1
|
Chen Y, Wang X, Chen S, Zhang M, Cheng Z, Zhang W, Liu D, Shan Y, Du G, Li W, Yang L, Wang J, Chu Y, Liu M. Evaluation of immune effect to recombinant potential protective antigens of Mycoplasma ovipneumoniae in mice. Microb Pathog 2025; 204:107555. [PMID: 40203960 DOI: 10.1016/j.micpath.2025.107555] [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: 11/20/2024] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Mycoplasma ovipneumoniae is a primary causative agent of pneumonia in ruminants, causing chronic non-progressive pneumonia in domestic sheep and goats, but leading to higher morbidity and mortality in bighorn sheep and wild small ruminants. This disease has become a widespread epidemic, resulting in significant losses to the sheep industry. In this study, we evaluated the immunogenicity and initial protective effects of four antigenic proteins of M. ovipneumoniae, namely Eno, EF-Tu, Ulad, and T4SS. These proteins were used to immunize BALB/c mice either individually or in a combination (rProteins group). The mice were intranasally infected with 109 CCU50/mL M. ovipneumoniae strain NJ01 twice, on days 28 and 30 after immunization. Among the four recombinant proteins, rEno demonstrated the most promising results in terms of inducing specific humoral and cellular immune responses. It also resulted in the lowest lung lesion scores and the lowest M. ovipneumoniae loads in the lungs and bronchoalveolar lavage fluid (BALF). Compared to the other three proteins, rEno provided superior protection. Furthermore, the rEno vaccine significantly reduced the inflammatory response in the lungs of mice, as evidenced by the evaluation of pro-inflammatory cytokines. The expression of IL-1β and NF-κB was significantly reduced, while the expression of IL-4 was significantly increased. In conclusion, the rEno vaccine elicited a favorable immunological response and conferred protection against M. ovipneumoniae. This finding presents a novel approach to controlling the global spread of this pathogen.
Collapse
MESH Headings
- Animals
- Mice, Inbred BALB C
- Mycoplasma ovipneumoniae/immunology
- Mycoplasma ovipneumoniae/genetics
- Pneumonia, Mycoplasma/prevention & control
- Pneumonia, Mycoplasma/immunology
- Pneumonia, Mycoplasma/microbiology
- Pneumonia, Mycoplasma/pathology
- Mice
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Cytokines/metabolism
- Lung/pathology
- Lung/microbiology
- Lung/immunology
- Recombinant Proteins/immunology
- Recombinant Proteins/genetics
- Antibodies, Bacterial/blood
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Bronchoalveolar Lavage Fluid/microbiology
- Female
- Immunity, Cellular
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Disease Models, Animal
- NF-kappa B/metabolism
- Interleukin-1beta/metabolism
- Immunity, Humoral
- Sheep
- Immunization
Collapse
Affiliation(s)
- Yi Chen
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Xiaonan Wang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Siyu Chen
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Mengjie Zhang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Zilong Cheng
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Wenwen Zhang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Diyue Liu
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China
| | - Yiyi Shan
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Gaimei Du
- College of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, 211169, PR China
| | - Wenliang Li
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Leilei Yang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, PR China
| | - Maojun Liu
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China.
| |
Collapse
|
2
|
Saha S, Sachivkina N, Kuznetsova O, Neborak E, Zhabo N. Elucidating the Role of Nrf2 Signaling Pathway in Mycoplasma Infections. FRONT BIOSCI-LANDMRK 2025; 30:28286. [PMID: 40302330 DOI: 10.31083/fbl28286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 05/02/2025]
Abstract
Mycoplasmas are the smallest cell-wall-less self-replicating prokaryotes. Mycoplasma species can be found within and outside cells as "silent parasites" that live intracellularly and as membrane surface parasites. The pathogen's impact on respiratory health seems primarily caused by its capacity to alter immune responses, cause airway inflammation, and damage epithelial barriers. Much progress has been made in understanding Mycoplasma-induced inflammation and oxidative stress. However, there are still issues in therapeutic management, such as the development of strains that are resistant to antibiotics, the shortcomings of the available diagnostic techniques, and possible long-term respiratory consequences. On the other hand, to combat oxidative stress, inflammation, and metabolic abnormalities, activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is becoming a more appealing therapeutic strategy. Nrf2 activation coordinates a thorough defense through its transcriptional targets, enabling adaptability and survival under a variety of cellular stressors. Nrf2 is regarded as a therapeutic target, and pharmacological Nrf2 activators have demonstrated protective effects in multiple pathological consequences and advantages in clinical trials. In this review, we discussed the rationale for targeting Nrf2 in a series of inflammatory responses caused by Mycoplasma species.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, 281406 Mathura, Uttar Pradesh, India
| | - Nadezhda Sachivkina
- Department of Microbiology V.S. Kiktenko, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Olga Kuznetsova
- Department of Biochemistry T.T. Berezov, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Neborak
- Department of Biochemistry T.T. Berezov, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Natallia Zhabo
- Department of Foreign Languages, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
3
|
Hensley MK, Dela Cruz CS. Host-Directed Adjunctive Therapies in Immunocompromised Patients with Pneumonia. Clin Chest Med 2025; 46:37-48. [PMID: 39890291 DOI: 10.1016/j.ccm.2024.10.003] [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] [Indexed: 02/03/2025]
Abstract
Immunocompromised (IC) hosts represent a unique patient population at risk for not only typical pathogens, but also opportunistic microorganisms. While antimicrobials remain the main treatment, new investigations have demonstrated the importance of host-response to pathogens. In this article, we highlight previously discovered and new areas of investigation for adjunctive host-response treatments for IC host pneumonia.
