1
|
Li D, Zheng H, Wang X, Li F, Wang H, Chen H, Shen C, Zhao S. Investigation of T lymphocyte subsets in children with Mycoplasma pneumoniae pneumonia. Immunol Res 2024; 73:24. [PMID: 39714538 DOI: 10.1007/s12026-024-09576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/19/2024] [Indexed: 12/24/2024]
Abstract
This study aims to characterize the majority of immune cell subsets in peripheral blood mononuclear cells in children with Mycoplasma pneumoniae pneumonia (MPP) by a 21-color flow cytometry panel. Patients who met the predetermined eligibility criteria for pneumonia diagnosis were recruited for the research study. Multi-color flow cytometry was conducted on the peripheral blood mononuclear cells of each patient group, which were then subjected to dimensionality reduction and cluster analysis. In our study, the proportion of activated CD4 + T cell and naïve CD8 + T in children with MPP was higher than that of children with non-MPP, and the proportion of CD8 + T cell and central memory CD8 + T cell in MPP children was lower. Central memory CD4 + T cell and activated CD4 + T cell in the severe MPP were higher than those in the mild MPP. The highest proportions of CD8 + T cell, CD8 + Tn cell, activated CD8 + T cell, and total activated T cell were observed in the pulmonary consolidation-mucous group when compared to the pulmonary consolidation-necrosis and bronchiolitis groups. In the pulmonary consolidation-necrosis group, the proportions of central memory CD4 + T cell and T helper 17 cell were higher than those in pulmonary consolidation-mucous and bronchiolitis groups. In the bronchiolitis group, the percentages of CD4 + T cell, naïve CD4 + T cell, and T helper 2 cell were higher than those in pulmonary consolidation-mucous and the pulmonary consolidation-necrosis groups. The T lymphocyte subsets were different among various groups, offering new insights into the immune system of pediatric patients with Mycoplasma pneumoniae pneumonia.
Collapse
Affiliation(s)
- Deze Li
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Huiwen Zheng
- Laboratory of Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Xiaotong Wang
- Laboratory of Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Feina Li
- Laboratory of Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Heng Wang
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Hao Chen
- Laboratory of Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Chen Shen
- Laboratory of Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
| | - Shunying Zhao
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Zuo Y, Zhang R, Li S. Reviewing advancement in Mycoplasma pneumoniae P30 adhesin protein provides insights for future diagnosis and treatment. Front Microbiol 2024; 15:1515291. [PMID: 39735188 PMCID: PMC11671514 DOI: 10.3389/fmicb.2024.1515291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Mycoplasma pneumoniae is a major pathogen that causes upper and lower respiratory tract infections in children, adolescents, and elderly individuals and can lead to pneumonia, intrapulmonary and extrapulmonary complications, and respiratory sequelae. M. pneumoniae must adhere to respiratory epithelial cells of a host for infection. The P1 and P30 proteins, as two adhesin proteins of M. pneumoniae, have attracted extensive attention from many researchers. In this paper, we present the latest research progress on the P30 protein in terms of structure and mutation typing, physiological function, clinical serological diagnosis and vaccine development in a literature review. This study deepens our knowledge on the pathogenesis of M. pneumoniae and is useful for diagnosing and preventing M. pneumoniae infection.
Collapse
Affiliation(s)
- Yingying Zuo
- Hengyang Medical School, University of South China, Hengyang, China
| | - Ru Zhang
- The Seventh Affiliated Hospital of University of South China, Changsha, China
| | - Shuihong Li
- Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
3
|
Zhang H, Sheng S, Li C, Bao X, Zhao L, Chen J, Guan P, Li X, Pan N, Liang Y, Wang X, Sun J, Wang X. Mucosal immunization with the lung Lactobacillus-derived amphiphilic exopolysaccharide adjuvanted recombinant vaccine improved protection against P. aeruginosa infection. PLoS Pathog 2024; 20:e1012696. [PMID: 39556597 DOI: 10.1371/journal.ppat.1012696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/25/2024] [Indexed: 11/20/2024] Open
Abstract
Respiratory infections caused by Pseudomonas aeruginosa are a major health problem globally. Current treatment for P. aeruginosa infections relies solely on antibiotics, but the rise of antibiotic-resistant strains necessitates an urgent need for a protective vaccine. Traditional parenteral vaccines, despite employing potent adjuvants aimed at serotype-dependent immunity, often fail to elicit the desired mucosal immune response. Thus, developing vaccines that target both localized mucosal and systemic immune responses represents a promising direction for future research on P. aeruginosa vaccination. In this study, we explored EPS301, the exopolysaccharide derived from the lung microbiota strain Lactobacillus plantarum WXD301, which exhibits excellent self-assembly properties, enabling the formation of homogeneous nanoparticles when encapsulating recombinant PcrV of P. aeruginosa, designated as EPS301@rPcrV. Notably, the EPS301 vector effectively enhanced antigen adhesion to the nasal and pulmonary mucosal tissues and prolonged antigen retention. Moreover, EPS301@rPcrV provided effective and sustained protection against P. aeruginosa pneumonia, surpassing the durability achieved with the "gold standard" cholera toxin adjuvant. The EPS301-adjuvanted vaccine formulation elicited robust mucosal IgA and Th17/γδ17 T cell responses, which exceeded those induced by the CTB-adjuvanted vaccination and were sustained for over 112 days. Additionally, Th 17 and γδ 17 resident memory T cells induced by EPS301@rPcrV were crucial for protection against P. aeruginosa challenge. Intriguingly, IL-17A knockout mice exhibited lower survival rates, impaired bacterial clearance ability, and exacerbated lung tissue damage upon EPS301 adjuvanted vaccination against P. aeruginosa-induced pneumonia, indicating an IL-17A-dependent protective mechanism. In conclusion, our findings provided direct evidence that EPS301@rPcrV mucosal vaccine is a promising candidate for future clinical application against P. aeruginosa-induced pulmonary infection.
Collapse
Affiliation(s)
- Haochi Zhang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
- The Spirit Jinyu Biological Pharmaceutical Co. Ltd, Hohhot, Inner Mongolia, China
| | - Shouxin Sheng
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| | - Chunhe Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| | - Xuemei Bao
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| | - Lixia Zhao
- The Spirit Jinyu Biological Pharmaceutical Co. Ltd, Hohhot, Inner Mongolia, China
| | - Jian Chen
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
- The Spirit Jinyu Biological Pharmaceutical Co. Ltd, Hohhot, Inner Mongolia, China
| | - Pingyuan Guan
- The Spirit Jinyu Biological Pharmaceutical Co. Ltd, Hohhot, Inner Mongolia, China
| | - Xiaoyan Li
- The Spirit Jinyu Biological Pharmaceutical Co. Ltd, Hohhot, Inner Mongolia, China
| | - Na Pan
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| | - Yanchen Liang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| | - Xueqi Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| | - Jingmin Sun
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| | - Xiao Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, P.R. China
| |
Collapse
|
4
|
Rodríguez-Míguez Y, Lozano-Ordaz V, Ortiz-Cabrera AE, Barrios-Payan J, Mata-Espinosa D, Huerta-Yepez S, Baay-Guzman G, Hernández-Pando R. Effect of IL-17A on the immune response to pulmonary tuberculosis induced by high- and low-virulence strains of Mycobacterium bovis. PLoS One 2024; 19:e0307307. [PMID: 39024223 PMCID: PMC11257284 DOI: 10.1371/journal.pone.0307307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
Tuberculosis (TB) is an infectious, chronic, and progressive disease occurring globally. Human TB is caused mainly by Mycobacterium tuberculosis (M. tuberculosis), while the main causative agent of bovine TB is Mycobacterium bovis (M. bovis). The latter is one of the most important cattle pathogens and is considered the main cause of zoonotic TB worldwide. The mechanisms responsible for tissue damage (necrosis) during post-primary TB remain elusive. Recently, IL-17A was reported to be important for protection against M. tuberculosis infection, but it is also related to the production of an intense inflammatory response associated with necrosis. We used two M. bovis isolates with different levels of virulence and high IL-17A production to study this important cytokine's contrasting functions in a BALB/c mouse model of pulmonary TB. In the first part of the study, the gene expression kinetics and cellular sources of IL-17A were determined by real time PCR and immunohistochemistry respectively. Non-infected lungs showed low production of IL-17A, particularly by the bronchial epithelium, while lungs infected with the low-virulence 534 strain showed high IL-17A expression on Day 3 post-infection, followed by a decrease in expression in the early stage of the infection and another increase during late infection, on Day 60, when very low bacillary burdens were found. In contrast, infection with the highly virulent strain 04-303 induced a peak of IL-17A expression on Day 14 of infection, 1 week before extensive pulmonary necrosis was seen, being lymphocytes and macrophages the most important sources. In the second part of the study, the contribution of IL-17A to immune protection and pulmonary necrosis was evaluated by suppressing IL-17A via the administration of specific blocking antibodies. Infection with M. bovis strain 534 and treatment with IL-17A neutralizing antibodies did not affect mouse survival but produced a significant increase in bacillary load and a non-significant decrease in inflammatory infiltrate and granuloma area. In contrast, mice infected with the highly virulent 04-303 strain and treated with IL-17A blocking antibodies showed a significant decrease in survival, an increase in bacillary loads on Day 24 post-infection, and significantly more and earlier necrosis. Our results suggest that high expression of IL-17A is more related to protection than necrosis in a mouse model of pulmonary TB induced by M. bovis strains.
Collapse
Affiliation(s)
- Yadira Rodríguez-Míguez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Vasti Lozano-Ordaz
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Angel E. Ortiz-Cabrera
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Jorge Barrios-Payan
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Dulce Mata-Espinosa
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Sara Huerta-Yepez
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México “Federico Gómez”, Mexico City, Mexico
| | - Guillermina Baay-Guzman
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México “Federico Gómez”, Mexico City, Mexico
| | - Rogelio Hernández-Pando
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| |
Collapse
|
5
|
Georgakopoulou VE, Lempesis IG, Sklapani P, Trakas N, Spandidos DA. Exploring the pathogenetic mechanisms of Mycoplasmapneumoniae (Review). Exp Ther Med 2024; 28:271. [PMID: 38765654 PMCID: PMC11097136 DOI: 10.3892/etm.2024.12559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/17/2024] [Indexed: 05/22/2024] Open
Abstract
Mycoplasmas, the smallest self-replicating prokaryotes without a cell wall, are the most prevalent and extensively studied species in humans. They significantly contribute to chronic respiratory tract illnesses and pneumonia, with children and adolescents being particularly vulnerable. Mycoplasma pneumoniae (M. pneumoniae) infections typically tend to be self-limiting and mild but can progress to severe or even life-threatening conditions in certain individuals. Extrapulmonary effects often occur without pneumonia, and both intrapulmonary and extrapulmonary complications operate through separate pathological mechanisms. The indirect immune-mediated damage of the immune system, vascular blockages brought on by vasculitis or thrombosis and direct harm from invasion or locally induced inflammatory cytokines are potential causes of extrapulmonary manifestations due to M. pneumoniae. Proteins associated with adhesion serve as the primary factor crucial for the pathogenicity of M. pneumoniae, relying on a specialized polarized terminal attachment organelle. The type and density of these host receptors significantly impact the adhesion and movement of M. pneumoniae, subsequently influencing the pathogenic mechanism and infection outcomes. Adjacent proteins are crucial for the proper assembly of the attachment organelle, with variations in the genetic domains of P1, P40 and P90 surfaces contributing to the variability of clinical symptoms and offering new avenues for developing vaccines against M. pneumoniae infections. M. pneumoniae causes oxidative stress within respiratory tract epithelial cells by adhering to host cells and releasing hydrogen peroxide and superoxide radicals. This oxidative stress enhances the vulnerability of host cells to harm induced by oxygen molecules. The lack of superoxide dismutase and catalase of bacteria allows it to hinder the catalase activity of the host cell, leading to the reduced breakdown of peroxides. Lung macrophages play a significant role in managing M. pneumoniae infection, identifying it via Toll-like receptor 2 and initiating the myeloid differentiation primary response gene 88-nuclear factor κΒ signaling cascade. However, the precise mechanisms enabling M. pneumoniae to evade intracellular host defenses remain unknown, necessitating further exploration of the pathways involved in intracellular survival. The present comprehensive review delves into the pathogenesis of M. pneumoniae infection within the pulmonary system and into extrapulmonary areas, outlining its impact.
