1
|
Huang S, Li X, Cao Y, Mou M, Li J, Zhuo K, Wang L, Zeng Z, Wei X, Tang C, Zhong M. TLR5 activation in respiratory epithelial cells orchestrate mucosal Th17 response through both indirect and direct pathways. Respir Res 2025; 26:104. [PMID: 40098159 PMCID: PMC11916947 DOI: 10.1186/s12931-025-03186-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Flagellin, a potent mucosal adjuvant administered via the intranasal route, has been widely recognized for its capacity to enhance immune responses against diverse pathogens. However, the effects and the underlying mechanisms by which flagellin modulates CD4+ T cell differentiation remain incompletely understood. METHODS Recombinant flagellin proteins, including full-length flagellin (SF) and a TLR5-binding deficient variant (SFΔ90-97), were produced and purified. An OT-II derived CD4+ T cell adoptive transfer model, a classical intranasal immunization model and dendritic cell (DC)-CD4+ T co-culturing system were used. The proliferation and differentiation of CD4+ T cells were analyzed using flow cytometry analysis. RNA sequencing and neutralizing antibody blocking experiments were performed to determine the essential cytokines involved in flagellin modulated Th17 differentiation. RESULTS Flagellin preferentially promotes Th17 cells differentiation. Respiratory epithelial cells (RECs), acting as sentinel cells, are the first to encounter exogenous stimuli during intranasal immunization. Flagellin stimulates the secretion of various soluble cytokines by binding to TLR5 on the surface of RECs, with GM-CSF facilitating the functional activation of airway DCs. GM-CSF-conditioned DCs exhibit upregulated IL-6 expression which in turn drives the polarization of naïve CD4+ T cells toward the Th17 phenotype. Furthermore, TLR5-regulated REC-derived IL-6 synergizes with TLR5-modulated DCs to amplify Th17 polarization signals, thereby enhancing the Th17 induction. CONCLUSION Flagellin preferentially induced a Th17-enhanced immune response and RECs were highlighted its essential roles during this process through both indirect and direct pathways. For indirect pathway, RECs modulate DC function through GM-CSF. Moreover, RECs directly contribute to Th17 differentiation by secreting IL-6.
Collapse
Affiliation(s)
- Sijian Huang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Xu Li
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
- Clinical Laboratory, Wuhan Asia General Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, 430056, China
| | - Yuan Cao
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
- Analytical & Testing Center, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Man Mou
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
- Department of Blood Transfusion, Wuhan Asia General Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, 430056, China
| | - Jianlun Li
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Kexing Zhuo
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Lijuan Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Zihang Zeng
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Xianghong Wei
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Chunlian Tang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China.
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, 430063, China.
| | - Maohua Zhong
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China.
| |
Collapse
|
2
|
Tian Y, Hu L, Huang Q, Qi J, Shen L, Wang G, Yu W, Hu T. A SARS-CoV-2 mucosal nanovaccine based on assembly of maltodextrin, STING agonist and polyethyleneimine. Int J Biol Macromol 2025; 294:139395. [PMID: 39756748 DOI: 10.1016/j.ijbiomac.2024.139395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/17/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
SARS-CoV-2 has the characteristics of strong transmission with severe morbidity and mortality. Protein-based vaccines have the properties of specificity, effectiveness and safety against SARS-CoV-2. Receptor-binding domain (RBD) homotrimer affords high protection efficacy against stringent lethal viral challenge. Mucosal immunity could block the infection that first infect and replicate in the upper airway mucosa. Due to the physical barriers of the mucosa, mucosal vaccines necessitated appropriate adjuvants and delivery system. In the present study, maltodextrin, PEI and 2',3'-cGAMP acted as the mucosal adjuvants and RBD trimer as the antigen. A mucosal nanovaccine was prepared by assembly of adjuvants and the antigen to a nanoparticle. The vaccine elicited strong serum RBD-specific IgG and IgA response, and mild mucosal IgA and IgG response in the respiratory tract. It stimulated strong neutralizing antibody response and high ACE2-blocking activity in the sera. It promoted the RBD-specific CD4+ and CD8+ T cells secreting IFN-γ, IL-4 and IL-17 A. Moreover, it elicited durable RBD-specific memory T and B memory cell response, activated the T and B cells, enhanced the cytotoxic T cell killing effect, and promoted the maturation of DCs. These findings suggested the clinical potential of the vaccine to combat against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yu Tian
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China; State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lijia Hu
- School of International Relations, Beijing Language and Culture University, Beijing 100083, China
| | | | - Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Guosheng Wang
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
3
|
Zhu Y, Yao ZC, Li S, Ma J, Wei C, Yu D, Stelzel JL, Ni BYX, Miao Y, Van Batavia K, Lu X, Lin J, Dai Y, Kong J, Shen R, Goodier KD, Liu X, Cheng L, Vuong I, Howard GP, Livingston NK, Choy J, Schneck JP, Doloff JC, Reddy SK, Hickey JW, Mao HQ. mRNA lipid nanoparticle-incorporated nanofiber-hydrogel composite generates a local immunostimulatory niche for cancer immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.633179. [PMID: 39975373 PMCID: PMC11838205 DOI: 10.1101/2025.01.27.633179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Hydrogel materials have emerged as versatile platforms for various biomedical applications. Notably, the engineered nanofiber-hydrogel composite (NHC) has proven effective in mimicking the soft tissue extracellular matrix, facilitating substantial recruitment of host immune cells and the formation of a local immunostimulatory microenvironment. Leveraging this feature, here we report an mRNA lipid nanoparticle (LNP)-incorporated NHC microgel matrix, termed LiNx, by incorporating LNPs loaded with mRNA encoding tumour antigens. Harnessing the potent transfection efficiency of LNPs in antigen-presenting cells (APCs), LiNx demonstrates remarkable immune cell recruitment, antigen expression and presentation, and cellular interaction. These attributes collectively create an immunostimulating milieu and yield a potent immune response achievable with a single dose, comparable to the conventional three-dose LNP immunization regimen. Further investigations reveal that the LiNx not only generates heightened Th1 and Th2 responses but also elicits a distinctive Type 17 T helper cell-mediated response pivotal for bolstering antitumour efficacy. Our findings elucidate the mechanism underlying LiNx's role in potentiating antigen-specific immune responses, presenting a new strategy for cancer immunotherapy.
Collapse
|
4
|
Zhang C, Li F, Yu X, Tian H, Li Y, Liu X, Liu W, Yu B, Qiao ZA, Yu X. Periodic mesoporous organosilica-loaded mincle agonists enhance the immunogenicity of COVID-19 subunit vaccines by dual activation of B cells and dendritic cells. Acta Biomater 2025; 193:362-376. [PMID: 39730102 DOI: 10.1016/j.actbio.2024.12.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Effective vaccination is crucial for intervening in the COVID-19 pandemic. However, with the continuous mutation of the SARS-CoV-2, existing vaccines including subunit vaccines cannot effectively prevent virus infections. Hence, there is an urgent need to enhance the immunogenicity of existing vaccines to induce a more potent and durable immune response. We previously found that periodic mesoporous organosilica (PMO) could act as a potential nanoadjuvant for subunit vaccines, eliciting potent antigen-specific germinal center (GC) responses by activating naïve B cells. In this study, we describe the design of PMO decorated with TDB, a potent Macrophage-induced C-type lectin (Mincle) agonist, to improve the adjuvanticity of PMO for COVID-19 vaccines. We found that the TDB@PMO adjuvant can effectively deliver antigens to lymph nodes and promote antigen uptake by immune cells. More importantly, the TDB@PMO adjuvant vaccine could activate the innate immune of both naïve B cells and dendritic cells via the Mincle signaling pathway, and further enhance the GC responses and resulting in potent SARS-CoV-2 specific humoral and cellular immune responses. Overall, we have developed an effective and safe nanoadjuvant platform, laying the foundation for the design and development of subunit vaccines against pathogens such as SARS-CoV-2. STATEMENT OF SIGNIFICANCE: Adjuvants play a crucial role in enhancing the effectiveness of vaccines by boosting the immune response. The emergence of highly mutated viruses, such as coronaviruses, has presented new requirements for adjuvant design. This work designed a nanoadjuvant platform, TDB@PMO, to enhance the immune response of the COVID-19 subunit vaccine. The result demonstrated that TDB@PMO nanoadjuvant can simultaneously boost the activation effects of B cells and DC cells through the Mincle signaling pathway. Furthermore, immunization with TDB@PMO-RBD nanoadjuvanted vaccine in mice significantly enhanced germinal center responses and antibody production, while also eliciting a robust antigen-specific T cell immune response in spleen. This design provided a reference for the development of next-generation virus subunit vaccines.
Collapse
Affiliation(s)
- Chunhe Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Haochen Tian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yiyang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinyao Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenmo Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Zhen-An Qiao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun, Jilin 130012, China.
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
5
|
Wang J, Hou Y, Mu L, Yang M, Ai X. Gut microbiota contributes to the intestinal and extraintestinal immune homeostasis by balancing Th17/Treg cells. Int Immunopharmacol 2024; 143:113570. [PMID: 39547012 DOI: 10.1016/j.intimp.2024.113570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Gut microbiota is generally considered to play an important role in host health due to its extensive immunomodulatory activities. Th17 and Treg cells are two important CD4+ T cell subsets involved in immune regulation, and their imbalance is closely tied to many immune diseases. Recently, abundant researches have highlighted the importance of gut microbiota in supporting intestinal and extraintestinal immunity through the balance of Th17 and Treg cells. Here, we presented a comprehensive review of these findings. This review first provided an overview of gut microbiota, along with Th17/Treg cell differentiation and cytokine production. Subsequently, the review summarized the regulatory effects of gut microbiota (in terms of species, components, and metabolites) on the Th17/Treg cell balance in the local intestines and extraintestinal organs, such as lung, liver, brain, kidney, and bone. Specifically, the Th17 and Treg cells that can be modulated by gut microbiota originate not only from the gut and extraintestinal organs, but also from peripheral blood and spleen. Then, the microbial therapeutics, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation (FMT), were also reviewed because of their therapeutic potentials in addressing intestinal and extraintestinal diseases via the Th17/Treg axis. Finally, the review discussed the clinical applications and future study prospects of microbial therapeutics by targeting the Th17/Treg cell balance. In conclusion, this review focused on elucidating the regulatory effects of gut microbiota in balancing Th17/Treg cells to maintain intestinal and extraintestinal immune homeostasis, contributing to the further development and promotion of microbial therapeutics.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Yaqin Hou
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Lifeng Mu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
6
|
Huang N, Ye L, Li H, Peng J, Wei H. Developmental patterns of intestinal group 3 innate lymphoid cells in piglets and their response to enterotoxigenic Escherichia coli infection. Vet Res 2024; 55:159. [PMID: 39695888 DOI: 10.1186/s13567-024-01418-3] [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: 10/06/2023] [Accepted: 09/22/2024] [Indexed: 12/20/2024] Open
Abstract
Diarrhoea and preweaning mortality in piglets are crucial factors impacting the economic sustainability of the swine industry. Pathogenic infections are among the main causes of diarrhea and mortality. Group 3 innate lymphoid cells (ILC3s) are crucial for safeguarding against pathogenic infections. However, knowledge regarding the development and function of ILC3s in suckling piglets is currently limited. Our findings demonstrate that the development of ILC3s in suckling piglets gradually progresses from day 1 to day 21, with a notable increase observed on day 28. Additionally, the development of NKp46+ILC3s and the production of interleukin (IL)-17A by ILC3s displayed consistent patterns with the changes observed in ILC3s. Notably, interferon (IFN)-γ levels significantly increased on day 14. Moreover, the production of IFN-γ by NKp46+ILC3s was greater than that by NKp46-ILC3s. Importantly, when piglets were subjected to a 4-h challenge with enterotoxigenic Escherichia coli, both the percentages of ILC3s significantly increased, accompanied by increased IL-22 production, highlighting their importance in maintaining intestinal health. The outcomes of this study provide valuable insights for future related research.
