1
|
Long H, He G, He J, Du TF, Feng P, Zhu C. The protective effect and immunomodulatory ability of orally administrated Lacticaseibacillus rhamnosus GG against Mycoplasma pneumoniae infection in BALB/c mice. PLoS One 2024; 19:e0312318. [PMID: 39453930 PMCID: PMC11508164 DOI: 10.1371/journal.pone.0312318] [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: 03/23/2024] [Accepted: 08/27/2024] [Indexed: 10/27/2024] Open
Abstract
Mycoplasma pneumoniae represents one of the significant etiologies of community-acquired pneumonia in pediatric patients. However, clinical treatment of M. pneumoniae infection in children has encountered challenges due to the escalating resistance to quinolones. Numerous studies have highlighted the potential of probiotic lactobacillus administration in boosting immune responses to bacterial and viral respiratory infections. In this study, the protective efficacy of pre-oral administration of Lacticaseibacillus rhamnosus GG (LGG), Limosilactobacillus reuteri F275, Lactiplantibacillus plantarum NCIMB 8826, L. plantarum S1 or L. plantarum S2 was evaluated in the BALB/c mice model; it was observed that among these five strains of lactobacillus, the supplementation of LGG exhibited the most significant protective effect against M. pneumoniae infection. Moreover, when administered orally, both live LGG and heat-inactivated LGG have demonstrated efficacy in reducing the burden of M. pneumoniae in the lungs and alleviating pulmonary inflammation. Oral supplementation with LGG resulted in the inhibition of neutrophil recruitment into the lungs and increased recruitment of alveolar macrophages in M. pneumoniae-infected mice. Additionally, LGG supplementation led to increased production of IL-10 and secretory IgA (sIgA), while suppressing the levels of IL-1β, IL-6, IL-17A, and TNF-α in the lungs of mice infected with M. pneumoniae. The data suggests that supplementation with LGG can modulate immune responses, decrease pathogen load, and alleviate inflammatory injury in the lungs of M. pneumoniae-infected mice.
Collapse
Affiliation(s)
- Huanbing Long
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Guiting He
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Jiarong He
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Ting feng Du
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Pengxiao Feng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Cuiming Zhu
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| |
Collapse
|
2
|
Ou X, Fang Z, Li W, Xia Y, Xia J, Zhang J. Novel role for integrin β4 in asthmatic children infected with Mycoplasma pneumoniae. J Asthma 2022; 60:1394-1401. [PMID: 36409462 DOI: 10.1080/02770903.2022.2149932] [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: 11/23/2022]
Abstract
Objectives: The regulatory role of integrin β4 (ITGB4) in asthmatic children infected with Mycoplasma pneumoniae (MP) was analyzed, and its potential molecular mechanisms and function were studied.Methods: We collected epidemiological data from 70,455 asthmatic children. The immunofluorescence assay was used to test 9 index IgMs against respiratory tract pathogens for 736 serum samples. Then, 98 children with severe asthma were treated via fiberoptic bronchoscope examination. During the surgery, the patients' lavage fluid was collected. Additionally, differences in transforming growth factor-beta (TGF-β) expression between the MP-infected and noninfected groups were examined. Experiments were performed using white blood cell counting methods and flow cytometry for 98 asthmatic children.Results: We tested 736 specimens, and the percentages of MP, RSV, and ADV infections were 50.27%, 11.68%, and 10.05%, respectively. The percentage of eosinophils was increased significantly in the AS-I-MP group, and their TGF-β expression levels were increased, which was related to tissue fibrosis. Furthermore, MP infection exacerbated the decreasing trend of ITGB4 expression in patients' blood compared with the noninfected group.Conclusions: There might be a chain reaction from MP infection to an increase in ITGB4, a decrease in TGF-β, a large accumulation of eosinophils and the development of asthma in children.