Collapse
Affiliation(s)
- Vasiliki Epameinondas Georgakopoulou
- Department of Pathophysiology, Laiko General Hospital, National and Kapodisttrian University of Athens, 11527 Athens, Greece
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, 11527 Athens, Greece
| | - Ioannis G. Lempesis
- Department of Pathophysiology, Laiko General Hospital, National and Kapodisttrian University of Athens, 11527 Athens, Greece
| | - Pagona Sklapani
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece
| | - Nikolaos Trakas
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
6
|
Giugni FR, Duarte-Neto AN, da Silva LFF, Monteiro RAA, Mauad T, Saldiva PHN, Dolhnikoff M. Younger age is associated with cardiovascular pathological phenotype of severe COVID-19 at autopsy. Front Med (Lausanne) 2024; 10:1327415. [PMID: 38259848 PMCID: PMC10801169 DOI: 10.3389/fmed.2023.1327415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction COVID-19 affects patients of all ages. There are few autopsy studies focusing on the younger population. We assessed an autopsy cohort aiming to understand how age influences pathological outcomes in fatal COVID-19. Methods This study included autopsied patients, aged 6 months to 83 years, with confirmed COVID-19 in 2020-2021. We collected tissue samples from deceased patients using a minimally invasive autopsy protocol and assessed pathological data following a systematic approach. Results Eighty-six patients were included, with a median age of 55 years (IQR 32.3-66.0). We showed that age was significantly lower in patients with acute heart ischemia (p = 0.004), myocarditis (p = 0.03) and lung angiomatosis (p < 0.001), and significantly higher in patients with exudative diffuse alveolar damage (p = 0.02), proliferative diffuse alveolar damage (p < 0.001), lung squamous metaplasia (p = 0.003) and lung viral atypia (p = 0.03), compared to patients without those findings. We stratified patients by their age and showed that cardiovascular findings were more prevalent in children and young adults. We performed principal component analysis and cluster of pathological variables, and showed that cardiovascular variables clustered and covariated together, and separated from pulmonary variables. Conclusion We showed that age modulates pathological outcomes in fatal COVID-19. Younger age is associated with cardiovascular abnormalities and older age with pulmonary findings.
Collapse
Affiliation(s)
- Fernando R. Giugni
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Amaro N. Duarte-Neto
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Fernando F. da Silva
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- SVOC - Serviço de Verificação de Óbitos da Capital, Universidade de São Paulo, São Paulo, Brazil
| | - Renata A. A. Monteiro
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Thais Mauad
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo H. N. Saldiva
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
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
|
8
|
Wang J, Li W, Li N, Wang B. Immunization with Multiple Virulence Factors Provides Maternal and Neonatal Protection against Group B Streptococcus Serotypes. Vaccines (Basel) 2023; 11:1459. [PMID: 37766135 PMCID: PMC10535937 DOI: 10.3390/vaccines11091459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Group B streptococcus (GBS) commonly colonizes the vaginal tract and is a leading cause of life-threatening neonatal infections and adverse pregnancy outcomes. No effective vaccine is clinically available. Conserved bacterial virulence factors, including those of GBS, have been employed as vaccine components. We investigated serotype-independent protection against GBS by intranasal immunization with six conserved GBS virulence factors (GBSV6). GBSV6 induced systemic and vaginal antibodies and T cell responses in mice. The immunity reduced mouse mortality and vaginal colonization by various GBS serotypes and protected newborn mice of immunized dams against GBS challenge. Intranasal GBSV6 immunization also provided long-lasting protective immunity and had advantages over intramuscular GBSV6 immunization regarding restricting vaginal GBS colonization. Our findings indicate that intranasal immunization targeting multiple conserved GBS virulence factors induces serotype-independent immunity, which protects against GBS infection systemically and vaginally in dams and prevents newborn death. The study presents valuable strategies for GBS vaccine development.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Varnotech Biopharm Ltd., Beijing 100176, China
| | - Wenbo Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Varnotech Biopharm Ltd., Beijing 100176, China
| | - Ning Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Beinan Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Yoon S, Lee G, Yu J, Lee K, Lee K, Si J, You HJ, Ko G. Distinct Changes in Microbiota-Mediated Intestinal Metabolites and Immune Responses Induced by Different Antibiotics. Antibiotics (Basel) 2022; 11:antibiotics11121762. [PMID: 36551419 PMCID: PMC9774394 DOI: 10.3390/antibiotics11121762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The cocktails of antibiotics are utilized to study the functions of microbiota. There have been studies on the alteration of not only the microbiota composition but also the host's metabolism or immunity. However, the bacterial species associated with these altered physiologic markers are still unclear. Therefore, we supplied mice with drinking water containing ampicillin (AMP), vancomycin (VAN), neomycin (NEO), or metronidazole (MET) to observe the effect of each antibiotic on helper T cells and inflammation-related gene expression and metabolism, including amino acid metabolism and changes in gut microbiota. We observed major changes in gut microbiota in mice treated with AMP and VAN, respectively, immediately after administration. The abundance of the genera Parabacteroides and Akkermansia increased in the AMP and VAN groups, while Prevotella almost disappeared from both groups. The compositional changes in intestinal metabolites in the AMP and VAN groups were more distinct than those in the NEO and MET groups, which was similar to the microbiome results. In particular, the most distinct changes were observed in amino acid related metabolism in AMP and VAN groups; the amounts of phenylalanine and tyrosine were increased in the AMP group while those were decreased in the VAN group. The changed amounts of intestinal amino acids in each of the AMP and VAN groups were correlated with increases in the abundance of the genera Parabacteroides and Akkermansia in the AMP and VAN groups, respectively. The most distinctive changes in intestinal gene expression were observed in the ileum, especially the expression Th17-related genes such as rorgt, il17a, and il17f, which decreased dramatically in the guts of most of the antibiotic-treated groups. These changes were also associated with a significant decrease in Prevotella in both the AMP and VAN groups. Taken together, these findings indicate that changes in gut microbiota as well as host physiology, including host metabolism and immunity, differ depending on the types of antibiotics, and the antibiotic-induced gut microbiota alteration has a correlation with host physiology such as host metabolic or immunological status. Thus, the immune and metabolic status of the host should be taken into account when administering antibiotics.
Collapse
Affiliation(s)
- Sunghyun Yoon
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Giljae Lee
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Republic of Korea
| | - Junsun Yu
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Kiuk Lee
- KoBioLabs, Inc., Seoul 13488, Republic of Korea
| | - Kyeongju Lee
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Jiyeon Si
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Natural Products Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun Ju You
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- KoBioLabs, Inc., Seoul 13488, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (H.J.Y.); (G.K.)
| | - GwangPyo Ko
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Republic of Korea
- KoBioLabs, Inc., Seoul 13488, Republic of Korea
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (H.J.Y.); (G.K.)
| |
Collapse
|
12
|
Blanc P, Liu Y, Reveneau N, Cavell B, Gorringe A, Renauld-Mongénie G. The role of bactericidal and opsonic activity in immunity against Bordetella pertussis. Expert Rev Vaccines 2022; 21:1727-1738. [PMID: 36369768 DOI: 10.1080/14760584.2022.2137145] [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: 11/13/2022]
Abstract
INTRODUCTION Pertussis vaccines have drastically reduced the disease burden in humans since their implementation. Despite their success, pertussis remains an important global public health challenge. Bordetella pertussis resurgence could be a result of greater surveillance combined with improved diagnosis methods, changes in Bordetella pertussis biology, vaccine schedules, and/or coverage. Additionally, mechanisms of protection conferred by acellular pertussis (aP) and whole-cell pertussis (wP) vaccines differ qualitatively. There are no clear immune correlates of protection for pertussis vaccines. Pertussis antigens can induce toxin neutralizing antibodies, block adherence or engage complement mediated phagocytic/bactericidal killing. AREAS COVERED We reviewed the existing evidence on antibody-mediated serum bactericidal and opsonophagocytic activity and discussed the relevance of these functional antibodies in the development of next-generation pertussis vaccines. EXPERT OPINION Current paradigm proposes that wP vaccines may confer greater herd protection than aP vaccines due to their enhanced clearance of bacteria from the nasopharynx in animal models. Functional antibodies may contribute to the reduction of nasal colonization, which differentiates aP and wP vaccines. Understanding the intrinsic differences in protective immune responses elicited by each class of vaccines will help to identify biomarkers that can be used as immunological end points in clinical trials.
Collapse
Affiliation(s)
- Pascal Blanc
- Research & Development, Sanofi, Marcy l'Etoile, France
| | - Yuanqing Liu
- Research & Development, Sanofi, Marcy l'Etoile, France
| | | | - Breeze Cavell
- Department of Research and Evaluation, United Kingdom (UK) Health Security Agency, Salisbury, UK
| | - Andrew Gorringe
- Department of Research and Evaluation, United Kingdom (UK) Health Security Agency, Salisbury, UK
| | | |
Collapse
|
13
|
Short KK, Lathrop SK, Davison CJ, Partlow HA, Kaiser JA, Tee RD, Lorentz EB, Evans JT, Burkhart DJ. Using Dual Toll-like Receptor Agonism to Drive Th1-Biased Response in a Squalene- and α-Tocopherol-Containing Emulsion for a More Effective SARS-CoV-2 Vaccine. Pharmaceutics 2022; 14:1455. [PMID: 35890352 PMCID: PMC9318334 DOI: 10.3390/pharmaceutics14071455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
A diversity of vaccines is necessary to reduce the mortality and morbidity of SARS-CoV-2. Vaccines must be efficacious, easy to manufacture, and stable within the existing cold chain to improve their availability around the world. Recombinant protein subunit vaccines adjuvanted with squalene-based emulsions such as AS03™ and MF59™ have a long and robust history of safe, efficacious use with straightforward production and distribution. Here, subunit vaccines were made with squalene-based emulsions containing novel, synthetic toll-like receptor (TLR) agonists, INI-2002 (TLR4 agonist) and INI-4001 (TLR7/8 agonist), using the recombinant receptor-binding domain (RBD) of SARS-CoV-2 S protein as an antigen. The addition of the TLR4 and TLR7/8 agonists, alone or in combination, maintained the formulation characteristics of squalene-based emulsions, including a sterile filterable droplet size (<220 nm), high homogeneity, and colloidal stability after months of storage at 4, 25, and 40 °C. Furthermore, the addition of the TLR agonists skewed the immune response from Th2 towards Th1 in immunized C57BL/6 mice, resulting in an increased production of IgG2c antibodies and a lower antigen-specific production of IL-5 with a higher production of IFNγ by lymphocytes. As such, incorporating TLR4 and TLR7/8 agonists into emulsions leveraged the desirable formulation and stability characteristics of emulsions and can induce Th1-type humoral and cell-mediated immune responses to combat the continued threat of SARS-CoV-2.
Collapse
Affiliation(s)
- Kristopher K. Short
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Stephanie K. Lathrop
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Clara J. Davison
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Haley A. Partlow
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Johnathan A. Kaiser
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Rebekah D. Tee
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Elizabeth B. Lorentz
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jay T. Evans
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - David J. Burkhart
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; (K.K.S.); (S.K.L.); (C.J.D.); (H.A.P.); (J.A.K.); (R.D.T.); (E.B.L.); (J.T.E.)
