
25
|
Chu DK, Jimenez-Saiz R, Verschoor CP, Walker TD, Goncharova S, Llop-Guevara A, Shen P, Gordon ME, Barra NG, Bassett JD, Kong J, Fattouh R, McCoy KD, Bowdish DM, Erjefält JS, Pabst O, Humbles AA, Kolbeck R, Waserman S, Jordana M. Indigenous enteric eosinophils control DCs to initiate a primary Th2 immune response in vivo. ACTA ACUST UNITED AC 2014; 211:1657-72. [PMID: 25071163 PMCID: PMC4113937 DOI: 10.1084/jem.20131800] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Eosinophils natively inhabit the small intestine, but a functional role for them there has remained elusive. Here, we show that eosinophil-deficient mice were protected from induction of Th2-mediated peanut food allergy and anaphylaxis, and Th2 priming was restored by reconstitution with il4(+/+) or il4(-/-) eosinophils. Eosinophils controlled CD103(+) dendritic cell (DC) activation and migration from the intestine to draining lymph nodes, events necessary for Th2 priming. Eosinophil activation in vitro and in vivo led to degranulation of eosinophil peroxidase, a granule protein whose enzymatic activity promoted DC activation in mice and humans in vitro, and intestinal and extraintestinal mouse DC activation and mobilization to lymph nodes in vivo. Further, eosinophil peroxidase enhanced responses to ovalbumin seen after immunization. Thus, eosinophils can be critical contributors to the intestinal immune system, and granule-mediated shaping of DC responses can promote both intestinal and extraintestinal adaptive immunity.
Collapse
Affiliation(s)
- Derek K Chu
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Rodrigo Jimenez-Saiz
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Christopher P Verschoor
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Tina D Walker
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Susanna Goncharova
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Alba Llop-Guevara
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Pamela Shen
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Melissa E Gordon
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Nicole G Barra
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Jennifer D Bassett
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Joshua Kong
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Ramzi Fattouh
- Clinical Microbiology, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario M5S 1A8, Canada
| | - Kathy D McCoy
- Maurice Müller Laboratories, Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM), University of Bern, 3008 Bern, Switzerland
| | - Dawn M Bowdish
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Jonas S Erjefält
- Department of Experimental Medical Science, Lund University, SE-22184 Lund, Sweden Department of Respiratory Medicine and Allergology, Lund University Hospital, SE-22185 Lund, Sweden
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Alison A Humbles
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MA 20878
| | - Roland Kolbeck
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MA 20878
| | - Susan Waserman
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Manel Jordana
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, and Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
26
|
Escherichia coli heat-labile detoxified enterotoxin modulates dendritic cell function and attenuates allergic airway inflammation. PLoS One 2014; 9:e90293. [PMID: 24637787 PMCID: PMC3956462 DOI: 10.1371/journal.pone.0090293] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 01/31/2014] [Indexed: 12/21/2022] Open
Abstract
Various mutant forms of Escherichia coli heat-labile enterotoxin (LT) have been used as a mucosal adjuvant for vaccines, as it enhances immune responses to specific antigens including antigen-specific IgA antibodies when administrated intranasally or orally. We hypothesized that a detoxified mutant form of LT, LTS61K, could modulate dendritic cell (DC) function and alleviate allergen-induced airway inflammation. Two protocols, preventative and therapeutic, were used to evaluate the effects of LTS61K in a Dermatophagoides pteronyssinus (Der p)-sensitized and challenged murine model of asthma. LTS61K or Der p-primed bone marrow-derived dendritic cells (BMDCs) were also adoptively transferred into Der p-sensitized and challenged mice. Intranasal inoculations with LTS61K or LTS61K/Der p decreased allergen-induced airway inflammation and alleviated systemic TH2-type immune responses. Bronchoalveolar lavage fluid (BALF) and sera from LTS61K/Der p-treated mice also had higher concentrations of Der p-specific immunoglobulin (Ig) A than those of other groups. In vitro, BMDCs stimulated with Der p underwent cellular maturation and secreted proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)α In contrast, Der p-stimulated BMDCs that were pretreated with LTS61K showed decreased IL-6 and TNFα production and were less mature. Intratracheal adoptive transfer of LTS61K- or LTS61K/Der p-primed BMDCs into Der p-sensitized mice reduced inflammatory cell infiltration and TH2-type chemokines in BALF and alleviated airway inflammation in treated mice. LTS61K influenced DC maturation and decreased inflammatory cytokine production. Moreover, LTS61K/Der p induced increased Der p-specific IgA production to decrease allergic TH2 cytokine responses and alleviated airway inflammation in Der p-sensitized mice. These results suggest that the immunomodulatory effects of LTS61K may have clinical applications for allergy and asthma treatment.