Collapse
Affiliation(s)
- Xia Ou
- School of Medicine, Kunming University of Science and Technology, Kunming, China
| | - Zhongyue Fang
- School of Medicine, Kunming University of Science and Technology, Kunming, China
| | - Weijie Li
- Department of Hospital Infection Management, Kunming Children Hospital, Kunming, China
| | - Yunxin Xia
- School of Medicine, Kunming University of Science and Technology, Kunming, China
| | - Jianming Xia
- The Department of Cardiovascular Medicine, FuWai Yunan Cardiovascular Hospital, Kunming, China
| | - Jihong Zhang
- School of Medicine, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
3
|
Vaccination with Mycoplasma pneumoniae membrane lipoproteins induces IL-17A driven neutrophilia that mediates Vaccine-Enhanced Disease. NPJ Vaccines 2022; 7:86. [PMID: 35906257 PMCID: PMC9336141 DOI: 10.1038/s41541-022-00513-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
Bacterial lipoproteins are an often-underappreciated class of microbe-associated molecular patterns with potent immunomodulatory activity. We previously reported that vaccination of BALB/c mice with Mycoplasma pneumoniae (Mp) lipid-associated membrane proteins (LAMPs) resulted in lipoprotein-dependent vaccine enhanced disease after challenge with virulent Mp, though the immune responses underpinning this phenomenon remain poorly understood. Herein, we report that lipoprotein-induced VED in a mouse model is associated with elevated inflammatory cytokines TNF-α, IL-1β, IL-6, IL-17A, and KC in lung lavage fluid and with suppurative pneumonia marked by exuberant neutrophilia in the pulmonary parenchyma. Whole-lung-digest flow cytometry and RNAScope analysis identified multiple cellular sources for IL-17A, and the numbers of IL-17A producing cells were increased in LAMPs-vaccinated/Mp-challenged animals compared to controls. Specific IL-17A or neutrophil depletion reduced disease severity in our VED model—indicating that Mp lipoproteins induce VED in an IL-17A-dependent manner and through exuberant neutrophil recruitment. IL-17A neutralization reduced levels of TNF-α, IL-1β, IL-6, and KC, indicating that IL-17A preceded other inflammatory cytokines. Surprisingly, we found that IL-17A neutralization impaired bacterial clearance, while neutrophil depletion improved it—indicating that, while IL-17A appears to confer both maladaptive and protective responses, neutrophils play an entirely maladaptive role in VED. Given that lipoproteins are found in virtually all bacteria, the potential for lipoprotein-mediated maladaptive inflammatory responses should be taken into consideration when developing vaccines against bacterial pathogens.
Collapse
|
4
|
Chen Y, Dong S, Tian L, Chen H, Chen J, He C. Combination of azithromycin and methylprednisolone alleviates Mycoplasma pneumoniae induced pneumonia by regulating miR‑499a‑5p/STAT3 axis. Exp Ther Med 2022; 24:578. [PMID: 35949317 PMCID: PMC9353499 DOI: 10.3892/etm.2022.11515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 06/10/2022] [Indexed: 11/06/2022] Open
Abstract
Mycoplasma pneumoniae (M. pneumoniae) is a contributing factor to community-acquired pneumonia in children. The present study sought to explain the underlying mechanism of azithromycin (AZM) combined with methylprednisolone (MP) in the treatment of M. pneumoniae infection. Peripheral blood samples were obtained from patients with M. pneumoniae and healthy volunteers for analysis. A549 cells were infected with M. pneumoniae to construct an in vitro cell model with M. pneumoniae, followed by treatment with AZM and MP. Cell Counting Kit-8 and TUNEL assays were conducted to detect cell viability and apoptosis. RT-qPCR was employed to measure the expression levels of microRNA (miR)-499a-5p and STAT3. Western blotting was performed to measure the expression of STAT3 and apoptosis-related proteins. Luciferase report assay was performed to verify the binding site between miR-499a-5p and STAT3. The production of inflammatory cytokines was determined using ELISA kits. The results exhibited the downregulated miR-499a-5p and dysregulated inflammatory cytokines in peripheral blood of patients and M. pneumoniae-infected A549 cells. AZM and MP treatment alone or combined significantly inhibited inflammatory response, cell viability loss and promoted apoptosis in A549 cells infected with M. pneumoniae, which was partly reversed by inhibition of miR-499a-5p. Furthermore, miR-499a-5p could negatively regulate its direct target STAT3. In addition, STAT3 is also regulated by AZM and MP. Collectively, the present results suggested that combination treatment of AZM and MP could inhibit M. pneumoniae infection-induced inflammation, cell viability loss and promoted apoptosis partly by regulating miR-499a-5p/STAT3 axis.