Collapse
Affiliation(s)
- Ningning Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Center of Cellular and Genetic Sciences, Henan Academy of Sciences, Zhengzhou, 450000, China
| | - Ling Ye
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hao Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
- Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.
| |
Collapse
|
7
|
Zeng Q, Yang Y, Liu Y, Li Z, Li P, Zhou Z. Fish IL-26 collaborates with IL-10R2 and IL-20R1 to enhance gut mucosal barrier during the antibacterial innate immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 161:105249. [PMID: 39154973 DOI: 10.1016/j.dci.2024.105249] [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: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/20/2024]
Abstract
IL-26 is a cytokine that is crucial for the maintenance and function of the gut mucosal barrier. IL-26 signaling pathway relies on a heterodimeric receptor complex, which is composed of two distinct subunits, IL-10R2 and IL-20R1. However, there are no reports on the antibacterial immunity of IL-26 and its receptors in fish. For this purpose, in this study we identified IL-26 and its receptors IL-10R2 and IL-20R1 in Carassius cuvieri × Carassius auratus red var. (named WR-IL-26, WR-IL10R2 and WR-IL20R1, respectively). Phylogenetic analysis confirmed the conservation of these genes, with shared structural motifs similar to those found in higher vertebrates. Upon exposure to Aeromonas hydrophila, a common fish pathogen, there was a significant upregulation of WR-IL-26, WR-IL10R2 and WR-IL20R1 in the gut, indicating a potential role in the immune response to infection. A co-immunoprecipitation assay revealed that WR-IL-26 formed complexes with WR-IL10R2 and WR-IL20R1. In vivo experiments demonstrated that administration of WR-IL-26 activated the JAK1-STAT3 signaling pathway and protected the gut mucosa barrier from A. hydrophila infection. Conversely, silencing WR-IL10R2 and WR-IL20R1 via RNA interference significantly attenuated the activation of WR-IL-26-mediated JAK1-STAT3 pathway. These results provided new insights into the role of IL-26 and its receptors in the gut mucosa barrier and could offer novel therapeutic strategies for managing bacterial infections in aquaculture.
Collapse
Affiliation(s)
- Qiongyao Zeng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ye Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yujun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Zhengwei Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Pingyuan Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Zejun Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, 524088, Guangdong, China.
| |
Collapse
|
8
|
Fachi JL, de Oliveira S, Gilfillan S, Antonova AU, Hou J, Vinolo MAR, Colonna M. NKp46 + ILC3s promote early neutrophil defense against Clostridioides difficile infection through GM-CSF secretion. Proc Natl Acad Sci U S A 2024; 121:e2416182121. [PMID: 39475653 PMCID: PMC11551360 DOI: 10.1073/pnas.2416182121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Clostridioides difficile infection (CDI) is a common cause of antibiotic-associated colitis. C. difficile proliferates and produces toxins that damage the colonic epithelium, leading to symptoms ranging from mild diarrhea to severe pseudomembranous colitis. The host's innate response to CDI occurs in two phases: an early phase in which neutrophils reduce the bacterial load and a late phase involving repair mechanisms to restore epithelial integrity. Group 3 innate lymphoid cells (ILC3s) are crucial in protecting the gut from CDI. Previous studies have shown that ILC3-derived IL-22 is essential in the late phase of CDI for epithelial repair and maintaining an intestinal microbiota that competes with C. difficile, preventing its expansion. Our study finds that ILC3s also protect during the early stages of CDI by sustaining neutrophils through GM-CSF. Less neutrophil production, accumulation, and activation was evident in ILC3-deficient mice than in wild-type (WT) mice, which led to exacerbated symptoms, impaired pathogen clearance, a compromised epithelial barrier, and increased mortality. The adoptive transfer of ILC3s into ILC3-deficient mice restored neutrophil responses and improved disease outcomes. Both in vitro and in vivo experiments revealed that GM-CSF production by ILC3s is crucial for neutrophil production and effective resistance during CDI. Using mice lacking NKp46+ ILC3s, we found that this subset significantly contributes to GM-CSF production in CDI. These findings highlight the critical role of the ILC3-neutrophil connection in early innate responses to CDI. Enhancing ILC3 production of GM-CSF could be a promising strategy for improving host defense against CDI and other enteric infections.
Collapse
Affiliation(s)
- José L. Fachi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| | - Sarah de Oliveira
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP13083-862, Brazil
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| | - JinChao Hou
- Department of Anesthesiology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou310052, China
| | - Marco A. R. Vinolo
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP13083-862, Brazil
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| |
Collapse
|
9
|
Mu X, Gu R, Tang M, Wu X, He W, Nie X. IL-17 in wound repair: bridging acute and chronic responses. Cell Commun Signal 2024; 22:288. [PMID: 38802947 PMCID: PMC11129447 DOI: 10.1186/s12964-024-01668-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024] Open
Abstract
Chronic wounds, resulting from persistent inflammation, can trigger a cascade of detrimental effects including exacerbating inflammatory cytokines, compromised blood circulation at the wound site, elevation of white blood cell count, increased reactive oxygen species, and the potential risk of bacterial infection. The interleukin-17 (IL-17) signaling pathway, which plays a crucial role in regulating immune responses, has been identified as a promising target for treating inflammatory skin diseases. This review aims to delve deeper into the potential pathological role and molecular mechanisms of the IL-17 family and its pathways in wound repair. The intricate interactions between IL-17 and other cytokines will be discussed in detail, along with the activation of various signaling pathways, to provide a comprehensive understanding of IL-17's involvement in chronic wound inflammation and repair.
Collapse
Affiliation(s)
- Xingrui Mu
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
| | - Rifang Gu
- School Medical Office, Zunyi Medical University, Zunyi, 563006, China
| | - Ming Tang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xingqian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
| | - Wenjie He
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China.
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China.
| |
Collapse
|
10
|
Fei X, Wang L, Dou YN, Fei F, Zhang Y, Lv W, He X, Wu X, Chao W, Chen H, Wei J, Gao D, Fei Z. Extracellular vesicle encapsulated Homer1a as novel nanotherapeutics against intracerebral hemorrhage in a mouse model. J Neuroinflammation 2024; 21:85. [PMID: 38582897 PMCID: PMC10999083 DOI: 10.1186/s12974-024-03088-6] [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: 10/14/2023] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
Homer1a and A2 astrocytes are involved in the regulation of inflammation induced by intracerebral hemorrhage (ICH). However, there is no anticipated treatment strategy based on the anti-inflammatory effect of Homer1a and A2 astrocytes. Here, we successfully induced A2 astrocytes in vitro, and then we report an efficient method to prepare Homer1a+ EVs derived from A2 astrocytes which making it more stable, safe, and targetable to injured neurons. Homer1a+ EVs promotes the conversion of A1 to A2 astrocytes in ICH mice. Homer1a+ EVs inhibits activation and nuclear translocation of NF-κB, thereby regulating transcription of IL-17A in neurons. Homer1a+ EVs inhibits the RAGE/NF-κB/IL-17 signaling pathway and the binding ability of IL-17A: IL17-AR and RAGE: DIAPH1. In addition, Homer1a+ EVs ameliorates the pathology, behavior, and survival rate in GFAPCreHomer1fl/-Homer1a± and NestinCreRAGEfl/fl ICH mice. Our study provides a novel insight and potential for the clinical translation of Homer1a+ EVs in the treatment of ICH.
Collapse
Affiliation(s)
- Xiaowei Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Li Wang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Ya-Nan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Weihao Lv
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Xin He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Wangshu Chao
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Hongqing Chen
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| | - Dakuan Gao
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| |
Collapse
|
11
|
Akhtar M, Basher SR, Nizam NN, Hossain L, Bhuiyan TR, Qadri F, Lundgren A. T helper cell responses in adult diarrheal patients following natural infection with enterotoxigenic Escherichia coli are primarily of the Th17 type. Front Immunol 2023; 14:1220130. [PMID: 37809062 PMCID: PMC10552643 DOI: 10.3389/fimmu.2023.1220130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
Background Infection with enterotoxigenic Escherichia coli (ETEC) gives rise to IgA antibodies against both the heat labile toxin (LT) and colonization factors (CFs), which are considered to synergistically protect against ETEC diarrhea. Since the development of ETEC-specific long lived plasma cells and memory B cells is likely to be dependent on T helper (Th) cells, we investigated if natural ETEC diarrhea elicits ETEC-specific Th cells and their relation to IgA responses. Methods Th cell subsets were analyzed in adult Bangladeshi patients hospitalized due to ETEC diarrhea by flow cytometric analysis of peripheral blood mononuclear cells (PBMCs) isolated from blood collected day 2, 7, 30 and 90 after hospitalization as well as in healthy controls. The LT- and CF-specific Th responses were determined by analysis of IL-17A and IFN-γ in antigen stimulated PBMC cultures using ELISA. ETEC-specific IgA secreted by circulating antibody secreting cells (plasmablasts) were analyzed by using the antibodies in lymphocyte supernatants (ALS) ELISA-based method and plasma IgA was also measured by ELISA. Results ETEC patients mounted significant ALS and plasma IgA responses against LTB and CFs on day 7 after hospitalization. ETEC patients had significantly elevated proportions of memory Th cells with a Th17 phenotype (CCR6+CXCR3-) in blood compared to controls, while frequencies of Th1 (CCR6-CXCR3+) or Th2 (CCR6-CXCR3-) cells were not increased. Antigen stimulation of PBMCs revealed IL-17A responses to LT, most clearly observed after stimulation with double mutant heat labile toxin (dmLT), but also with LT B subunit (LTB), and to CS6 in samples from patients with LT+ or CS6+ ETEC bacteria. Some individuals also mounted IFN-γ responses to dmLT and LTB. Levels of LTB specific IgA antibodies in ALS, but not plasma samples correlated with both IL-17A (r=0.5, p=0.02) and IFN-γ (r=0.6, p=0.01) responses to dmLT. Conclusions Our results show that ETEC diarrhea induces T cell responses, which are predominantly of the Th17 type. The correlations between IL-17A and IFN-g and intestine-derived plasmablast responses support that Th responses may contribute to the development of protective IgA responses against ETEC infection. These observations provide important insights into T cell responses that need to be considered in the evaluation of advanced ETEC vaccine candidates.
Collapse
Affiliation(s)
- Marjahan Akhtar
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Salima Raiyan Basher
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Nuder Nower Nizam
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Lazina Hossain
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Anna Lundgren
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
12
|
Thirugnanam S, Walker EM, Schiro F, Aye PP, Rappaport J, Rout N. Enhanced IL-17 Producing and Maintained Cytolytic Effector Functions of Gut Mucosal CD161 +CD8 + T Cells in SIV-Infected Rhesus Macaques. Viruses 2023; 15:1944. [PMID: 37766350 PMCID: PMC10535321 DOI: 10.3390/v15091944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Previous studies have indicated that the loss of CD161-expressing CD4+ Th17 cells is linked to the progression of chronic HIV. These cells are significantly depleted in peripheral blood and gut mucosa of HIV-infected individuals, contributing to inflammation and disruption of the gut barrier. However, the impact of HIV infection on CD161-expressing CD8+ T cells remain unclear. Here, we examined the functions of peripheral blood and mucosal CD161+CD8+ T cells in the macaque model of HIV infection. In contrast to the significant loss of CD161+CD4+ T cells, CD161+CD8+ T cell frequencies were maintained in blood and gut during chronic SIV infection. Furthermore, gut CD161+CD8+ T cells displayed greater IL-17 production and maintained Th1-type and cytolytic functions, contrary to impaired IL-17 and granzyme-B production in CD161+CD4+ T cells of SIV-infected macaques. These results suggest that augmented Th17-type effector functions of CD161+CD8+ T cells during SIV infection is a likely mechanism to compensate for the sustained loss of gut mucosal Th17 cells. Targeting the cytokine and cytolytic effector functions of CD161+CD8+ T cells in the preclinical setting of chronic SIV infection with antiretroviral therapy has implications in the restoration of gut barrier disruption in persons with HIV infection.
Collapse
Affiliation(s)
- Siva Thirugnanam
- Tulane National Primate Research Center, Covington, LA 70433, USA; (S.T.); (P.P.A.); (J.R.)
| | - Edith M. Walker
- Tulane National Primate Research Center, Covington, LA 70433, USA; (S.T.); (P.P.A.); (J.R.)
| | - Faith Schiro
- Tulane National Primate Research Center, Covington, LA 70433, USA; (S.T.); (P.P.A.); (J.R.)
| | - Pyone P. Aye
- Tulane National Primate Research Center, Covington, LA 70433, USA; (S.T.); (P.P.A.); (J.R.)
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA 70433, USA; (S.T.); (P.P.A.); (J.R.)
| | - Namita Rout
- Tulane National Primate Research Center, Covington, LA 70433, USA; (S.T.); (P.P.A.); (J.R.)