Collapse
Affiliation(s)
- Matthew K Hensley
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Charles S Dela Cruz
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Fan L, Xu N, Guo Y, Li L. Enhanced insights into the neutrophil-driven immune mechanisms during Mycoplasma pneumoniae infection. Heliyon 2024; 10:e38950. [PMID: 39524902 PMCID: PMC11550053 DOI: 10.1016/j.heliyon.2024.e38950] [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: 05/15/2024] [Revised: 09/10/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Mycoplasma pneumoniae (MP) infections represent a significant component of community-acquired pneumonia, especially in children, invoking a complex neutrophil-mediated immune response, crucial for host defense. This review consolidates current knowledge on the role of neutrophils in MP infection, focusing on their recruitment, migration and activation, as well as the molecular mechanisms underpinning these processes. Significant findings indicate that specific bacterial components, notably CARDS toxin and lipoproteins, intensify neutrophil recruitment via signaling pathways, including the IL-23/IL-17 axis and G-CSF. Furthermore, neutrophils engage in a series of responses, including phagocytosis, degranulation and NETosis, to combat infection effectively. However, dysregulated neutrophil activity can lead to exacerbated lung injury, highlighting the delicate balance required in neutrophil responses. Age and immunodeficiency also emerge as critical factors influencing the severity of MP infections. This review emphasizes the dual role of neutrophils in both defending against and exacerbating MP infections, suggesting that targeted therapeutic strategies could mitigate the adverse effects while enhancing beneficial neutrophil functions.
Collapse
Affiliation(s)
- Lu Fan
- Department of Respiratory Medicine, Affiliated Children's Hospital of Jiangnan University, Wuxi, 214000, China
| | - Nuo Xu
- Department of Respiratory Medicine, Affiliated Children's Hospital of Jiangnan University, Wuxi, 214000, China
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214000, China
| | - Yun Guo
- Department of Respiratory Medicine, Affiliated Children's Hospital of Jiangnan University, Wuxi, 214000, China
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214000, China
| | - Ling Li
- Department of Respiratory Medicine, Affiliated Children's Hospital of Jiangnan University, Wuxi, 214000, China
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214000, China
| |
Collapse
|
5
|
Wang C, Wen J, Yan Z, Zhou Y, Gong Z, Luo Y, Li Z, Zheng K, Zhang H, Ding N, Wang C, Zhu C, Wu Y, Lei A. Suppressing neutrophil itaconate production attenuates Mycoplasma pneumoniae pneumonia. PLoS Pathog 2024; 20:e1012614. [PMID: 39499730 PMCID: PMC11567624 DOI: 10.1371/journal.ppat.1012614] [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: 04/29/2024] [Revised: 11/15/2024] [Accepted: 09/24/2024] [Indexed: 11/07/2024] Open
Abstract
Mycoplasma pneumoniae is a common cause of community-acquired pneumonia in which neutrophils play a critical role. Immune-responsive gene 1 (IRG1), responsible for itaconate production, has emerged as an important regulator of inflammation and infection, but its role during M. pneumoniae infection remains unknown. Here, we reveal that itaconate is an endogenous pro-inflammatory metabolite during M. pneumoniae infection. Irg1 knockout (KO) mice had lower levels of bacterial burden, lactate dehydrogenase (LDH), and pro-inflammatory cytokines compared with wild-type (WT) controls after M. pneumoniae infection. Neutrophils were the major cells producing itaconate during M. pneumoniae infection in mice. Neutrophil counts were positively correlated with itaconate concentrations in bronchoalveolar lavage fluid (BALF) of patients with severe M. pneumoniae pneumonia. Adoptive transfer of Irg1 KO neutrophils, or administration of β-glucan (an inhibitor of Irg1 expression), significantly attenuated M. pneumoniae pneumonia in mice. Mechanistically, itaconate impaired neutrophil bacterial killing and suppressed neutrophil apoptosis via inhibiting mitochondrial ROS. Moreover, M. pneumoniae induced Irg1 expression by activating NF-κB and STAT1 pathways involving TLR2. Our data thus identify Irg1/itaconate pathway as a potential therapeutic target for the treatment of M. pneumoniae pneumonia.