Collapse
Affiliation(s)
- Yongli Chen
- Department of Paediatrics, Wuhan Fourth Hospital, Hankou, Wuhan, Hubei 430034, P.R. China
| | - Shanwu Dong
- Department of Paediatrics, Wuhan Fourth Hospital, Hankou, Wuhan, Hubei 430034, P.R. China
| | - Lin Tian
- Department of Paediatrics, Wuhan Fourth Hospital, Hankou, Wuhan, Hubei 430034, P.R. China
| | - Haishan Chen
- Department of Paediatrics, Wuhan Fourth Hospital, Hankou, Wuhan, Hubei 430034, P.R. China
| | - Jing Chen
- Department of Paediatrics, Wuhan Fourth Hospital, Hankou, Wuhan, Hubei 430034, P.R. China
| | - Chunzhi He
- Department of Paediatrics, Wuhan Fourth Hospital, Hankou, Wuhan, Hubei 430034, P.R. China
| |
Collapse
|
5
|
Absence of Association between Previous Mycoplasma pneumoniae Infection and Subsequent Myasthenia Gravis: A Nationwide Population-Based Matched Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147677. [PMID: 34300128 PMCID: PMC8306290 DOI: 10.3390/ijerph18147677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 02/05/2023]
Abstract
Mycoplasma pneumoniae (M. pneumoniae) is not only one of the most common pathogenic bacteria for respiratory infection but also a trigger for many autoimmune diseases. Its infection process shared many similarities with the pathogenesis of myasthenia gravis (MG) at cellular and cytokine levels. Recent case reports demonstrated patients present with MG after M. pneumoniae infection. However, no epidemiological studies ever looked into the association between the two. Our study aimed to investigate the relationship between M. pneumoniae infection and subsequent development of MG. In this population-based retrospective cohort study, the risk of MG was analyzed in patients who were newly diagnosed with M. pneumoniae infection between 2000 and 2013. A total of 2428 M. pneumoniae patients were included and matched with the non-M. pneumoniae control cohort at a 1:4 ratio by age, sex, and index date. Cox proportional hazards regression analysis was applied to analyze the risk of MG development after adjusting for sex, age, and comorbidities, with hazard ratios and 95% confidence intervals. The incidence rates of MG in the non-M. pneumoniae and M. pneumoniae cohorts were 0.96 and 1.97 per 10,000 person-years, respectively. Another case-control study of patients with MG (n = 515) was conducted to analyze the impact of M. pneumoniae on MG occurrence as a sensitivity analysis. The analysis yielded consistent absence of a link between M. pneumoniae and MG. Although previous studies have reported that M. pneumoniae infection and MG may share associated immunologic pathways, we found no statistical significance between M. pneumoniae infection and subsequent development of MG in this study.
Collapse
|
6
|
Luo Y, Li C, Zhou Z, Gong Z, Zhu C, Lei A. Biological functions of IL-17-producing cells in mycoplasma respiratory infection. Immunology 2021; 164:223-230. [PMID: 33930194 DOI: 10.1111/imm.13346] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/07/2021] [Accepted: 04/18/2021] [Indexed: 12/17/2022] Open
Abstract
Mycoplasmas are the smallest and simplest bacteria that lack a cell wall but have the capability of self-replication. Among them, Mycoplasma pneumoniae is one of the most common causes of community-acquired pneumonia. The hallmark of mycoplasma respiratory diseases is the persistence of lung inflammation that involves both innate and adaptive immune responses. In recent years, a growing body of evidence demonstrates that IL-17 plays an important role in respiratory mycoplasma infection, and associates with the pathologic outcomes of infection, such as pneumonitis and asthma. Numerous studies have shown that a variety of cells, in particular Th17 cells, in the lung can secrete IL-17 during respiratory mycoplasma infection. In this article, we review the biological functions of distinct IL-17-producing cells in mycoplasma respiratory infection with a focus on the effect of IL-17 on the outcomes of infection.