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
13
|
Caspe SG, Ewing DA, Livingstone M, Underwood C, Milne E, Sargison ND, Wattegedera SR, Longbottom D. The Immune Response in the Uteri and Placentae of Chlamydia abortus-Infected Ewes and Its Association with Pregnancy Outcomes. Pathogens 2023; 12:846. [PMID: 37375536 PMCID: PMC10303648 DOI: 10.3390/pathogens12060846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
The enzootic abortion of ewes, caused by the bacterium Chlamydia abortus (C. abortus), is one of the main causes of abortion in sheep. There are multiple contributory factors, including chlamydial growth, host immune response, and hormonal balance, that result in different pregnancy outcomes, such as abortion, the birth of weak lambs that may die, or healthy lambs. This study aimed to determine the relationship between phenotypical patterns of immune cell infiltration and different pregnancy outcomes in twin-bearing sheep (both lambs born dead; one alive and one dead; both alive) when experimentally infected with C. abortus. Both the sheep uteri and placentae were collected after parturition. All samples were analysed for specific immune cell features, including cell surface antigens and the T-regulatory (Treg) cell-associated transcription factor and cytokines, by immunohistochemistry and in situ hybridisation. Some of these immunological antigens were evaluated in ovine reproductive tissues for the first time. Differential patterns of T helper/Treg cells revealed significant group effects in the placentae. It suggests the potential role that the balance of lymphocyte subsets may play in affecting different pregnancy outcomes in C. abortus-infected sheep. The present study provides novel detailed information about the immune responses observed at the maternofoetal interface in sheep at the time of pre-term abortion or lambing.
Collapse
Affiliation(s)
- Sergio Gaston Caspe
- Moredun Research Institute, Penicuik EH26 0PZ, UK
- Estación Experimental Mercedes, Instituto Nacional de Tecnología Agropecuaria (INTA), Corrientes W3400, Argentina
| | | | | | | | - Elspeth Milne
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH10 5HF, UK
| | - Neil Donald Sargison
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH10 5HF, UK
| | | | | |
Collapse
|
14
|
Zhang Y, Feng X, Chen J, Liu J, Wu J, Tan H, Mi Z, Rong P. Controversial role of ILC3s in intestinal diseases: A novelty perspective on immunotherapy. Front Immunol 2023; 14:1134636. [PMID: 37063879 PMCID: PMC10090672 DOI: 10.3389/fimmu.2023.1134636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
ILC3s have been identified as crucial immune regulators that play a role in maintaining host homeostasis and modulating the antitumor response. Emerging evidence supports the idea that LTi cells play an important role in initiating lymphoid tissue development, while other ILC3s can promote host defense and orchestrate adaptive immunity, mainly through the secretion of specific cytokines and crosstalk with other immune cells or tissues. Additionally, dysregulation of ILC3-mediated overexpression of cytokines, changes in subset abundance, and conversion toward other ILC subsets are closely linked with the occurrence of tumors and inflammatory diseases. Regulation of ILC3 cytokines, ILC conversion and LTi-induced TLSs may be a novel strategy for treating tumors and intestinal or extraintestinal inflammatory diseases. Herein, we discuss the development of ILCs, the biology of ILC3s, ILC plasticity, the correlation of ILC3s and adaptive immunity, crosstalk with the intestinal microenvironment, controversial roles of ILC3s in intestinal diseases and potential applications for treatment.
Collapse
Affiliation(s)
- Yunshu Zhang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuefei Feng
- Department of Government & Public Administration, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Juan Chen
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahao Liu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianmin Wu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongpei Tan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| |
Collapse
|
15
|
Yi EJ, Kim YI, Song JH, Ko HJ, Chang SY. Intranasal immunization with curdlan induce Th17 responses and enhance protection against enterovirus 71. Vaccine 2023; 41:2243-2252. [PMID: 36863926 DOI: 10.1016/j.vaccine.2023.01.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/28/2022] [Accepted: 01/31/2023] [Indexed: 03/04/2023]
Abstract
Mucosal surfaces are in contact with the external environment and protect the body from infection by various microbes. To prevent infectious diseases at the first line of defense, the establishment of pathogen-specific mucosal immunity by mucosal vaccine delivery is needed. Curdlan, a 1,3-β-glucan has a strong immunostimulatory effect when delivered as a vaccine adjuvant. Here, we investigated whether intranasal administration of curdlan and antigen (Ag) could induce sufficient mucosal immune responses and protect against viral infections. Intranasal co-administration of curdlan and OVA increased OVA-specific IgG and IgA Abs in both serum and mucosal secretions. In addition, intranasal co-administration of curdlan and OVA induced the differentiation of OVA-specific Th1/Th17 cells in the draining lymph nodes. To investigate the protective immunity of curdlan against viral infection, intranasal co-administration of curdlan with recombinant VP1 of EV71 C4a was administered and showed enhanced protection against enterovirus 71 in a passive serum transfer model using neonatal hSCARB2 mice, although intranasal administration of VP1 plus curdlan increased VP1-specific helper T cells responses but not mucosal IgA. Next, Mongolian gerbils were intranasally immunized with curdlan plus VP1, and they had effective protection against EV71 C4a infection, while decreasing viral infection and tissue damage by inducing Th17 responses. These results indicated that intranasal curdlan with Ag improved Ag-specific protective immunity by enhancing mucosal IgA and Th17 against viral infection. Our results suggest that curdlan is an advantageous candidate as a mucosal adjuvant and delivery vehicle for the development of mucosal vaccines.
Collapse
Affiliation(s)
- Eun-Je Yi
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Young-In Kim
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea; AI-Superconvergence KIURI Translational Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Jae-Hyoung Song
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea.
| |
Collapse
|
16
|
Earnest DJ, Burns S, Pandey S, Mani KK, Sohrabji F. Sex differences in the diathetic effects of shift work schedules on circulating cytokine levels and pathological outcomes of ischemic stroke during middle age. Neurobiol Sleep Circadian Rhythms 2022; 13:100079. [PMID: 35800977 PMCID: PMC9253906 DOI: 10.1016/j.nbscr.2022.100079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022] Open
Abstract
Shift work is associated with increased risk for vascular disease, including stroke- and cardiovascular-related mortality. However, evidence from these studies is inadequate to distinguish the effect of altered circadian rhythms in isolation from other risk factors for stroke associated with shift work (e.g., smoking, poor diet, lower socioeconomic status). Thus, the present study examined the diathetic effects of exposure to shifted LD cycles during early adulthood on circadian rhythmicity, inflammatory signaling and ischemic stroke pathology during middle age, when stroke risk is high and outcomes are more severe. Entrainment of circadian activity was stable in all animals maintained on a fixed light:dark 12:12 cycle but was severely disrupted during exposure to shifted LD cycles (12hr advance/5d). Following treatment, circadian entrainment in the shifted LD group was distinguished by increased daytime activity and decreased rhythm amplitude that persisted into middle-age. Circadian rhythm desynchronization in shifted LD males and females was accompanied by significant elevations in circulating levels of the inflammatory cytokine IL-17A and gut-derived inflammatory mediator lipopolysaccharide (LPS) during the post-treatment period. Middle-cerebral artery occlusion, 3 months after exposure to shifted LD cycles, resulted in greater post-stroke mortality in shifted LD females. In surviving subjects, sensorimotor performance, assessed 2- and 5-days post-stroke, was impaired in males of both treatment groups, whereas in females, recovery of function was observed in fixed but not shifted LD rats. Overall, these results indicate that early exposure to shifted LD cycles promotes an inflammatory phenotype that amplifies stroke impairments, specifically in females, later in life. Early exposure to shifted LD cycles alters circadian entrainment of the activity rhythm that persists into middle age. In conjunction with circadian dysregulation, shift work-like schedules promote the induction of key inflammatory mediators. In females, exposure to shift work-like schedules amplifies functional impairments caused by strokes arising later in life. Circadian dysregulation during shift work is a hysteretic risk factor in the overall severity of ischemic strokes. Shift work-related circadian dysregulation affects stroke outcomes independent of lifestyle vascular disease risk factors.
Collapse
|
17
|
Cao H, Diao J, Liu H, Liu S, Liu J, Yuan J, Lin J. The Pathogenicity and Synergistic Action of Th1 and Th17 Cells in Inflammatory Bowel Diseases. Inflamm Bowel Dis 2022; 29:818-829. [PMID: 36166586 DOI: 10.1093/ibd/izac199] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 12/09/2022]
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis and Crohn's disease, are characterized by chronic idiopathic inflammation of gastrointestinal tract. Although the pathogenesis of IBD remains unknown, intestinal immune dysfunction has been considered as the core pathogenesis. In the intestinal immune system, T helper 1 (Th1) and Th17 cells are indispensable for intestine homeostasis via preventing pathogenic bacteria invasion, regulating metabolism and functions of intestinal epithelial cells (IECs), and promoting IEC self-renewal. However, during the development of IBD, Th1 and Th17 cells acquire the pathogenicity and change from the maintainer of intestinal homeostasis to the destroyer of intestinal mucosa. Because of coexpressing interferon-γ and interleukin-17A, Th17 cells with pathogenicity are named as pathogenic Th17 cells. In disease states, Th1 cells impair IEC programs by inducing IEC apoptosis, recruiting immune cells, promoting adhesion molecules expression of IECs, and differentiating to epithelial cell adhesion molecule-specific interferon γ-positive Th1 cells. Pathogenic Th17 cells induce IEC injury by triggering IBD susceptibility genes expression of IECs and specifically killing IECs. In addition, Th1 and pathogenic Th17 cells could cooperate to induce colitis. The evidences from IBD patients and animal models demonstrate that synergistic action of Th1 and pathogenic Th17 cells occurs in the diseases development and aggravates the mucosal inflammation. In this review, we focused on Th1 and Th17 cell programs in homeostasis and intestine inflammation and specifically discussed the impact of Th1 and Th17 cell pathogenicity and their synergistic action on the onset and the development of IBD. We hoped to provide some clues for treating IBD.
Collapse
Affiliation(s)
- Hui Cao
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Diao
- Department of Pediatrics, Yueyang Hospital of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huosheng Liu
- Department of Acupuncture and Moxibustion, Shanghai Jiading Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Suxian Liu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Liu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianye Yuan
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiang Lin
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
18
|
Laumaea AE, Lewin A, Chatterjee D, Marchitto L, Ding S, Gendron‐Lepage G, Goyette G, Allard M, Simard C, Tremblay T, Perreault J, Duerr R, Finzi A, Bazin R. COVID-19 vaccine humoral response in frequent platelet donors with plateletpheresis-associated lymphopenia. Transfusion 2022; 62:1779-1790. [PMID: 35919021 PMCID: PMC9539235 DOI: 10.1111/trf.17037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Plateletpheresis involves platelet separation and collection from whole blood while other blood cells are returned to the donor. Because platelets are replaced faster than red blood cells, as many as 24 donations can be done annually. However, some frequent apheresis platelet donors (>20 donations annually) display severe plateletpheresis-associated lymphopenia; in particular, CD4+ T but not B cell numbers are decreased. COVID-19 vaccination thereby provides a model to assess whether lymphopenic platelet donors present compromised humoral immune responses. STUDY DESIGN AND METHODS We assessed vaccine responses following 2 doses of COVID-19 vaccination in a cohort of 43 plateletpheresis donors with a range of pre-vaccination CD4+ T cell counts (76-1537 cells/μl). In addition to baseline T cell measurements, antibody binding assays to full-length Spike and the Receptor Binding Domain (RBD) were performed pre- and post-vaccination. Furthermore, pseudo-particle neutralization and antibody-dependent cellular cytotoxicity assays were conducted to measure antibody functionality. RESULTS Participants were stratified into two groups: <400 CD4/μl (n = 27) and ≥ 400 CD4/μl (n = 16). Following the first dose, 79% seroconverted within the <400 CD4/μl group compared to 87% in the ≥400 CD4/μl group; all donors were seropositive post-second dose with significant increases in antibody levels. Importantly differences in CD4+ T cell levels minimally impacted neutralization, Spike recognition, and IgG Fc-mediated effector functions. DISCUSSION Overall, our results indicate that lymphopenic plateletpheresis donors do not exhibit significant immune dysfunction; they have retained the T and B cell functionality necessary for potent antibody responses after vaccination.
Collapse
Affiliation(s)
- Annemarie Eare Laumaea
- Centre de Recherche du CHUMMontréalCanada
- Département de Microbiologie, Infectiologie et ImmunologieUniversité de MontréalMontréalQuébecCanada
- Héma‐QuébecAffaires Médicales et InnovationQuébecCanada
| | - Antoine Lewin
- Héma‐QuébecAffaires Médicales et InnovationMontréalQuébecCanada
| | | | - Lorie Marchitto
- Centre de Recherche du CHUMMontréalCanada
- Département de Microbiologie, Infectiologie et ImmunologieUniversité de MontréalMontréalQuébecCanada
| | | | | | | | | | - Carl Simard
- Héma‐QuébecAffaires Médicales et InnovationQuébecCanada
| | - Tony Tremblay
- Héma‐QuébecAffaires Médicales et InnovationQuébecCanada
| | | | - Ralf Duerr
- Department of MicrobiologyNew York University School of MedicineNew York CityNew YorkUSA
| | - Andrés Finzi
- Centre de Recherche du CHUMMontréalCanada
- Département de Microbiologie, Infectiologie et ImmunologieUniversité de MontréalMontréalQuébecCanada
| | - Renée Bazin
- Héma‐QuébecAffaires Médicales et InnovationQuébecCanada
| |
Collapse
|
19
|
Abstract
Over the past decades, tremendous success in the treatment of psoriasis has been achieved using biologics, such as neutralizing antibodies against TNF/TNFR, IL-23, and IL-17A/IL-17RA. Although psoriatic skin lesions appear to resolve after treatment with these biologics, lesions often recur after therapy is discontinued or during therapy. Memory T cells residing in the skin have been considered as the major driver of psoriasis relapse. However, whether structural cells in the skin such as keratinocytes and fibroblasts are involved in the relapse of psoriasis is unknown. In this review, we outline the therapeutic rationale of biologics used in the treatment of psoriasis, summarize different clinical features of psoriasis relapse on the basis of preclinical and clinical data, and specifically discuss how memory T cells and structural cells in the skin are involved in psoriasis relapse. Finally, we discuss the future challenges in the basic or clinical research on psoriasis.