Collapse
Affiliation(s)
- Cui Wang
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Jun Wen
- Department of pediatrics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zijun Yan
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Yujun Zhou
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Zhande Gong
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Ying Luo
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Zhenkui Li
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Kang Zheng
- Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, Hunan, China
| | - Haijun Zhang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Nan Ding
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Chuan Wang
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Yimou Wu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Aihua Lei
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| |
Collapse
|
6
|
Cui X, Xiang Q, Huang Y, Ji Q, Hu Z, Shi T, Bao G, Liu Y. Mixed Th1/Th2/Th17 Responses Induced by Plant Oil Adjuvant-Based B. bronchiseptica Vaccine in Mice, with Mechanisms Unraveled by RNA-Seq, 16S rRNA and Metabolomics. Vaccines (Basel) 2024; 12:1182. [PMID: 39460348 PMCID: PMC11512391 DOI: 10.3390/vaccines12101182] [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: 09/05/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The current Bordetella bronchiseptica (Bb) vaccine, when adjuvanted with alum, does not elicit adequate robust cellular immunity or effective antibody defense against Bb attacks. Unfortunately, antibiotic treatment generally represents an ineffective strategy due to the development of resistance against a broad range of antibiotics. METHODS The present study was designed to investigate the immune response, protective capabilities and underlying mechanisms of a plant oil-based adjuvant E515 formulated with inactivated Bb antigen as a potential vaccine candidate against Bordetella bronchiseptica. RESULTS Immunization studies revealed that a combination of SO, VE and GS (E515) exhibited a good synergistic adjuvant effect. The E515 adjuvanted Bb vaccine was proven to be highly efficacious and induced a mixed Th1/Th2/Th17 immune response in mice, leading to a significant increase in Bb-specific IgG, IgG1 and IgG2a antibodies, proliferative lymphocyte responses and cytokine levels (by lymphocytes and serum) and effectively induced responses by CD4+ TE, TM cells and B cells. The E515 adjuvant significantly enhanced the immune protection provided by the Bb vaccine in a mice model, as indicated by a reduced bacterial burden in the lungs. Multi-omics sequencing analysis revealed that E515 functions as an adjuvant by modulating critical pathways, including cytokine-cytokine receptor interaction, the IL-17 signaling pathway and the chemokine signaling pathway. This modulation also included interactions with beneficial species of bacteria including Alistipes, Odoribacter and Colidextribacter, as well as energy and lipid-related metabolites, thus highlighting its role as an immunomodulatory agent. CONCLUSION Collectively, our results demonstrate the huge potential of E515-Bb vaccine candidates, thus highlighting the vegetable oil original adjuvant E515 as a promising agent for the development of new veterinary vaccines.
Collapse
Affiliation(s)
- Xuemei Cui
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Qiuju Xiang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
- College of Animal Science and Technology·College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Yee Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Quanan Ji
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Zizhe Hu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Tuanyuan Shi
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Guolian Bao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| | - Yan Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (X.C.); (Q.X.); (Y.H.); (Q.J.); (Z.H.); (T.S.)
| |
Collapse
|
7
|
Karatas O, Akcakavak G. An immunohistochemical study on the evaluation of mast cell, interleukin 17 and interleukin 1β profile in contagious caprine pleuropneumonia. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2024; 15:515-521. [PMID: 39588464 PMCID: PMC11585848 DOI: 10.30466/vrf.2024.2023580.4184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/07/2024] [Indexed: 11/27/2024]
Abstract
Contagious caprine pleuropneumonia (CCPP) in goats is defined as a highly contagious and rapidly spreading mycoplasmal disease that is now among the leading causes of major economic losses on many continents (Asia, Africa and the Middle East). In this study, we aimed to evaluate immunohistochemically mast cells (MCs) profile and local interleukin (IL)-17 and IL-1β protein expressions in naturally infected CCPP according to the course of the inflammation (peracute-acute, subacute-chronic). The material of the study consisted of 40 naturally infected CCPP and 6 healthy control goat lung tissues. Appropriate samples were taken from the necropsied goats and subjected to histopathological and immunohistochemical examination. In the histopathological examination of the samples, it was determined that 29 samples had a peracute-acute course and 11 had a subacute-chronic course. In immuno-histochemical examination, MC profile and local IL-17 and IL-1β protein expressions were evaluated in the peracute-acute and subacute-chronic course. Immunohistochemically, significant increases in MC number, local IL-17 and IL-1β scores were detected in the peracute-acute course compared to the control group. There were significant decreases in the relevant scores in the subacute-chronic course compared to the peracute-acute course. Current findings indicated that MC, IL-17, and IL-1β expressions played important roles in the pathogenesis of infection in naturally infected CCPP, especially in the peracute-acute course. Additionally, MC profile was evaluated for the first time in naturally infected CCPP.
Collapse
Affiliation(s)
- Ozhan Karatas
- Department of Pathology, Faculty of Veterinary Medicine, Sivas Cumhuriyet University, Sivas, Türkiye;
| | - Gokhan Akcakavak
- Department of Pathology, Faculty of Veterinary Medicine, Aksaray University, Aksaray, Türkiye.
| |
Collapse
|
8
|
Ahn JH, Jung DH, Kim DY, Lee TS, Kim YJ, Lee YJ, Seo IS, Kim WG, Cho YJ, Shin SJ, Park JH. Impact of IL-1β on lung pathology caused by Mycobacterium abscessus infection and its association with IL-17 production. Microbes Infect 2024; 26:105351. [PMID: 38724000 DOI: 10.1016/j.micinf.2024.105351] [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: 02/08/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024]
Abstract
Mycobacterium abscessus (MAB), a non-tuberculous mycobacterium (NTM), causes chronic pulmonary inflammation in humans. The NLRP3 inflammasome is a multi-protein complex that triggers IL-1β maturation and pyroptosis through the cleavage of caspase-1. In this study, we investigated the roles of NLRP3 and IL-1β in the host's defense against MAB. The IL-1β production by MAB was completely abolished in NLRP3, but not NLRC4, deficient macrophages. The NLRP3 inflammasome components, which are ASC and caspase-1 were also found to be essential for IL-1β production in response to MAB. NLRP3 and IL-1β deficiency did not affect the intracellular growth of MAB in macrophages, and the bacterial burden in lungs of NLRP3- and IL-1β-deficient mice was also comparable to the burden observed in WT mice. In contrast, IL-1β deficiency ameliorated lung pathology in MAB-infected mice. Notably, the lung homogenates of IL-1β-deficient mice had reduced levels of IL-17, but not IFN-γ and IL-4 when compared with WT counterparts. Furthermore, in vitro co-culture analysis showed that IL-1β signaling was essential for IL-17 production in response to MAB. Finally, we observed that the anti-IL-17 antibody administration moderately mitigated MAB-induced lung pathology. These findings indicated that IL-1β production contribute to MAB-induced lung pathology via the elevation of IL-17 production.