Collapse
|
27
|
Jiao J, Dragomir AC, Kocabayoglu P, Rahman AH, Chow A, Hashimoto D, Leboeuf M, Kraus T, Moran T, Carrasco-Avino G, Friedman SL, Merad M, Aloman C. Central role of conventional dendritic cells in regulation of bone marrow release and survival of neutrophils. THE JOURNAL OF IMMUNOLOGY 2014; 192:3374-82. [PMID: 24591364 DOI: 10.4049/jimmunol.1300237] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neutrophils are the most abundant cell type in the immune system and play an important role in the innate immune response. Using a diverse range of mouse models with either defective dendritic cell (DC) development or conditional DC depletion, we provide in vivo evidence indicating that conventional DCs play an important role in the regulation of neutrophil homeostasis. Flk2, Flt3L, and Batf3 knockout mice, which have defects in DC development, have increased numbers of liver neutrophils in the steady state. Conversely, neutrophil frequency is reduced in DC-specific PTEN knockout mice, which have an expansion of CD8(+) and CD103(+) DCs. In chimeric CD11c-DTR mice, conventional DC depletion results in a systemic increase of neutrophils in peripheral organs in the absence of histological inflammation or an increase in proinflammatory cytokines. This effect is also present in splenectomized chimeric CD11c-DTR mice and is absent in chimeric mice with 50% normal bone marrow. In chimeric CD11c-DTR mice, diphtheria toxin treatment results in enhanced neutrophil trafficking from the bone marrow into circulation and increased neutrophil recruitment. Moreover, there is an increased expression of chemokines/cytokines involved in neutrophil homeostasis and reduced neutrophil apoptosis. These data underscore the role of the DC pool in regulating the neutrophil compartment in nonlymphoid organs.
Collapse
Affiliation(s)
- Jingjing Jiao
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Mavropoulos A, Bogdanos DP, Liaskos C, Orfanidou T, Simopoulou T, Zafiriou E, Sakkas LI, Rigopoulou EI. Flow cytometric detection of p38 MAPK phosphorylation and intracellular cytokine expression in peripheral blood subpopulations from patients with autoimmune rheumatic diseases. J Immunol Res 2014; 2014:671431. [PMID: 24741615 PMCID: PMC3987974 DOI: 10.1155/2014/671431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 11/13/2013] [Indexed: 12/11/2022] Open
Abstract
Flow cytometric analysis of p38 mitogen-activated protein kinase (p38 MAPK) signaling cascade is optimally achieved by methanol permeabilization protocols. Such protocols suffer from the difficulties to accurately detect intracellular cytokines and surface epitopes of infrequent cell subpopulations, which are removed by methanol. To overcome these limitations, we have modified methanol-based phosphoflow protocols using several commercially available antibody clones suitable for surface antigens, intracellular cytokines, and p38 MAPK. These included markers of B cells (CD19, CD20, and CD22), T cells (CD3, CD4, and CD8), NK (CD56 and CD7), and dendritic cells (CD11c). We have also tested surface markers of costimulatory molecules, such as CD27. We have successfully determined simultaneous expression of IFN- γ , as well as IL-10, and phosphorylated p38 in cell subsets. The optimized phosphoflow protocol has also been successfully applied in peripheral blood mononuclear cells or purified cell subpopulations from patients with various autoimmune diseases. In conclusion, our refined phosphoflow cytometric approach allows simultaneous detection of p38 MAPK activity and intracellular cytokine expression and could be used as an important tool to study signaling cascades in autoimmunity.
Collapse
Affiliation(s)
- Athanasios Mavropoulos
- Department of Rheumatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research & Technology Hellas (CE.R.T.H.)/Institute for Research and Technology-Thessaly (I.RE.TE.TH), 41222 Larissa, Greece
| | - Dimitrios P. Bogdanos
- Department of Rheumatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research & Technology Hellas (CE.R.T.H.)/Institute for Research and Technology-Thessaly (I.RE.TE.TH), 41222 Larissa, Greece
- Division of Transplantation Immunology and Mucosal Biology, Institute of Liver Studies, King's College London School of Medicine King's College Hospital, London SE5 9RS, UK
| | - Christos Liaskos
- Department of Rheumatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research & Technology Hellas (CE.R.T.H.)/Institute for Research and Technology-Thessaly (I.RE.TE.TH), 41222 Larissa, Greece
| | - Timoklia Orfanidou
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research & Technology Hellas (CE.R.T.H.)/Institute for Research and Technology-Thessaly (I.RE.TE.TH), 41222 Larissa, Greece
| | - Theodora Simopoulou
- Department of Rheumatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Efterpi Zafiriou
- Department of Dermatology, School of Health Sciences, Faculty of Medicine, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Lazaros I. Sakkas
- Department of Rheumatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Eirini I. Rigopoulou
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Thessaly, Biopolis, 41110 Larissa, Greece
| |
Collapse
|
29
|
Cherif MS, Shuaibu MN, Kodama Y, Kurosaki T, Helegbe GK, Kikuchi M, Ichinose A, Yanagi T, Sasaki H, Yui K, Tien NH, Karbwang J, Hirayama K. Nanoparticle formulation enhanced protective immunity provoked by PYGPI8p-transamidase related protein (PyTAM) DNA vaccine in Plasmodium yoelii malaria model. Vaccine 2014; 32:1998-2006. [PMID: 24440206 DOI: 10.1016/j.vaccine.2014.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/23/2013] [Accepted: 01/02/2014] [Indexed: 11/15/2022]
Abstract
We have previously reported the new formulation of polyethylimine (PEI) with gamma polyglutamic acid (γ-PGA) nanoparticle (NP) to have provided Plasmodium yoelii merozoite surface protein-1 (PyMSP-1) plasmid DNA vaccine with enhanced protective cellular and humoral immunity in the lethal mouse malaria model. PyGPI8p-transamidase-related protein (PyTAM) was selected as a possible candidate vaccine antigen by using DNA vaccination screening from 29 GPI anchor and signal sequence motif positive genes picked up using web-based bioinformatics tools; though the observed protection was not complete. Here, we observed augmented protective effect of PyTAM DNA vaccine by using PEI and γ-PGA complex as delivery system. NP-coated PyTAM plasmid DNA immunized mice showed a significant survival rate from lethal P. yoelii challenge infection compared with naked PyTAM plasmid or with NP-coated empty plasmid DNA group. Antigen-specific IgG1 and IgG2b subclass antibody levels, proportion of CD4 and CD8T cells producing IFN-γ in the splenocytes and IL-4, IFN-γ, IL-12 and TNF-α levels in the sera and in the supernatants from ex vivo splenocytes culture were all enhanced by the NP-coated PyTAM DNA vaccine. These data indicates that NP augments PyTAM protective immune response, and this enhancement was associated with increased DC activation and concomitant IL-12 production.