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
14
|
Chen S, Zhang X, Yang C, Wang S, Shen H. Essential role of IL-17 in acute exacerbation of pulmonary fibrosis induced by non-typeable Haemophilus influenzae. Theranostics 2022; 12:5125-5137. [PMID: 35836804 PMCID: PMC9274745 DOI: 10.7150/thno.74809] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/14/2022] [Indexed: 01/12/2023] Open
Abstract
Background: Acute exacerbation (AE) of idiopathic pulmonary fibrosis (IPF) has a poor prognosis and lacks effective therapy. Animal models that mimic AE-IPF can greatly accelerate investigation of its pathogenesis and development of effective therapy. However, there are few reports of animal models of AE-IPF caused by bacteria. Thus, our study aimed to establish a mouse model of bacterium-induced AE-IPF and explore the potential pathogenic mechanism of AE-IPF. Methods: Mice were instilled intranasally with bleomycin (BLM) followed by non-typeable Haemophilus influenzae (NTHi) strain NT127. Murine survival, bacterial load, body weight and pulmonary histopathological changes were evaluated. We analyzed the T cell and inflammatory cell responses in the lungs. Results: Infection with 107 CFU NT127 triggered AE in mice with PF induced by 30 μg BLM. Compared with BLM-instilled mice, the BLM/NT127-treated mice showed more obvious airway inflammation, lower survival rate, higher inflammatory cell response, and increased proportions and numbers of IL-17+CD4+, IL-17+ γδ T, IL-22+CD4+ and regulatory T (Treg) cells in lungs. γδ T cells were the predominant source of IL-17. IL-17 gene knockout mice with AE-IPF had quicker body weight recovery, milder pulmonary inflammation and fibrosis, stronger IL-22+CD4+T, TGF-β+ γδ T and Treg cell responses, and weaker neutrophil and eosinophil responses than wild-type mice with AE-IPF. Conclusions: NTHi infection after BLM-induced IPF can cause AE-IPF in a murine model. This novel model can be used to investigate the pathogenesis of AE-IPF and develop new therapies for AE-IPF caused by bacteria. IL-17 is essential for the development of AE-IPF, and it may be a new therapeutic target for bacteria-induced AE-IPF.
Collapse
Affiliation(s)
- Shengsen Chen
- Department of Endoscopy (the bronchoscope group), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA
| | - Xinyun Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA.,Department of Infectious Diseases, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Cheng Yang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA.,Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shi Wang
- Department of Endoscopy (the bronchoscope group), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.,✉ Corresponding authors: Shi Wang, Department of Endoscopy (the bronchoscope group), Zhejiang Cancer Hospital, No. 1 Banshandong Road, Hangzhou 310022, China. E-mail: ; Hao Shen, Department of Microbiology, University of Pennsylvania Perelman School of Medicine, 3610 Hamilton Walk, Philadelphia 19104, USA. E-mail:
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA.,✉ Corresponding authors: Shi Wang, Department of Endoscopy (the bronchoscope group), Zhejiang Cancer Hospital, No. 1 Banshandong Road, Hangzhou 310022, China. E-mail: ; Hao Shen, Department of Microbiology, University of Pennsylvania Perelman School of Medicine, 3610 Hamilton Walk, Philadelphia 19104, USA. E-mail:
| |
Collapse
|
15
|
The role of Th17 cells: explanation of relationship between periodontitis and COPD? Inflamm Res 2022; 71:1011-1024. [PMID: 35781342 DOI: 10.1007/s00011-022-01602-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/13/2022] [Indexed: 11/05/2022] Open
Abstract
Periodontitis and chronic obstructive pulmonary disease (COPD) are chronic inflammatory diseases with common risk factors, such as long-term smoking, age, and social deprivation. Many observational studies have shown that periodontitis and COPD are correlated. Moreover, they share a common pathophysiological process involving local accumulation of inflammatory cells and cytokines and damage of soft tissues. The T helper 17 (Th17) cells and the related cytokines, interleukin (IL)-17, IL-22, IL-1β, IL-6, IL-23, and transforming growth factor (TGF)-β, play a crucial regulatory role during the pathophysiological process. This paper reviewed the essential roles of Th17 lineage in the occurrence of periodontitis and COPD. The gaps in the study of their common pathological mechanism were also evaluated to explore future research directions. Therefore, this review can provide study direction for the association between periodontitis and COPD and new ideas for the clinical diagnosis and treatment of the two diseases.
Collapse
|
16
|
Correia A, Alves P, Fróis-Martins R, Teixeira L, Vilanova M. Protective Effect against Neosporosis Induced by Intranasal Immunization with Neospora caninum Membrane Antigens Plus Carbomer-Based Adjuvant. Vaccines (Basel) 2022; 10:vaccines10060925. [PMID: 35746533 PMCID: PMC9230871 DOI: 10.3390/vaccines10060925] [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: 05/02/2022] [Revised: 06/04/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Neospora caninum is an obligate intracellular protozoan responsible for abortion and stillbirths in cattle. We previously developed a mucosal vaccination approach using N. caninum membrane proteins and CpG adjuvant that conferred long-term protection against neosporosis in mice. Here, we have extended this approach by alternatively using the carbomer-based adjuvant Carbigen™ in the immunizing preparation. Immunized mice presented higher proportions and numbers of memory CD4+ and CD8+ T cells. Stimulation of spleen, lungs and liver leukocytes with parasite antigens induced a marked production of IFN-γ and IL-17A and, less markedly, IL-4. This balanced response was also evident in that both parasite-specific IgG1 and IgG2c were raised by immunization, together with specific intestinal IgA. Upon intraperitoneal infection with N. caninum, immunized mice presented lower parasitic burdens than sham-immunized controls. In the infected immunized mice, memory CD4+ T cells predominantly expressed T-bet and RORγt, and CD8+ T cells expressing T-bet were found increased. While spleen, lungs and liver leukocytes of both immunized and sham-immunized infected animals produced high amounts of IFN-γ, only the cells from immunized mice responded with high IL-17A production. Since in cattle both IFN-γ and IL-17A have been associated with protective mechanisms against N. caninum infection, the elicited cytokine profile obtained using CarbigenTM as adjuvant indicates that it could be worth exploring for bovine neosporosis vaccination.
Collapse
Affiliation(s)
- Alexandra Correia
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 4050-313 Porto, Portugal; (A.C.); (P.A.)
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal
| | - Pedro Alves
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 4050-313 Porto, Portugal; (A.C.); (P.A.)
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland;
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Luzia Teixeira
- UMIB—Unidade Multidisciplinar de Investigação Biomédica, ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-290 Porto, Portugal
| | - Manuel Vilanova
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 4050-313 Porto, Portugal; (A.C.); (P.A.)
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
17
|
Jiang Z, Zhou R, Leung PHM, Deng Z, Li S. An attenuated multiple genetic mutant of Mycoplasma pneumoniae imparts good immuno-protection against M. pneumoniae pneumonia in BALB/c mice. Microb Pathog 2022; 165:105463. [PMID: 35240287 DOI: 10.1016/j.micpath.2022.105463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/19/2022]
Abstract
Mycoplasma pneumoniae (M. pneumoniae) is the causative agent of both upper and lower respiratory infections that can lead to pneumonia, extrapulmonary complications and devastating sequela. With the increasing rate of macrolide-resistant strains, the severe clinical consequence of refractory mycoplasma pneumonia in children health calls for the need of vaccine research for this pathogen. In this report, the immunomodulatory effectiveness of a live attenuated M. pneumoniae vaccine was evaluated. The vaccine strain was a mutant strain of M. pneumoniae, MUT129, obtained after multiple passages of M129 strain in PPLO broth. The SNP/InDel detection results showed that mutations were present in genes encoding the adhesion organelle-associated proteins and lipoproteins of M. pneumoniae MUT129. Upon intranasal challenge of BALB/c mice with 1 × 107 CFU of MUT129, there were very small amount of Mycoplasma antigens and almost no M. pneumoniae present in the lung tissues of BALB/c mice. Besides, there was almost no inflammatory cell infiltration in the lung tissue. Results of the M. pneumoniae challenge study showed that mice immunized with MUT129 presented with less inflammation, lower detectable number of M. pneumoniae in the lungs when compared with the unimmunized mice. These results indicated that the live attenuated vaccine can efficiently prevent the proliferation of M. pneumonia in the lungs, reduce but not completely prevent the pulmonary inflammatory response.
Collapse
Affiliation(s)
- Zhulin Jiang
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - Runjie Zhou
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - Polly H M Leung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Zhongliang Deng
- Hengyang Medical College, University of South China, Hengyang, 421001, China.
| | - Shuihong Li
- Hengyang Medical College, University of South China, Hengyang, 421001, China.
| |
Collapse
|
18
|
Leibel SL, Sun X. COVID-19 in Early Life: Infants and Children Are Affected Too. Physiology (Bethesda) 2021; 36:359-366. [PMID: 34704855 PMCID: PMC8560374 DOI: 10.1152/physiol.00022.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/02/2021] [Accepted: 08/08/2021] [Indexed: 12/12/2022] Open
Abstract
Compared with adults, children are less likely infected with SARS-CoV-2 and are often asymptomatic when infected. However, infection in children can lead to severe disease. The pandemic affects the lives of all children, especially those with lower socioeconomic status. This review highlights the physiological impacts of COVID-19 in early life.
Collapse
Affiliation(s)
- Sandra L Leibel
- Department of Pediatrics, University of California at San Diego, La Jolla, California
| | - Xin Sun
- Department of Pediatrics, University of California at San Diego, La Jolla, California
- Department of Biological Sciences, University of California at San Diego, La Jolla, California
| |
Collapse
|
19
|
Al-Suhaimi EA, Aljafary MA, Alkhulaifi FM, Aldossary HA, Alshammari T, AL-Qaaneh A, Aldahhan R, Alkhalifah Z, Gaymalov ZZ, Shehzad A, Homeida AM. Thymus Gland: A Double Edge Sword for Coronaviruses. Vaccines (Basel) 2021; 9:1119. [PMID: 34696231 PMCID: PMC8539924 DOI: 10.3390/vaccines9101119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023] Open
Abstract
The thymus is the main lymphoid organ that regulates the immune and endocrine systems by controlling thymic cell proliferation and differentiation. The gland is a primary lymphoid organ responsible for generating mature T cells into CD4+ or CD8+ single-positive (SP) T cells, contributing to cellular immunity. Regarding humoral immunity, the thymic plasma cells almost exclusively secrete IgG1 and IgG3, the two main complement-fixing effector IgG subclasses. Deformity in the thymus can lead to inflammatory diseases. Hassall's corpuscles' epithelial lining produces thymic stromal lymphopoietin, which induces differentiation of CDs thymocytes into regulatory T cells within the thymus medulla. Thymic B lymphocytes produce immunoglobulins and immunoregulating hormones, including thymosin. Modulation in T cell and naive T cells decrement due to thymus deformity induce alteration in the secretion of various inflammatory factors, resulting in multiple diseases. Influenza virus activates thymic CD4+ CD8+ thymocytes and a large amount of IFNγ. IFNs limit virus spread, enhance macrophages' phagocytosis, and promote the natural killer cell restriction activity against infected cells. Th2 lymphocytes-produced cytokine IL-4 can bind to antiviral INFγ, decreasing the cell susceptibility and downregulating viral receptors. COVID-19 epitopes (S, M, and N proteins) with ≥90% identity to the SARS-CoV sequence have been predicted. These epitopes trigger immunity for antibodies production. Boosting the immune system by improving thymus function can be a therapeutic strategy for preventing virus-related diseases. This review aims to summarize the endocrine-immunoregulatory functions of the thymus and the underlying mechanisms in the prevention of COVID-19.
Collapse
Affiliation(s)
- Ebtesam A. Al-Suhaimi
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| | - Meneerah A. Aljafary
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| | - Fadwa M. Alkhulaifi
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| | - Hanan A. Aldossary
- Epidemic Diseases Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; or
| | - Thamer Alshammari
- Genetic Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (T.A.); (A.A.-Q.); (Z.A.)
| | - Ayman AL-Qaaneh
- Genetic Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (T.A.); (A.A.-Q.); (Z.A.)