Collapse
Affiliation(s)
- Jae-Hun Ahn
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Yeon Kim
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yeong-Jun Kim
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yun-Ji Lee
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - In-Su Seo
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Wan-Gyu Kim
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Young Jin Cho
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea; NODCURE, INC., 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea.
| |
Collapse
|
9
|
Rosales RS, Risco D, García-Nicolás O, Pallarés FJ, Ramírez AS, Poveda JB, Nicholas RAJ, Salguero FJ. Differential Gene Expression in Porcine Lung Compartments after Experimental Infection with Mycoplasma hyopneumoniae. Animals (Basel) 2024; 14:1290. [PMID: 38731294 PMCID: PMC11083927 DOI: 10.3390/ani14091290] [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: 03/10/2024] [Revised: 04/20/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Mycoplasma hyopneumoniae (Mhyo) is the causative agent of porcine enzootic pneumonia (EP), as well as one of the main pathogens involved in the porcine respiratory disease complex. The host-pathogen interaction between Mhyo and infected pigs is complex and not completely understood; however, improving the understanding of these intricacies is essential for the development of effective control strategies of EP. In order to improve our knowledge about this interaction, laser-capture microdissection was used to collect bronchi, bronchi-associated lymphoid tissue, and lung parenchyma from animals infected with different strains of Mhyo, and mRNA expression levels of different molecules involved in Mhyo infection (ICAM1, IL-8, IL-10, IL-23, IFN-α, IFN-γ, TGF-β, and TNF-α) were analyzed by qPCR. In addition, the quantification of Mhyo load in the different lung compartments and the scoring of macroscopic and microscopic lung lesions were also performed. Strain-associated differences in virulence were observed, as well as the presence of significant differences in expression levels of cytokines among lung compartments. IL-8 and IL-10 presented the highest upregulation, with limited differences between strains and lung compartments. IFN-α was strongly downregulated in BALT, implying a relevant role for this cytokine in the immunomodulation associated with Mhyo infections. IL-23 was also upregulated in all lung compartments, suggesting the potential involvement of a Th17-mediated immune response in Mhyo infections. Our findings highlight the relevance of Th1 and Th2 immune response in cases of EP, shedding light on the gene expression levels of key cytokines in the lung of pigs at a microscopic level.
Collapse
Affiliation(s)
- Rubén S. Rosales
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Veterinary Faculty, University of Las Palmas de Gran Canaria, Trasmontaña s/n, 35416 Arucas, Spain; (R.S.R.); (A.S.R.); (J.B.P.)
| | - David Risco
- Unidad de Histología y Anatomía Patológica, Departamento de Medicina Animal, Veterinary Faculty, University of Extremadura, Avenida de la Universidad, s/n, 10003 Cáceres, Spain
| | - Obdulio García-Nicolás
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Francisco J. Pallarés
- Pathology and Immunology Group (UCO-PIG), Department of Anatomy and Comparative Pathology and Toxicology, UIC Zoonosis y Enfermedades Emergentes ENZOEM, University of Córdoba, International Excellence Agrifood Campus “CeiA3”, 14001 Córdoba, Spain;
| | - Ana S. Ramírez
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Veterinary Faculty, University of Las Palmas de Gran Canaria, Trasmontaña s/n, 35416 Arucas, Spain; (R.S.R.); (A.S.R.); (J.B.P.)
| | - José B. Poveda
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Veterinary Faculty, University of Las Palmas de Gran Canaria, Trasmontaña s/n, 35416 Arucas, Spain; (R.S.R.); (A.S.R.); (J.B.P.)