Collapse
Affiliation(s)
- Mahamoud Sama Cherif
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Institut National de Santé Publique, Université de Conakry, Guinea
| | - Mohammed Nasir Shuaibu
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | | | | | - Gideon Kofi Helegbe
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Mihoko Kikuchi
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Akitoyo Ichinose
- Electron Microscopy Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Tetsuo Yanagi
- Animal Research Center for Tropical Medicine, Nagasaki, Japan
| | - Hitoshi Sasaki
- Department of Hospital Pharmacy, Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Nguyen Huy Tien
- Department of Product Development, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Juntra Karbwang
- Department of Product Development, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.
| |
Collapse
|
30
|
McCormick S, Shaler CR, Xing Z. Pulmonary mucosal dendritic cells in T-cell activation: implications for TB therapy. Expert Rev Respir Med 2014; 5:75-85. [DOI: 10.1586/ers.10.81] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
31
|
Shin MK, Lee WJ, Jung MH, Cha SB, Shin SW, Yoo A, Kim DH, Yoo HS. Oral immunization of mice with Saccharomyces cerevisiae expressing a neutralizing epitope of ApxIIA exotoxin from Actinobacillus pleuropneumoniae induces systemic and mucosal immune responses. Microbiol Immunol 2014; 57:417-25. [PMID: 23773020 DOI: 10.1111/1348-0421.12053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 03/21/2013] [Accepted: 04/02/2013] [Indexed: 12/24/2022]
Abstract
An oral delivery system based on ApxIIA#5-expressed on Saccharomyces cerevisiae was studied for its potential to induce immune responses in mice. Murine bone marrow-derived dendritic cells (DCs) stimulated in vitro with ApxIIA#5-expressed on S. cerevisiae upregulated the expression of maturation and activation markers, leading to production of tumor necrosis factor-α, interleukin (IL)-1β, IL-12p70 and IL-10. Presentation of these activated DCs to cluster of differentiation CD4+ T cells collected from mice that had been orally immunized with the ApxIIA#5-expressed on S. cerevisiae elicited specific T-cell proliferation. In addition, the orally immunized mice had stronger antigen-specific serum IgG and IgA antibody responses and larger numbers of antigen-specific IgG and IgA antibody-secreting cells in their spleens, Peyer's patches and lamina propria than did those immunized with vector-only S. cerevisiae or those not immunized. Furthermore, oral immunization induced T helper 1-type immune responses mediated via increased serum concentrations of IgG2a and an increase predominantly of IFN-γ-producing cells in their spleens and lamina propria. Our findings suggest that surface-displayed ApxIIA#5-expressed on S. cerevisiae may be a promising candidate for an oral vaccine delivery system for eliciting systemic and mucosal immunity.
Collapse
Affiliation(s)
- Min-Kyoung Shin
- Department of Infectious Disease, College of Veterinary Medicine, Brain Korea 21 Program for Veterinary Science, Seoul National University, Gwanak 599 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kumamoto Y, Linehan M, Weinstein JS, Laidlaw BJ, Craft JE, Iwasaki A. CD301b⁺ dermal dendritic cells drive T helper 2 cell-mediated immunity. Immunity 2013; 39:733-43. [PMID: 24076051 DOI: 10.1016/j.immuni.2013.08.029] [Citation(s) in RCA: 299] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 07/29/2013] [Indexed: 12/24/2022]
Abstract
Unlike other types of T helper (Th) responses, whether the development of Th2 cells requires instruction from particular subset of dendritic cells (DCs) remains unclear. By using an in vivo depletion approach, we have shown that DCs expressing CD301b were required for the generation of Th2 cells after subcutaneous immunization with ovalbumin (OVA) along with papain or alum. CD301b⁺ DCs are distinct from epidermal or CD207⁺ dermal DCs (DDCs) and were responsible for transporting antigen injected subcutaneously with Th2-type adjuvants. Transient depletion of CD301b⁺ DCs resulted in less effective accumulation and decreased expression of CD69 by polyclonal CD4⁺ T cells in the lymph node. Moreover, despite intact cell division and interferon-γ production, CD301b⁺ DC depletion led to blunted interleukin-4 production by OVA-specific OT-II transgenic CD4⁺ T cells and significantly impaired Th2 cell development upon infection with Nippostrongylus brasiliensis. These results reveal CD301b⁺ DDCs as the key mediators of Th2 immunity.
