1
|
Konovalovas A, Armalytė J, Klimkaitė L, Liveikis T, Jonaitytė B, Danila E, Bironaitė D, Mieliauskaitė D, Bagdonas E, Aldonytė R. Insights into respiratory microbiome composition and systemic inflammatory biomarkers of bronchiectasis patients. Microbiol Spectr 2024; 12:e0414423. [PMID: 39535197 PMCID: PMC11619244 DOI: 10.1128/spectrum.04144-23] [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: 12/07/2023] [Accepted: 07/17/2024] [Indexed: 11/16/2024] Open
Abstract
The human microbiomes, including the ones present in the respiratory tract, are described and characterized in an increasing number of studies. However, the composition and the impact of the healthy and/or impaired microbiome on pulmonary health and its interaction with the host tissues remain enigmatic. In chronic airway diseases, bronchiectasis stands out as a progressive condition characterized by microbial colonization and infection. In this study, we aimed to investigate the microbiome of the lower airways and lungs of bronchiectasis patients together with their serum cytokine and chemokine content, and gain novel insights into the pathogenesis of bronchiectasis. The microbiome of 47 patients was analyzed by sequencing of full-length 16S rRNA gene using amplicon sequencing Oxford Nanopore technologies. Their serum inflammatory mediators content was quantified in parallel. Several divergently composed microbiome groups were identified and characterized, the majority of patients displayed one dominant bacterial species, whereas others had a more diverse microbiome. The analysis of systemic immune biomarkers revealed two distinct inflammatory response groups, i.e., low and high response groups, each associated with a specific array of clinical symptoms, microbial composition, and diversity. Moreover, we have identified some microbiome compositions associated with high inflammatory response, i.e., high levels of pro- and anti-inflammatory cytokines, whereas other microbiomes were in correlation with low inflammatory responses. Although bronchiectasis pathogenetic mechanisms remain to be elucidated, it is clear that addressing microbiome composition in the airways is a valuable resource not only for diagnosis but also for personalized disease management. IMPORTANCE The population of microorganisms on/in the human body resides in distinct local microbiomes, including the respiratory microbiome. It remains unclear what defines a healthy and a diseased respiratory microbiome. We investigated the respiratory microbiome in chronic pulmonary infectious disease, i.e., bronchiectasis, and researched correlations between microbiome composition, systemic inflammatory biomarkers, and disease characteristics. The bronchoalveolar microbiome of 47 patients was sequenced, and their serum inflammatory mediators were quantified. The microbiomes were grouped based on their content and diversity. In addition, patients were also grouped into low- and high-response groups according to their inflammatory biomarkers' levels. Certain microbiome compositions, mainly single-species dominated, were associated with high levels of inflammatory cytokines, whereas others correlated with low inflammatory response and remained diverse. We conclude that respiratory microbiome composition is a valuable resource for the diagnostics and personalized management of bronchiectasis, which may include preserving microbiome diversity and introducing possible probiotics.
Collapse
Affiliation(s)
- Aleksandras Konovalovas
- Life Sciences Center, Institute of Biosciences, Vilnius University, Vilnius, Lithuania
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Julija Armalytė
- Life Sciences Center, Institute of Biosciences, Vilnius University, Vilnius, Lithuania
| | - Laurita Klimkaitė
- Life Sciences Center, Institute of Biosciences, Vilnius University, Vilnius, Lithuania
| | - Tomas Liveikis
- Life Sciences Center, Institute of Biosciences, Vilnius University, Vilnius, Lithuania
| | - Brigita Jonaitytė
- Clinic of Chest Diseases, Immunology, and Allergology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Edvardas Danila
- Clinic of Chest Diseases, Immunology, and Allergology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Centre of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Daiva Bironaitė
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | | | - Edvardas Bagdonas
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Rūta Aldonytė
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|
2
|
Wang H, Yu L, Cheng L, Guo Z. The roles of lncRNAs in Th17-associated diseases, with special focus on JAK/STAT signaling pathway. Clin Exp Med 2023; 23:3349-3359. [PMID: 37743424 DOI: 10.1007/s10238-023-01181-3] [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: 03/27/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023]
Abstract
One of the most crucial T cell subsets in a variety of autoimmune and chronic inflammatory illnesses is T helper (Th) 17 cells. Th17 cells appear to have an essential role in the clearance of extracellular pathogens during infections. However, Th17 cells are also involved in inflammation and have been implicated in the pathogenesis of several autoimmune diseases and human inflammatory conditions. Due to the involvement of Th17 cells in the onset of Th17-associated diseases, understanding molecular mechanisms of Th17 cell functions may open the door to developing tailored therapies to address these difficult disorders. However, the molecular mechanisms governing Th17 differentiation in various diseases are still not well understood. The JAK/STAT signaling pathway plays a critical role in immune responses and has been linked to various aspects of Th17 cell differentiation and function. In this article, we conducted a comprehensive review of various molecular mechanisms (JAK/STAT, microRNAs, etc.), that can affect the differentiation of Th17 cells in various Th17-associated diseases.
Collapse
Affiliation(s)
- Han Wang
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Lanlan Yu
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Li Cheng
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Zhigang Guo
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, 130031, China.
| |
Collapse
|
3
|
Saadh MJ, Arellano MTC, Saini RS, Amin AH, Sharma N, Arias-Gonzáles JL, Alsandook T, Cotrina-Aliaga JC, Akhavan-Sigari R. Molecular mechanisms of long non-coding RNAs in differentiation of T Helper17 cells. Int Immunopharmacol 2023; 123:110728. [PMID: 37572506 DOI: 10.1016/j.intimp.2023.110728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
T helper (Th) 17 cells are one of the most important T cell subsets in a number of autoimmune and chronic inflammatory diseases. During infections, Th17 cells appear to play an important role in the clearance of extracellular pathogens. Th17 cells, on the other hand, are engaged in inflammation and have been linked to the pathophysiology of a number of autoimmune illnesses and human inflammatory disorders. A diverse group of RNA molecules known as lncRNAs serve critical functions in gene expression regulation. They may interact with a wide range of molecules, including DNA, RNA, and proteins, and have a complex structure. LncRNAs, which have restricted or no protein-coding activity, are implicated in a number of illnesses due to their regulatory impact on a variety of biological processes such as cell proliferation, apoptosis, and differentiation. Several lncRNAs have been associated with Th7 cell development in the context of immune cell differentiation. In this article, we cover new studies on the involvement of lncRNAs in Th17 cell differentiation in a variety of disorders, including auto-immune diseases, malignancies, asthma, heart disease, and infections.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan.
| | | | | | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Nidhi Sharma
- Department of Computer Engineering & Application, GLA University, Mathura, India.
| | | | - Tahani Alsandook
- Dentistry Department, Al-Turath University College, Baghdad, Iraq.