Collapse
|
20
|
de Costa A. The appendix‐mucosal immunity and tolerance in the gut: consequences for the syndromes of appendicitis and its epidemiology. ANZ J Surg 2022; 92:653-660. [PMID: 35152541 PMCID: PMC9304207 DOI: 10.1111/ans.17522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/06/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
The cause of appendicitis is unknown. A review is presented across diverse sources relating to the biology of the appendix and its perturbations. A mechanistic model of the function of the appendix is presented, and its application to the syndromes and consequences of appendicitis is described.
Collapse
Affiliation(s)
- Alan de Costa
- College of Medicine and Dentistry James Cook University, Cairns Clinical School, Cairns Hospital Cairns Queensland Australia
| |
Collapse
|
21
|
Sinha P, Dash M, Bhatkoti B, Krishnan L. Epithelial herpes simplex keratitis in a patient on treatment with secukinumab for psoriasis: An effect of interleukin-17 blockade? Indian J Dermatol Venereol Leprol 2021; 88:225-227. [PMID: 34877851 DOI: 10.25259/ijdvl_496_2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/01/2021] [Indexed: 01/18/2023]
Affiliation(s)
- Preema Sinha
- Department of Dermatology, Base Hospital, Lucknow, Uttar Pradesh, India
| | - Mahashweta Dash
- Department of Dermatology, Base Hospital, Lucknow, Uttar Pradesh, India
| | - Bhupesh Bhatkoti
- Department of Ophthalmology, Command Hospital, Lucknow, Uttar Pradesh, India
| | | |
Collapse
|
22
|
Harada N, Okamura Y, Kono T, Sakai M, Hikima JI. Identification of two interleukin 17 receptor C (IL-17RC) genes and their binding activities to three IL-17A/F ligands in the Japanese medaka, Oryzias latipes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 124:104179. [PMID: 34171369 DOI: 10.1016/j.dci.2021.104179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 06/13/2023]
Abstract
In mammals, interleukin (IL)-17 receptor C (IL-17RC) and IL-17RA mediate IL-17A and IL-17F signaling to produce mucin, antimicrobial peptides, and maintain healthy intestinal flora. However, IL-17RC signaling in fish remains unclear. In this study, three il17rc transcripts (il17rca1, il17rca2, and il17rcb) from the Japanese medaka (Oryzias latipes) were cloned; il17rca1 and il17rca2 mRNAs were alternatively spliced from il17rca pre-mRNA as transcript variants. The il17rca and il17rcb genes were located on chromosomes 7 and 5, respectively. Teleost clades containing medaka il17rca and il17rcb clustered separately from the tetrapod clade. In adult tissues, il17rca1 expression was significantly higher than il17rca2 and il17rcb. Conversely, il17rcb expression was significantly higher in embryos and larvae. These expression patterns changed following infection with Edwardsiella piscicida and Aeromonas hydrophila. Furthermore, an immunoprecipitation assay using recombinant IL-17RCs and rIL-17A/Fs suggested that, in teleosts, three ligands could function in signaling through two IL-17RCs.
Collapse
Affiliation(s)
- Nanaki Harada
- International Course of Agriculture, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Yo Okamura
- Interdisciplinary Graduate School of Agriculture and Engineering, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Tomoya Kono
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Masahiro Sakai
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Jun-Ichi Hikima
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan.
| |
Collapse
|
23
|
Pandiyan P, McCormick TS. Regulation of IL-17A-Producing Cells in Skin Inflammatory Disorders. J Invest Dermatol 2021; 142:867-875. [PMID: 34561088 DOI: 10.1016/j.jid.2021.06.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
This review focuses on the IL-17A family of cytokines produced by T lymphocytes and other immune cells and how they are involved in cutaneous pathogenic responses. It will also discuss cutaneous dysbiosis and FOXP3+ regulatory T cells in the context of inflammatory conditions linked to IL-17 responses in the skin. Specifically, it will review key literature on chronic mucocutaneous candidiasis and psoriasis.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| | - Thomas S McCormick
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
24
|
Jangra S, Landers JJ, Rathnasinghe R, O’Konek JJ, Janczak KW, Cascalho M, Kennedy AA, Tai AW, Baker JR, Schotsaert M, Wong PT. A Combination Adjuvant for the Induction of Potent Antiviral Immune Responses for a Recombinant SARS-CoV-2 Protein Vaccine. Front Immunol 2021; 12:729189. [PMID: 34603303 PMCID: PMC8481386 DOI: 10.3389/fimmu.2021.729189] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/30/2021] [Indexed: 01/03/2023] Open
Abstract
Several SARS-CoV-2 vaccines have received EUAs, but many issues remain unresolved, including duration of conferred immunity and breadth of cross-protection. Adjuvants that enhance and shape adaptive immune responses that confer broad protection against SARS-CoV-2 variants will be pivotal for long-term protection as drift variants continue to emerge. We developed an intranasal, rationally designed adjuvant integrating a nanoemulsion (NE) that activates TLRs and NLRP3 with an RNA agonist of RIG-I (IVT DI). The combination adjuvant with spike protein antigen elicited robust responses to SARS-CoV-2 in mice, with markedly enhanced TH1-biased cellular responses and high virus-neutralizing antibody titers towards both homologous SARS-CoV-2 and a variant harboring the N501Y mutation shared by B1.1.7, B.1.351 and P.1 variants. Furthermore, passive transfer of vaccination-induced antibodies protected naive mice against heterologous viral challenge. NE/IVT DI enables mucosal vaccination, and has the potential to improve the immune profile of a variety of SARS-CoV-2 vaccine candidates to provide effective cross-protection against future drift variants.
Collapse
MESH Headings
- Adaptive Immunity/immunology
- Adjuvants, Immunologic/pharmacology
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19 Vaccines/immunology
- Chlorocebus aethiops
- Cross Protection/immunology
- DEAD Box Protein 58
- HEK293 Cells
- Humans
- Immunity, Humoral/immunology
- Immunization, Passive
- Mice
- Mice, Inbred C57BL
- Receptors, Immunologic/agonists
- Recombinant Proteins/immunology
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Vaccination
- Vaccines, Synthetic/immunology
- Vero Cells
Collapse
Affiliation(s)
- Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jeffrey J. Landers
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Raveen Rathnasinghe
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jessica J. O’Konek
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Katarzyna W. Janczak
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Marilia Cascalho
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrew A. Kennedy
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrew W. Tai
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
- Medicine Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - James R. Baker
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Pamela T. Wong
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
25
|
Cornacchia MG, Sangineto M, Villani R, Cavallone F, Di Gioia G, Cicciomessere P, Serviddio G. High prevalence of false positive SARS-CoV2 serology in a cohort of patients with liver autoimmune diseases. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Aim
Monitoring the prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) immunization in patients with autoimmune diseases is of particular concern to understand their response to the infection and to the vaccine. In fact, the immunological disorder and the immunosuppressive therapies could affect the serological response. SARS-CoV2 serological tests potentially provide this information, although they were rapidly commercialized with internal verifications. Here, we analysed the seroprevalence to SARS-CoV2 in a cohort of patients with liver autoimmune diseases.
Methods
From May to December 2020, a cohort of patients affected by primary biliary cholangitis (PBC), autoimmune hepatitis (AIH) and PBC/AIH overlap syndrome were screened with (reverse transcription-polymerase chain reaction) RT-PCR of nasopharyngeal swabs, rapid antigenic test and chemiluminescent serological test during routine follow-up.
Results
The analysis of 42 patients was carried out: 18 (42.85%) PBC, 12 (28.57%) AIH and 12 (28.57%) PBC/AIH overlap syndromes. Only 2 patients (4.76%) resulted positive to the RNA, antigen and antibody detection tests, hence affected by SARS-CoV2 infection. 14 subjects out of 40 negative cases presented a positive serology for SARS-CoV2 antibodies, hence with a false positivity in the 35% of cases without infection. Among these, 6 (42.86%) patients presented only immunoglobulin (Ig)M positivity, 6 (42.86%) patients presented positivity for only IgG and 2 (14.28%) patients were positive to both IgM and IgG.
Notably, the presence of autoantibodies did not correlate with the serological false positivity, highlighting that there is no cross-reactivity with autoantibodies. The presence of polyclonal hypergammaglobulinemia did not interfere with the serological test as well.
Interestingly, the patients with false positive serology showed higher levels of gamma-glutamyltransferase (GGT) and C-reactive protein (CRP).
Conclusions
Patients with liver autoimmune diseases present a high rate of false positive SARS-CoV2 serology. Therefore, new strategies are needed to study the serological response in this patient category.
Collapse
Affiliation(s)
- Maria Giulia Cornacchia
- University Center for Liver Disease Research and Treatment (C.U.R.E.), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71121 Foggia, Puglia, Italy
| | - Moris Sangineto
- University Center for Liver Disease Research and Treatment (C.U.R.E.), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71121 Foggia, Puglia, Italy
| | - Rosanna Villani
- University Center for Liver Disease Research and Treatment (C.U.R.E.), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71121 Foggia, Puglia, Italy
| | - Francesco Cavallone
- University Center for Liver Disease Research and Treatment (C.U.R.E.), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71121 Foggia, Puglia, Italy
| | - Giuseppe Di Gioia
- University Center for Liver Disease Research and Treatment (C.U.R.E.), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71121 Foggia, Puglia, Italy
| | - Paola Cicciomessere
- University Center for Liver Disease Research and Treatment (C.U.R.E.), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71121 Foggia, Puglia, Italy
| | - Gaetano Serviddio
- University Center for Liver Disease Research and Treatment (C.U.R.E.), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71121 Foggia, Puglia, Italy
| |
Collapse
|
26
|
Jangra S, Landers JJ, Rathnasinghe R, O'Konek JJ, Janczak KW, Cascalho M, Kennedy AA, Tai AW, Baker JR, Schotsaert M, Wong PT. A Combination Adjuvant for the Induction of Potent Antiviral Immune Responses for a Recombinant SARS-CoV-2 Protein Vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.18.431484. [PMID: 33619480 PMCID: PMC7899444 DOI: 10.1101/2021.02.18.431484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Several SARS-CoV-2 vaccines have received EUAs, but many issues remain unresolved, including duration of conferred immunity and breadth of cross-protection. Adjuvants that enhance and shape adaptive immune responses that confer broad protection against SARS-CoV-2 variants will be pivotal for long-term protection. We developed an intranasal, rationally designed adjuvant integrating a nanoemulsion (NE) that activates TLRs and NLRP3 with an RNA agonist of RIG-I (IVT DI). The combination adjuvant with spike protein antigen elicited robust responses to SARS-CoV-2 in mice, with markedly enhanced T H 1-biased cellular responses and high virus-neutralizing antibody titers towards both homologous SARS-CoV-2 and a variant harboring the N501Y mutation shared by B1.1.7, B.1.351 and P.1 variants. Furthermore, passive transfer of vaccination-induced antibodies protected naive mice against heterologous viral challenge. NE/IVT DI enables mucosal vaccination, and has the potential to improve the immune profile of a variety of SARS-CoV-2 vaccine candidates to provide effective cross-protection against future drift variants.