Collapse
Affiliation(s)
- Yosuke Kumamoto
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
33
|
Obata T, Shibata N, Goto Y, Ishikawa I, Sato S, Kunisawa J, Kiyono H. Critical role of dendritic cells in T cell retention in the interfollicular region of Peyer's patches. THE JOURNAL OF IMMUNOLOGY 2013; 191:942-8. [PMID: 23772027 DOI: 10.4049/jimmunol.1200636] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Peyer's patches (PPs) simultaneously initiate active and quiescent immune responses in the gut. The immunological function is achieved by the rigid regulation of cell distribution and trafficking, but how the cell distribution is maintained remains to be elucidated. In this study, we show that binding of stromal cell-derived lymphoid chemokines to conventional dendritic cells (cDCs) is essential for the retention of naive CD4(+) T cells in the interfollicular region (IFR) of PPs. Transitory depletion of CD11c(high) cDCs in mice rapidly impaired the IFR structure in the PPs without affecting B cell follicles or germinal centers, lymphoid chemokine production from stromal cells, or the immigration of naive T cells into the IFRs of PPs. The cDC-orchestrated retention of naive T cells was mediated by heparinase-sensitive molecules that were expressed on cDCs and bound the lymphoid chemokine CCL21 produced from stromal cells. These data collectively reveal that interactions among cDCs, stromal cells, and naive T cells are necessary for the formation of IFRs in the PPs.
Collapse
Affiliation(s)
- Takashi Obata
- Division of Mucosal Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Gustafsson T, Hua YJ, Dahlgren MW, Livingston M, Johansson-Lindbom B, Yrlid U. Direct interaction between cholera toxin and dendritic cells is required for oral adjuvant activity. Eur J Immunol 2013; 43:1779-88. [DOI: 10.1002/eji.201242867] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Revised: 04/18/2013] [Accepted: 04/30/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Tobias Gustafsson
- Department of Microbiology and Immunology; Institute of Biomedicine; University of Gothenburg; Gothenburg; Sweden
| | - Yeu-Jiann Hua
- Department of Microbiology and Immunology; Institute of Biomedicine; University of Gothenburg; Gothenburg; Sweden
| | | | - Megan Livingston
- Department of Microbiology and Immunology; Institute of Biomedicine; University of Gothenburg; Gothenburg; Sweden
| | | | - Ulf Yrlid
- Department of Microbiology and Immunology; Institute of Biomedicine; University of Gothenburg; Gothenburg; Sweden
| |
Collapse
|
35
|
Kaparakis-Liaskos M, Tate MD, Price JD, Pearse M, Wijburg OLC. Increased antigen specific T cell numbers in the absence of altered migration or division rates as a result of mucosal cholera toxin administration. PLoS One 2013; 8:e59934. [PMID: 23544110 PMCID: PMC3609821 DOI: 10.1371/journal.pone.0059934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 02/22/2013] [Indexed: 11/18/2022] Open
Abstract
Cholera toxin (CT) is a mucosal adjuvant capable of inducing strong immune responses to co-administered antigens following oral or intranasal immunization of mice. To date, the direct effect of CT on antigen-specific CD4(+) T cell migration and proliferation profiles in vivo is not well characterized. In this study, the effect of CT on the migration pattern and proliferative responses of adoptively transferred, CD4(+) TCR transgenic T cells in orally or intranasally vaccinated mice, was analyzed by flow cytometry. GFP-expressing or CFSE-labeled OT-II lymphocytes were adoptively transferred to naïve C57BL/6 mice, and mice were subsequently vaccinated with OVA with or without CT via the oral or intranasal route. CT did not alter the migration pattern of antigen-specific T cells, regardless of the route of immunization, but increased the number of transgenic CD4(+) T cells in draining lymphoid tissue. This increase in the number of transgenic CD4(+) T cells was not due to cells undergoing more rounds of cellular division in vivo, suggesting that CT may exert an indirect adjuvant effect on CD4(+) T cells. The findings reported here suggest that CT functions as a mucosal adjuvant by increasing the number of antigen specific CD4(+) T cells independent of their migration pattern or kinetics of cellular division.