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland.
| |
Collapse
|
4
|
Li Y, Yang Y, Chen D, Wang Y, Zhang X, Li W, Chen S, Wong SM, Shen M, Akerley BJ, Shen H. Memory Th17 cell-mediated protection against lethal secondary pneumococcal pneumonia following influenza infection. mBio 2023; 14:e0051923. [PMID: 37222516 PMCID: PMC10470593 DOI: 10.1128/mbio.00519-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/11/2023] [Indexed: 05/25/2023] Open
Abstract
Streptococcus pneumoniae (Sp) frequently causes secondary pneumonia after influenza A virus (IAV) infection, leading to high morbidity and mortality worldwide. Concomitant pneumococcal and influenza vaccination improves protection against coinfection but does not always yield complete protection. Impaired innate and adaptive immune responses have been associated with attenuated bacterial clearance in influenza virus-infected hosts. In this study, we showed that preceding low-dose IAV infection caused persistent Sp infection and suppression of bacteria-specific T-helper type 17 (Th17) responses in mice. Prior Sp infection protected against subsequent IAV/Sp coinfection by improving bacterial clearance and rescuing bacteria-specific Th17 responses in the lungs. Furthermore, blockade of IL-17A by anti-IL-17A antibodies abrogated the protective effect of Sp preinfection. Importantly, memory Th17 responses induced by Sp preinfection overcame viral-driven Th17 inhibition and provided cross-protection against different Sp serotypes following coinfection with IAV. These results indicate that bacteria-specific Th17 memory cells play a key role in providing protection against IAV/Sp coinfection in a serotype-independent manner and suggest that a Th17-based vaccine would have excellent potential to mitigate disease caused by coinfection. IMPORTANCE Streptococcus pneumoniae (Sp) frequently causes secondary bacterial pneumonia after influenza A virus (IAV) infection, leading to increased morbidity and mortality worldwide. Current pneumococcal vaccines induce highly strain-specific antibody responses and provide limited protection against IAV/Sp coinfection. Th17 responses are broadly protective against Sp single infection, but whether the Th17 response, which is dramatically impaired by IAV infection in naïve mice, might be effective in immunization-induced protection against pneumonia caused by coinfection is not known. In this study, we have revealed that Sp-specific memory Th17 cells rescue IAV-driven inhibition and provide cross-protection against subsequent lethal coinfection with IAV and different Sp serotypes. These results indicate that a Th17-based vaccine would have excellent potential to mitigate disease caused by IAV/Sp coinfection.
Collapse
Affiliation(s)
- Yong Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Shanghai Institute of Immunology, Shanghai Jiaotong University, Shanghai, China
| | - Ying Yang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dafan Chen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Xinyun Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenchao Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shengsen Chen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Endoscopy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Sandy M. Wong
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Mengwen Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Emergency Medical, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Brian J. Akerley
- Department of Cell and Molecular Biology, Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Zhang X, Yang Y, Chen S, Li W, Li Y, Akerley BJ, Shao L, Zhang W, Shen H, Abt MC. Antigen-specific memory Th17 cells promote cross-protection against nontypeable Haemophilus influenzae after mild influenza A virus infection. Mucosal Immunol 2023; 16:153-166. [PMID: 36736665 DOI: 10.1016/j.mucimm.2023.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
Secondary bacterial pneumonia after influenza A virus (IAV) infection is the leading cause of hospitalization and death associated with IAV infection worldwide. Nontypeable Haemophilus influenzae (NTHi) is one of the most common causes of secondary bacterial pneumonia. Current efforts to develop vaccines against NTHi infection focus on inducing antibodies but are hindered by antigenic diversity among NTHi strains. Therefore, we investigated the contribution of the memory T helper type 17 (Th17) response in protective immunity against IAV/NTHi coinfection. We observed that even a mild IAV infection impaired the NTHi-specific Th17 response and increased morbidity and mortality compared with NTHi monoinfected mice. However, pre-existing memory NTHi-specific Th17 cells induced by a previous NTHi infection overcame IAV-driven Th17 inhibition and were cross-protective against different NTHi strains. Last, mice immunized with a NTHi protein that induced a strong Th17 memory response were broadly protected against diverse NTHi strains after challenge with coinfection. These results indicate that vaccination that limits IAV infection to mild disease may be insufficient to eliminate the risk of a lethal secondary bacterial pneumonia. However, NTHi-specific memory Th17 cells provide serotype-independent protection despite an ongoing IAV infection and demonstrate the advantage of developing broadly protective Th17-inducing vaccines against secondary bacterial pneumonia.
Collapse
Affiliation(s)
- Xinyun Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Ying Yang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - ShengSen Chen
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA; Department of Endoscopy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wenchao Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA; Shanghai Institute of Immunology, Shanghai Jiaotong University, Shanghai, China; Department of Immunology and Rheumatology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yong Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA; Shanghai Institute of Immunology, Shanghai Jiaotong University, Shanghai, China
| | - Brian J Akerley
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Linyun Shao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA.
| | - Michael C Abt
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA.
| |
Collapse
|
6
|
Muruganandah V, Kupz A. Immune responses to bacterial lung infections and their implications for vaccination. Int Immunol 2021; 34:231-248. [PMID: 34850883 DOI: 10.1093/intimm/dxab109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/28/2021] [Indexed: 11/14/2022] Open
Abstract
The pulmonary immune system plays a vital role in protecting the delicate structures of gaseous exchange against invasion from bacterial pathogens. With antimicrobial resistance becoming an increasing concern, finding novel strategies to develop vaccines against bacterial lung diseases remains a top priority. In order to do so, a continued expansion of our understanding of the pulmonary immune response is warranted. Whilst some aspects are well characterised, emerging paradigms such as the importance of innate cells and inducible immune structures in mediating protection provide avenues of potential to rethink our approach to vaccine development. In this review, we aim to provide a broad overview of both the innate and adaptive immune mechanisms in place to protect the pulmonary tissue from invading bacterial organisms. We use specific examples from several infection models and human studies to depict the varying functions of the pulmonary immune system that may be manipulated in future vaccine development. Particular emphasis has been placed on emerging themes that are less reviewed and underappreciated in vaccine development studies.