- Clinical Pharmacy Services Division, Pharmacy Services Department, Johns Hopkins Aramco Healthcare (JHAH), Dhahran 31311, Saudi Arabia
| | - Razan Aldahhan
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Zahra Alkhalifah
- Genetic Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (T.A.); (A.A.-Q.); (Z.A.)
| | - Zagit Z. Gaymalov
- Earlystage OÜ, Lasnamäe Linnaosa, Sepapaja tn 6, Harju Maakond, 15551 Tallinn, Estonia;
| | - Adeeb Shehzad
- Clinical Pharmacy Research Department, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Abdelgadir M. Homeida
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| |
Collapse
|
20
|
Biryukov S, Dankmeyer JL, Shamsuddin Z, Velez I, Rill NO, Rosario-Acevedo R, Klimko CP, Shoe JL, Hunter M, Ward MD, Cazares LH, Fetterer DP, Bozue JA, Worsham PL, Cote CK, Amemiya K. Impact of Toll-Like Receptor-Specific Agonists on the Host Immune Response to the Yersinia pestis Plague rF1V Vaccine. Front Immunol 2021; 12:726416. [PMID: 34512658 PMCID: PMC8430260 DOI: 10.3389/fimmu.2021.726416] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
Relatively recent advances in plague vaccinology have produced the recombinant fusion protein F1-V plague vaccine. This vaccine has been shown to readily protect mice from both bubonic and pneumonic plague. The protection afforded by this vaccine is solely based upon the immune response elicited by the F1 or V epitopes expressed on the F1-V fusion protein. Accordingly, questions remain surrounding its efficacy against infection with non-encapsulated (F1-negative) strains. In an attempt to further optimize the F1-V elicited immune response and address efficacy concerns, we examined the inclusion of multiple toll-like receptor agonists into vaccine regimens. We examined the resulting immune responses and also any protection afforded to mice that were exposed to aerosolized Yersinia pestis. Our data demonstrate that it is possible to further augment the F1-V vaccine strategy in order to optimize and augment vaccine efficacy.
Collapse
Affiliation(s)
- Sergei Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Zain Shamsuddin
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Ivan Velez
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Nathaniel O. Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Raysa Rosario-Acevedo
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jennifer L. Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Michael D. Ward
- Molecular Biology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Lisa H. Cazares
- Molecular Biology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - David P. Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Joel A. Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Patricia L. Worsham
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Kei Amemiya
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| |
Collapse
|
21
|
Tomioka H, Tatano Y, Shimizu T, Sano C. Immunoadjunctive Therapy against Bacterial Infections Using Herbal Medicines Based on Th17 Cell-mediated Protective Immunity. Curr Pharm Des 2021; 27:3949-3962. [PMID: 34102961 DOI: 10.2174/1381612827666210608143449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/27/2021] [Indexed: 11/22/2022]
Abstract
One of the major health concerns in the world is the global increase in intractable bacterial infectious diseases due to the emergence of multi- and extensively drug-resistant bacterial pathogens as well as an increase in compromised hosts around the world. Particularly, in the case of mycobacteriosis, the high incidence of tuberculosis in developing countries, resurgence of tuberculosis in industrialized countries, and increase in the prevalence of Mycobacterium avium complex infections are important worldwide health concerns. However, the development of novel antimycobacterial drugs is currently making slow progress. Therefore, it is considered that devising improved administration protocols for clinical treatment against refractory mycobacteriosis using existing chemotherapeutics is more practical than awaiting the development of new antimycobacterial drugs. The regulation of host immune responses using immunoadjunctive agents may increase the efficacy of antimicrobial treatment against mycobacteriosis. The same situations also exist in cases of intractable infectious diseases due to common bacteria other than mycobacteria. The mild and long-term up-regulation of host immune reactions in hosts with intractable chronic bacterial infections, using herbal medicines and medicinal plants, may be beneficial for such immunoadjunctive therapy. This review describes the current status regarding basic and clinical studies on therapeutic regimens using herbal medicines, useful for the clinical treatment of patients with intractable bacterial infections. In particular, we focus on immunoadjunctive effects of herbal medicines on the establishment and manifestation of host antibacterial immunity related to the immunological roles of Th17 cell lineages.
Collapse
Affiliation(s)
- Haruaki Tomioka
- Department of Basic Medical Science for Nursing, Department of Contemporary Psychology, Yasuda Women's University, Hiroshima, Japan
| | - Yutaka Tatano
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Fukuoka, Japan
| | - Toshiaki Shimizu
- Department of Nutrition Administration, Yasuda Women's University, Hiroshima,, Japan
| | - Chiaki Sano
- Department of Community Medicine Management, Shimane University School of Medicine, Izumo, Japan
| |
Collapse
|
22
|
Di Giorgio A, Hartleif S, Warner S, Kelly D. COVID-19 in Children With Liver Disease. Front Pediatr 2021; 9:616381. [PMID: 33777864 PMCID: PMC7991080 DOI: 10.3389/fped.2021.616381] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/19/2021] [Indexed: 12/15/2022] Open
Abstract
Background: The global pandemic caused by novel Coronavirus SARS-CoV-2 disease (COVID-19) is a major threat to the general population and for patients with pre-existing chronic conditions. We report data concerning SARS-CoV-2 infection in children with chronic liver disease (CLD). Methods: A literature review using the online database PubMed was performed to summarize available findings on the association between pre-existing liver disease and COVID-19 infection in children. Results: Children with COVID-19 have preserved effector and immunosuppressive components resulting in a milder disease compared to adults. The most common hepatic manifestation is an elevation of hepatic transaminases. Liver damage may be directly caused by viral infection of liver cells, by medications or by the chronic hypoxia seen in COVID-19 patients. A multicenter study reported that the majority of children with a CLD remained healthy during the outbreak. Similarly, studies reported that children on immunosuppressive treatment, including patients with autoimmune liver disease (AILD) and liver transplantation (LT), maintained good health during the outbreak without experiencing major complications even if infected with COVID-19. Conclusion: COVID-19-related liver injury presents with a mild elevation of transaminases, although its clinical significance is unclear. Children with CLD, including those with AILD and post-LT, do not have an increased risk for severe disease course of SARS-CoV-2 infection with little or no liver dysfunction. These data highlight the necessity to ensure normal standards of care while adhering to national Covid-19 guidelines, and particularly to maintain immunosuppressive medication to prevent relapse or rejection. Further research is required to evaluate the differences in clinical course between immunosuppressed adults and children and in particular whether asymptomatic infection is a concern.
Collapse
Affiliation(s)
- Angelo Di Giorgio
- Paediatric Liver, Gastroenterology and Transplantation, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Steffen Hartleif
- Paediatric Gastroenterology and Hepatology, University Children's Hospital, University of Tübingen, Tübingen, Germany
| | - Suzan Warner
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- The Liver Unit, Birmingham Women's and Children's Hospital, University of Birmingham, Birmingham, United Kingdom
| | - Deirdre Kelly
- The Liver Unit, Birmingham Women's and Children's Hospital, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
23
|
Kroemer A, Belyayev L, Khan K, Loh K, Kang J, Duttargi A, Dhani H, Sadat M, Aguirre O, Gusev Y, Bhuvaneshwar K, Kallakury B, Cosentino C, Houlihan B, Diaz J, Moturi S, Yazigi N, Kaufman S, Subramanian S, Hawksworth J, Girlanda R, Robson SC, Matsumoto CS, Zasloff M, Fishbein TM. Rejection of intestinal allotransplants is driven by memory T helper type 17 immunity and responds to infliximab. Am J Transplant 2021; 21:1238-1254. [PMID: 32882110 PMCID: PMC8049508 DOI: 10.1111/ajt.16283] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
Intestinal transplantation (ITx) can be life-saving for patients with advanced intestinal failure experiencing complications of parenteral nutrition. New surgical techniques and conventional immunosuppression have enabled some success, but outcomes post-ITx remain disappointing. Refractory cellular immune responses, immunosuppression-linked infections, and posttransplant malignancies have precluded widespread ITx application. To shed light on the dynamics of ITx allograft rejection and treatment resistance, peripheral blood samples and intestinal allograft biopsies from 51 ITx patients with severe rejection, alongside 37 stable controls, were analyzed using immunohistochemistry, polychromatic flow cytometry, and reverse transcription-PCR. Our findings inform both immunomonitoring and treatment. In terms of immunomonitoring, we found that while ITx rejection is associated with proinflammatory and activated effector memory T cells in the blood, evidence of treatment efficacy can only be found in the allograft itself, meaning that blood-based monitoring may be insufficient. In terms of treatment, we found that the prominence of intra-graft memory TNF-α and IL-17 double-positive T helper type 17 (Th17) cells is a leading feature of refractory rejection. Anti-TNF-α therapies appear to provide novel and safer treatment strategies for refractory ITx rejection; with responses in 14 of 14 patients. Clinical protocols targeting TNF-α, IL-17, and Th17 warrant further testing.
Collapse
Affiliation(s)
- Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Leonid Belyayev
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC,Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Katrina Loh
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC,Department of Gastroenterology, Hepatology and Nutrition, Children’s National Medical Center, Washington, DC
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Anju Duttargi
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Harmeet Dhani
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Mohammed Sadat
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Oswaldo Aguirre
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC
| | - Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC
| | - Bhaskar Kallakury
- Department of Pathology, MedStar Georgetown University Hospital, Washington, DC
| | - Christopher Cosentino
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Brenna Houlihan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Jamie Diaz
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC,Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| | - Sangeetha Moturi
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Nada Yazigi
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Stuart Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Sukanya Subramanian
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Jason Hawksworth
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC,Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| | - Raffaele Girlanda
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Simon C. Robson
- Departments of Anesthesiology and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Cal S. Matsumoto
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Michael Zasloff
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| | - Thomas M. Fishbein
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
24
|
Mycoplasma pneumoniae Infections: Pathogenesis and Vaccine Development. Pathogens 2021; 10:pathogens10020119. [PMID: 33503845 PMCID: PMC7911756 DOI: 10.3390/pathogens10020119] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 12/16/2022] Open
Abstract
Mycoplasma pneumoniae is a major causative agent of community-acquired pneumonia which can lead to both acute upper and lower respiratory tract inflammation, and extrapulmonary syndromes. Refractory pneumonia caused by M. pneumonia can be life-threatening, especially in infants and the elderly. Here, based on a comprehensive review of the scientific literature related to the respective area, we summarize the virulence factors of M. pneumoniae and the major pathogenic mechanisms mediated by the pathogen: adhesion to host cells, direct cytotoxicity against host cells, inflammatory response-induced immune injury, and immune evasion. The increasing rate of macrolide-resistant strains and the harmful side effects of other sensitive antibiotics (e.g., respiratory quinolones and tetracyclines) in young children make it difficult to treat, and increase the health risk or re-infections. Hence, there is an urgent need for development of an effective vaccine to prevent M. pneumoniae infections in children. Various types of M. pneumoniae vaccines have been reported, including whole-cell vaccines (inactivated and live-attenuated vaccines), subunit vaccines (involving M. pneumoniae protein P1, protein P30, protein P116 and CARDS toxin) and DNA vaccines. This narrative review summarizes the key pathogenic mechanisms underlying M. pneumoniae infection and highlights the relevant vaccines that have been developed and their reported effectiveness.
Collapse
|
25
|
Wang Y, Cheng X, Wan C, Wei J, Gao C, Zhang Y, Zeng H, Peng L, Luo P, Lu D, Zou Q, Gu J. Development of a Chimeric Vaccine Against Pseudomonas aeruginosa Based on the Th17-Stimulating Epitopes of PcrV and AmpC. Front Immunol 2021; 11:601601. [PMID: 33552056 PMCID: PMC7859429 DOI: 10.3389/fimmu.2020.601601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022] Open
Abstract
Pulmonary infection caused by Pseudomonas aeruginosa (PA) has created an urgent need for an efficient vaccine, but the protection induced by current candidates is limited, partially because of the high variability of the PA genome. Antigens targeting pulmonary Th17 responses are able to provide antibody-independent and broad-spectrum protection; however, little information about Th17-stimulating antigens in PA is available. Herein, we identified two novel PA antigens that effectively induce Th17-dependent protection, namely, PcrV (PA1706) and AmpC (PA4110). Compared to intramuscular immunization, intranasal immunization enhanced the protection of rePcrV due to activation of a Th17 response. The Th17-stimulating epitopes of PcrV and AmpC were identified, and the recombinant protein PVAC was designed and generated by combining these Th17-stimulating epitopes. PVAC was successfully produced in soluble form and elicited broad protective immunity against PA. Our results provide an alternative strategy for the development of Th17-based vaccines against PA and other pathogens.