| | | | - Francisco J. Salguero
- School of Veterinary Medicine, University of Surrey, Daphne Jackson Rd, Guildford GU2 7AL, UK;
| |
Collapse
|
10
|
Mills JL, Lepletier A, Ozberk V, Dooley J, Kaden J, Calcutt A, Huo Y, Hicks A, Zaid A, Good MF, Pandey M. Disruption of IL-17-mediated immunosurveillance in the respiratory mucosa results in invasive Streptococcus pyogenes infection. Front Immunol 2024; 15:1351777. [PMID: 38576622 PMCID: PMC10991685 DOI: 10.3389/fimmu.2024.1351777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/22/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Streptococcus pyogenes is a Gram-positive pathogen that causes a significant global burden of skin pyoderma and pharyngitis. In some cases, infection can lead to severe invasive streptococcal diseases. Previous studies have shown that IL-17 deficiency in mice (IL-17-/-) can reduce S. pyogenes clearance from the mucosal surfaces. However, the effect of IL-17 on the development of severe invasive streptococcal disease has not yet been assessed. Methods Here, we modeled single or repeated non-lethal intranasal (IN) S. pyogenes M1 strain infections in immunocompetent and IL-17-/- mice to assess bacterial colonization following a final IN or skin challenge. Results Immunocompetent mice that received a single S. pyogenes infection showed long-lasting immunity to subsequent IN infection, and no bacteria were detected in the lymph nodes or spleens. However, in the absence of IL-17, a single IN infection resulted in dissemination of S. pyogenes to the lymphoid organs, which was accentuated by repeated IN infections. In contrast to what was observed in the respiratory mucosa, skin immunity did not correlate with the systemic levels of IL-17. Instead, it was found to be associated with the activation of germinal center responses and accumulation of neutrophils in the spleen. Discussion Our results demonstrated that IL-17 plays a critical role in preventing invasive disease following S. pyogenes infection of the respiratory tract.
Collapse
Affiliation(s)
- Jamie-Lee Mills
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Ailin Lepletier
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Victoria Ozberk
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Jessica Dooley
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Jacqualine Kaden
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Ainslie Calcutt
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Yongbao Huo
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Allan Hicks
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Ali Zaid
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Michael F. Good
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Manisha Pandey
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
11
|
Liu Y, Ouyang Y, You W, Liu W, Cheng Y, Mai X, Shen Z. Physiological roles of human interleukin-17 family. Exp Dermatol 2024; 33:e14964. [PMID: 37905720 DOI: 10.1111/exd.14964] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023]
Abstract
Interleukin-17 s (IL-17s) are well-known proinflammatory cytokines, and their antagonists perform excellently in the treatment of inflammatory skin diseases such as psoriasis. However, their physiological functions have not been given sufficient attention by clinicians. IL-17s can protect the host from extracellular pathogens, maintain epithelial integrity, regulate cognitive processes and modulate adipocyte activity through distinct mechanisms. Here, we present a systematic review concerning the physiological functions of IL-17s. Our goal is not to negate the therapeutic effect of IL-17 antagonists, but to ensure their safe use and reasonably explain the possible adverse events that may occur in their application.
Collapse
Affiliation(s)
- Yucong Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ye Ouyang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wanchun You
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wenqi Liu
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yufan Cheng
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xinming Mai
- Medical School, Shenzhen University, Shenzhen, China
| | - Zhu Shen
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
da Costa HHM, Bielavsky M, Orts DJB, Araujo S, Adriani PP, Nogueira JS, Astray RM, Pandey RP, Lancellotti M, Cunha-Junior JP, Prudencio CR. Production of Recombinant Zika Virus Envelope Protein by Airlift Bioreactor as a New Subunit Vaccine Platform. Int J Mol Sci 2023; 24:13955. [PMID: 37762254 PMCID: PMC10531330 DOI: 10.3390/ijms241813955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 09/29/2023] Open
Abstract
The Zika Virus (ZIKV) is an emerging arbovirus of great public health concern, particularly in the Americas after its last outbreak in 2015. There are still major challenges regarding disease control, and there is no ZIKV vaccine currently approved for human use. Among many different vaccine platforms currently under study, the recombinant envelope protein from Zika Virus (rEZIKV) constitutes an alternative option for vaccine development and has great potential for monitoring ZIKV infection and antibody response. This study describes a method to obtain a bioactive and functional rEZIKV using an E. coli expression system, with the aid of a 5-L airlift bioreactor and following an automated fast protein liquid chromatography (FPLC) protocol, capable of obtaining high yields of approximately 20 mg of recombinant protein per liter of bacterium cultures. The purified rEZIKV presented preserved antigenicity and immunogenicity. Our results show that the use of an airlift bioreactor for the production of rEZIKV is ideal for establishing protocols and further research on ZIKV vaccines bioprocess, representing a promising system for the production of a ZIKV envelope recombinant protein-based vaccine candidate.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Monica Bielavsky
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Diego J. B. Orts
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Laboratory of Cardiobiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, São Paulo 04023-062, Brazil
| | - Sergio Araujo
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Patrícia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Renato M. Astray
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
- Multi-Purpose Laboratory Butantan Institute, São Paulo 05503-900, Brazil
| | - Ramendra P. Pandey
- School of Health Sciences and Technology, UPES University, Dehradun 248007, Uttarakhand, India
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences—FCF, University of Campinas—UNICAMP, Campinas 13083-871, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
13
|
Yekani M, Memar MY. Immunologic biomarkers for bacterial meningitis. Clin Chim Acta 2023; 548:117470. [PMID: 37419301 DOI: 10.1016/j.cca.2023.117470] [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: 12/23/2022] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Meningitis is defined as the inflammation of the meninges that is most often caused by various bacterial and viral pathogens, and is associated with high rates of mortality and morbidity. Early detection of bacterial meningitis is essential to appropriate antibiotic therapy. Alterations in immunologic biomarkers levels have been considered the diagnostic approach in medical laboratories for the identifying of infections. The early increasing immunologic mediators such as cytokines and acute phase proteins (APPs) during bacterial meningitis have made they significant indicators for laboratory diagnosis. Immunology biomarkers showed wide variable sensitivity and specificity values that influenced by different reference values, selected a certain cutoff point, methods of detection, patient characterization and inclusion criteria, as well as etiology of meningitis and time of CSF or blood specimens' collection. This study provides an overview of different immunologic biomarkers as diagnostic markers for the identification of bacterial meningitis and their efficiencies in the differentiating of bacterial from viral meningitis.