Collapse
Affiliation(s)
- Visai Muruganandah
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4878, Australia
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4878, Australia
| |
Collapse
|
7
|
Xiang L, Meng X. Emerging cellular and molecular interactions between the lung microbiota and lung diseases. Crit Rev Microbiol 2021; 48:577-610. [PMID: 34693852 DOI: 10.1080/1040841x.2021.1992345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With the discovery of the lung microbiota, its study in both pulmonary health and disease has become a vibrant area of emerging research interest. Thus far, most studies have described the lung microbiota composition in lung disease quite well, and some of these studies indicated alterations in lung microbial communities related to the onset and development of lung disease and vice versa. However, the underlying mechanisms, particularly the cellular and molecular links, are still largely unknown. In this review, we highlight the current progress in the complex cellular and molecular mechanisms by which the lung microbiome interacts with immune homeostasis and pulmonary disease pathogenesis to advance our understanding of the elaborate function of the lung microbiota in lung disease. We hope that this work can attract more attention to this still-young yet very promising field to facilitate the identification of new therapeutic targets and provide more innovative therapies. Additional accurate standard-based methodologies and technological breakthroughs are critical to propel the field forward to ultimately achieve the goal of maintaining respiratory health.
Collapse
Affiliation(s)
- Li Xiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Mehata AK, Viswanadh MK, Priya V, Vikas, Muthu MS. Harnessing immunological targets for COVID-19 immunotherapy. Future Virol 2021. [PMID: 34447458 PMCID: PMC8375415 DOI: 10.2217/fvl-2021-0048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/16/2021] [Indexed: 12/22/2022]
Abstract
COVID-19 is an infectious and highly contagious disease caused by SARS-CoV-2. The immunotherapy strategy has a great potential to develop a permanent cure against COVID-19. Innate immune cells are in constant motion to scan molecular alteration to cells led by microbial infections throughout the body and helps in clearing invading viruses. Harnessing immunological targets for removing viral infection, generally based on the principle of enhancing the T-cell and protective immune responses. Currently-approved COVID-19 vaccines are mRNA encapsulated in liposomes that stimulate the host immune system to produce antibodies. Given the vital role of innate immunity, harnessing these immune responses opens up new hope for the generation of long-lasting and protective immunity against COVID-19.
Collapse
Affiliation(s)
- Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Matte Kasi Viswanadh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Vishnu Priya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Vikas
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| |
Collapse
|
9
|
Kadry NA, Porsch EA, Shen H, St Geme JW. Immunization with HMW1 and HMW2 adhesins protects against colonization by heterologous strains of nontypeable Haemophilus influenzae. Proc Natl Acad Sci U S A 2021; 118:e2019923118. [PMID: 34344825 PMCID: PMC8364133 DOI: 10.1073/pnas.2019923118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) is a common cause of localized respiratory tract disease and results in significant morbidity. The pathogenesis of NTHi disease begins with nasopharyngeal colonization, and therefore, the prevention of colonization represents a strategy to prevent disease. The NTHi HMW1 and HMW2 proteins are a family of conserved adhesins that are present in 75 to 80% of strains and have been demonstrated to play a critical role in colonization of the upper respiratory tract in rhesus macaques. In this study, we examined the vaccine potential of HMW1 and HMW2 using a mouse model of nasopharyngeal colonization. Immunization with HMW1 and HMW2 by either the subcutaneous or the intranasal route resulted in a strain-specific antibody response associated with agglutination of bacteria and restriction of bacterial adherence. Despite the specificity of the antibody response, immunization resulted in protection against colonization by both the parent NTHi strain and heterologous strains expressing distinct HMW1 and HMW2 proteins. Pretreatment with antibody against IL-17A eliminated protection against heterologous strains, indicating that heterologous protection is IL-17A dependent. This work demonstrates the vaccine potential of the HMW1 and HMW2 proteins and highlights the importance of IL-17A in protection against diverse NTHi strains.
Collapse
Affiliation(s)
- Nadia A Kadry
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Eric A Porsch
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Joseph W St Geme
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104;
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
10
|
Zhao B, Yang J, He B, Li X, Yan H, Liu S, Yang Y, Zhou D, Liu B, Fan X, Zhong M, Zhang E, Zhang F, Zhang Y, Chen YQ, Jiang S, Yan H. A safe and effective mucosal RSV vaccine in mice consisting of RSV phosphoprotein and flagellin variant. Cell Rep 2021; 36:109401. [PMID: 34289371 DOI: 10.1016/j.celrep.2021.109401] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/03/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of serious acute lower respiratory tract infection in infants and the elderly. The lack of a licensed RSV vaccine calls for the development of vaccines with other targets and vaccination strategies. Here, we construct a recombinant protein, designated P-KFD1, comprising RSV phosphoprotein (P) and the E.-coli-K12-strain-derived flagellin variant KFD1. Intranasal immunization with P-KFD1 inhibits RSV replication in the upper and lower respiratory tract and protects mice against lung disease without vaccine-enhanced disease (VED). The P-specific CD4+ T cells provoked by P-KFD1 intranasal (i.n.) immunization either reside in or migrate to the respiratory tract and mediate protection against RSV infection. Single-cell RNA sequencing (scRNA-seq) and carboxyfluorescein succinimidyl ester (CFSE)-labeled cell transfer further characterize the Th1 and Th17 responses induced by P-KFD1. Finally, we find that anti-viral protection depends on either interferon-γ (IFN-γ) or interleukin-17A (IL-17A). Collectively, P-KFD1 is a promising safe and effective mucosal vaccine candidate for the prevention of RSV infection.