Collapse
|
27
|
Pérez-Cruz M, Koné B, Porte R, Carnoy C, Tabareau J, Gosset P, Trottein F, Sirard JC, Pichavant M, Gosset P. The Toll-Like Receptor 5 agonist flagellin prevents Non-typeable Haemophilus influenzae-induced infection in cigarette smoke-exposed mice. PLoS One 2021; 16:e0236216. [PMID: 33784296 PMCID: PMC8009382 DOI: 10.1371/journal.pone.0236216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 02/25/2021] [Indexed: 01/13/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality worldwide. The major bacterial cause of COPD exacerbations is non-typeable Haemophilus influenzae (NTHi). 25 to over 80% of cases are associated with NTHi. This susceptibility to infection involves a defective production of interleukin (IL)-22 which plays an important role in mucosal defense. Prophylactic administration of flagellin, a Toll-like receptor 5 (TLR5) agonist, protects healthy mice against respiratory pathogenic bacteria. We hypothesized that TLR5-mediated stimulation of lung immunity might prevent COPD exacerbations. Mice chronically exposed to cigarette smoke (CS), which presented COPD symptoms, were infected with NTHi and intraperitoneally treated with recombinant flagellin following a prophylactic or therapeutic protocol. Compared with control, cigarette smoke-exposed mice treated with flagellin showed a lower bacterial load in the airways, the lungs and the blood. This protection was associated with an early neutrophilia, a lower production of pro-inflammatory cytokines and an increased IL-22 production. Flagellin treatment decreased the recruitment of inflammatory cells and the lung damages related to exacerbation. Morover, the protective effect of flagellin against NTHi was altered by treatment with anti-IL-22 blocking antibodies in cigarette smoke-exposed mice and in Il22-/- mice. The effect of flagellin treatment did not implicated the anti-bacterial peptides calgranulins and defensin-β2. This study shows that stimulation of innate immunity by a TLR5 ligand is a potent antibacterial treatment in CS-exposed mice, suggesting innovative therapeutic strategies against acute exacerbation in COPD.
Collapse
Affiliation(s)
- Magdiel Pérez-Cruz
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Bachirou Koné
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Rémi Porte
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Christophe Carnoy
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Julien Tabareau
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Pierre Gosset
- Service d’Anatomo-pathologie, Hôpital Saint Vincent de Paul, Lille, France
| | - François Trottein
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Jean-Claude Sirard
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Muriel Pichavant
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Philippe Gosset
- Univ. Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL—Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
28
|
Schmidt S, Sassu EL, Vatzia E, Pierron A, Lagler J, Mair KH, Stadler M, Knecht C, Spergser J, Dolezal M, Springer S, Theuß T, Fachinger V, Ladinig A, Saalmüller A, Gerner W. Vaccination and Infection of Swine With Salmonella Typhimurium Induces a Systemic and Local Multifunctional CD4 + T-Cell Response. Front Immunol 2021; 11:603089. [PMID: 33584671 PMCID: PMC7874209 DOI: 10.3389/fimmu.2020.603089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/25/2020] [Indexed: 01/14/2023] Open
Abstract
The gram-negative facultative intracellular bacteria Salmonella Typhimurium (STM) often leads to subclinical infections in pigs, but can also cause severe enterocolitis in this species. Due to its high zoonotic potential, the pathogen is likewise dangerous for humans. Vaccination with a live attenuated STM strain (Salmoporc) is regarded as an effective method to control STM infections in affected pig herds. However, information on the cellular immune response of swine against STM is still scarce. In this study, we investigated the T-cell immune response in pigs that were vaccinated twice with Salmoporc followed by a challenge infection with a virulent STM strain. Blood- and organ-derived lymphocytes (spleen, tonsils, jejunal and ileocolic lymph nodes, jejunum, ileum) were stimulated in vitro with heat-inactivated STM. Subsequently, CD4+ T cells present in these cell preparations were analyzed for the production of IFN-γ, TNF-α, and IL-17A by flow cytometry and Boolean gating. Highest frequencies of STM-specific cytokine-producing CD4+ T cells were found in lamina propria lymphocytes of jejunum and ileum. Significant differences of the relative abundance of cytokine-producing phenotypes between control group and vaccinated + infected animals were detected in most organs, but dominated in gut and lymph node-residing CD4+ T cells. IL-17A producing CD4+ T cells dominated in gut and gut-draining lymph nodes, whereas IFN-γ/TNF-α co-producing CD4+ T cells were present in all locations. Additionally, the majority of cytokine-producing CD4+ T cells had a CD8α+CD27- phenotype, indicative of a late effector or effector memory stage of differentiation. In summary, we show that Salmonella-specific multifunctional CD4+ T cells exist in vaccinated and infected pigs, dominate in the gut and most likely contribute to protective immunity against STM in the pig.
Collapse
Affiliation(s)
- Selma Schmidt
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Elena L Sassu
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Eleni Vatzia
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Alix Pierron
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Julia Lagler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.,Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Kerstin H Mair
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Christian Knecht
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Joachim Spergser
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Marlies Dolezal
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | | | - Tobias Theuß
- Ceva Innovation Center GmbH, Dessau-Roßlau, Germany
| | | | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Wilhelm Gerner
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
29
|
Saleh M, Friedl A, Srivastava M, Secombes CJ, El-Matbouli M. Modulation of local and systemic immune responses in brown trout (Salmo trutta) following exposure to Myxobolus cerebralis. FISH & SHELLFISH IMMUNOLOGY 2020; 106:844-851. [PMID: 32891791 DOI: 10.1016/j.fsi.2020.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 06/11/2023]
Abstract
Myxobolus cerebralis, the etiological agent of Whirling Disease (WD), is a freshwater myxozoan parasite with considerable economic and ecological relevance for salmonids. There are differences in disease susceptibility between species and strains of salmonids. Recently, we have reported that the suppressor of cytokine signaling SOCS1 and SOCS3 are key in modulating rainbow trout (Oncorhynchus mykiss) immune responses and that resistant fish apparently exhibit effective Th17 cell response after exposure to M. cerebralis. It is unclear whether such molecules and pathways are also involved in the immune response of M. cerebralis infected brown trout (Salmo trutta). Hence, this study aimed to explore their role during immune modulation in infected brown trout, which is considered resistant to this parasite. Fish were exposed to the triactinomyxon (TAM) stages of M. cerebralis and quantitative real-time PCR (RT-qPCR) was carried out to examine local (caudal fin) and systemic (head kidney, spleen) immune transcriptional changes associated with WD over time in infected and control fish. All of the immune genes in the three tissues studied were differentially expressed in infected fish at multiple time points. Brown trout reduced the parasite load and demonstrated effective immune responses, likely by keeping pro-inflammatory and anti-inflammatory cytokines in balance whilst stimulating efficient Th17-mediated immunity. This study increases knowledge on the brown trout immune response to M. cerebralis and helps us to understand the underlying mechanisms of WD resistance.
Collapse
Affiliation(s)
- Mona Saleh
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Austria.
| | - Adina Friedl
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Austria.
| | - Mitaly Srivastava
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Austria.
| | - Christopher J Secombes
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Scotland, UK.
| | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Austria.
| |
Collapse
|
30
|
Koné B, Pérez‐Cruz M, Porte R, Hennegrave F, Carnoy C, Gosset P, Trottein F, Sirard J, Pichavant M, Gosset P. Boosting the IL-22 response using flagellin prevents bacterial infection in cigarette smoke-exposed mice. Clin Exp Immunol 2020; 201:171-186. [PMID: 32324274 PMCID: PMC7366752 DOI: 10.1111/cei.13445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/27/2020] [Accepted: 04/10/2020] [Indexed: 12/29/2022] Open
Abstract
The progression of chronic obstructive pulmonary disease (COPD), a lung inflammatory disease being the fourth cause of death worldwide, is marked by acute exacerbations. These episodes are mainly caused by bacterial infections, frequently due to Streptococcus pneumoniae. This susceptibility to infection involves a defect in interleukin (IL)-22, which plays a pivotal role in mucosal defense mechanism. Administration of flagellin, a Toll-like receptor 5 (TLR-5) agonist, can protect mice and primates against respiratory infections in a non-pathological background. We hypothesized that TLR-5-mediated stimulation of innate immunity might improve the development of bacteria-induced exacerbations in a COPD context. Mice chronically exposed to cigarette smoke (CS), mimicking COPD symptoms, are infected with S. pneumoniae, and treated in a preventive and a delayed manner with flagellin. Both treatments induced a lower bacterial load in the lungs and blood, and strongly reduced the inflammation and lung lesions associated with the infection. This protection implicated an enhanced production of IL-22 and involved the recirculation of soluble factors secreted by spleen cells. This is also associated with higher levels of the S100A8 anti-microbial peptide in the lung. Furthermore, human mononuclear cells from non-smokers were able to respond to recombinant flagellin by increasing IL-22 production while active smoker cells do not, a defect associated with an altered IL-23 production. This study shows that stimulation of innate immunity by a TLR-5 ligand reduces CS-induced susceptibility to bacterial infection in mice, and should be considered in therapeutic strategies against COPD exacerbations.
Collapse
Affiliation(s)
- B. Koné
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - M. Pérez‐Cruz
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - R. Porte
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - F. Hennegrave
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - C. Carnoy
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - P. Gosset
- Service d’Anatomo‐pathologieHôpital Saint Vincent de PaulLilleFrance
| | - F. Trottein
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - J.‐C. Sirard
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - M. Pichavant
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| | - P. Gosset
- Université de LilleCNRSInsermCHU LilleInstitut Pasteur de LilleLilleFrance
| |
Collapse
|
31
|
Kim HJ, Lee SH, Hong SJ. Antibiotics-Induced Dysbiosis of Intestinal Microbiota Aggravates Atopic Dermatitis in Mice by Altered Short-Chain Fatty Acids. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:137-148. [PMID: 31743970 PMCID: PMC6875482 DOI: 10.4168/aair.2020.12.1.137] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/23/2019] [Accepted: 10/03/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Alterations in the intestinal microbiota in early life affects the development of atopic dermatitis (AD) in humans. This study aimed to further investigate the effects of gut dysbiosis in early life in an ovalbumin (OVA)-induced mouse model of AD. METHODS The AD mouse model was developed by serial OVA sensitization and mice were treated with an antibiotic cocktail in their drinking water for 2 weeks before primary sensitization. Probiotics (Lactobacillus rhamnosus, 1 × 10⁹ CFU) or 100 μL of fresh fecal supernatant were orally administered daily from 1 week before the first sensitization until the end of the study. RESULTS The AD mice which received antibiotics had significantly aggravated phenotypes, including clinical score, transepidermal water loss, and histopathology, compared to those treated with healthy feces or probiotics. Total systemic immunoglobulin E production and skin interleukin (IL) 4 levels were significantly increased in the antibiotic-treated mice compared to the other groups. Antibiotic treatment also increased the levels of IL17 and group 3 innate lymphoid cells (ILC3) in the gut and significantly suppressed the production of short-chain fatty acids (SCFAs) and decreased the number FOXP3⁺ cells. CONCLUSIONS Our results suggest that the status of the gut microbiota in early life in the mouse may play a crucial role in AD development through intestinal SCFA production through regulate the numbers of CD4⁺IL17⁺/CD4⁺FOXP3⁺ regulatory T cells and ILC3s.
Collapse
Affiliation(s)
- Ha Jung Kim
- Department of Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Seung Hwa Lee
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Environmental Health Center, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
32
|
Liu F, Lee SA, Riordan SM, Zhang L, Zhu L. Effects of Anti-Cytokine Antibodies on Gut Barrier Function. Mediators Inflamm 2019; 2019:7028253. [PMID: 31780866 PMCID: PMC6875247 DOI: 10.1155/2019/7028253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
Anti-cytokine antibodies are used in treating chronic inflammatory diseases and autoimmune diseases such as inflammatory bowel disease and rheumatic diseases. Patients with these diseases often have a compromised gut barrier function, suggesting that anti-cytokine antibodies may contribute to the re-establishment of gut barrier integrity, in addition to their immunomodulatory effects. This paper reviews the effects of anti-cytokine antibodies on gut barrier function and their mechanisms.