Collapse
Affiliation(s)
- Ying Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xin Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chuang Wan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jinning Wei
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chen Gao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yi Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Liusheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jiang Gu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
26
|
Li N, Fan X, Xu M, Zhou Y, Wang B. Flu Virus Attenuates Memory Clearance of Pneumococcus via IFN-γ-Dependent Th17 and Independent Antibody Mechanisms. iScience 2020; 23:101767. [PMID: 33251497 PMCID: PMC7683269 DOI: 10.1016/j.isci.2020.101767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 06/16/2020] [Accepted: 10/30/2020] [Indexed: 12/25/2022] Open
Abstract
Bacterial coinfection is a major cause of influenza-associated mortality. Most people have experienced infections with bacterial pathogens commonly associated with influenza A virus (IAV) coinfection before IAV exposure; however, bacterial clearance through the immunological memory response (IMR) in coinfected patients is inefficient, suggesting that the IMR to bacteria is impaired during IAV infection. Adoptive transfer of CD4+ T cells from mice that had experienced bacterial infection into IAV-infected mice revealed that memory protection against bacteria was weakened in the latter. Additionally, memory Th17 cell responses were impaired due to an IFN-γ-dependent reduction in Th17 cell proliferation and delayed migration of CD4+ T cells into the lungs. A bacterium-specific antibody-mediated memory response was also substantially reduced in coinfected mice, independently of IFN-γ. These findings provide additional perspectives on the pathogenesis of coinfection and suggest additional strategies for the treatment of defective antibacterial immunity and the design of bacterial vaccines against coinfection. Memory protection against bacteria was impaired in coinfection Memory Th17 response to bacteria was reduced by IAV-induced IFN-γ The Th17 reduction was caused by impeded Th17 proliferation and migration Bacteria-specific antibody was reduced in coinfection independent of IFN-γ
Collapse
Affiliation(s)
- Ning Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Fan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meiyi Xu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ya Zhou
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Beinan Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
27
|
Pierce CA, Preston-Hurlburt P, Dai Y, Aschner CB, Cheshenko N, Galen B, Garforth SJ, Herrera NG, Jangra RK, Morano NC, Orner E, Sy S, Chandran K, Dziura J, Almo SC, Ring A, Keller MJ, Herold KC, Herold BC. Immune responses to SARS-CoV-2 infection in hospitalized pediatric and adult patients. Sci Transl Med 2020; 12:scitranslmed.abd5487. [PMID: 32958614 PMCID: PMC7658796 DOI: 10.1126/scitranslmed.abd5487] [Citation(s) in RCA: 261] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022]
Abstract
Compared to adults, young people with COVID-19 have milder disease. Pierce et al. compared immune responses in hospitalized adult and young patients with COVID-19 to identify potential contributing mechanisms. In the first week after hospitalization, circulating IL-17A and IFN-γ concentrations were inversely related to age. More than 3 weeks later, CD4+ T cell responses to viral spike protein were higher in adult compared to younger patients. Neutralizing antibody titers were also higher in adults and correlated positively with age and negatively with IL-17A and IFN-γ. These findings suggest that the poor outcome in adults is not caused by a failure to generate adaptive immune responses. Children and youth infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have milder disease than do adults, and even among those with the recently described multisystem inflammatory syndrome, mortality is rare. The reasons for the differences in clinical manifestations are unknown but suggest that age-dependent factors may modulate the antiviral immune response. We compared cytokine, humoral, and cellular immune responses in pediatric (children and youth, age <24 years) (n = 65) and adult (n = 60) patients with coronavirus disease 2019 (COVID-19) at a metropolitan hospital system in New York City. The pediatric patients had a shorter length of stay, decreased requirement for mechanical ventilation, and lower mortality compared to adults. The serum concentrations of interleukin-17A (IL-17A) and interferon-γ (IFN-γ), but not tumor necrosis factor–α (TNF-α) or IL-6, were inversely related to age. Adults mounted a more robust T cell response to the viral spike protein compared to pediatric patients as evidenced by increased expression of CD25+ on CD4+ T cells and the frequency of IFN-γ+ CD4+ T cells. Moreover, serum neutralizing antibody titers and antibody-dependent cellular phagocytosis were higher in adults compared to pediatric patients with COVID-19. The neutralizing antibody titer correlated positively with age and negatively with IL-17A and IFN-γ serum concentrations. There were no differences in anti-spike protein antibody titers to other human coronaviruses. Together, these findings demonstrate that the poor outcome in hospitalized adults with COVID-19 compared to children may not be attributable to a failure to generate adaptive immune responses.
Collapse
Affiliation(s)
- Carl A Pierce
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Yile Dai
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Clare Burn Aschner
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Natalia Cheshenko
- Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Benjamin Galen
- Department of Medicine, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Scott J Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Natalia G Herrera
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nicholas C Morano
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Erika Orner
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Sharlene Sy
- Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - James Dziura
- Department of Emergency Medicine and Biostatistics, Yale University, New Haven, CT 06520, USA
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Aaron Ring
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Marla J Keller
- Department of Medicine, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA. .,Department of Emergency Medicine and Biostatistics, Yale University, New Haven, CT 06520, USA
| | - Betsy C Herold
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA. .,Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
28
|
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants, and is associated with the development of severe lung inflammation. The pathogenesis of NEC-induced lung injury remains unknown, yet infiltrating immune cells may play a role. In support of this possibility, we now show that NEC in mice and humans was associated with the development of profound lung injury that was characterized by an influx of Th17 cells and a reduction in T regulatory lymphocytes (Tregs). Importantly, the adoptive transfer of CD4 T cells isolated from lungs of mice with NEC into the lungs of immune incompetent mice (Rag1 mice) induced profound inflammation in the lung, while the depletion of Tregs exacerbated NEC induced lung injury, demonstrating that imbalance of Th17/Treg in the lung is required for the induction of injury. In seeking to define the mechanisms involved, the selective deletion of toll-like receptor 4 (TLR4) from the Sftpc1 pulmonary epithelial cells reversed lung injury, while TLR4 activation induced the Th17 recruiting chemokine (C-C motif) ligand 25 (CCL25) in the lungs of mice with NEC. Strikingly, the aerosolized inhibition of both CCL25 and TLR4 and the administration of all trans retinoic acid restored Tregs attenuated NEC-induced lung injury. In summary, we show that TLR4 activation in Surfactant protein C-1 (Sftpc1) cells disrupts the Treg/Th17 balance in the lung via CCL25 leading to lung injury after NEC and reveal that inhibition of TLR4 and stabilization of Th17/Treg balance in the neonatal lung may prevent this devastating complication of NEC.
Collapse
|
29
|
Zhao K, Li R, Wu X, Zhao Y, Wang T, Zheng Z, Zeng S, Ding X, Nie H. Clinical features in 52 patients with COVID-19 who have increased leukocyte count: a retrospective analysis. Eur J Clin Microbiol Infect Dis 2020; 39:2279-2287. [PMID: 32651736 PMCID: PMC7351641 DOI: 10.1007/s10096-020-03976-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022]
Abstract
Recent reports have showed that a proportion of patients with Coronavirus Disease 2019 (COVID-19) presented elevated leukocyte count. Clinical data about these patients is scarce. We aimed to evaluate the clinical findings of patients with COVID-19 who have increased leukocyte at admission. We retrospectively collected the clinical data on the 52 patients who have increased leukocyte count at admission from the 619 patients with confirmed COVID-19 who had pneumonia with abnormal features on chest CT scan in Renmin Hospital of Wuhan University in Wuhan, China, from February 3 to March 3, 2020. The mean age of the 52 patients with increased leukocyte count was 64.7 (SD 11.4) years, 32 (61.5%) were men and 47 (90.4%) had fever. Compared with the patients with non-increased leukocyte count, the patients with increased leukocyte count were significantly older (P < 0.01), were more likely to have underlying chronic diseases (P < 0.01), more likely to develop critically illness (P < 0.01), more likely to admit to an ICU (P < 0.01), more likely to receive mechanical ventilation (P < 0.01), had higher rate of death (P < 0.01) and the blood levels of neutrophil count and the serum concentrations of CRP and IL-6 were significantly increased, (P < 0.01). The older patients with COVID-19 who had underlying chronic disorders are more likely to develop leukocytosis. These patients are more likely to develop critical illness, with a high admission to an ICU and a high mortality rate.
Collapse
Affiliation(s)
- Kaochang Zhao
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Ruiyun Li
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Xiaojun Wu
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Yang Zhao
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Tao Wang
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Zhishui Zheng
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Shaolin Zeng
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Xuhong Ding
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Hanxiang Nie
- Department of Respiratory & Critical Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|
30
|
Xing X, Bi S, Fan X, Jin M, Liu W, Wang B. Intranasal Vaccination With Multiple Virulence Factors Promotes Mucosal Clearance of Streptococcus suis Across Serotypes and Protects Against Meningitis in Mice. J Infect Dis 2020; 220:1679-1687. [PMID: 31287878 DOI: 10.1093/infdis/jiz352] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Streptococcus suis is an emerging zoonotic agent. Its natural habitat is the tonsils, which are the main portals of S. suis entry into the bloodstream of pigs. The remarkable variability of the bacteria and complex pathogenic mechanisms make the development of a vaccine a difficult task. METHOD Five conserved virulence factors involved in critical events of S. suis pathogenesis were combined and used as an intranasal vaccine (V5). The effect of V5 was investigated with intranasal and systemic challenge models. RESULTS V5 induced antibody and T-cell responses at the mucosal site and systemically. The immunity promoted clearance of S. suis from the nasopharynx independent of S. suis serotypes and reduced lethality after systemic challenge with S. suis serotype 2. Moreover, mice that survived sepsis from intravenous infection developed meningitis, whereas none of these mice showed neuropathological symptoms after V5 receipt. CONCLUSION Intranasal immunization with multiple conserved virulence factors decreases S. suis colonization at the nasopharynx across serotypes and inhibits the dissemination of the bacteria in the host. The protective mucosal immunity effects would potentially reduce the S. suis reservoir and prevent S. suis disease in pigs.
Collapse
Affiliation(s)
- Xinxin Xing
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing
| | - Shuai Bi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences
| | - Xin Fan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences
| | - Meilin Jin
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Wenjun Liu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences
| | - Beinan Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences
| |
Collapse
|
31
|
Leveraging -omics for asthma endotyping. J Allergy Clin Immunol 2020; 144:13-23. [PMID: 31277743 DOI: 10.1016/j.jaci.2019.05.015] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
Asthma is a highly heterogeneous disease, often manifesting with wheeze, dyspnea, chest tightness, and cough as prominent symptoms. The eliciting factors, natural history, underlying molecular biology, and clinical management of asthma vary highly among affected subjects. Because of this variation, many efforts have gone into subtyping asthma. Endotypes are subtypes of disease based on distinct pathophysiologic mechanisms. Endotypes can be clinically useful because they organize our mechanistic understanding of heterogeneous diseases and can direct treatment toward modalities that are likely to be the most effective. Asthma endotyping can be shaped by clinical features, laboratory parameters, and/or -omics approaches. We discuss the application of -omics approaches, including transcriptomics, epigenomics, microbiomics, metabolomics, and proteomics, to asthma endotyping. -Omics approaches have provided supporting evidence for many existing endotyping paradigms and also suggested novel ways to conceptualize asthma endotypes. Although endotypes based on single -omics approaches are relatively common, their integrated multi-omics application to asthma endotyping has been more limited thus far. We discuss paths forward to integrate multi-omics with clinical features and laboratory parameters to achieve the goal of precise asthma endotypes.