Collapse
Affiliation(s)
- Maria Kaparakis-Liaskos
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Clayton, Victoria, Australia
| | - Michelle D. Tate
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Clayton, Victoria, Australia
| | - Jason D. Price
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Odilia L. C. Wijburg
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
36
|
Utriainen L, Firmin D, Wright P, Cerovic V, Breban M, McInnes I, Milling S. Expression of HLA-B27 causes loss of migratory dendritic cells in a rat model of spondylarthritis. ARTHRITIS AND RHEUMATISM 2012; 64:3199-209. [PMID: 22674414 PMCID: PMC3553565 DOI: 10.1002/art.34561] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE In rats transgenic for human HLA-B27 and β(2) -microglobulin (B27-transgenic rats), colitis and peripheral inflammation develop spontaneously. Therefore, B27-transgenic rats provide a model of spondylarthritis. Because inflammation in these rats requires CD4+ T lymphocytes and involves intestinal pathology, we hypothesized that dendritic cells (DCs) that migrate from the intestine and control CD4+ T cell differentiation would be aberrant in B27-transgenic rats. METHODS Migrating intestinal lymph DCs were collected via thoracic duct cannulation from B27-transgenic and control (HLA-B7-transgenic or nontransgenic) rats. The phenotypes of these DCs and of mesenteric lymph node DCs were assessed by flow cytometry. The ability of DCs to differentiate from bone marrow precursors in vitro was also assessed. RESULTS Lymph DCs showed increased activation and, strikingly, lacked the specific DC population that is important for maintaining tolerance to self-antigens. This population of DCs was also depleted from the mesenteric lymph nodes of B27-transgenic rats. Furthermore, in vitro culture of DCs from bone marrow precursors revealed a defect in the ability of B27-transgenic rats to produce DCs of the migratory phenotype, although the DCs that were generated induced enhanced interleukin-17 (IL-17) production from naive CD4+ T cells. CONCLUSION We describe 2 different mechanisms by which HLA-B27 may contribute to inflammatory disease: increased apoptotic death of B27-transgenic DCs that normally function to maintain immunologic tolerance and enhanced IL-17 production from CD4+ T cells stimulated by the surviving B27-transgenic DCs.
Collapse
Affiliation(s)
| | | | | | | | - Maxime Breban
- Institut Cochin, Université Paris Descartes, and CNRS (UMR 8104), INSERM U1016, Paris, France, and Hôpital Ambroise Paré, AP-HP, and Université de Versailles Saint Quentin en Yvelines, Boulogne-Billancourt, France
| | | | | |
Collapse
|
37
|
Rhee JH, Lee SE, Kim SY. Mucosal vaccine adjuvants update. Clin Exp Vaccine Res 2012; 1:50-63. [PMID: 23596577 PMCID: PMC3623511 DOI: 10.7774/cevr.2012.1.1.50] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 05/25/2012] [Accepted: 06/15/2012] [Indexed: 12/16/2022] Open
Abstract
Mucosal vaccination, capable of inducing protective immune responses both in the mucosal and systemic immune compartments, has many advantages and is regarded as a blue ocean in the vaccine industry. Mucosal vaccines can offer lower costs, better accessability, needle-free delivery, and higher capacity of mass immunizations during pandemics. However, only very limited number of mucosal vaccines was approved for human use in the market yet. Generally, induction of immune responses following mucosal immunization requires the co-administration of appropriate adjuvants that can initiate and support the effective collaboration between innate and adaptive immunity. Classically, adjuvant researches were rather empirical than keenly scientific. However, during last several years, fundamental scientific achievements in innate immunity have been translated into the development of new mucosal adjuvants. This review focuses on recent developments in the concepts of adjuvants and innate immunity, mucosal immunity with special interest of vaccine development, and basic and applied researches in mucosal adjuvant.
Collapse
Affiliation(s)
- Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. ; Department of Microbiology and Research Institute of Vibrio Infections, Chonnam National University Medical School, Gwangju, Korea
| | | | | |
Collapse
|
38
|
Lycke N. Recent progress in mucosal vaccine development: potential and limitations. Nat Rev Immunol 2012; 12:592-605. [DOI: 10.1038/nri3251] [Citation(s) in RCA: 495] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
The A subunit of Escherichia coli heat-labile enterotoxin functions as a mucosal adjuvant and promotes IgG2a, IgA, and Th17 responses to vaccine antigens. Infect Immun 2012; 80:2426-35. [PMID: 22526674 DOI: 10.1128/iai.00181-12] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) produces both heat-labile (LT) and heat-stable (ST) enterotoxins and is a major cause of diarrhea in infants in developing countries and in travelers to those regions. In addition to inducing fluid secretion, LT is a powerful mucosal adjuvant capable of promoting immune responses to coadministered antigens. In this study, we examined purified A subunit to further understand the toxicity and adjuvanticity of LT. Purified A subunit was enzymatically active but sensitive to proteolytic degradation and unable to bind gangliosides, and even in the presence of admixed B subunit, it displayed low cyclic AMP (cAMP) induction and no enterotoxicity. Thus, the AB5 structure plays a key role in protecting the A subunit from proteolytic degradation and in delivering the enzymatic signals required for secretion. In contrast, the A subunit alone was capable of activating dendritic cells and enhanced immune responses to multiple antigens following intranasal immunization; therefore, unlike toxicity, LT adjuvanticity is not dependent on the AB5 holotoxin structure or the presence of the B subunit. However, immune responses were maximal when signals were received from both subunits either in an AB5 structure or with A and B admixed. Furthermore, the quality of the immune response (i.e., IgG1/IgG2 balance and mucosal IgA and IL-17 secretion) was determined by the presence of an A subunit, revealing for the first time induction of Th17 responses with the A subunit alone. These results have important implications for understanding ETEC pathogenesis, unraveling immunologic responses induced by LT-based adjuvants, and developing new mucosal vaccines.