Collapse
Affiliation(s)
- Fang Liu
- Department of General Surgery and Central Lab, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Seul A. Lee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Stephen M. Riordan
- Gastrointestinal and Liver Unit, Prince of Wales Hospital, University of New South Wales, Sydney, NSW, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lixin Zhu
- Department of General Surgery and Central Lab, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
33
|
Immunization of BLT Humanized Mice Redirects T Cell Responses to Gag and Reduces Acute HIV-1 Viremia. J Virol 2019; 93:JVI.00814-19. [PMID: 31375576 DOI: 10.1128/jvi.00814-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/24/2019] [Indexed: 12/21/2022] Open
Abstract
BLT (bone marrow-liver-thymus) humanized mice, which reconstitute a functional human immune system, develop prototypic human virus-specific CD8+ T cell responses following infection with human immunodeficiency virus type 1 (HIV-1). We explored the utility of the BLT model for HIV-1 vaccine development by immunizing BLT mice against the conserved viral Gag protein, utilizing a rapid prime-boost protocol of poly(lactic-co-glycolic) acid microparticles and a replication-defective herpes simplex virus (HSV) recombinant vector. After HIV-1 challenge, the mice developed broad, proteome-wide gamma interferon-positive (IFN-γ+) T cell responses against HIV-1 that reached magnitudes equivalent to what is observed in HIV-1-infected individuals. The functionality of these responses was underscored by the consistent emergence of escape mutations in multiple CD8+ T cell epitopes during the course of infection. Although prechallenge vaccine-induced responses were largely undetectable, the Gag immunization increased both the magnitude and the kinetics of anamnestic Gag-specific T cell responses following HIV-1 infection, and the magnitude of these postchallenge Gag-specific responses was inversely correlated with acute HIV-1 viremia. Indeed, Gag immunization was associated with a modest but significant 0.5-log reduction in HIV-1 viral load when analyzed across four experimental groups of BLT mice. Notably, the HSV vector induced elevated plasma concentrations of polarizing cytokines and chemotactic factors, including interleukin-12p70 (IL-12p70) and MIP-1α, which were positively correlated with the magnitude of Gag-specific responses. Overall, these results support the ability of BLT mice to recapitulate human pathogen-specific T cell responses and to respond to immunization; however, additional improvements to the model are required to develop a robust system for testing HIV-1 vaccine efficacy.IMPORTANCE Advances in the development of humanized mice have raised the possibility of a small-animal model for preclinical testing of an HIV-1 vaccine. Here, we describe the capacity of BLT humanized mice to mount broadly directed HIV-1-specific human T cell responses that are functionally active, as indicated by the rapid emergence of viral escape mutations. Although immunization of BLT mice with the conserved viral Gag protein did not result in detectable prechallenge responses, it did increase the magnitude and kinetics of postchallenge Gag-specific T cell responses, which was associated with a modest but significant reduction in acute HIV-1 viremia. Additionally, the BLT model revealed immunization-associated increases in the plasma concentrations of immunomodulatory cytokines and chemokines that correlated with more robust T cell responses. These data support the potential utility of the BLT humanized mouse for HIV-1 vaccine development but suggest that additional improvements to the model are warranted.
Collapse
|
34
|
Vitale S, Santarlasci V, Camarca A, Picascia S, Pasquale AD, Maglio M, Maggi E, Cosmi L, Annunziato F, Troncone R, Auricchio R, Gianfrani C. The intestinal expansion of TCRγδ + and disappearance of IL4 + T cells suggest their involvement in the evolution from potential to overt celiac disease. Eur J Immunol 2019; 49:2222-2234. [PMID: 31553811 DOI: 10.1002/eji.201948098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 07/26/2019] [Indexed: 01/03/2023]
Abstract
Celiac disease (CD) is characterized by a spectrum of intestinal inflammatory lesions. Most patients have villous atrophy (overt-CD), while others have a morphologically normal mucosa, despite the presence of CD-specific autoantibodies (potential-CD). As the mechanism responsible for villous atrophy is not completely elucidated, we investigated biomarkers specific for the different celiac lesions. Phenotype and cytokine production of intestinal mucosa cells were analyzed by flow cytometry in gut biopsies of children with overt- or potential-CD and in healthy controls. Density of TCRγδ+ T cells was found markedly enhanced in intestinal mucosa of children with overt-CD compared to potential-CD or controls. By contrast, very few IL4+ T cells infiltrated the mucosa with villous atrophy compared to morphologically normal mucosa. IL4+ T cells were classical CD4+ T-helper cells (CD161- ), producing or not IFN-γ, and negative for IL17A. Our study demonstrated that the transition to villous atrophy in CD patients is characterized by increased density of TCRγδ+ T cells, and concomitant disappearance of IL4+ cells. These findings suggest that immunomodulatory mechanisms are active in potential-CD to counteract the inflammatory cascade responsible of villous atrophy. Further studies are required to validate the use of IL4+ and TCRγδ+ T cells as biomarkers of the different CD forms.
Collapse
Affiliation(s)
- Serena Vitale
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | | | | | - Stefania Picascia
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Angela Di Pasquale
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Mariantonia Maglio
- Department of Translational Medicine & European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Enrico Maggi
- Immunology Department, Pediatric Hospital Bambino Gesù, IRCCS, Rome
| | - Lorenzo Cosmi
- Denothe Center, University of Florence, Florence, Italy
| | | | - Riccardo Troncone
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Renata Auricchio
- Department of Translational Medicine & European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy.,Department of Translational Medicine & European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| |
Collapse
|
35
|
Golpour A, Bereswill S, Heimesaat MM. Antimicrobial and Immune-Modulatory Effects of Vitamin D Provide Promising Antibiotics-Independent Approaches to Tackle Bacterial Infections - Lessons Learnt from a Literature Survey. Eur J Microbiol Immunol (Bp) 2019; 9:80-87. [PMID: 31662886 PMCID: PMC6798578 DOI: 10.1556/1886.2019.00014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/24/2019] [Indexed: 12/19/2022] Open
Abstract
Antimicrobial multidrug-resistance (MDR) constitutes an emerging threat to global health and makes the effective prevention and treatment of many, particularly severe infections challenging, if not impossible. Many antibiotic classes have lost antimicrobial efficacy against a plethora of infectious agents including bacterial species due to microbial acquisition of distinct resistance genes. Hence, the development of novel anti-infectious intervention strategies including antibiotic-independent approaches is urgently needed. Vitamins such as vitamin D and vitamin D derivates might be such promising molecular candidates to combat infections caused by bacteria including MDR strains. Using the Pubmed database, we therefore performed an in-depth literature survey, searching for publications on the antimicrobial effect of vitamin D directed against bacteria including MDR strains. In vitro and clinical studies between 2009 and 2019 revealed that vitamin D does, in fact, possess antimicrobial properties against both Gram-positive and Gram-negative bacterial species, whereas conflicting results could be obtained from in vivo studies. Taken together, the potential anti-infectious effects for the antibiotic-independent application of vitamin D and/or an adjunct therapy in combination with antibiotic compounds directed against infectious diseases such as tuberculosis, H. pylori infections, or skin diseases, for instance, should be considered and further investigated in more detail.
Collapse
Affiliation(s)
- Ainoosh Golpour
- Institute of Microbiology, Infectious Diseases and Immunology Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
36
|
Rodriguez-Nicolas A, Jiménez P, Carmona FD, Martín J, Matas Cobos AM, Ruiz-Cabello F, Redondo-Cerezo E. Association between Genetic Polymorphisms of Inflammatory Response Genes and Acute Pancreatitis. Immunol Invest 2019; 48:585-596. [PMID: 31044631 DOI: 10.1080/08820139.2019.1576729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inflammation plays a central role in the pathophysiology of acute pancreatitis (AP). We hypothesized that changes in the function of key components of the inflammatory cascade, caused by genetic polymorphisms, could determine the development and/or severity of AP. We studied the following polymorphisms in 269 patients: IL23R rs11209026, TNF rs1800629, RIPK2 rs42490, NOD2 rs9302752, MCP1 rs1024611 and NFKB1 rs28362491. The rs11209026 A allele was related to the presence of AP (p = 0.007261; OR = 1 .523). Epistasis analysis revealed that AP susceptibility was increased by interaction between IL23R rs11209026 and TNF rs1800629 (p = 1.205 × 10-5; ORinteraction = 4.031). The rs42490-G allele was associated with an increased risk of severe pancreatitis (p = 0.01583; OR = 2.736), severe or moderately severe pancreatitis (p = 0.04206; OR = 1.609), and death (p = 0.03226; OR = 3.010). In conclusion, these results point to a plausible role for genetic polymorphisms in IL23R and RIPK2 in the development and severity of AP.
Collapse
Affiliation(s)
- Antonio Rodriguez-Nicolas
- a Servicio de Análisis Clínicos e Inmunología, UGC de Laboratorio Clínico , Hospital Universitario Virgen de las Nieves , Granada , Spain
- b Programa de doctorado en Biomedicina , Universidad de Granada , Granada , Spain
| | - Pilar Jiménez
- a Servicio de Análisis Clínicos e Inmunología, UGC de Laboratorio Clínico , Hospital Universitario Virgen de las Nieves , Granada , Spain
| | - F David Carmona
- c Departamento de Genética e Instituto de Biotecnología , Universidad de Granada , Granada , Spain
| | - Javier Martín
- d Instituto de Parasitología y Biomedicina López Neyra , CSIC , Granada , Spain
| | - Ana M Matas Cobos
- e Servicio de Aparato Digestivo , Hospital Universitario Virgen de las Nieves , Granada , Spain
| | - Francisco Ruiz-Cabello
- a Servicio de Análisis Clínicos e Inmunología, UGC de Laboratorio Clínico , Hospital Universitario Virgen de las Nieves , Granada , Spain
- f Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) , Granada , Spain
| | - Eduardo Redondo-Cerezo
- e Servicio de Aparato Digestivo , Hospital Universitario Virgen de las Nieves , Granada , Spain
- f Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) , Granada , Spain
| |
Collapse
|
37
|
Zazueta-Favela D, Donis-Maturano L, Licea-Navarro AF, Bernáldez-Sarabia J, Dan KWL, Cota-Arce JM, Escobedo G, De León-Nava MA. Marine peptides as immunomodulators: Californiconus californicus-derived synthetic conotoxins induce IL-10 production by regulatory T cells (CD4+Foxp3+). Immunopharmacol Immunotoxicol 2019; 41:463-468. [DOI: 10.1080/08923973.2019.1641114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Daniela Zazueta-Favela
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education of Ensenada (CICESE), Baja California, Mexico
| | - Luis Donis-Maturano
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education of Ensenada (CICESE), Baja California, Mexico
| | - Alexei F. Licea-Navarro
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education of Ensenada (CICESE), Baja California, Mexico
| | - Johanna Bernáldez-Sarabia
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education of Ensenada (CICESE), Baja California, Mexico
| | - Kee W. L. Dan
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education of Ensenada (CICESE), Baja California, Mexico
| | - Julián M. Cota-Arce
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education of Ensenada (CICESE), Baja California, Mexico
| | - Galileo Escobedo
- Laboratory for Proteomics and Metabolomics, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City, Mexico
| | - Marco A. De León-Nava
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education of Ensenada (CICESE), Baja California, Mexico
| |
Collapse
|
38
|
Stehle C, Hernández DC, Romagnani C. Innate lymphoid cells in lung infection and immunity. Immunol Rev 2019; 286:102-119. [PMID: 30294964 DOI: 10.1111/imr.12712] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022]
Abstract
In recent years, innate lymphoid cells (ILCs) have emerged as key mediators of protection and repair of mucosal surfaces during infection. The lung, a dynamic mucosal tissue that is exposed to a plethora of microbes, is a playground for respiratory infection-causing pathogens which are not only a major cause of fatalities worldwide, but are also associated with comorbidities and decreased quality of life. The lung provides a rich microenvironment to study ILCs in the context of innate protection mechanisms within the airways, unraveling their distinct functions not only in health but also in disease. In this review, we discuss how pulmonary ILCs play a role in protection against viral, parasitic, bacterial, and fungal challenge, along with the mechanisms underlying this ILC-mediated immunity.
Collapse
Affiliation(s)
- Christina Stehle
- Innate Immunity, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | | | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma-Forschungszentrum, Berlin, Germany.,Medical Department I, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
39
|
Saleh M, Montero R, Kumar G, Sudhagar A, Friedl A, Köllner B, El-Matbouli M. Kinetics of local and systemic immune cell responses in whirling disease infection and resistance in rainbow trout. Parasit Vectors 2019; 12:249. [PMID: 31113489 PMCID: PMC6528198 DOI: 10.1186/s13071-019-3505-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/14/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Whirling disease (WD), caused by the myxozoan parasite Myxobolus cerebralis, is responsible for high mortalities in rainbow trout hatcheries and natural populations. To elucidate how resistant and susceptible rainbow trout strains respond to early invasion, a well-established model of WD was used to demonstrate the kinetics of local and systemic immune responses in two rainbow trout strains, the susceptible American Trout Lodge (TL) and the more resistant German Hofer strain (HO). METHODS Parasite load and cellular immune responses were compared across several time points after M. cerebralis exposure to elucidate the kinetics of immune cells in resistant and susceptible rainbow trout in response to early invasion. In the course of the 20 days following exposure, leukocyte kinetics was monitored by flow cytometry in the caudal fin (CF), head kidney (HK) and spleen (SP). For the analysis of the leukocyte composition, cells were stained using a set of monoclonal antibodies with known specificity for distinct subpopulations of rainbow trout leukocytes. RESULTS Experiments indicated general increases of CF, HK and SP myeloid cells, while decreases of B cells and T cells in the SP and HK were observed at several time points in the TL strain. On the other hand, in the HO strain, increases of T cells were dominant in CF, HK and SP at multiple time points. The differences between HO and TL were most distinct at 2, 4, 12 and 48 hours post-exposure (hpe) as well as at 4 days post-exposure (dpe), with the vast majority of innate immune response cells having higher values in the susceptible TL strain. Alteration of the leukocyte populations with augmented local cellular responses and excessive immune reactions likely lead to subsequent host tissue damage and supports parasite invasion and development in TL. CONCLUSIONS The findings of this study highlight the significance of effective local and systemic immune reaction and indicate proper activation of T lymphocytes critical for host resistance during M. cerebralis infection. The present study provides insights into the cellular basis of protective immune responses against M. cerebralis and can help us to elucidate the mechanisms underlying the variation in resistance to WD.