Collapse
|
32
|
Sakuma M, Khan MAS, Yasuhara S, Martyn JA, Palaniyar N. Mechanism of pulmonary immunosuppression: extrapulmonary burn injury suppresses bacterial endotoxin-induced pulmonary neutrophil recruitment and neutrophil extracellular trap (NET) formation. FASEB J 2019; 33:13602-13616. [PMID: 31577450 PMCID: PMC6894048 DOI: 10.1096/fj.201901098r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Pulmonary immunosuppression often occurs after burn injury (BI). However, the reasons for BI-induced pulmonary immunosuppression are not clearly understood. Neutrophil recruitment and neutrophil extracellular trap (NET) formation (NETosis) are important components of a robust pulmonary immune response, and we hypothesized that pulmonary inflammation and NETosis are defective after BI. To test this hypothesis, we established a mouse model with intranasal LPS instillation in the presence or absence of BI (15% of body surface burn) and determined the degree of immune cell infiltration, NETosis, and the cytokine levels in the airways and blood on d 2. Presence of LPS recruited monocytes and large numbers of neutrophils to the airways and induced NETosis (citrullinated histone H3, DNA, myeloperoxidase). By contrast, BI significantly reduced LPS-mediated leukocyte recruitment and NETosis. This BI-induced immunosuppression is attributable to the reduction of chemokine (C-C motif) ligand (CCL) 2 (monocyte chemoattractant protein 1) and CCL3 (macrophage inflammatory protein 1α). BI also suppressed LPS-induced increase in IL-17A, IL-17C, and IL-17E/IL-25 levels in the airways. Therefore, BI-mediated reduction in leukocyte recruitment and NETosis in the lungs are attributable to these cytokines. Regulating the levels of some of these key cytokines represents a potential therapeutic option for mitigating BI-mediated pulmonary immunosuppression.-Sakuma, M., Khan, M. A. S., Yasuhara, S., Martyn, J. A., Palaniyar, N. Mechanism of pulmonary immunosuppression: extrapulmonary burn injury suppresses bacterial endotoxin-induced pulmonary neutrophil recruitment and neutrophil extracellular trap (NET) formation.
Collapse
Affiliation(s)
- Miyuki Sakuma
- Massachusetts General Hospital and Shriners Hospitals for Children–Boston, Harvard Medical School, Boston, Massachusetts, USA
| | - Mohammed A. S. Khan
- Massachusetts General Hospital and Shriners Hospitals for Children–Boston, Harvard Medical School, Boston, Massachusetts, USA
| | - Shingo Yasuhara
- Massachusetts General Hospital and Shriners Hospitals for Children–Boston, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeevendra A. Martyn
- Massachusetts General Hospital and Shriners Hospitals for Children–Boston, Harvard Medical School, Boston, Massachusetts, USA
| | - Nades Palaniyar
- Massachusetts General Hospital and Shriners Hospitals for Children–Boston, Harvard Medical School, Boston, Massachusetts, USA
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, Institute of Medical Sciences, Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Bedke N, Swindle EJ, Molnar C, Holt PG, Strickland DH, Roberts GC, Morris R, Holgate ST, Davies DE, Blume C. A method for the generation of large numbers of dendritic cells from CD34+ hematopoietic stem cells from cord blood. J Immunol Methods 2019; 477:112703. [PMID: 31711888 PMCID: PMC6983936 DOI: 10.1016/j.jim.2019.112703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) play a central role in regulating innate and adaptive immune responses. It is well accepted that their regulatory functions change over the life course. In order to study DCs function during early life it is important to characterize the function of neonatal DCs. However, the availability of neonatal DCs is limited due to ethical reasons or relative small samples of cord blood making it difficult to perform large-scale experiments. Our aim was to establish a robust protocol for the generation of neonatal DCs from cord blood derived CD34+ hematopoietic stem cells. For the expansion of DC precursor cells we used a cytokine cocktail containing Flt-3 L, SCF, TPO, IL-3 and IL-6. The presence of IL-3 and IL-6 in the first 2 weeks of expansion culture was essential for the proliferation of DC precursor cells expressing CD14. After 4 weeks in culture, CD14+ precursor cells were selected and functional DCs were generated in the presence of GM-CSF and IL-4. Neonatal DCs were then stimulated with Poly(I:C) and LPS to mimic viral or bacterial infections, respectively. Poly(I:C) induced a higher expression of the maturation markers CD80, CD86 and CD40 compared to LPS. In line with literature data using cord blood DCs, our Poly(I:C) matured neonatal DCs cells showed a higher release of IL-12p70 compared to LPS matured neonatal DCs. Additionally, we demonstrated a higher release of IFN-γ, TNF-α, IL-1β and IL-6, but lower release of IL-10 in Poly(I:C) matured compared to LPS matured neonatal DCs derived from cord blood CD34+ hematopoietic stem cells. In summary, we established a robust protocol for the generation of large numbers of functional neonatal DCs. In line with previous studies, we showed that neonatal DCs generated form CD34+ cord blood progenitors have a higher inflammatory potential when exposed to viral than bacterial related stimuli. A robust protocol for the generation of high numbers of neonatal dendritic cells. IL-3 and IL-6 are crucial for the proliferation of cord blood CD34+ progenitors. Neonatal DCs have a higher inflammatory potential when exposed to viral stimuli. LPS induces higher release of IL-10 in neonatal DCs compared to Poly(I:C).
Collapse
Affiliation(s)
- Nicole Bedke
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Emily J Swindle
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Camelia Molnar
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Patrick G Holt
- Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, Perth, Australia
| | - Deborah H Strickland
- Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, Perth, Australia
| | - Graham C Roberts
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ruth Morris
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Stephen T Holgate
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Donna E Davies
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Cornelia Blume
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
34
|
Majd AMM, Faghihzadeh S, Pourfarzam S, Eghtedardoost M, Jamali D, Mirsharif ES, Dilmaghanian R, Ghazanfari T. Serum and sputum levels of IL-17, IL-21, TNFα and mRNA expression of IL-17 in sulfur mustard lung tissue with long term pulmonary complications (28 years after sulfur mustard exposure). Int Immunopharmacol 2019; 76:105828. [DOI: 10.1016/j.intimp.2019.105828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 01/16/2023]
|
35
|
Yang JQ, Kalim KW, Li Y, Zheng Y, Guo F. Ablation of RhoA impairs Th17 cell differentiation and alleviates house dust mite-triggered allergic airway inflammation. J Leukoc Biol 2019; 106:1139-1151. [PMID: 31260596 DOI: 10.1002/jlb.3a0119-025rrr] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 01/10/2023] Open
Abstract
Asthma is a heterogeneous chronic airway inflammation in which Th2 and Th17 cells are key players in its pathogenesis. We have reported that RhoA of Rho GTPases orchestrated glycolysis for Th2 cell differentiation and allergic airway inflammation by the use of a conditional RhoA-deficient mouse line. However, the role of RhoA in Th17 cells remains to be elucidated. In this study, we investigated the effects of RhoA deficiency on Th17 cells in the context of ex vivo cell culture systems and an in vivo house dust mites (HDM)-induced allergic airway inflammation. We found that RhoA deficiency inhibited Th17 differentiation and effector cytokine secretion, which was associated with the downregulations of Stat3 and Rorγt, key Th17 transcription factors. Furthermore, loss of RhoA markedly suppressed Th17 and neutrophil-involved airway inflammation induced by HDM in mice. The infiltrating inflammatory cells in the lungs and bronchoalveolar lavage (BAL) fluids were dramatically reduced in conditional RhoA-deficient mice. Th17 as well as Th2 effector cytokines were suppressed in the airways at both protein and mRNA levels. Interestingly, Y16, a specific RhoA inhibitor, was able to recapitulate the most phenotypes of RhoA genetic deletion in Th17 differentiation and allergic airway inflammation. Our data demonstrate that RhoA is a key regulator of Th17 cell differentiation and function. RhoA might serve as a potential novel therapeutic target for asthma and other inflammatory disorders.
Collapse
Affiliation(s)
- Jun-Qi Yang
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasitic and Vector Control, Jiangsu Institute of Parasitic Diseases and Public Health Research Center, Jiangnan University, Wuxi, Jiangsu, China
| | - Khalid W Kalim
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yuan Li
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
36
|
Associations Between Inflammatory Endotypes and Clinical Presentations in Chronic Rhinosinusitis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 7:2812-2820.e3. [PMID: 31128376 DOI: 10.1016/j.jaip.2019.05.009] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is a heterogeneous disease characterized by mucosal inflammation in the nose and paranasal sinuses. Inflammation in CRS is also heterogeneous and is mainly characterized by type 2 (T2) inflammation, but subsets of patients show type 1 (T1) and type 3 (T3) inflammation. Whether inflammatory endotypes are associated with clinical phenotypes has yet to be explored in detail. OBJECTIVE To identify associations between inflammatory endotypes and clinical presentations in CRS. METHODS We compared 121 patients with nonpolypoid CRS (CRSsNP) and 134 patients with polypoid CRS (CRSwNP) and identified inflammatory endotypes using markers including IFN-γ (T1), eosinophil cationic protein (T2), Charcot-Leyden crystal galectin (T2), and IL-17A (T3). We collected clinical parameters from medical and surgical records and examined whether there were any associations between endotype and clinical features. RESULTS The presence of nasal polyps, asthma comorbidity, smell loss, and allergic mucin was significantly associated with the presence of T2 endotype in all patients with CRS. The T1 endotype was significantly more common in females, and the presence of pus was significantly associated with T3 endotype in all patients with CRS. We further analyzed these associations in CRSsNP and CRSwNP separately and found that smell loss was still associated with T2 endotype and pus with the T3 endotype in both CRSsNP and CRSwNP. Importantly, patients with CRS with T2 and T3 mixed endotype tended to have clinical presentations shared by both T2 and T3 endotypes. CONCLUSIONS Clinical presentations are directly associated with inflammatory endotypes in CRS. Identification of inflammatory endotypes may allow for more precise and personalized medical treatments in CRS.
Collapse
|
37
|
Östling J, van Geest M, Schofield JPR, Jevnikar Z, Wilson S, Ward J, Lutter R, Shaw DE, Bakke PS, Caruso M, Dahlen SE, Fowler SJ, Horváth I, Krug N, Montuschi P, Sanak M, Sandström T, Sun K, Pandis I, Auffray C, Sousa AR, Guo Y, Adcock IM, Howarth P, Chung KF, Bigler J, Sterk PJ, Skipp PJ, Djukanović R, Vaarala O. IL-17-high asthma with features of a psoriasis immunophenotype. J Allergy Clin Immunol 2019; 144:1198-1213. [PMID: 30998987 DOI: 10.1016/j.jaci.2019.03.027] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 03/06/2019] [Accepted: 03/18/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The role of IL-17 immunity is well established in patients with inflammatory diseases, such as psoriasis and inflammatory bowel disease, but not in asthmatic patients, in whom further study is required. OBJECTIVE We sought to undertake a deep phenotyping study of asthmatic patients with upregulated IL-17 immunity. METHODS Whole-genome transcriptomic analysis was performed by using epithelial brushings, bronchial biopsy specimens (91 asthmatic patients and 46 healthy control subjects), and whole blood samples (n = 498) from the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes (U-BIOPRED) cohort. Gene signatures induced in vitro by IL-17 and IL-13 in bronchial epithelial cells were used to identify patients with IL-17-high and IL-13-high asthma phenotypes. RESULTS Twenty-two of 91 patients were identified with IL-17, and 9 patients were identified with IL-13 gene signatures. The patients with IL-17-high asthma were characterized by risk of frequent exacerbations, airway (sputum and mucosal) neutrophilia, decreased lung microbiota diversity, and urinary biomarker evidence of activation of the thromboxane B2 pathway. In pathway analysis the differentially expressed genes in patients with IL-17-high asthma were shared with those reported as altered in psoriasis lesions and included genes regulating epithelial barrier function and defense mechanisms, such as IL1B, IL6, IL8, and β-defensin. CONCLUSION The IL-17-high asthma phenotype, characterized by bronchial epithelial dysfunction and upregulated antimicrobial and inflammatory response, resembles the immunophenotype of psoriasis, including activation of the thromboxane B2 pathway, which should be considered a biomarker for this phenotype in further studies, including clinical trials targeting IL-17.