Collapse
Affiliation(s)
- Bali Zhao
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jingyi Yang
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Bing He
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Xian Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Hu Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shuning Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Dihan Zhou
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Bowen Liu
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xuxu Fan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Maohua Zhong
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; 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, Wuhan 430065, China
| | - Ejuan Zhang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fan Zhang
- The Core Facility and Technical Support, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yue Zhang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yao-Qing Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Shibo Jiang
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Huimin Yan
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
11
|
The Adjuvants Polyphosphazene (PCEP) and a Combination of Curdlan Plus Leptin Promote a Th17-Type Immune Response to an Intramuscular Vaccine in Mice. Vaccines (Basel) 2021; 9:vaccines9050507. [PMID: 34069081 PMCID: PMC8156850 DOI: 10.3390/vaccines9050507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022] Open
Abstract
Our aim was to determine whether polyphosphazene (PCEP), Curdlan (β-glucan, a dectin-1 agonist), and Leptin could act as adjuvants to promote a Th17-type adaptive immune response in mice. Mice were vaccinated via the intramuscular route then boosted three weeks later with Ovalbumin plus: PCEP, Leptin, Curdlan, PCEP+Curdlan, Curdlan+Leptin, or saline. Mice vaccinated with OVA+PCEP and OVA+Curdlan+Leptin showed significantly higher frequency of antigen-specific CD4+ T cells secreting IL-17 relative to OVA-vaccinated mice. No formulation increased the frequency of CD4+ T cells secreting IL-4 or IFNγ. Since activation of innate immunity precedes the development of adaptive immunity, we wished to establish whether induction of Th17-type immunity could be predicted from in vitro experiments and/or from the local cytokine environment after immunization with adjuvants alone. Elevated IL-6 and TGFβ with reduced secretion of IL-12 is a cytokine milieu known to promote differentiation of Th17-type immunity. We injected the immunostimulants or saline buffer into murine thigh muscles and measured acute local cytokine production. PCEP induced significant production of IL-6 and reduced IL-12 production in muscle but it did not lead to elevated TGFβ production. Curdlan+Leptin injected into muscle induced significant production of TGFβ and IL-17 but not IL-6 or IL-12. We also stimulated splenocytes with media or PCEP, Leptin, Curdlan, PCEP+Curdlan, Curdlan+Leptin, PCEP+Leptin, and PCEP+Curdlan+Leptin and measured cytokine production. PCEP stimulation of splenocytes failed to induce significant production of IL-6, IL-12, TGFβ, or IL-17 and therefore ex vivo splenocyte stimulation failed to predict the increased frequency of Th17-type T cells in response to the vaccine. Curdlan-stimulated splenocytes produced Th1-type, inducing cytokine, IL-12. Curdlan+/-PCEP stimulated TGF-β production and Curdlan+Leptin+/- PCEP induced secretion of IL-17. We conclude that PCEP as well as Curdlan+Leptin are Th17-type vaccine adjuvants in mice but that cytokines produced in response to these adjuvants in muscle after injection or in ex vivo cultured splenocytes did not predict their role as a Th17-type adjuvant. Together, these data suggest that the cytokine environments induced by these immunostimulants did not predict induction of an antigen-specific Th17-type adaptive immune response. This is the first report of these adjuvants inducing a Th17-type adaptive immune response.
Collapse
|
12
|
Saliu F, Rizzo G, Bragonzi A, Cariani L, Cirillo DM, Colombo C, Daccò V, Girelli D, Rizzetto S, Sipione B, Cigana C, Lorè NI. Chronic infection by nontypeable Haemophilus influenzae fuels airway inflammation. ERJ Open Res 2021; 7:00614-2020. [PMID: 33778054 PMCID: PMC7983230 DOI: 10.1183/23120541.00614-2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) is commonly isolated from airways of patients suffering from chronic respiratory diseases, such as COPD or cystic fibrosis (CF). However, to what extent NTHi long-term infection contributes to the lung inflammatory burden during chronic airway disease is still controversial. Here, we exploited human respiratory samples from a small cohort of CF patients and found that patients chronically infected with NTHi had significantly higher levels of interleukin (IL)-8 and CXCL1 than those who were not infected. To better define the impact of chronic NTHi infection in fuelling inflammatory response in chronic lung diseases, we developed a new mouse model using both laboratory and clinical strains. Chronic NTHi infection was associated with chronic inflammation of the lung, characterised by recruitment of neutrophils and cytokine release keratinocyte-derived chemokine (KC), macrophage inflammatory protein 2 (MIP-2), granulocyte colony-stimulating factor (G-CFS), IL-6, IL-17A and IL-17F) at 2 and 14 days post-infection. An increased burden of T-cell-mediated response (CD4+ and γδ cells) and higher levels of pro-matrix metalloproteinase 9 (pro-MMP9), known to be associated with tissue remodelling, were observed at 14 days post-infection. Of note we found that both CD4+IL-17+ cells and levels of IL-17 cytokines were enriched in mice at advanced stages of NTHi chronic infection. Moreover, by immunohistochemistry we found CD3+, B220+ and CXCL-13+ cells localised in bronchus-associated lymphoid tissue-like structures at day 14. Our results demonstrate that chronic NTHi infection exerts a pro-inflammatory activity in the human and murine lung and could therefore contribute to the exaggerated burden of lung inflammation in patients at risk. The pathological impact of long-term infection by nontypeable Haemophilus influenzae (NTHi) is still debated. Chronic NTHi infection fuels lung inflammation in human samples and in a new mouse model of bacterial long-term persistence.https://bit.ly/3lvyvge
Collapse
Affiliation(s)
- Fabio Saliu
- IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Emerging bacterial pathogens Unit, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy.,IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Infections and cystic fibrosis unit, Milan, Italy
| | - Giulia Rizzo
- Università Vita-Salute San Raffaele, Milan, Italy.,IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Infections and cystic fibrosis unit, Milan, Italy
| | - Alessandra Bragonzi
- IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Infections and cystic fibrosis unit, Milan, Italy
| | - Lisa Cariani
- Cystic Fibrosis Microbiology Laboratory, Fondazione IRCCS Ca' Granda, Milan, Italy
| | - Daniela M Cirillo
- IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Emerging bacterial pathogens Unit, Milan, Italy
| | - Carla Colombo
- Cystic Fibrosis Regional Reference Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Valeria Daccò
- Cystic Fibrosis Regional Reference Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Daniela Girelli
- Cystic Fibrosis Microbiology Laboratory, Fondazione IRCCS Ca' Granda, Milan, Italy
| | - Sara Rizzetto
- Cystic Fibrosis Microbiology Laboratory, Fondazione IRCCS Ca' Granda, Milan, Italy
| | - Barbara Sipione
- IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Infections and cystic fibrosis unit, Milan, Italy
| | - Cristina Cigana
- IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Infections and cystic fibrosis unit, Milan, Italy
| | - Nicola I Lorè
- IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Emerging bacterial pathogens Unit, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy.,IRCCS San Raffaele Scientific Institute, Division of Immunology, Transplantation, and Infectious Diseases, Infections and cystic fibrosis unit, Milan, Italy
| |
Collapse
|
13
|
Robledo-Avila FH, Ruiz-Rosado JDD, Partida-Sanchez S, Brockman KL. A Bacterial Epigenetic Switch in Non-typeable Haemophilus influenzae Modifies Host Immune Response During Otitis Media. Front Cell Infect Microbiol 2020; 10:512743. [PMID: 33194779 PMCID: PMC7644868 DOI: 10.3389/fcimb.2020.512743] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 09/15/2020] [Indexed: 01/15/2023] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) causes multiple diseases of the human airway and is a predominant bacterial pathogen of acute otitis media and otitis media in which treatment fails. NTHi utilizes a system of phase variable epigenetic regulation, termed the phasevarion, to facilitate adaptation and survival within multiple sites of the human host. The NTHi phasevarion influences numerous disease-relevant phenotypes such as biofilm formation, antibiotic resistance, and opsonization. We have previously identified an advantageous selection for a specific phasevarion status, which significantly affects severity and chronicity of experimental otitis media. In this study, we utilized pure cultures of NTHi variants in which modA was either locked ON or locked OFF, and thus modA was unable to phase vary. These locked variants were used to assess the progression of experimental otitis media and define the specific immune response induced by each subpopulation. Although the initial disease caused by each subpopulation was similar, the immune response elicited by each subpopulation was unique. The modA2 OFF variant induced significantly greater activation of macrophages both in vitro and within the middle ear during disease. In contrast, the modA2 ON variant induced a greater neutrophil extracellular trap response, which led to greater killing of the modA2 ON variant. These data suggest that not only does the NTHi phasevarion facilitate adaptation, but also allows the bacteria to alter immune responses during disease. Understanding these complex bacterial-host interactions and the regulation of bacterial factors responsible is critical to the development of better diagnostic, treatment, and preventative strategies for these bacterial pathogens.
Collapse
Affiliation(s)
- Frank H Robledo-Avila
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Juan de Dios Ruiz-Rosado
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Biochemistry and Immunology, National Technological Institute of Oaxaca, Oaxaca, Mexico
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Kenneth L Brockman
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
14
|
Li W, Chen W, Huang S, Yao G, Tang X, Sun L. Mesenchymal stem cells prevent overwhelming inflammation and reduce infection severity via recruiting CXCR3 + regulatory T cells. Clin Transl Immunology 2020; 9:e1181. [PMID: 33014369 PMCID: PMC7526004 DOI: 10.1002/cti2.1181] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022] Open
Abstract
Objectives Mesenchymal stem cells (MSCs) have shown great potential in treating autoimmune diseases (ADs). Unlike the traditional immunosuppressants, which inadvertently impair patients' antimicrobial immunity, MSCs reduce the incidence and duration of respiratory infection. However, the underlying mechanisms are unknown. Methods To investigate how MSCs regulate the lung immunity and improve the defence against respiratory infection, we infected MSC‐treated wild‐type and lupus‐prone mice with Haemophilus influenzae intranasally and determined the clearance of bacteria. Tissue damage and inflammatory cytokines were measured by H&E staining and ELISA separately. Immune cell subsets in the tissues were analysed by flow cytometry. Results MSC pretreatment prevented overwhelming inflammation and accelerated bacterial clearance in both wild‐type and lupus‐prone mice. Tregs increased dramatically in the lung after MSC treatment. Adoptive transfer of Tregs isolated from MSC‐treated mice offered similar protection, while deletion of Tregs abrogated the protective effects of MSCs. The majority of the intravenously injected MSCs were engulfed by lung phagocytes, which in turn produced CXCL9 and CXCL10 and recruited tremendous CXCR3+ Tregs into the lung. Compared with their CXCR3− counterparts, CXCR3+ Tregs displayed enhanced proliferation and stronger inhibitory functions. Neutralisation of CXCL9 and CXCL10 significantly downregulated the migration of CXCR3+ Tregs and eliminated the benefits of MSC pretreatment. Conclusion Here, we showed that by recruiting CXCR3+ Tregs, MSC treatment restricted the overactivation of inflammatory responses and prevented severe symptoms caused by infection. By discovering this novel property of MSCs, our study sheds light on optimising long‐term immunosuppressive regimen for autoimmune diseases and other immune disorders.