Collapse
Affiliation(s)
- Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
14
|
Korta A, Kula J, Gomułka K. The Role of IL-23 in the Pathogenesis and Therapy of Inflammatory Bowel Disease. Int J Mol Sci 2023; 24:10172. [PMID: 37373318 DOI: 10.3390/ijms241210172] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Interleukin-23 (IL-23) is a proinflammatory cytokine produced mainly by macrophages and antigen-presenting cells (APCs) after antigenic stimulation. IL-23 plays a significant role as a mediator of tissue damage. Indeed, the irregularities in IL-23 and its receptor signaling have been implicated in inflammatory bowel disease. IL-23 interacts with both the innate and adaptive immune systems, and IL-23/Th17 appears to be involved in the development of chronic intestinal inflammation. The IL-23/Th17 axis may be a critical driver of this chronic inflammation. This review summarizes the main aspects of IL-23's biological function, cytokines that control cytokine production, effectors of the IL-23 response, and the molecular mechanisms associated with IBD pathogenesis. Although IL-23 modulates and impacts the development, course, and recurrence of the inflammatory response, the etiology and pathophysiology of IBD are not completely understood, but mechanism research shows huge potential for clinical applications as therapeutic targets in IBD treatment.
Collapse
Affiliation(s)
- Aleksandra Korta
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Julia Kula
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| |
Collapse
|
15
|
Xue Y, Wang M, Han H. Interaction between alveolar macrophages and epithelial cells during Mycoplasma pneumoniae infection. Front Cell Infect Microbiol 2023; 13:1052020. [PMID: 37113130 PMCID: PMC10126420 DOI: 10.3389/fcimb.2023.1052020] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
Mycoplasma pneumoniae, as one of the most common pathogens, usually causes upper respiratory tract infections and pneumonia in humans and animals. It accounts for 10% to 40% of community-acquired pneumonia in children. The alveolar epithelial cells (AECs) are the first barrier against pathogen infections, triggering innate immune responses by recruiting and activating immune cells when pathogens invade into the lung. Alveolar macrophages (AMs) are the most plentiful innate immune cells in the lung, and are the first to initiate immune responses with pathogens invasion. The cross-talk between the alveolar epithelium and macrophages is necessary to maintain physiological homeostasis and to eradicate invaded pathogen by regulating immune responses during Mycoplasma pneumoniae infections. This review summarizes the communications between alveolar macrophages and epithelial cells during Mycoplasma pneumoniae infections, including cytokines-medicated communications, signal transduction by extracellular vesicles, surfactant associated proteins-medicated signal transmission and establishment of intercellular gap junction channels.
Collapse
Affiliation(s)
- Yazhi Xue
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mengyao Wang
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hongbing Han
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
16
|
Pyung YJ, Park DJ, Kim CG, Yun CH. Remodeling and Restraining Lung Tissue Damage Through the Regulation of Respiratory Immune Responses. Tissue Eng Regen Med 2023; 20:329-339. [PMID: 36763280 PMCID: PMC9913030 DOI: 10.1007/s13770-022-00516-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/13/2022] [Accepted: 12/26/2022] [Indexed: 02/11/2023] Open
Abstract
Tissue damage caused by various stimuli under certain conditions, such as biological and environmental cues, can actively induce systemic and/or local immune responses. Therefore, understanding the immunological perspective would be critical to not only regulating homeostasis of organs and tissues but also to restrict and remodel their damage. Lungs serve as one of the key immunological organs, and thus, in the present article, we focus on the innate and adaptive immune systems involved in remodeling and engineering lung tissue. Innate immune cells are known to react immediately to damage. Macrophages, one of the most widely studied types of innate immune cells, are known to be involved in tissue damage and remodeling, while type 2 innate lymphoid cells (ILC2s) have recently been revealed as an important cell type responsible for tissue remodeling. On the other hand, adaptive immune cells are also involved in damage control. In particular, resident memory T cells in the lung prevent prolonged disease that causes tissue damage. In this review, we first outlined the structure of the respiratory system with biological and environmental cues and the innate/adaptive immune responses in the lung. It is our hope that understanding an immunological perspective for tissue remodeling and damage control in the lung will be beneficial for stakeholders in this area.
Collapse
Affiliation(s)
- Young Jin Pyung
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Da-Jeong Park
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol Gyun Kim
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
- Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea.
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-Do, 25354, Republic of Korea.