Collapse
Affiliation(s)
- Mona Saleh
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ruth Montero
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Gokhlesh Kumar
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Arun Sudhagar
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Adina Friedl
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Bernd Köllner
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
40
|
Acquisition of specific antibodies and their influence on cell-mediated immune response in neonatal cord blood after maternal pertussis vaccination during pregnancy. Vaccine 2019; 37:2569-2579. [PMID: 30955978 DOI: 10.1016/j.vaccine.2019.03.070] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 11/20/2022]
Abstract
Maternal immunization with pertussis acellular vaccine (Tdap) is an intervention that provides protection to newborns. However, it has been reported that high maternal antibody levels may adversely affect the immune response of infants after active immunization. In this study, we evaluated neonatal passive acquisition of pertussis-specific antibodies and their influence on the neonatal cell-mediated immune response. Pregnant women were either vaccinated with Tdap vaccine (case group, n = 66) or received no vaccine (control group, n = 101). Whole-cell Bordetella pertussis (Bp), pertussis toxin (PT), filamentous hemagglutinin (FHA) and pertactin (PRN)-specific serum IgG were quantified in paired maternal-cord sera, and Bp- and PT-specific IgA were evaluated in colostrum by ELISA. Ex vivo neonatal blood lymphocyte responsiveness after Bp stimulation was assessed in case (n = 17) and control (n = 15) groups using flow cytometry to detect proliferation, cytokine production and activation phenotype of lymphocytes in the context of high specific IgG acquired after maternal vaccination. Anti-Bp, PT, FHA and PRN IgG concentrations in maternal and cord sera from case group were higher than those in control group with positive correlation indexes in both groups for all pertussis antigens. The control group presented higher placental transfer ratios of specific antibodies and, in the case group, vaccination between 26 and 31 gestation weeks was associated with the best placental transfer ratios. Specific IgA concentrations in colostrum were not affected by vaccine status. Whole blood assays revealed that newborns responded to Bp stimulation with higher expression of CD40L, CD69 and CD4+ T cell proliferation compared to unstimulated cells, and a lower Th1 response, while a preserved Th2 response compared to adults, but there were no differences between the neonatal groups for any of the studied parameters. Our results indicate that higher pertussis-specific IgG levels in newborn sera after maternal vaccination do not affect the neonatal ex vivo cell-mediated immune response.
Collapse
|
41
|
Neumann C, Blume J, Roy U, Teh PP, Vasanthakumar A, Beller A, Liao Y, Heinrich F, Arenzana TL, Hackney JA, Eidenschenk C, Gálvez EJC, Stehle C, Heinz GA, Maschmeyer P, Sidwell T, Hu Y, Amsen D, Romagnani C, Chang HD, Kruglov A, Mashreghi MF, Shi W, Strowig T, Rutz S, Kallies A, Scheffold A. c-Maf-dependent T reg cell control of intestinal T H17 cells and IgA establishes host-microbiota homeostasis. Nat Immunol 2019; 20:471-481. [PMID: 30778241 DOI: 10.1038/s41590-019-0316-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
Foxp3+ regulatory T cells (Treg cells) are crucial for the maintenance of immune homeostasis both in lymphoid tissues and in non-lymphoid tissues. Here we demonstrate that the ability of intestinal Treg cells to constrain microbiota-dependent interleukin (IL)-17-producing helper T cell (TH17 cell) and immunoglobulin A responses critically required expression of the transcription factor c-Maf. The terminal differentiation and function of several intestinal Treg cell populations, including RORγt+ Treg cells and follicular regulatory T cells, were c-Maf dependent. c-Maf controlled Treg cell-derived IL-10 production and prevented excessive signaling via the kinases PI(3)K (phosphatidylinositol-3-OH kinase) and Akt and the metabolic checkpoint kinase complex mTORC1 (mammalian target of rapamycin) and expression of inflammatory cytokines in intestinal Treg cells. c-Maf deficiency in Treg cells led to profound dysbiosis of the intestinal microbiota, which when transferred to germ-free mice was sufficient to induce exacerbated intestinal TH17 responses, even in a c-Maf-competent environment. Thus, c-Maf acts to preserve the identity and function of intestinal Treg cells, which is essential for the establishment of host-microbe symbiosis.
Collapse
Affiliation(s)
- Christian Neumann
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Jonas Blume
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Urmi Roy
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Peggy P Teh
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Ajithkumar Vasanthakumar
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Alexander Beller
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Yang Liao
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Frederik Heinrich
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | | | - Jason A Hackney
- Department of Bioinformatics and Computational Biology, Genentech, San Francisco, CA, USA
| | - Celine Eidenschenk
- Department of Biochemical and Cellular Pharmacology, Genentech, San Francisco, CA, USA
| | - Eric J C Gálvez
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Christina Stehle
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Gitta A Heinz
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Patrick Maschmeyer
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Tom Sidwell
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Yifang Hu
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Derk Amsen
- Sanquin Research, Department of Hematopoiesis and Landsteiner Laboratory, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Chiara Romagnani
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
- Medical Department I, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hyun-Dong Chang
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Andrey Kruglov
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Mir-Farzin Mashreghi
- German Rheumatism Research Center (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Wei Shi
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Till Strowig
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sascha Rutz
- Department of Cancer Immunology, Genentech, San Francisco, CA, USA.
| | - Axel Kallies
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts-Universität zu Kiel & Universitätsklinik Schleswig Holstein, Kiel, Germany.
| |
Collapse
|
42
|
Lange J, Ganesh S, Meier S, Kay JK, Crookenden MA, Walker CG, Mitchell MD, Loor JJ, Roche JR, Heiser A. Far-off and close-up feeding levels affect immunological performance in grazing dairy cows during the transition period. J Anim Sci 2019; 97:192-207. [PMID: 30428048 PMCID: PMC6313127 DOI: 10.1093/jas/sky427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Indexed: 12/12/2022] Open
Abstract
During the peripartum period, dairy cows often have signs of inflammation. Various stresses, including infectious and metabolic diseases, have been discussed as causative for this inflammation. In this study, expression profiles for 17 immune markers were measured in whole blood preparations from 78 dairy cows over a time frame starting 1 wk before calving to 4 wk after calving. Additionally, the effects of far-off and close-up feeding on immune function of dairy cows during the peripartum period were investigated. Cows were assigned to 1 of 2 feeding levels in late lactation to achieve a low and high BCS at the time of dry-off (approximately 4.25 and 5.0 on a 10-point scale). Following dry-off, both herds were managed to achieve a BCS of 5.0 one month before calving; this involved controlled feeding (i.e., maintenance) and over-feeding of ME during the far-off dry period. Within each far-off feeding-level treatment, cows were offered 65, 90, or 120% of their precalving ME requirements for 3 wk precalving in a 2 × 3 factorial arrangement. Analysis of gene expression profiles from blood cells revealed effects of time indicating that the transition cow's immune system counteracts the peripartum inflammation, whereas later postcalving it becomes activated to provide protection against postpartum infections. Far-off feeding affected (P < 0.05) the expression of 2 of the investigated genes at calving. Interleukin-6 (IL-6) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) expression in unstimulated, peripheral leukocytes were lower (P < 0.05) in animals from the Far-Off_Over-fed group compared with the Far-Off_Control-fed group. Close-up feeding had several effects on gene expression, indicating that immune function in Feed120 animals was distinct from the Feed90 and Feed65. In conclusion, feeding management precalving becomes an important intervention to ensure immunocompetence at and after calving.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Murray D Mitchell
- Centre for Clinical Research, University of Queensland, Royal Brisbane and Women’s Hospital Campus, Herston, Australia
| | - Juan J Loor
- Department of Animal Sciences, University of Illinois, Urbana, IL
| | | | | |
Collapse
|
43
|
Sviridova AA, Melnikov MV, Belousova OO, Rogovskii VS, Pashenkov MV, Boyko AN. Serotonergic system as a therapeutic target in multiple sclerosis. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:64-72. [DOI: 10.17116/jnevro20191192264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Paudel S, Ghimire L, Jin L, Baral P, Cai S, Jeyaseelan S. NLRC4 suppresses IL-17A-mediated neutrophil-dependent host defense through upregulation of IL-18 and induction of necroptosis during Gram-positive pneumonia. Mucosal Immunol 2019; 12:247-257. [PMID: 30279514 PMCID: PMC6301100 DOI: 10.1038/s41385-018-0088-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/15/2018] [Accepted: 09/04/2018] [Indexed: 02/04/2023]
Abstract
Gram-positive pathogens, including Staphylococcus aureus, cause necrotizing pneumonia. The central feature of S. aureus pneumonia is toxin-induced necroptosis of immune and resident cells, which impedes host defense. However, the role of the NLRC4 in the lung following S. aureus infection remains elusive. Here, we demonstrate that S. aureus activates the NLRC4 to drive necroptosis and IL-18 production, which impaired IL-17A-dependent neutrophil-mediated host susceptibility. In particular, Nlrc4-/- mice exhibit reduced necroptosis, enhanced neutrophil influx into the lungs, decreased bacterial burden, and improved host survival. Loss of NLRC4 signaling in both hematopoietic and non-hematopoietic cells contributes to the host protection against S. aureus pneumonia. Secretion of IL-17A by γδ T cells is essential for neutrophil recruitment into the lungs of Nlrc4-/- mice following infection. Moreover, treatment of wild-type mice with necroptosis inhibitors or genetic ablation of MLKL and IL-18 improves host defense against S. aureus infection, which is associated with increased IL-17A+γδ T cells and neutrophils. Taken together, these novel findings reveal that S. aureus activates the NLRC4 to dampen IL-17A-dependent neutrophil accumulation through induction of necroptosis and IL-18. Thus, modulating the function of the NLRC4 may be an attractive therapeutic approach for treating S. aureus infections.
Collapse
Affiliation(s)
- Sagar Paudel
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Experimental Infectious Disease Research, School of Veterinary Medicine, Louisiana State University (LSU), Baton Rouge, Louisiana, USA, 70803
| | - Laxman Ghimire
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Experimental Infectious Disease Research, School of Veterinary Medicine, Louisiana State University (LSU), Baton Rouge, Louisiana, USA, 70803
| | - Liliang Jin
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Experimental Infectious Disease Research, School of Veterinary Medicine, Louisiana State University (LSU), Baton Rouge, Louisiana, USA, 70803
| | - Pankaj Baral
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Experimental Infectious Disease Research, School of Veterinary Medicine, Louisiana State University (LSU), Baton Rouge, Louisiana, USA, 70803
| | - Shanshan Cai
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Experimental Infectious Disease Research, School of Veterinary Medicine, Louisiana State University (LSU), Baton Rouge, Louisiana, USA, 70803
| | - Samithamby Jeyaseelan
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Experimental Infectious Disease Research, School of Veterinary Medicine, Louisiana State University (LSU), Baton Rouge, Louisiana, USA, 70803.,Section of Pulmonary and Critical Care, Department of Medicine, LSU Health Sciences Center, New Orleans, Louisiana, USA, 70112.,Address Correspondence: Dr. Samithamby Jeyaseelan, Laboratory of Lung Biology, Department of Pathobiological Sciences, Louisiana State University (LSU), Baton Rouge, Louisiana, USA, 70803. Phone: +1 225 578 9524;
| |
Collapse
|
45
|
Kim D, McAlees JW, Bischoff LJ, Kaur D, Houshel LK, Gray J, Hargis J, Davis X, Dudas PL, Deshmukh H, Lewkowich IP. Combined administration of anti-IL-13 and anti-IL-17A at individually sub-therapeutic doses limits asthma-like symptoms in a mouse model of Th2/Th17 high asthma. Clin Exp Allergy 2018; 49:317-330. [PMID: 30353972 DOI: 10.1111/cea.13301] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/17/2018] [Accepted: 10/08/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Recent studies have demonstrated that Th2 responses have the ability to antagonize Th17 responses. In mouse models of allergic asthma, blockade of Th2-effector cytokines results in elaboration of Th17 responses and associated increases in pulmonary neutrophilia. While these can be controlled by simultaneous blockade of Th17-associated effector cytokines, clinical trials of anti-IL-17/IL-17RA blocking therapies have demonstrated increased of risk of bacterial and fungal infections. Identification of minimally effective doses of cytokine-blocking therapies with the goal of reducing the potential emergence of infection-related complications is a translationally relevant goal. OBJECTIVE In the current report, we examine whether combined blockade of IL-13 and IL-17A, at individually sub-therapeutic levels, can limit the development of allergic asthma while sparing expression of IL-17A-associated anti-microbial effectors. METHODS House dust mite was given intratracheally to A/J mice. Anti-IL-13 and anti-IL-17A antibodies were administered individually, or concomitantly at sub-therapeutic doses. Airway hyper-reactivity, lung inflammation, magnitude of Th2- and Th17-associated cytokine production and expression of IL-13- and IL-17A-induced genes in the lungs was assessed. RESULTS Initial dosing studies identified sub-therapeutic levels of IL-13 and IL-17A blocking mAbs that have a limited effect on asthma parameters and do not impair responses to microbial products or infection. Subsequent studies demonstrated that combined sub-therapeutic dosing with IL-13 and IL-17A blocking mAbs resulted in significant improvement in airway hyperresponsiveness (AHR) and expression of IL-13-induced gene expression. Importantly, these doses neither exacerbated nor inhibited production of Th17-associated cytokines, or IL-17A-associated gene expression. CONCLUSION This study suggests that combining blockade of individual Th2 and Th17 effector cytokines, even at individually sub-therapeutic levels, may be sufficient to limit disease development while preserving important anti-microbial pathways. Such a strategy may therefore have reduced potential for adverse events associated with blockade of these pathways.