Collapse
Affiliation(s)
- Wenchao Li
- Department of Rheumatology and Immunology The Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing China
| | - Weiwei Chen
- Department of Rheumatology and Immunology The Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing China
| | - Saisai Huang
- Department of Rheumatology and Immunology The Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing China
| | - Genhong Yao
- Department of Rheumatology and Immunology The Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology The Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing China
| | - Lingyun Sun
- Department of Rheumatology and Immunology The Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing China
| |
Collapse
|
15
|
Smith KA, Hiyoshi A, Burkill S, Bahmanyar S, Öckinger J, Alfredsson L, Olsson T, Montgomery S. Hospital diagnosed pneumonia before age 20 years and multiple sclerosis risk. BMJ Neurol Open 2020; 2:e000044. [PMID: 33681783 PMCID: PMC7903180 DOI: 10.1136/bmjno-2020-000044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/27/2020] [Accepted: 05/10/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction Respiratory inflammation has been proposed as a risk factor for MS. This study aims to determine if hospital-diagnosed pneumonia in adolescence (before age 20 years) is associated with subsequent multiple sclerosis (MS). Methods This case-control study included incident MS cases after age 20 years identified using the Swedish national registers. Cases were matched with 10 general population controls by age, sex and region. Pneumonia diagnoses were identified between 0–5, 6–10, 11–15 and 16–20 years of age. Conditional logistic regression models adjusted for infectious mononucleosis (IM) and education calculated ORs with 95% CIs. Urinary tract infections (UTIs), a common complication of MS, before age 20 years were included as a control diagnosis for reverse causation. Results There were 6109 cases and 49 479 controls included. Pneumonia diagnosed between age 11–15 years was associated with subsequent MS (adj OR 2.00, 95% CI 1.22 to 3.27). Although not statistically significant, sensitivity analyses showed similar magnitude associations of pneumonia between age 11–15 years and MS. No statistically significant associations with MS for pneumonia at other age groups were observed. Adjustment for IM had no notable effect on associations, but was statistically significantly associated with MS. UTIs were not associated with MS. Conclusion Pneumonia at 11–15 years of age was associated with MS, suggesting a possible role for inflammation of the respiratory system in the aetiology of MS during a period of susceptibility in adolescence. Further research on respiratory infections prior to MS onset should be conducted to replicate this finding and determine explanatory causal mechanisms.
Collapse
Affiliation(s)
- Kelsi A Smith
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ayako Hiyoshi
- Clinical Epidemiology and Biostatistics, Örebro Universitet-Campus USÖ, Orebro, Sweden.,Department of Public Health Sciences, Stockholm University, Stockholm, Sweden
| | - Sarah Burkill
- Centre for Pharmacoepidemiology, Karolinska Institutet, Stockholm, Sweden.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Shahram Bahmanyar
- Centre for Pharmacoepidemiology, Karolinska Institutet, Stockholm, Sweden.,Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Johan Öckinger
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Occupational and Environmental Medicine, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Scott Montgomery
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Clinical Epidemiology and Biostatistics, Örebro Universitet-Campus USÖ, Orebro, Sweden
| |
Collapse
|
16
|
Campuzano A, Zhang H, Ostroff GR, Dos Santos Dias L, Wüthrich M, Klein BS, Yu JJ, Lara HH, Lopez-Ribot JL, Hung CY. CARD9-Associated Dectin-1 and Dectin-2 Are Required for Protective Immunity of a Multivalent Vaccine against Coccidioides posadasii Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:3296-3306. [PMID: 32358020 PMCID: PMC7323849 DOI: 10.4049/jimmunol.1900793] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 04/15/2020] [Indexed: 12/11/2022]
Abstract
Coccidioides species are fungal pathogens that can cause a widely varied clinical manifestation from mild pulmonary symptom to disseminated, life-threatening disease. We have previously created a subunit vaccine by encapsulating a recombinant coccidioidal Ag (rCpa1) in glucan-chitin particles (GCPs) as an adjuvant-delivery system. The GCP-rCpa1 vaccine has shown to elicit a mixed Th1 and Th17 response and confers protection against pulmonary coccidioidomycosis in mice. In this study, we further delineated the vaccine-induced protective mechanisms. Depletion of IL-17A in vaccinated C57BL/6 mice prior to challenge abrogated the protective efficacy of GCP-rCpa1 vaccine. Global transcriptome and Ingenuity Pathway Analysis of murine bone marrow-derived macrophages after exposure to this vaccine revealed the upregulation of proinflammatory cytokines (TNF-α, IL-6, and IL-1β) that are associated with activation of C-type lectin receptors (CLR) Dectin-1- and Dectin-2-mediated CARD9 signaling pathway. The GCP formulation of rCpa1 bound soluble Dectin-1 and Dectin-2 and triggered ITAM signaling of corresponding CLR reporter cells. Furthermore, macrophages that were isolated from Dectin-1 -/-, Dectin-2 -/-, and CARD9 -/- mice significantly reduced production of inflammatory cytokines in response to the GCP-rCpa1 vaccine compared with those of wild-type mice. The GCP-rCpa1 vaccine had significantly reduced protective efficacy in Dectin-1 -/-, Dectin-2 -/-, and CARD9 -/- mice that showed decreased acquisition of Th cells in Coccidioides-infected lungs compared with vaccinated wild-type mice, especially Th17 cells. Collectively, we conclude that the GCP-rCpa1 vaccine stimulates a robust Th17 immunity against Coccidioides infection through activation of the CARD9-associated Dectin-1 and Dectin-2 signal pathways.