- Interdisciplinary Programs in Agricultural Genomics, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
17
|
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
|
18
|
The trehalose glycolipid C18Brar promotes antibody and T-cell immune responses to Mannheimia haemolytica and Mycoplasma ovipneumoniae whole cell antigens in sheep. PLoS One 2023; 18:e0278853. [PMID: 36656850 PMCID: PMC9851559 DOI: 10.1371/journal.pone.0278853] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/23/2022] [Indexed: 01/20/2023] Open
Abstract
Bronchopneumonia is a common respiratory disease in livestock. Mannheimia haemolytica is considered the main causative pathogen leading to lung damage in sheep, with Mycoplasma ovipneumoniae and ParaInfluenza virus type 3, combined with adverse physical and physiological stress, being predisposing factors. A balance of humoral and cellular immunity is thought to be important for protection against developing respiratory disease. In the current study, we compared the ability of the trehalose glycolipid adjuvant C18Brar (C18-alkylated brartemicin analogue) and three commercially available adjuvant systems i.e., Quil-A, Emulsigen-D, and a combination of Quil-A and aluminium hydroxide gel, to stimulate antibody and cellular immune responses to antigens from inactivated whole cells of M. haemolytica and M. ovipneumoniae in sheep. C18Brar and Emulsigen-D induced the strongest antigen-specific antibody responses to both M. haemolytica and M. ovipneumoniae, while C18Brar and Quil-A promoted the strongest antigen-specific IL-17A responses. The expression of genes with known immune functions was determined in antigen-stimulated blood cultures using Nanostring nCounter technology. The expression levels of CD40, IL22, TGFB1, and IL2RA were upregulated in antigen-stimulated blood cultures from animals vaccinated with C18Brar, which is consistent with T-cell activation. Collectively, the results demonstrate that C18Brar can promote both antibody and cellular responses, notably Th17 immune responses in a ruminant species.
Collapse
|
19
|
Paroli M, Caccavale R, Fiorillo MT, Spadea L, Gumina S, Candela V, Paroli MP. The Double Game Played by Th17 Cells in Infection: Host Defense and Immunopathology. Pathogens 2022; 11:pathogens11121547. [PMID: 36558881 PMCID: PMC9781511 DOI: 10.3390/pathogens11121547] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
T-helper 17 (Th17) cells represent a subpopulation of CD4+ T lymphocytes that play an essential role in defense against pathogens. Th17 cells are distinguished from Th1 and Th2 cells by their ability to produce members of the interleukin-17 (IL-17) family, namely IL-17A and IL-17F. IL-17 in turn induces several target cells to synthesize and release cytokines, chemokines, and metalloproteinases, thereby amplifying the inflammatory cascade. Th17 cells reside predominantly in the lamina propria of the mucosa. Their main physiological function is to maintain the integrity of the mucosal barrier against the aggression of infectious agents. However, in an appropriate inflammatory microenvironment, Th17 cells can transform into immunopathogenic cells, giving rise to inflammatory and autoimmune diseases. This review aims to analyze the complex mechanisms through which the interaction between Th17 and pathogens can be on the one hand favorable to the host by protecting it from infectious agents, and on the other hand harmful, potentially generating autoimmune reactions and tissue damage.
Collapse
Affiliation(s)
- Marino Paroli
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| | - Rosalba Caccavale
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Spadea
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
| | - Stefano Gumina
- Department of Anatomy, Histology, Legal Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
| | - Vittorio Candela
- Department of Anatomy, Histology, Legal Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Pia Paroli
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
20
|
Gavitt TD, Mara AB, Goodridge ML, Ozyck RG, Reinhardt E, Miller JM, Hunte M, Tulman ER, Frasca Jr S, Silbart LK, Geary SJ, Szczepanek SM. B cells oppose Mycoplasma pneumoniae vaccine enhanced disease and limit bacterial colonization of the lungs. NPJ Vaccines 2022; 7:130. [PMID: 36310317 PMCID: PMC9618410 DOI: 10.1038/s41541-022-00556-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Development of an effective vaccine for Mycoplasma pneumoniae has been hindered by reports of Vaccine Enhanced Disease (VED) in test subjects vaccinated and challenged in studies conducted in the 1960s. The exact mechanism of disease exacerbation has yet to be fully described, but host immune responses to Lipid-Associated Membrane Proteins (LAMPs) lipoprotein lipid moieties have been implicated. LAMPs-induced exacerbation appears to involve helper T cell recall responses, due in part to their influence on neutrophil recruitment and subsequent inflammatory responses in the lung. Herein, we characterized the functions of host B cell responses to M. pneumoniae LAMPs and delipidated-LAMPs (dLAMPs) by conducting passive transfer and B cell depletion studies to assess their contribution to disease exacerbation or protection using a BALB/c mouse model. We found that antibody responses to M. pneumoniae LAMPs and dLAMPs differ in magnitude, but not in isotype or subclass. Passive transfer, dLAMP denaturation, and monoclonal antibody studies indicate that antibodies do not cause VED, but do appear to contribute to control of bacterial loads in the lungs. Depletion of B cells prior to LAMPs-vaccination results in significantly enhanced pathology in comparison to B cell competent controls, suggesting a possible regulatory role of B cells distinct from antibody secretion. Taken together, our findings suggest that B cell antibody responses to M. pneumoniae contribute to, but are insufficient for protection against challenge on their own, and that other functional properties of B cells are necessary to limit exacerbation of disease in LAMPs-vaccinated mice after infection.