Collapse
Affiliation(s)
- Jörgen Östling
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Marleen van Geest
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - James P R Schofield
- Centre for Proteomic Research, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Zala Jevnikar
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Susan Wilson
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom; Histochemistry Research Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jonathan Ward
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Rene Lutter
- AUMC, Department of Experimental Immunology, University of Amsterdam, Amsterdam, The Netherlands; AUMC, Department of Respiratory Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Dominick E Shaw
- Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom
| | - Per S Bakke
- Institute of Medicine, University of Bergen, Bergen, Norway
| | - Massimo Caruso
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sven-Erik Dahlen
- Centre for Allergy Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephen J Fowler
- Respiratory and Allergy Research Group, University of Manchester, Manchester, United Kingdom
| | - Ildikó Horváth
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine Hannover, Hannover, Germany
| | - Paolo Montuschi
- Faculty of Medicine, Catholic University of the Sacred Heart, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Marek Sanak
- Laboratory of Molecular Biology and Clinical Genetics, Medical College, Jagiellonian University, Krakow, Poland
| | - Thomas Sandström
- Department of Medicine, Department of Public Health and Clinical Medicine Respiratory Medicine Unit, Umeå University, Umeå, Sweden
| | - Kai Sun
- Data Science Institute, Imperial College, London, United Kingdom
| | - Ioannis Pandis
- Data Science Institute, Imperial College, London, United Kingdom
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Université de Lyon, Lyon, France
| | - Ana R Sousa
- Respiratory Therapeutic Unit, GlaxoSmithKline, Stockley Park, United Kingdom
| | - Yike Guo
- Data Science Institute, Imperial College, London, United Kingdom
| | - Ian M Adcock
- Experimental Studies, Airways Disease Section, National Heart & Lung institute, Imperial College London, London, United Kingdom
| | - Peter Howarth
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Kian Fan Chung
- Experimental Studies, Airways Disease Section, National Heart & Lung institute, Imperial College London, London, United Kingdom
| | | | - Peter J Sterk
- AUMC, Department of Respiratory Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Skipp
- Centre for Proteomic Research, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Ratko Djukanović
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom.
| | - Outi Vaarala
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
38
|
Immunodeficiency in Patients with Acute Exacerbation of Chronic Obstructive Pulmonary Disease. Inflammation 2019; 41:1582-1589. [PMID: 30047000 DOI: 10.1007/s10753-018-0830-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD), characterized by progressive airway inflammation and irreversible airflow limitation, leads to serious decline in life quality. The acute exacerbation of COPD (AECOPD) results in high healthcare costs as well as a significant mortality rate. The most common cause of acute exacerbation is infection. Immune deficiency, which induces dysfunction of anti-infection, plays an important role in the pathogenesis of acute exacerbation. As described in this review, the immune dysfunction in patients with AECOPD can be a major focus of efforts to therapeutic strategy.
Collapse
|
39
|
Jafarzadeh A, Nemati M. Therapeutic potentials of ginger for treatment of Multiple sclerosis: A review with emphasis on its immunomodulatory, anti-inflammatory and anti-oxidative properties. J Neuroimmunol 2018; 324:54-75. [PMID: 30243185 DOI: 10.1016/j.jneuroim.2018.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/24/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is characterized by chronic inflammatory response-induced demyelination of the neurons and degeneration of the axons within the central nervous system (CNS). A complex network of immunopathological-, inflammatory- and oxidative parameters involve in the development and advancement of MS. The anti-inflammatory, immunomodulatory and anti-oxidative characteristics of the ginger and several of its components have been indicated in some of experimental and clinical investigations. The possible therapeutic potentials of ginger and its ingredients in the treatment of MS may exert mainly through the regulation of the Th1-, Th2-, Th9-, Th17-, Th22- and Treg cell-related immune responses, down-regulation of the B cell-related immune responses, modulation of the macrophages-related responses, modulation of the production of pro- and anti-inflammatory cytokines, down-regulation of the arachidonic acid-derived mediators, interfering with the toll like receptor-related signaling pathways, suppression of the inflammasomes, down-regulation of the oxidative stress, reduction of the adhesion molecules expression, and down-regulation of the expression of the chemokines and chemokine receptors. This review aimed to provide a comprehensive knowledge regarding the immunomodulatory-, anti-inflammatory and anti-oxidative properties of ginger and its components, and highlight novel insights into the possible therapeutic potentials of this plant for treatment of MS. The review encourages more investigations to consider the therapeutic potentials of ginger and its effective components for managing of MS.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Maryam Nemati
- Department of Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
40
|
Liu C, Pan X, Xia B, Chen F, Jin Y, Bai F, Priebe G, Cheng Z, Jin S, Wu W. Construction of a Protective Vaccine Against Lipopolysaccharide-Heterologous Pseudomonas aeruginosa Strains Based on Expression Profiling of Outer Membrane Proteins During Infection. Front Immunol 2018; 9:1737. [PMID: 30093906 PMCID: PMC6070602 DOI: 10.3389/fimmu.2018.01737] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/13/2018] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous opportunistic pathogen, which causes infectious disease in patients with cystic fibrosis and compromised immunity. P. aeruginosa is difficult to eradicate because of its intrinsic resistance to most traditional antibiotics as well as acquired resistance mechanisms after decades of antibiotic usage. A full understanding of the P. aeruginosa pathogenesis mechanisms is necessary for the development of novel prevention and treatment strategies. To identify novel vaccine candidates, here we comprehensively examined the expression levels of all the known outer membrane proteins in two P. aeruginosa strains in a murine acute pneumonia model. OprH was one of the most highly expressed proteins during infection. In addition, OprH is known to be highly immunogenic and accessible by host proteins. Thus, it was chosen as a vaccine candidate. To further identify vaccine candidates, 34 genes highly expressed during infection were evaluated for their contributions in virulence by testing individual transposon insertion mutants. Among them, fpvA, hasR, and foxA were found essential for bacterial virulence and therefore included in vaccine construction. Immunization with a mixture of FpvA, HasR, and FoxA rendered no protection, however, while immunization by OprH refolded in liposomes elicited specific opsonic antibodies and conferred protection against two lipopolysaccharide-heterologous P. aeruginosa strains (PA14 and PA103). Overall, by studying the expression profile of the P. aeruginosa outer membrane proteins during infection, we identified OprH as a potential vaccine candidate for the prevention of lung infection by P. aeruginosa.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Bin Xia
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Fei Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Gregory Priebe
- Division of Critical Care Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Anaesthesia, Harvard Medical School, Boston, MA, United States
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouguang Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China.,Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
41
|
De Luca A, Pariano M, Cellini B, Costantini C, Villella VR, Jose SS, Palmieri M, Borghi M, Galosi C, Paolicelli G, Maiuri L, Fric J, Zelante T. The IL-17F/IL-17RC Axis Promotes Respiratory Allergy in the Proximal Airways. Cell Rep 2018; 20:1667-1680. [PMID: 28813677 DOI: 10.1016/j.celrep.2017.07.063] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/17/2017] [Accepted: 07/23/2017] [Indexed: 12/13/2022] Open
Abstract
The interleukin 17 (IL-17) cytokine and receptor family is central to antimicrobial resistance and inflammation in the lung. Mice lacking IL-17A, IL-17F, or the IL-17RA subunit were compared with wild-type mice for susceptibility to airway inflammation in models of infection and allergy. Signaling through IL-17RA was required for efficient microbial clearance and prevention of allergy; in the absence of IL-17RA, signaling through IL-17RC on epithelial cells, predominantly by IL-17F, significantly exacerbated lower airway Aspergillus or Pseudomonas infection and allergic airway inflammation. In contrast, following infection with the upper respiratory pathogen Staphylococcus aureus, the IL-17F/IL-17RC axis mediated protection. Thus, IL-17A and IL-17F exert distinct biological effects during pulmonary infection; the IL-17F/IL-17RC signaling axis has the potential to significantly worsen pathogen-associated inflammation of the lower respiratory tract in particular, and should be investigated further as a therapeutic target for treating pathological inflammation in the lung.
Collapse
Affiliation(s)
- Antonella De Luca
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Barbara Cellini
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Valeria Rachela Villella
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Shyam Sushama Jose
- Center for Translational Medicine, International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic
| | - Melissa Palmieri
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Claudia Galosi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Giuseppe Paolicelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy; Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Jan Fric
- Center for Translational Medicine, International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic
| | - Teresa Zelante
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
42
|
Magiri R, Lai K, Chaffey A, Zhou Y, Pyo HM, Gerdts V, Wilson HL, Mutwiri G. Intradermal immunization with inactivated swine influenza virus and adjuvant polydi(sodium carboxylatoethylphenoxy)phosphazene (PCEP) induced humoral and cell-mediated immunity and reduced lung viral titres in pigs. Vaccine 2018; 36:1606-1613. [PMID: 29454517 DOI: 10.1016/j.vaccine.2018.02.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/16/2017] [Accepted: 02/02/2018] [Indexed: 12/15/2022]
Abstract
Swine influenza virus is endemic worldwide and it is responsible for significant economic losses to the swine industry. A vaccine that stimulates a rapid and long-lasting protective immune response to prevent this infection is highly sought. Poly[di(sodium carboxylatoethylphenoxy)-phosphazene (PCEP) has demonstrated adjuvant activity when formulated as part of multiple vaccines in mice and pigs. In this study we examined the magnitude and type of immune response induced in pigs vaccinated via the intramuscular or intradermal routes with inactivated swine influenza virus (SIV) H1N1 vaccine formulated with PCEP. Intradermal administration of PCEP-adjuvanted inactivated SIV vaccine stimulated significant anti-SIV antibody titres, increased neutralizing antibodies, and significantly reduced lung virus load with limited reduction of gross lung lesions after challenge with virulent H1N1 relative to control animals. These results indicate that PCEP may be effective as a vaccine adjuvant against swine influenza viruses in pigs and should be considered a potential candidate adjuvant for future swine intradermal influenza vaccines.