Collapse
Affiliation(s)
- Althea Campuzano
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Hao Zhang
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Gary R Ostroff
- Program in Molecular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Lucas Dos Santos Dias
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI 53792
| | - Marcel Wüthrich
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI 53792
| | - Bruce S Klein
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI 53792
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Humberto H Lara
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Jose L Lopez-Ribot
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249
| | - Chiung-Yu Hung
- South Texas Center for Emerging Infectious Diseases, Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249;
| |
Collapse
|
17
|
Morris MC, Chapman TJ, Pichichero ME, Broderick G. Immune Network Modeling Predicts Specific Nasopharyngeal and Peripheral Immune Dysregulation in Otitis-Prone Children. Front Immunol 2020; 11:1168. [PMID: 32595639 PMCID: PMC7301607 DOI: 10.3389/fimmu.2020.01168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/12/2020] [Indexed: 11/18/2022] Open
Abstract
Acute otitis media (AOM) pathogenesis involves nasopharyngeal colonization by potential otopathogens and a viral co-infection. Stringently-defined otitis prone (sOP) children show characteristic patterns of immune dysfunction. We hypothesized that otitis proneness is largely a result of altered signaling between immune components that are otherwise competent, resulting in increased susceptibility to infection by bacterial otopathogens. To test this, we constructed a regulatory immune network model linking immune cells and signaling elements known to be involved in AOM and/or dysregulated in sOP children. The alignment of immune response mechanisms with data from in vivo and in vitro experimental observations produced 82 putative immune network models, each describing variants of immune regulatory networks consistent with available observations. Analysis of these models suggested that new measurements of serum levels of IL-4 and CXCL8 could refine competing models and resulted in the elimination of 38 of the models. Further analysis of the remaining 44 models suggested specific deviations in the predicted regulation of nasopharyngeal and peripheral immunity during response to AOM. Specifically, immune responses active in sOP children during AOM were characterized by early and constitutive activation of pro-inflammatory signaling in the nasopharynx and a Th2- and Treg-dominated profile in the periphery. We conclude that sOP children have altered regulation of key immune mediators during both health and pathogenesis. This altered regulation may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Matthew C. Morris
- Center for Clinical Systems Biology, Research Institute, Rochester General Hospital, Rochester, NY, United States
| | - Timothy J. Chapman
- Center for Infectious Diseases and Immunology, Research Institute, Rochester General Hospital, Rochester, NY, United States
| | - Michael E. Pichichero
- Center for Infectious Diseases and Immunology, Research Institute, Rochester General Hospital, Rochester, NY, United States
| | - Gordon Broderick
- Center for Clinical Systems Biology, Research Institute, Rochester General Hospital, Rochester, NY, United States
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| |
Collapse
|
18
|
Hynes GM, Hinks TSC. The role of interleukin-17 in asthma: a protective response? ERJ Open Res 2020; 6:00364-2019. [PMID: 32494573 PMCID: PMC7248344 DOI: 10.1183/23120541.00364-2019] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
While there now exist effective treatments for type 2 high, eosinophilic asthma, there are no specific therapies for 40–50% of people with asthma with other phenotypes, which result from poorly understood underlying pathological mechanisms. One such pathology is neutrophilic inflammation, which has been associated with interleukin (IL)-17 family cytokines. Human genetic studies identified IL-17 polymorphisms associated with asthma; in murine models of allergic airways disease, IL-17A contributes to airway hyperresponsiveness, and in humans, elevated airway IL-17A levels are repeatedly observed in severe asthma. However, the directionality of this association is unknown, and the assumption that IL-17 cytokines drive disease pathology remains speculative. Here, we explore the evidence underlying the relationship between IL-17 and asthma, we review lessons learned from investigating IL-17 in other inflammatory diseases, and discuss the possibility that IL-17 may even be protective in asthma rather than pathogenic. We also critically examine the newly proposed paradigm of a reciprocal relationship between type 2 and type 17 airways inflammation. In summary, we suggest an association between IL-17 and asthma, but research is needed examining the diverse functions of these cytokines, their longitudinal stability, their response to clinical interventions, and for mechanistic studies determining whether they are protective or pathogenic. IL-17 cytokines have been implicated in neutrophilic asthma by genetic, murine and human data. Here, previous studies are critiqued and the assumption their dominant role is pathogenic rather than protective of airway epithelial barrier integrity is challenged.http://bit.ly/3axB4Zs
Collapse
Affiliation(s)
- Gareth M Hynes
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, Nuffield Dept of Medicine, Experimental Medicine, University of Oxford, Oxford, UK
| | - Timothy S C Hinks
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, Nuffield Dept of Medicine, Experimental Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
19
|
Pichichero ME. Immunologic dysfunction contributes to the otitis prone condition. J Infect 2020; 80:614-622. [PMID: 32205139 DOI: 10.1016/j.jinf.2020.03.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 12/11/2022]
Abstract
Acute Otitis Media (AOM) is a multifactorial disease occurring mostly in young children who are immunologically naïve to AOM pathogens. This review focuses on work from Rochester NY, USA over the past 12 years among young children who had AOM infections microbiologically-confirmed by tympanocentesis, so called "stringently-defined". Among stringently-defined otitis prone children deficiencies in fundamental immune defense mechanisms have been identified that contribute to the propensity of young children to experience recurrent AOM. Dysfunction in innate immune responses that cause an immunopathological impact in the nasopharynx have been discovered including inadequate proinflammatory cytokine response and poor epithelial cell repair. Adaptive immunity defects in B cell function and immunologic memory resulting in low levels of antibody to otopathogen-specific antigens allows repeated infections. CD4+ and CD8+ T cell function and memory defects significantly contribute. The immune profile of an otitis prone child resembles that of a neonate through the first year of life. Immunologic deficits in otitis prone children cause them to be unusually vulnerable to viral upper respiratory infections and respond inadequately to routine pediatric vaccines.
Collapse
Affiliation(s)
- Michael E Pichichero
- Center for Infectious Diseases and Immunology, Rochester General Hospital Research Institute, Rochester, NY.
| |
Collapse
|
20
|
Lung Microbiome in Asthma: Current Perspectives. J Clin Med 2019; 8:jcm8111967. [PMID: 31739446 PMCID: PMC6912699 DOI: 10.3390/jcm8111967] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/20/2022] Open
Abstract
A growing body of evidence implicates the human microbiome as a potentially influential player actively engaged in shaping the pathogenetic processes underlying the endotypes and phenotypes of chronic respiratory diseases, particularly of the airways. In this article, we specifically review current evidence on the characteristics of lung microbiome, and specifically the bacteriome, the modes of interaction between lung microbiota and host immune system, the role of the “lung–gut axis”, and the functional effects thereof on asthma pathogenesis. We also attempt to explore the possibilities of therapeutic manipulation of the microbiome, aiming at the establishment of asthma prevention strategies and the optimization of asthma treatment.