Collapse
|
40
|
Replicating adenovirus-simian immunodeficiency virus (SIV) vectors efficiently prime SIV-specific systemic and mucosal immune responses by targeting myeloid dendritic cells and persisting in rectal macrophages, regardless of immunization route. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:629-37. [PMID: 22441384 DOI: 10.1128/cvi.00010-12] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Although priming with replicating adenovirus type 5 host range mutant (Ad5hr)-human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) recombinants, followed by HIV/SIV envelope boosting, has proven highly immunogenic, resulting in protection from SIV/simian-human immunodeficiency virus (SHIV) challenges, Ad5hr recombinant distribution, replication, and persistence have not been examined comprehensively in nonhuman primates. We utilized Ad5hr-green fluorescent protein and Ad5hr-SIV recombinants to track biodistribution and immunogenicity following mucosal priming of rhesus macaques by the intranasal/intratracheal, sublingual, vaginal, or rectal route. Ad recombinants administered by all routes initially targeted macrophages in bronchoalveolar lavage (BAL) fluid and rectal tissue, later extending to myeloid dendritic cells in BAL fluid with persistent expression in rectal mucosa 25 weeks after the last Ad immunization. Comparable SIV-specific immunity, including cellular responses, serum binding antibody, and mucosal secretory IgA, was elicited among all groups. The ability of the vector to replicate in multiple mucosal sites irrespective of delivery route, together with the targeting of macrophages and professional antigen-presenting cells, which provide potent immunogenicity at localized sites of virus entry, warrants continued use of replicating Ad vectors.
Collapse
|
41
|
Westlund J, Livingston M, Fahlén-Yrlid L, Oldenborg PA, Yrlid U. CD47-deficient mice have decreased production of intestinal IgA following oral immunization but a maintained capacity to induce oral tolerance. Immunology 2012; 135:236-44. [PMID: 22070457 DOI: 10.1111/j.1365-2567.2011.03536.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Signal regulatory protein α (SIRPα/CD172a), expressed by myeloid cells including CD11b(+) dendritic cells, interacts with ubiquitously expressed CD47 to mediate cell-cell signalling and therefore, may be pivotal in the development of tolerance or immunity. We show that in mice deficient in CD47 (CD47(-/-) ) the cellularity in gut-associated lymphoid tissues is reduced by 50%. In addition, the frequency of CD11b(+) CD172a(+) dendritic cells is significantly reduced in the gut and mesenteric lymph nodes, but not in Peyer's patches. Activation of ovalbumin (OVA)-specific CD4(+) T cells in the mesenteric lymph nodes after feeding OVA is reduced in CD47(-/-) mice compared with wild-type however, induction of oral tolerance is maintained. The addition of cholera toxin generated normal serum anti-OVA IgG and IgA titres but resulted in reduced intestinal anti-OVA IgA in CD47(-/-) mice. Replacing the haematopoietic compartment in CD47(-/-) mice with wild-type cells restored neither the cellularity in gut-associated lymphoid tissues nor the capacity to produce intestinal anti-OVA IgA following immunization. This study demonstrates that CD47 signalling is dispensable for oral tolerance induction, whereas the expression of CD47 by non-haematopoietic cells is required for intestinal IgA B-cell responses. This suggests that differential CD4 T cell functions control tolerance and enterotoxin-induced IgA immunity in the gut.
Collapse
Affiliation(s)
- Jessica Westlund
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
42
|
Bemark M, Boysen P, Lycke NY. Induction of gut IgA production through T cell-dependent and T cell-independent pathways. Ann N Y Acad Sci 2012; 1247:97-116. [PMID: 22260403 DOI: 10.1111/j.1749-6632.2011.06378.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gut immune system protects against mucosal pathogens, maintains a mutualistic relationship with the microbiota, and establishes tolerance against food antigens. This requires a balance between immune effector responses and induction of tolerance. Disturbances of this strictly regulated balance can lead to infections or the development inflammatory diseases and allergies. Production of secretory IgA is a unique effector function at mucosal surfaces, and basal mechanisms regulating IgA production have been the focus of much recent research. These investigations have aimed at understanding how long-term IgA-mediated mucosal immunity can best be achieved by oral or sublingual vaccination, or at analyzing the relationship between IgA production, the composition of the gut microbiota, and protection from allergies and autoimmunity. This research has lead to a better understanding of the IgA system; but at the same time seemingly conflicting data have been generated. Here, we discuss how gut IgA production is controlled, with special focus on how differences between T cell-dependent and T cell-independent IgA production may explain some of these discrepancies.