Collapse
Affiliation(s)
- Ying Luo
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China.,Changsha Central Hospital, University of South China, Changsha, China
| | - Cheng Li
- Changsha Central Hospital, University of South China, Changsha, China
| | - Zhou Zhou
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Zhande Gong
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Cuiming Zhu
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Aihua Lei
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| |
Collapse
|
7
|
Tamiya S, Yoshikawa E, Suzuki K, Yoshioka Y. Susceptibility Analysis in Several Mouse Strains Reveals Robust T-Cell Responses After Mycoplasma pneumoniae Infection in DBA/2 Mice. Front Cell Infect Microbiol 2021; 10:602453. [PMID: 33520736 PMCID: PMC7839406 DOI: 10.3389/fcimb.2020.602453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/27/2020] [Indexed: 02/03/2023] Open
Abstract
Mycoplasma pneumoniae (Mp) is a highly contagious respiratory pathogen responsible for human community-acquired pneumonia. The number of antibiotic-resistant Mp strains is increasing; therefore, to develop novel therapeutics, it is crucial to precisely understand the pathogenesis of mycoplasma pneumonia. Herein, we examined the susceptibility and response to Mp among eight inbred mouse strains. Following infection, the bacterial load in the bronchoalveolar lavage fluid (BALF) from DBA/2 mice was higher than that in the other tested strains such as BALB/c mice, which are frequently used in Mp research. In contrast, the numbers of CD45+ immune cells and neutrophils in BALF were comparable between BALB/c and DBA/2 mice, with lower numbers observed in C57BL/6J and CBA/N mice than in BALB/c mice. Among the tested strains, the BALF level of interleukin 12 subunit p40 was highest in DBA/2 mice; however, significant differences in other cytokines levels were not observed between BALB/c and DBA/2 mice. After Mp infection, Mp-specific Th1 and Th17 responses were significantly enhanced in DBA/2 mice when compared with BALB/c mice. Furthermore, prior infection with Mp increased the number of neutrophils in BALF after the reinfection of DBA/2 mice through an Mp-specific CD4+ T cell-dependent mechanism. Thus, DBA/2 may be an appropriate strain for evaluating Mp infection. Moreover, a comparison of responses revealed by various inbred mouse strains could be useful for elucidating the pathogenesis of Mycoplasma pneumonia.
Collapse
Affiliation(s)
- Shigeyuki Tamiya
- Laboratory of Nano-design for innovative drug development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.,Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Eisuke Yoshikawa
- Laboratory of Nano-design for innovative drug development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.,Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Koichiro Suzuki
- The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for innovative drug development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.,Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
8
|
Lee CH, Choi Y, Seo SY, Kim SH, Kim IH, Kim SW, Lee ST, Lee SO. Addition of probiotics to antibiotics improves the clinical course of pneumonia in young people without comorbidities: a randomized controlled trial. Sci Rep 2021; 11:926. [PMID: 33441702 PMCID: PMC7806890 DOI: 10.1038/s41598-020-79630-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022] Open
Abstract
This study was aimed at investigating the clinical efficacy of probiotics in pneumonia patients. To this end, we enrolled 80 participants diagnosed with pneumonia at Naval Pohang Hospital, Pohang, Korea, from May 2016 to January 2017. The participants were randomly assigned to the control and probiotic groups depending on whether they received probiotics. All participants clinically improved but 22.6% of the participants complained of abnormal stool habits after pneumonia treatment. In comparison, fever duration was significantly shorter in the probiotic group, and the group exhibited an improved general condition. The probiotic group also showed better stool characteristics according to the Bristol stool scale (P = 0.009). Notably, the serum hs-CRP levels were significantly lower in the probiotic group at 2 weeks of treatment (P = 0.015), and all participants in the probiotic group achieved their levels within the normal range. Flow cytometry was used to analyze T-helper 17 (Th17) cells and regulatory T cells (Tregs). Tregs were promoted and the Th17 cell/Treg ratio was suppressed after 2 weeks of treatment in the probiotic group (P = 0.007 and 0.037, respectively). This study demonstrated that probiotics improved clinical symptoms and normalized inflammatory biomarker levels in patients with pneumonia. Early infection and inflammation recovery may be due to the immunomodulatory effects of probiotics by facilitating the subset of Tregs and suppressing the Th17 cell/Treg ratio.
Collapse
Affiliation(s)
- Chang Hun Lee
- Department of Internal Medicine, Naval Pohang Hospital, Pohang, South Korea.,Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Yunjung Choi
- Division of Rheumatology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Seung Young Seo
- Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Seong-Hun Kim
- Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - In Hee Kim
- Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Sang Wook Kim
- Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Soo Teik Lee
- Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Seung Ok Lee
- Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea.