Collapse
Affiliation(s)
- Dasom Kim
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jaclyn W McAlees
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lindsay J Bischoff
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Davinder Kaur
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lauren K Houshel
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jerilyn Gray
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Julie Hargis
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Xenia Davis
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Paul L Dudas
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Hitesh Deshmukh
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Ian P Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
46
|
Khatoon J, Prasad KN, Rai RP, Shukla SK, Krishnani N, Ghoshal UC. Expression levels of A disintegrin and metalloproteases (ADAMs), and Th17-related cytokines and their association with Helicobacter pylori infection in patients with gastroduodenal diseases. Pathog Dis 2018; 76:5145580. [PMID: 30371773 DOI: 10.1093/femspd/fty078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/25/2018] [Indexed: 02/06/2023] Open
Abstract
Expression levels of A disintegrin and metalloproteases (ADAMs) (10 and 17) and Th17-related cytokines [interleukin (IL) 17A, IL-17F, IL-33, IL-23, IL-23R] were investigated by quantitative real time polymerase chain reaction in gastric biopsies of patients with different gastroduodenal pathologies in the presence and absence of Helicobacter pylori infection. Patients with gastric cancer (GC) (n = 70, intestinal-type 38 and diffuse type 32), peptic ulcer disease [n = 50, duodenal ulcer (DU) 16 and gastric ulcer (GU) 34] and functional dyspepsia (n = 120) were included in the study. Further, the expression levels of ADAMs and Th17 cytokines were correlated with H. pylori cytotoxin-associated genes pathogenicity island (cagPAI) status. Expression levels of ADAMs (10 and 17) and Th17-related cytokines (IL-17A, IL-23, IL-23R) were significantly higher in H. pylori-positive than in H. pylori-negative gastric biopsies. Significant increase in ADAM17 and Th17 cytokines (IL-17A and IL-23) expressions was observed in patients with GU and intestinal-type GC in the presence of H. pylori infection and in strains harbouring intact cagPAI. Expression levels of IL-17A, IL-23 and ADAM17 were strongly correlated with GU and intestinal-type GC and weakly with DU and diffuse-type GC in the presence of H. pylori infection. Higher expression levels of ADAM17 and Th17 cytokines (IL-17A and IL-23), and their strong correlation with GU and intestinal-type GC patients in the presence of H. pylori and its intact cagPAI status, suggest a possible role of strain specificity in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Jahanarah Khatoon
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Kashi Nath Prasad
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Ravi Prakash Rai
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Sanket Kumar Shukla
- Department of Medicine, Center of Translational Medicine, Thomas Jefferson University, Philadelphia, PA-19107 USA
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Uday Chand Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| |
Collapse
|
47
|
Park HE, Park HT, Jung YH, Yoo HS. Gene expression profiles of immune-regulatory genes in whole blood of cattle with a subclinical infection of Mycobacterium avium subsp. paratuberculosis. PLoS One 2018; 13:e0196502. [PMID: 29698503 PMCID: PMC5919679 DOI: 10.1371/journal.pone.0196502] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Johne’s disease is a chronic wasting disease of ruminants caused by Mycobacterium avium subsp. paratuberculosis (MAP), resulting in inflammation of intestines and persistent diarrhea. The initial host response against MAP infections is mainly regulated by the Th1 response, which is characterized by the production of IFN-γ. With the progression of disease, MAP can survive in the host through the evasion of the host’s immune response by manipulating the host immune response. However, the host response during subclinical phases has not been fully understood. Immune regulatory genes, including Th17-derived cytokines, interferon regulatory factors, and calcium signaling-associated genes, are hypothesized to play an important role during subclinical phases of Johne’s disease. Therefore, the present study was conducted to analyze the expression profiles of immune regulatory genes during MAP infection in whole blood. Different expression patterns of genes were identified depending on the infection stages. Downregulation of IL-17A, IL-17F, IL-22, IL-26, HMGB1, and IRF4 and upregulation of PIP5K1C indicate suppression of the Th1 response due to MAP infection and loss of granuloma integrity. In addition, increased expression of IRF5 and IRF7 suggest activation of IFN-α/β signaling during subclinical stages, which induced indoleamine 2,3-dioxygenase mediated depletion of tryptophan metabolism. Increased expression of CORO1A indicate modulation of calcium signaling, which enhanced the survival of MAP. Taken together, distinct host gene expression induced by MAP infection indicates enhanced survival of MAP during subclinical stages.
Collapse
Affiliation(s)
- Hyun-Eui Park
- Department of Infectious Disease, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hong-Tae Park
- Department of Infectious Disease, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Young Hoon Jung
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Han Sang Yoo
- Department of Infectious Disease, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
- * E-mail:
| |
Collapse
|
48
|
Li Z, Kang H, You Q, Ossa F, Mead P, Quinton M, Karrow NA. In vitro bioassessment of the immunomodulatory activity of Saccharomyces cerevisiae components using bovine macrophages and Mycobacterium avium ssp. paratuberculosis. J Dairy Sci 2018; 101:6271-6286. [PMID: 29655556 DOI: 10.3168/jds.2017-13805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/24/2018] [Indexed: 12/18/2022]
Abstract
The yeast Saccharomyces cerevisiae and its components are used for the prevention and treatment of enteric disease in different species; therefore, they may also be useful for preventing Johne's disease, a chronic inflammatory bowel disease of ruminants caused by Mycobacterium avium ssp. paratuberculosis (MAP). The objective of this study was to identify potential immunomodulatory S. cerevisiae components using a bovine macrophage cell line (BOMAC). The BOMAC phagocytic activity, reactive oxygen species production, and immune-related gene (IL6, IL10, IL12p40, IL13, IL23), transforming growth factor β, ARG1, CASP1, and inducible nitric oxide synthase expression were investigated when BOMAC were cocultured with cell wall components from 4 different strains (A, B, C, and D) and 2 forms of dead yeast from strain A. The BOMAC phagocytosis of mCherry-labeled MAP was concentration-dependently attenuated when BOMAC were cocultured with yeast components for 6 h. Each yeast derivative also induced a concentration-dependent increase in BOMAC reactive oxygen species production after a 6-h exposure. In addition, BOMAC mRNA expression of the immune-related genes was investigated after 6 and 24 h of exposure to yeast components. All yeast components were found to regulate the immunomodulatory genes of BOMAC; however, the response varied among components and over time. The in vitro bioassessment studies reported here suggest that dead yeast and its cell wall components may be useful for modulating macrophage function before or during MAP infection.
Collapse
Affiliation(s)
- Z Li
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - H Kang
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Q You
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - F Ossa
- Lallemand Inc., Montréal, QC, Canada, H4P 2R2
| | - P Mead
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - M Quinton
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - N A Karrow
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1.
| |
Collapse
|
49
|
The influence of biogenic amines on Th17-mediated immune response in multiple sclerosis. Mult Scler Relat Disord 2018; 21:19-23. [DOI: 10.1016/j.msard.2018.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 12/13/2022]
|
50
|
Gourdin N, Bossennec M, Rodriguez C, Vigano S, Machon C, Jandus C, Bauché D, Faget J, Durand I, Chopin N, Tredan O, Marie JC, Dubois B, Guitton J, Romero P, Caux C, Ménétrier-Caux C. Autocrine Adenosine Regulates Tumor Polyfunctional CD73 +CD4 + Effector T Cells Devoid of Immune Checkpoints. Cancer Res 2018; 78:3604-3618. [PMID: 29559470 DOI: 10.1158/0008-5472.can-17-2405] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 02/08/2018] [Accepted: 03/16/2018] [Indexed: 11/16/2022]
Abstract
The production of CD73-derived adenosine (Ado) by Tregs has been proposed as a resistance mechanism to anti-PD-1 therapy in murine tumor models. We reported that human Tregs express the ectonucleotidase CD39, which generates AMP from ATP, but do not express the AMPase CD73. In contrast, CD73 defined a subset of effector CD4+ T cells (Teffs) enriched in polyfunctional Th1.17 cells characterized by expression of CXCR3, CCR6, and MDR1, and production of IL17A/IFNγ/IL22/GM-CSF. CD39+ Tregs selectively targeted CD73+ Teffs through cooperative degradation of ATP into Ado inhibiting and restricting the ability of CD73+ Teffs to secrete IL17A. CD73+ Teffs infiltrating breast and ovarian tumors were functionally blunted by Tregs expressing upregulated levels of CD39 and ATPase activity. Moreover, tumor-infiltrating CD73+ Teffs failed to express inhibitory immune checkpoints, suggesting that CD73 might be selected under pressure from immune checkpoint blockade therapy and thus may represent a nonredundant target for restoring antitumor immunity.Significance: Polyfunctional CD73+ T-cell effectors lacking other immune checkpoints are selectively targeted by CD39 overexpressing Tregs that dominate the breast tumor environment. Cancer Res; 78(13); 3604-18. ©2018 AACR.
Collapse
Affiliation(s)
- Nicolas Gourdin
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,Innovation and Translational Research Department, Centre Léon Bérard, Lyon, France
| | - Marion Bossennec
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France
| | - Céline Rodriguez
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,Innovation and Translational Research Department, Centre Léon Bérard, Lyon, France
| | - Selena Vigano
- Ludwig Cancer Research Center, Department of Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Christelle Machon
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Laboratoire de Biochimie et Toxicologie, Pierre-Bénite, France.,Université de Lyon, Université Lyon 1, ISPB Faculté de pharmacie, Laboratoire de Chimie Analytique, Lyon, France
| | - Camilla Jandus
- Ludwig Cancer Research Center, Department of Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - David Bauché
- Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,TGF-β and Immuno-evasion Department of Immunology Virology and Inflammation, INSERM U1052, Cancer Research Center of Lyon, Lyon, France.,TGF-β and Immuno-evasion, Tumor immunology Program, DKFZ, Heidelberg, Germany
| | - Julien Faget
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,Innovation and Translational Research Department, Centre Léon Bérard, Lyon, France
| | - Isabelle Durand
- Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,Cytometry platform, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France
| | - Nicolas Chopin
- Centre Léon Bérard, Medical Oncology Department, Lyon, France
| | - Olivier Tredan
- Centre Léon Bérard, Medical Oncology Department, Lyon, France
| | - Julien C Marie
- Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,TGF-β and Immuno-evasion Department of Immunology Virology and Inflammation, INSERM U1052, Cancer Research Center of Lyon, Lyon, France.,TGF-β and Immuno-evasion, Tumor immunology Program, DKFZ, Heidelberg, Germany
| | - Bertrand Dubois
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France
| | - Jérôme Guitton
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Laboratoire de Biochimie et Toxicologie, Pierre-Bénite, France.,Université de Lyon, Université Lyon 1, ISPB Faculté de pharmacie, Laboratoire de Toxicologie, Lyon, France
| | - Pedro Romero
- Ludwig Cancer Research Center, Department of Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Christophe Caux
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,Innovation and Translational Research Department, Centre Léon Bérard, Lyon, France
| | - Christine Ménétrier-Caux
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France. .,Team 11, Immunology Virology Inflammation (IVI) Department, INSERM U-1052, Cancer Research Center of Lyon, Lyon, France.,Innovation and Translational Research Department, Centre Léon Bérard, Lyon, France
| |
Collapse
|