Collapse
|
21
|
Singh AK, Curtiss R, Sun W. A Recombinant Attenuated Yersinia pseudotuberculosis Vaccine Delivering a Y. pestis YopE Nt138-LcrV Fusion Elicits Broad Protection against Plague and Yersiniosis in Mice. Infect Immun 2019; 87:e00296-19. [PMID: 31331960 PMCID: PMC6759313 DOI: 10.1128/iai.00296-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/18/2019] [Indexed: 12/22/2022] Open
Abstract
In this study, a novel recombinant attenuated Yersinia pseudotuberculosis PB1+ strain (χ10069) engineered with ΔyopK ΔyopJ Δasd triple mutations was used to deliver a Y. pestis fusion protein, YopE amino acid 1 to 138-LcrV (YopENt138-LcrV), to Swiss Webster mice as a protective antigen against infections by yersiniae. χ10069 bacteria harboring the pYA5199 plasmid constitutively synthesized the YopENt138-LcrV fusion protein and secreted it via the type 3 secretion system (T3SS) at 37°C under calcium-deprived conditions. The attenuated strain χ10069(pYA5199) was manifested by the establishment of controlled infection in different tissues without developing conspicuous signs of disease in histopathological analysis of microtome sections. A single-dose oral immunization of χ10069(pYA5199) induced strong serum antibody titers (log10 mean value, 4.2), secretory IgA in bronchoalveolar lavage (BAL) fluid from immunized mice, and Yersinia-specific CD4+ and CD8+ T cells producing high levels of tumor necrosis factor alpha (TNF-α), gamma interferon (IFN-γ), and interleukin 2 (IL-2), as well as IL-17, in both lungs and spleens of immunized mice, conferring comprehensive Th1- and Th2-mediated immune responses and protection against bubonic and pneumonic plague challenges, with 80% and 90% survival, respectively. Mice immunized with χ10069(pYA5199) also exhibited complete protection against lethal oral infections by Yersinia enterocolitica WA and Y. pseudotuberculosis PB1+. These findings indicated that χ10069(pYA5199) as an oral vaccine induces protective immunity to prevent bubonic and pneumonic plague, as well as yersiniosis, in mice and would be a promising oral vaccine candidate for protection against plague and yersiniosis for human and veterinary applications.
Collapse
Affiliation(s)
- Amit K Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Wei Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| |
Collapse
|
22
|
Infection with Opportunistic Bacteria Triggers Severe Pulmonary Inflammation in Lupus-Prone Mice. Mediators Inflamm 2019; 2019:1701367. [PMID: 31565032 PMCID: PMC6745158 DOI: 10.1155/2019/1701367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/03/2019] [Accepted: 07/22/2019] [Indexed: 12/05/2022] Open
Abstract
Infection is a common cause of hospitalization and mortality in patients with systemic lupus erythematosus (SLE). How the underlying immune dysfunctions affect the antimicrobial immunity remains largely unknown. In the present study, employing the pulmonary infection model, we determined the antimicrobial defence of lupus-prone mice. After infecting with opportunistic bacterium Haemophilus influenzae (Hi), lupus-prone mice (B6/lpr) exhibited inefficient bacterial elimination and recovered slowly. They generated severer inflammation at the early stage of infection, as excessive accumulation of neutrophils and enhanced production of proinflammatory cytokines were observed in the lung. In addition, a large number of apoptotic cells were detected in the lungs of B6/lpr mice. For adaptive immune responses, B6/lpr mice were capable to generate enough protective Hi-specific Th17 cells. They evoked stronger Hi-specific γδ T17 response in both lungs and spleens. Unexpectedly, both CD4 and γδ T cells from lupus-prone mice showed deficiency in IFN-γ production. For humoral immune responses, compared with those of WT mice, the concentrations of Hi-specific IgA, IgM, and IgG, especially IgG, were significantly higher in the B6/lpr mice. Our findings suggest that lupus mice are capable to generate antibacterial immune responses; however, the overwhelming inflammation and overactivated immune responses increase the severity of infection.
Collapse
|
23
|
Su YC, Jalalvand F, Thegerström J, Riesbeck K. The Interplay Between Immune Response and Bacterial Infection in COPD: Focus Upon Non-typeable Haemophilus influenzae. Front Immunol 2018; 9:2530. [PMID: 30455693 PMCID: PMC6230626 DOI: 10.3389/fimmu.2018.02530] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating respiratory disease and one of the leading causes of morbidity and mortality worldwide. It is characterized by persistent respiratory symptoms and airflow limitation due to abnormalities in the lower airway following consistent exposure to noxious particles or gases. Acute exacerbations of COPD (AECOPD) are characterized by increased cough, purulent sputum production, and dyspnea. The AECOPD is mostly associated with infection caused by common cold viruses or bacteria, or co-infections. Chronic and persistent infection by non-typeable Haemophilus influenzae (NTHi), a Gram-negative coccobacillus, contributes to almost half of the infective exacerbations caused by bacteria. This is supported by reports that NTHi is commonly isolated in the sputum from COPD patients during exacerbations. Persistent colonization of NTHi in the lower airway requires a plethora of phenotypic adaptation and virulent mechanisms that are developed over time to cope with changing environmental pressures in the airway such as host immuno-inflammatory response. Chronic inhalation of noxious irritants in COPD causes a changed balance in the lung microbiome, abnormal inflammatory response, and an impaired airway immune system. These conditions significantly provide an opportunistic platform for NTHi colonization and infection resulting in a "vicious circle." Episodes of large inflammation as the consequences of multiple interactions between airway immune cells and NTHi, accumulatively contribute to COPD exacerbations and may result in worsening of the clinical status. In this review, we discuss in detail the interplay and crosstalk between airway immune residents and NTHi, and their effect in AECOPD for better understanding of NTHi pathogenesis in COPD patients.
Collapse
Affiliation(s)
- Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Farshid Jalalvand
- Department of Biology, Centre for Bacterial Stress Response and Persistence, University of Copenhagen, Copenhagen, Denmark
| | - John Thegerström
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
24
|
Iwanaga N, Kolls JK. Updates on T helper type 17 immunity in respiratory disease. Immunology 2018; 156:3-8. [PMID: 30260473 DOI: 10.1111/imm.13006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022] Open
Abstract
Interleukin-17 (IL-17)-producing cells play a critical role in mucosal immunity including the respiratory tract. This review will highlight recent advances in our understanding of these cells in mucosal immunity in the lung as well as their potential pathogenic roles in respiratory diseases. The IL-17-producing cells include γδ T cells, natural killer cells, group 3 innate lymphoid cells, and T helper type 17 (Th17) cells. There have been recent advances in our understanding of these cell populations in the lung as well as emerging data on how these cells are regulated in the lung. Moreover, Th17 cells may be a key component of tissue-resident memory cells that may be acquired over time or elicited by mucosal immunization that provides the host with enhanced immunity against certain pathogens.
Collapse
Affiliation(s)
- Naoki Iwanaga
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|