Collapse
Affiliation(s)
- Mats Bemark
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| | | | | |
Collapse
|
43
|
Abstract
The innate immune system consists of multiple cell types that express germline-encoded pattern recognition receptors that recognize pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). Allergens are frequently found in forms and mixtures that contain PAMPs and DAMPs. The innate immune system is interposed between the external environment and the internal acquired immune system. It is also an integral part of the airways, gut, and skin. These tissues face continuous exposure to allergens, PAMPs, and DAMPs. Interaction of allergens with the innate immune system normally results in immune tolerance but, in the case of allergic disease, this interaction induces recurring and/or chronic inflammation as well as the loss of immunologic tolerance. Upon activation by allergens, the innate immune response commits the acquired immune response to a variety of outcomes mediated by distinct T-cell subsets, such as T-helper 2, regulatory T, or T-helper 17 cells. New studies highlighted in this review underscore the close relationship between allergens, the innate immune system, and the acquired immune system that promotes homeostasis versus allergic disease.
Collapse
Affiliation(s)
- Michael Minnicozzi
- Asthma, Allergy and Inflammation Branch, Division of Allergy, Immunology, and Transplantation, Department of Health and Human Services, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-6601, USA
| | | | | |
Collapse
|
44
|
Kushwah R, Oliver JR, Wu J, Chang Z, Hu J. Elf3 regulates allergic airway inflammation by controlling dendritic cell-driven T cell differentiation. THE JOURNAL OF IMMUNOLOGY 2011; 187:4639-53. [PMID: 21948981 DOI: 10.4049/jimmunol.1101967] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Elf3 belongs to the Ets family of transcription factors and has been implicated in inflammation. Elf3 is highly expressed in the lungs, and Elf3(-/-) mice are impaired in IL-6 production after intranasal LPS exposure. To identify the role of Elf3 in Th17-driven pulmonary inflammation, we have performed epicutaneous sensitization of Elf3(-/-) mice with OVA followed by airway OVA challenge and have identified Elf3(-/-) mice to be impaired in induction of Th17 response, attributable to impairment of IL-6 production by dendritic cells (DCs). However, increased serum levels of OVA-specific IgG1 and IgE were observed, pointing toward an exaggerated Th2 response. To study Th2 response, we performed i.p. sensitization of Elf3(-/-) mice with OVA and confirmed loss of Elf3 to result in an aggravated Th2 response, characterized by increased generation of IL-4-producing T cells, increased levels of OVA-specific IgE and IgG1 Ab titers, and increased serum levels of Th2 cytokines, together with extensive inflammation and mucus production in airways. Elf3(-/-) DCs were impaired in priming Th1 differentiation, which, in turn, promoted Th2 differentiation. This was mediated by the ability of Elf3(-/-) DCs to undergo hypermaturation but secrete significantly lower levels of IL-12 in response to inflammatory stimuli. The impairment of IL-12 production was due to impairment of IL-12p40 gene induction in Elf3(-/-) DCs in response to inflammatory stimuli. Taken together, our study identifies a novel function of Elf3 in regulating allergic airway inflammation by regulating DC-driven Th1, Th2, and Th17 differentiation.
Collapse
Affiliation(s)
- Rahul Kushwah
- Physiology and Experimental Medicine Research Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | | | | | |
Collapse
|
45
|
Duthie MS, Windish HP, Fox CB, Reed SG. Use of defined TLR ligands as adjuvants within human vaccines. Immunol Rev 2011; 239:178-96. [PMID: 21198672 DOI: 10.1111/j.1600-065x.2010.00978.x] [Citation(s) in RCA: 326] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our improved understanding of how innate immune responses can be initiated and how they can shape adaptive B- and T-cell responses is having a significant impact on vaccine development by directing the development of defined adjuvants. Experience with first generation vaccines, as well as rapid advances in developing defined vaccines containing Toll-like receptor ligands (TLRLs), indicate that an expanded number of safe and effective vaccines containing such molecules will be available in the future. In this review, we outline current knowledge regarding TLRs, detailing the different cell types that express TLRs, the various signaling pathways TLRs utilize, and the currently known TLRLs. We then discuss the current status of TLRLs within vaccine development programs, including the importance of appropriate formulation, and how recent developments can be used to better define the mechanisms of action of vaccines. Finally, we introduce the possibility of using TLRLs, either in combination or with non-TLRLs, to synergistically potentiate vaccine-induced responses to provide not only prophylactic, but therapeutic protection against infectious diseases and cancer.