Collapse
Affiliation(s)
- Royford Magiri
- Vaccinology & Immunotherapeutic Program, School of Public Health at the University of Saskatchewan, Canada; Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada
| | - Ken Lai
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada
| | - Alyssa Chaffey
- Vaccinology & Immunotherapeutic Program, School of Public Health at the University of Saskatchewan, Canada; Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada
| | - Yan Zhou
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada
| | - Hyun-Mi Pyo
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada
| | - Volker Gerdts
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada
| | - Heather L Wilson
- Vaccinology & Immunotherapeutic Program, School of Public Health at the University of Saskatchewan, Canada; Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada.
| | - George Mutwiri
- Vaccinology & Immunotherapeutic Program, School of Public Health at the University of Saskatchewan, Canada; Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Canada
| |
Collapse
|
43
|
Jafarzadeh A, Larussa T, Nemati M, Jalapour S. T cell subsets play an important role in the determination of the clinical outcome of Helicobacter pylori infection. Microb Pathog 2018; 116:227-236. [PMID: 29407232 DOI: 10.1016/j.micpath.2018.01.040] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/20/2018] [Accepted: 01/26/2018] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori (H. pylori) is one of the most prevalent human pathogen and a persistent infection with this bacterium causes common pathologies, such as gastritis or peptic ulcers, and also less common but more serious pathologies, such as gastric cancer or gastric mucosa-associated lymphoid tissue (MALT) lymphoma. The clinical outcome of gastrointestinal infection sustained by H. pylori is determined by the reciprocal interactions between virulence factors of the bacterium and host factors, including immune response genes. Although H. pylori induces a strong immune response, the bacterium is not eliminated. The eradication failure could be attributed to the bacterial capability to regulate helper T (Th) cell-related responses. H. pylori specific CD4+ T cells play a fundamental role in regulating host immunity and immunopathologic events. It has been documented that Th1, Th2, Th9, Th17, Th22 and T regulatory (Treg) cells, separately or in coordination with each other, can affect the outcome of the infection sustained by of H. pylori. Some studies indicated that both Th1 and Th17 cells may be protective or pathogenic, whereas Treg and Th2 cells perform anti-inflammatory impacts during H. pylori infection. This review gathers recent information regarding the association of the CD4+ T cells-mediated immunological responses and the clinical consequence of H. pylori infection.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Tiziana Larussa
- Department of Health Science, University of Catanzaro "Magna Graecia", 88100 Catanzaro, Italy
| | - Maryam Nemati
- Department of Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shila Jalapour
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
44
|
Abstract
Whooping cough is a highly contagious, acute respiratory disease, caused by the Gram-negative bacterium Bordetella pertussis (Bp). Despite the introduction and widespread use of vaccines starting in the 1950s pertussis cases continue to be reported, with a significant global impact. The role of specific virulence factors in disease and the immune mechanisms associated with protection following natural infection or vaccination are still not completely understood. The recently-developed baboon model of clinical pertussis provides a valuable tool for the study of pertussis. Baboons infected with B. pertussis exhibit all of the manifestations of human pertussis including paroxysmal coughing, mucus production, leukocytosis and transmission. The establishment of this model provides the opportunity to address unanswered questions about the natural progression of this disease and host responses to infection and vaccination in a very relevant model. In this review, we present an overview of our knowledge of pertussis along with recent advances resulting from use of the baboon model. Remaining questions and future research directions are discussed. We hope that the knowledge gained through use of the baboon model of pertussis and clinical studies will allow the development of more efficacious vaccines, conferring long lasting protection against disease and transmission.
Collapse
Affiliation(s)
- Marta V Pinto
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Tod J Merkel
- Laboratory of Respiratory and Special Pathogens, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA.
| |
Collapse
|
45
|
Abstract
Currently used vaccines have had major effects on eliminating common infections, largely by duplicating the immune responses induced by natural infections. Now vaccinology faces more complex problems, such as waning antibody, immunosenescence, evasion of immunity by the pathogen, deviation of immunity by the microbiome, induction of inhibitory responses, and complexity of the antigens required for protection. Fortunately, vaccine development is now incorporating knowledge from immunology, structural biology, systems biology and synthetic chemistry to meet these challenges. In addition, international organisations are developing new funding and licensing pathways for vaccines aimed at pathogens with epidemic potential that emerge from tropical areas.
Collapse
Affiliation(s)
| | - Marta V Pinto
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Manish Sadarangani
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver BC, Canada.
| | - Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
46
|
Nemati M, Malla N, Yadav M, Khorramdelazad H, Jafarzadeh A. Humoral and T cell-mediated immune response against trichomoniasis. Parasite Immunol 2018; 40. [PMID: 29266263 DOI: 10.1111/pim.12510] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022]
Abstract
Trichomonas vaginalis (T. vaginalis) infection leads to the synthesis of specific antibodies in the serum and local secretions. The profile of T. vaginalis-specific antibodies and T cell-mediated immune responses may influence the outcome of infection, towards parasite elimination, persistence or pathological reactions. Studies have indicated that Th1-, Th17- and Th22 cell-related cytokines may be protective or pathogenic, whereas Th2- and Treg cell-related cytokines can exert anti-inflammatory effects during T. vaginalis infection. A number of T. vaginalis-related components such as lipophosphoglycan (TvLPG), α-actinin, migration inhibitory factor (TvMIF), pyruvate:ferredoxin oxidoreductase (PFO), legumain-1 (TvLEGU-1), adhesins and cysteine proteases lead to the induction of specific antibodies. T. vaginalis has acquired several strategies to evade the humoral immune responses such as degradation of immunoglobulins by cysteine proteases, antigenic variation and killing of antibody-producing B cells. The characterization of the T. vaginalis-specific antibodies to significant immunogenic molecules and formulation of strategies to promote their induction in vaginal mucosa may reveal their potential protective effects against trichomoniasis. In this review, we discuss the current understanding of antibody and T cell-mediated immune responses to T. vaginalis and highlight novel insights into the possible role of immune responses in protection against parasite.
Collapse
Affiliation(s)
- M Nemati
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Department of Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - N Malla
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - M Yadav
- Dr. B. R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - H Khorramdelazad
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - A Jafarzadeh
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
47
|
Xue J, Wang Y, Chen C, Zhu X, Zhu H, Hu Y. Effects of Th17 cells and IL-17 in the progression of cervical carcinogenesis with high-risk human papillomavirus infection. Cancer Med 2017; 7:297-306. [PMID: 29277958 PMCID: PMC5806118 DOI: 10.1002/cam4.1279] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 01/08/2023] Open
Abstract
The existence of Th17 cells and IL‐17 was recently shown in several types of infectious diseases, but their distribution and functions in cervical lesions with high‐risk human papillomavirus (HPV) infection have not been fully elucidated. In this study, the frequency of Th17 cells in peripheral blood samples obtained from 28 cervical squamous cell carcinoma patients, 26 CIN1 patients, 30 CIN2 patients, 29 CIN3 patients, 25 high‐risk HPV‐infected women with normal cervical cytology, and 30 healthy controls was determined by flow cytometry. Besides, the levels of IL‐17 in peripheral blood samples as well as in supernatant of cervical tissue homogenate were assessed by enzyme‐linked immunosorbent assay (ELISA) simultaneously. We found that during the disease progression of cervical lesions, the proportion of Th17 cells in the total CD4+ cells showed a gradually increased tendency compared with the controls (P < 0.05). Moreover, levels of IL‐17 in serum and supernatant of cervical tissue homogenate showed the same tendency as the proportion of Th17 cells (P < 0.05). When compared in pairs, the levels of IL‐17 in supernatant differed significantly among the study groups and the control group (P < 0.05), but no significant difference was observed in serum (P > 0.05). In conclusions, the results indicate that Th17 cells and IL‐17 may play a role of immune enhancement in the infection of high‐risk HPV especially in the cervical microenvironment, which contribute to the disease progression of its associated cervical lesions.
Collapse
Affiliation(s)
- JiSen Xue
- The Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - YuLi Wang
- The Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Cheng Chen
- The Department of Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - XueJie Zhu
- The Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Hua Zhu
- The Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yan Hu
- The Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
48
|
Le Rouzic O, Pichavant M, Frealle E, Guillon A, Si-Tahar M, Gosset P. Th17 cytokines: novel potential therapeutic targets for COPD pathogenesis and exacerbations. Eur Respir J 2017; 50:1602434. [PMID: 29025886 DOI: 10.1183/13993003.02434-2016] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/14/2017] [Indexed: 12/31/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease of the airways caused mainly by cigarette smoke exposure. COPD progression is marked by exacerbations of the disease, often associated with infections. Recent data show the involvement in COPD pathophysiology of interleukin (IL)-17 and IL-22, two cytokines that are important in the control of lung inflammation and infection. During the initiation and progression of the disease, increased IL-17 secretion causes neutrophil recruitment, leading to chronic inflammation, airways obstruction and emphysema. In the established phase of COPD, a defective IL-22 response facilitates pathogen-associated infections and disease exacerbations. Altered production of these cytokines involves a complex network of immune cells and dysfunction of antigen-presenting cells. In this review, we describe current knowledge on the involvement of IL-17 and IL-22 in COPD pathophysiology at steady state and during exacerbations, and discuss implications for COPD management and future therapeutic approaches.
Collapse
Affiliation(s)
- Olivier Le Rouzic
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
- Service de Pneumologie Immunologie et Allergologie, CHU Lille, Lille, France
| | - Muriel Pichavant
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Emilie Frealle
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
- Laboratoire de Parasitologie et Mycologie Médicale, CHU Lille, Lille, France
| | - Antoine Guillon
- Service de Réanimation Polyvalente, CHRU de Tours, Tours, France
- Inserm, U1100 - Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université François Rabelais, Tours, France
| | - Mustapha Si-Tahar
- Inserm, U1100 - Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université François Rabelais, Tours, France
| | - Philippe Gosset
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
| |
Collapse
|
49
|
Vaccination with a recombinant OprL fragment induces a Th17 response and confers serotype-independent protection against Pseudomonas aeruginosa infection in mice. Clin Immunol 2017; 183:354-363. [PMID: 28970186 DOI: 10.1016/j.clim.2017.09.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/06/2017] [Accepted: 09/25/2017] [Indexed: 01/06/2023]
Abstract
Pseudomonas aeruginosa (PA) is the major causative agent of nosocomial infection. Despite of adequate use of antibiotics, it still represents a major challenge in controlling PA infection. The local pulmonary Th17 response plays an important protective role against PA infection. And the Th17-mediated protection is antibody independent, so we hypothesized that it would be an optimal strategy of a vaccine for PA control to induce an effective Th17 response. Herein we report the successful production of a recombinant fragment of the OprL (reOprL) of PA. Purified reOprL forms homogeneous monomers in solution and vaccination with reOprL elicited a remarkable Th17 response. In addition, reOprL vaccination conferred effective serotype-independent protection against PA infection, which relied on the Th17 response. Our data suggest that reOprL is a good candidate for the future development of Th17 immunity based PA vaccines.
Collapse
|
50
|
Yang AJ, Xi K, Yao YQ, Ding YX, Yang LJ, Cui H. Effect of Qingfei Mixture () on pediatric mycoplasma pneumoniae pneumonia with phlegm heat obstructing Fei (Lung) syndrome. Chin J Integr Med 2017; 23:681-688. [PMID: 28861888 DOI: 10.1007/s11655-017-2409-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore the effect and mechanism of Qingfei Mixture (), a Chinese medicine, in treating mycoplasma pneumonia (MP) in MP patients and rat model METHODS: A total of 46 MP children with phlegm heat obstructing Fei (Lung) syndrome were randomly assigned to two groups by the method of random number table, with 23 children in each group. The control group was treated with intravenous infusion of azithromycin; the treatment group received intravenous infusion of azithromycin and oral administration of Qingfei Mixture. The treatment course was 7 days. Major symptoms and minor symptoms were observed and scored before and after treatments. A rat model of MP was also established. A total of 120 wistar rats were randomly divided into 5 groups: a normal group, infection group, Qingfei Mixture treatment group, azithromycin treatment group, and Qingfei Mixture + azithromycin treatment group. Each group contained 24 rats, from which every 6 were euthanatized 1, 3, 7 and 14 days after infection. MP DNA in pulmonary tissue homogenates was detected using real-time fluorescence quantitative polymerase chain reaction. Pathology was assessed after hematoxylin (HE) staining and lung tissue pathology scores were determined in pulmonary tissue. Transmission electron microscopic detection and electronic image analysis were performed on lung tissue 3 days after infection. Interleukin (IL)-17 was detected in serum using enzymelinked immunosorbent assay (ELISA) 7 days after infection. RESULTS In the clinical study, both control and the treatment group showed improved results on removing symptoms of phlegm heat syndrome compared to the control group (P<0.05). In animal experiments, On the 7th day after MP infection, as detected by electron microscopy, the pulmonary capillary basement membranes of the azithromycin + Qingfei Mixture treatment group were much thinner than those of the azithromycin or Qingfei mixture treatment groups (P<0.05). The level of serum IL-17 in the azithromycin + Qingfei Mixture treatment group was lower than that in the azithromycin or Qingfei Mixture groups (P<0.01). CONCLUSION Both Qingfei Mixture and azithromycin have therapeutic effects on mycoplasma pneumoniae pneumonia, but the combination of both agents had the greatest effect.
Collapse
Affiliation(s)
- Ai-Jun Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Kun Xi
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yan-Qing Yao
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying-Xue Ding
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Li-Jun Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Hong Cui
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|