| |
Collapse
|
9
|
Xu X, Sheng Y, Yang L, Zhou H, Tang L, Du L. Immunological Features of Pediatric Interstitial Pneumonia Due to Mycoplasma pneumoniae. Front Pediatr 2021; 9:651487. [PMID: 33959573 PMCID: PMC8093394 DOI: 10.3389/fped.2021.651487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/24/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Inflammatory response, oxidative stress, and immunologic mechanism are involved in the pathogenesis of Mycoplasma pneumoniae pneumonia (MPP). However, the role of immune system of pediatric interstitial pneumonia due to M. pneumoniae infections remains poorly understood. The aim of this study was to analyze the immunologic features of pediatric interstitial pneumonia due to Mycoplasma pneumoniae (M. pneumoniae). Methods: A retrospective study was conducted on a primary cohort of children with MPP. Propensity score analysis was performed to match interstitial pneumonia and pulmonary consolidation children. Results: The clinical characteristics strongly associated with the development of interstitial pneumonia were boys, age >5 years, wheezing history, hydrothorax free, lymphocytes (>3.0 × 109/L), CD19+ (>0.9 × 109/L), CD3+ (>2.5 × 109/L), CD4+ (>1.5 × 109/L), CD8+ (>0.9 × 109/L), interleukin-6 (IL-6, <30 pg/ml), IL-10 (<6 pg/ml), and interferon-γ (IFN-γ, <15 pg/ml). After propensity score analysis, children with interstitial pneumonia showed significantly higher CD19+, CD3+, and CD4+ T cell counts, and lower serum IL-6, IL-10, and IFN-γ levels. The final regression model showed that only CD4+ T cells (>1.5 × 109/L, OR = 2.473), IFN-γ (<15 pg/ml, OR = 2.250), and hydrothorax free (OR = 14.454) were correlated with the development of interstitial pneumonia among children with MPP. Conclusions: The M. pneumoniae-induced interstitial pneumonia showed increased CD4+ T cells and lower serum IFN-γ level. Specific immunologic profiles could be involved in the development of pediatric interstitial pneumonia due to M. pneumoniae infections.
Collapse
Affiliation(s)
- Xuefeng Xu
- Department of Rheumatology Immunology & Allergy, National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pulmonary Medicine, National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanjian Sheng
- Department of Pulmonary Medicine, National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Yang
- Department of Radiology, National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haichun Zhou
- Department of Radiology, National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanfang Tang
- Department of Pulmonary Medicine, National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lizhong Du
- Department of Neonatology, National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Tamiya S, Yoshikawa E, Ogura M, Kuroda E, Suzuki K, Yoshioka Y. Vaccination using inactivated Mycoplasma pneumoniae induces detrimental infiltration of neutrophils after subsequent infection in mice. Vaccine 2020; 38:4979-4987. [PMID: 32536549 DOI: 10.1016/j.vaccine.2020.05.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/18/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022]
Abstract
Mycoplasma pneumoniae (Mp) is one of the most common causes of community-acquired pneumonia. Given the emergence and high rates of antibiotic-resistant Mp strains, vaccines that prevent the pneumonia and secondary complications due to Mp infection are urgently needed. Although several studies have shown the protective efficacy of Mp vaccines in human clinical trials, some reports suggest that vaccination against Mp exacerbates disease upon subsequent Mp challenge. Therefore, to develop optimal vaccines against Mp, understanding the immune responses that contribute to post-vaccination exacerbation of inflammation is crucial. Here we examined whether Mp vaccination might exacerbate pneumonia after subsequent Mp infection in mice. We found that vaccination with inactivated Mp plus aluminum salts as an adjuvant induced Mp-specific IgG, Th1 cells, and Th17 cells. Toll-like receptor 2 signaling contributed to the induction of an Mp-specific IgG response and was necessary for Mp-specific Th17-cell-but not Th1-cell-responses in vaccinated mice. In addition, vaccination with inactivated Mp plus aluminum salts suppressed the number of Mp organisms in the bronchoalveolar lavage fluid, indicating that vaccination can reduce Mp infection. However, the numbers of total immune cells and neutrophils in bronchoalveolar lavage fluid after Mp challenge did not differ between vaccinated mice and non-vaccinated control mice. Furthermore, depletion of CD4+ T cells prior to Mp challenge decreased pulmonary neutrophil infiltration in vaccinated mice, suggesting that Th1 or Th17 cells (or both) are responsible for the vaccination-induced neutrophil infiltration. These results suggest that, despite reducing Mp infection, vaccination of mice by using inactivated Mp fails to suppress inflammation, such as neutrophil infiltration into the lung, after subsequent Mp infection.