Collapse
|
46
|
The subcellular location of antigen expressed by adenoviral vectors modifies adaptive immunity but not dependency on cross-presenting dendritic cells. Eur J Immunol 2011; 41:2185-96. [DOI: 10.1002/eji.201041009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 03/06/2011] [Accepted: 04/29/2011] [Indexed: 11/07/2022]
|
47
|
Smith KA, Hochweller K, Hämmerling GJ, Boon L, MacDonald AS, Maizels RM. Chronic helminth infection promotes immune regulation in vivo through dominance of CD11cloCD103- dendritic cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:7098-109. [PMID: 21576507 DOI: 10.4049/jimmunol.1003636] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Gastrointestinal helminth infections are extremely prevalent in many human populations and are associated with downmodulated immune responsiveness. In the experimental model system of Heligmosomoides polygyrus, a chronic infection establishes in mice, accompanied by a modulated Th2 response and increased regulatory T cell (Treg) activity. To determine if dendritic cell (DC) populations in the lymph nodes draining the intestine are responsible for the regulatory effects of chronic infection, we first identified a population of CD11c(lo) nonplasmacytoid DCs that expand after chronic H. polygyrus infection. The CD11c(lo) DCs are underrepresented in magnetic bead-sorted preparations and spared from deletion in CD11c-diptheria toxin receptor mice. After infection, CD11c(lo) DCs did not express CD8, CD103, PDCA, or Siglec-H and were poorly responsive to TLR stimuli. In DC/T cell cocultures, CD11c(lo) DCs from naive and H. polygyrus-infected mice could process and present protein Ag, but induced lower levels of Ag-specific CD4(+) T cell proliferation and effector cytokine production, and generated higher percentages of Foxp3(+) T cells in the presence of TGF-β. Treg generation was also dependent on retinoic acid receptor signaling. In vivo, depletion of CD11c(hi) DCs further favored the dominance of the CD11c(lo) DC phenotype. After CD11c(hi) DC depletion, effector responses were inhibited dramatically, but the expansion in Treg numbers after H. polygyrus infection was barely compromised, showing a significantly higher regulatory/effector CD4(+) T cell ratio compared with that of CD11c(hi) DC-intact animals. Thus, the proregulatory environment of chronic intestinal helminth infection is associated with the in vivo predominance of a newly defined phenotype of CD11c(lo) tolerogenic DCs.
Collapse
Affiliation(s)
- Katherine A Smith
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | | | | | | | | | | |
Collapse
|
48
|
Lawson LB, Norton EB, Clements JD. Defending the mucosa: adjuvant and carrier formulations for mucosal immunity. Curr Opin Immunol 2011; 23:414-20. [PMID: 21511452 DOI: 10.1016/j.coi.2011.03.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 03/25/2011] [Indexed: 01/10/2023]
Abstract
A majority of infectious microorganisms either colonize or cross mucosal surfaces to enter the host. A major goal in vaccine design is to induce a protective, lasting immune response against potential pathogens at mucosal surfaces. In addition, mucosal vaccines can offer needle-free delivery, thereby improving accessibility, safety, and cost-effectiveness. Challenges to successful mucosal vaccination include poor induction of mucosal immunity, limited understanding of protective mechanisms and crosstalk between mucosal compartments, and the availability of safe, effective mucosal adjuvants and delivery systems. This review focuses on some key advances in the field of mucosal vaccinology within the past 2-3 years, including reports on promising new formulations and investigations into the mechanisms of established mucosal adjuvants and/or particulate carrier systems.
Collapse
Affiliation(s)
- Louise B Lawson
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, United States
| | | | | |
Collapse
|
49
|
Mucosal adjuvants and long-term memory development with special focus on CTA1-DD and other ADP-ribosylating toxins. Mucosal Immunol 2010; 3:556-66. [PMID: 20844480 DOI: 10.1038/mi.2010.54] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The ultimate goal for vaccination is to stimulate protective immunological memory. Protection against infectious diseases not only relies on the magnitude of the humoral immune response, but more importantly on the quality and longevity of it. Adjuvants are critical components of most non-living vaccines. Although little attention has been given to qualitative aspects of the choice of vaccine adjuvant, emerging data demonstrate that this function may be central to vaccine efficacy. In this review we describe efforts to understand more about how adjuvants influence qualitative aspects of memory development. We describe recent advances in understanding how vaccines induce long-lived plasma and memory B cells, and focus our presentation on the germinal center reaction. As mucosal vaccination requires powerful adjuvants, we have devoted much attention to the adenosine diphosphate (ADP)-ribosylating cholera toxin and the CTA1-DD adjuvants as examples of how mucosal adjuvants can influence induction of long-term memory.
Collapse
|
50
|
Abstract
Research has yielded an abundance of vaccine candidates against mucosal infections, but only few mucosal vaccines have been registered for human use. Extensive research is being carried out to identify new and safe adjuvants for mucosal immunization, novel delivery systems, including live vectors and reporter molecules for tissue- and cell-specific targeting of vaccine antigens. If these candidates are to reach those in need, several lessons from clinical and field research carried out under resource-poor settings must be considered. These lessons include the need to develop new vaccines that can be administered topically onto the skin or to the mucosa, without needles or expensive delivery devices. Such topical vaccines must be able to protect all age groups at risk, be safe and effective in immunocompromised people, and be able to contain epidemics following complex emergencies. The anatomical compartmentalization of immune responses imposes constraints on the selection of topical route(s) of vaccine administration and on strategies for measuring these responses, especially in young infants. Thus, the selection of any particular route of immunization is critical when designing and formulating vaccines against organ-specific infections.
Collapse
Affiliation(s)
- C Czerkinsky
- International Vaccine Institute, Seoul, South Korea.
| | | |
Collapse
|