Collapse
Affiliation(s)
- Shigeyuki Tamiya
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Eisuke Yoshikawa
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Monami Ogura
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Etsushi Kuroda
- Department of Immunology and Medical Zoology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan; Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, NIBIOHN, Ibaraki, Osaka, Japan
| | - Koichiro Suzuki
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan; BIKEN Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
11
|
Interleukin-17 mediates lung injury by promoting neutrophil accumulation during the development of contagious caprine pleuropneumonia. Vet Microbiol 2020; 243:108651. [PMID: 32273025 DOI: 10.1016/j.vetmic.2020.108651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/05/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
Contagious caprine pleuropneumonia (CCPP) is a highly contagious infectious disease of goats caused by Mycoplasma capricolum subspecies capripneumoniae (Mccp). CCPP outbreaks usually result in high morbidity and mortality of the affected goats, making this disease a major cause of economic losses to goat producers globally. However, the pathogenesis of CCPP remains unclear. Here, we show that IL-17-driven neutrophil accumulation is involved in the lung damage in CCPP goats. During CCPP development, intense inflammatory infiltrates could be observed in the injured lungs. Specifically, neutrophils were observed to be present within the alveoli. Increased IL-17 release drove the excessive influx of neutrophils into the lung, as IL-17 effectively stimulated the production of neutrophil chemoattractants from lung epithelial cells following Mccp infection. Our data highlight a critical role of IL-17-driven neutrophil accumulation in the pathogenesis of CCPP and suggest that IL-17 may potentially be a useful immunotherapeutic target for the treatment of CCPP.
Collapse
|
12
|
Interleukin-17A Exacerbates Disease Severity in BALB/c Mice Susceptible to Lung Infection with Mycoplasma pulmonis. Infect Immun 2018; 86:IAI.00292-18. [PMID: 29986888 DOI: 10.1128/iai.00292-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/29/2018] [Indexed: 12/18/2022] Open
Abstract
Mycoplasmas are atypical bacteria that disrupt the immune response to promote respiratory tract infections and secondary complications. However, not every immunologic response that protects or damages the host during mycoplasma infection is known. Interleukin-17A (IL-17A) is elevated in individuals infected with mycoplasmas, but how IL-17A and its cellular sources dictate disease outcome remains unclear. Here, IL-17A is hypothesized to worsen disease in individuals susceptible to mycoplasma infection. Thus, monoclonal anti-IL-17A antibodies were given to disease-susceptible BALB/c mice and disease-resistant C57BL/6 mice infected with Mycoplasma pulmonis Neutralizing the function of IL-17A using anti-IL-17A antibodies reduced disease severity during M. pulmonis infection in BALB/c, but not C57BL/6, mice. Neutralizing IL-17A also reduced the incidence of neutrophilic lung lesions during infection in BALB/c mice. Reduced pathology occurred without impacting the bacterial burden, demonstrating that IL-17A is not required for mycoplasma clearance. The main source of IL-17A throughout infection in BALB/c mice was CD4+ T cells, and neutralizing IL-17A after infiltration of the lungs by T cells reduced disease severity, identifying the Th17 response as a herald of late mycoplasma pathology in susceptible mice. Neutralizing IL-17A did not further reduce disease during M. pulmonis infection in BALB/c mice depleted of neutrophils, suggesting that IL-17A requires the presence of pulmonary neutrophils to worsen respiratory pathology. IL-17A is a pathological element of murine respiratory mycoplasma infection. Using monoclonal antibodies to neutralize IL-17A could reduce disease severity during mycoplasma infection in humans and domesticated animals.
Collapse
|
13
|
Chackelevicius CM, Gambaro SE, Tiribelli C, Rosso N. Th17 involvement in nonalcoholic fatty liver disease progression to non-alcoholic steatohepatitis. World J Gastroenterol 2016; 22:9096-9103. [PMID: 27895397 PMCID: PMC5107591 DOI: 10.3748/wjg.v22.i41.9096] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/22/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
The nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome. NAFLD encompasses a wide histological spectrum ranging from benign simple steatosis to non-alcoholic steatohepatitis (NASH). Sustained inflammation in the liver is critical in this process. Hepatic macrophages, including liver resident macropaghes (Kupffer cells), monocytes infiltrating the injured liver, as well as specific lymphocytes subsets play a pivotal role in the initiation and perpetuation of the inflammatory response, with a major deleterious impact on the progression of fatty liver to fibrosis. During the last years, Th17 cells have been involved in the development of inflammation not only in liver but also in other organs, such as adipose tissue or lung. Differentiation of a naïve T cell into a Th17 cell leads to pro-inflammatory cytokine and chemokine production with subsequent myeloid cell recruitment to the inflamed tissue. Th17 response can be mitigated by T regulatory cells that secrete anti-inflammatory cytokines. Both T cell subsets need TGF-β for their differentiation and a characteristic plasticity in their phenotype may render them new therapeutic targets. In this review, we discuss the role of the Th17 pathway in NAFLD progression to NASH and to liver fibrosis analyzing different animal models of liver injury and human studies.
Collapse
|