Collapse
Affiliation(s)
- Tyler D. Gavitt
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Arlind B. Mara
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Meagan L. Goodridge
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA
| | - Rosemary Grace Ozyck
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA
| | - Emily Reinhardt
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,Connecticut Veterinary Medical Diagnostic Laboratory, Storrs, CT 06238 USA
| | - Jeremy M. Miller
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Morgan Hunte
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,grid.417555.70000 0000 8814 392XPresent Address: Sanofi, Meriden, CT 06450 USA
| | - Edan R. Tulman
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Salvatore Frasca Jr
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA
| | - Lawrence K. Silbart
- grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Department of Allied of Health Sciences, University of Connecticut, Storrs, CT 06238 USA
| | - Steven J. Geary
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Steven M. Szczepanek
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| |
Collapse
|
21
|
Ma X, Liu Z, Yu Y, Jiang Y, Wang C, Zuo Z, Ling S, He M, Cao S, Wen Y, Zhao Q, Wu R, Huang X, Zhong Z, Peng G, Gu Y. Microsporum gypseum Isolated from Ailuropoda melanoleuca Provokes Inflammation and Triggers Th17 Adaptive Immunity Response. Int J Mol Sci 2022; 23:ijms231912037. [PMID: 36233337 PMCID: PMC9570494 DOI: 10.3390/ijms231912037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/04/2022] [Accepted: 10/08/2022] [Indexed: 12/02/2022] Open
Abstract
Microsporum gypseum causes dermatomycoses in giant pandas (Ailuropoda melanoleuca). This study aimed to investigate the immune response of M. gypseum following deep infection. The degree of damage to the heart, liver, spleen, lungs, and kidneys was evaluated using tissue fungal load, organ index, and histopathological methods. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) detected the mRNA expression of receptors and cytokines in the lung, and immunofluorescence staining and flow cytometry, were used to assess immune cells in the lung. The results indicated that conidia mainly colonized the lungs and caused serious injury with M. gypseum infection. Furthermore, dectin-1, TLR-2, and TLR-4 played a role in recognizing M. gypseum cells. Numerous inflammatory cells, mainly macrophages, dendritic cells, polymorphonuclear neutrophils, and inflammatory cytokines (TGF-β, TNF-α, IL-1β, IL-6, IL-10, IL-12, and IL-23), were activated in the early stages of infection. With the high expression of IL-22, IL-17A, and IL-17F, the Th17 pathway exerted an adaptive immune response to M. gypseum infection. These results can potentially aid in the diagnosis and treatment of diseases caused by M. gypseum in giant pandas.
Collapse
Affiliation(s)
- Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhen Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yaozhang Jiang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chengdong Wang
- China Conservation and Research Center for the Giant Panda, Chengdu 611800, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shanshan Ling
- China Conservation and Research Center for the Giant Panda, Chengdu 611800, China
| | - Ming He
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- China Conservation and Research Center for the Giant Panda, Chengdu 611800, China
| | - Sanjie Cao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yiping Wen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui Wu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaobo Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: ; Tel.: +86-18190681226
| |
Collapse
|
22
|
Pesce B, Ribeiro CH, Larrondo M, Ramos V, Soto L, Catalán D, Aguillón JC. TNF-α Affects Signature Cytokines of Th1 and Th17 T Cell Subsets through Differential Actions on TNFR1 and TNFR2. Int J Mol Sci 2022; 23:ijms23169306. [PMID: 36012570 PMCID: PMC9408897 DOI: 10.3390/ijms23169306] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor necrosis factor (TNF)-α is a pleiotropic cytokine implicated in the etiology of several autoimmune diseases, including rheumatoid arthritis (RA). TNF-α regulates diverse effector functions through the activation of TNF-α receptor (TNFR)1 and TNFR2. Although the detrimental role of this cytokine has been addressed in distinct disease settings, the effects of TNF-α on cytokine production by isolated CD4+ T helper type 1 (Th1) and Th17 cells, two T cell subpopulations that contribute to the pathogenesis of RA, have not been completely elucidated. Here, we show that TNF-α promotes a reduction and expansion in the frequency of both T cell subsets producing IFN-γ and IL-17, respectively. Selective blockade of TNFR1 or TNFR2 on Th1 and Th17 cells revealed that TNFR2 mediates the decrease in IFN-γ production, while signaling through both receptors augments IL-17 production. We also demonstrate that Th1, but not Th17 cells from RA patients present lower levels of TNFR1 compared to healthy controls, whereas TNFR2 expression on both T cell types is similar between patients and controls. Since TNF-α receptors levels in RA patients are not significantly changed by the therapeutic blockade of TNF-α, we propose that targeting TNFR2 may represent an alternative strategy to normalize the levels of key cytokines that contribute to RA pathogenesis.
Collapse
Affiliation(s)
- Bárbara Pesce
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Laboratorio MED.UCHILE-FACS, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Carolina H. Ribeiro
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Milton Larrondo
- Hospital Clínico, Universidad de Chile, Santiago 8380453, Chile
| | - Verónica Ramos
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Hospital Clínico, Universidad de Chile, Santiago 8380453, Chile
| | - Lilian Soto
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Hospital Clínico, Universidad de Chile, Santiago 8380453, Chile
| | - Diego Catalán
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Correspondence: (D.C.); (J.C.A.)
| | - Juan Carlos Aguillón
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Correspondence: (D.C.); (J.C.A.)
| |
Collapse
|
23
|
Vaccination with Mycoplasma pneumoniae membrane lipoproteins induces IL-17A driven neutrophilia that mediates Vaccine-Enhanced Disease. NPJ Vaccines 2022; 7:86. [PMID: 35906257 PMCID: PMC9336141 DOI: 10.1038/s41541-022-00513-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
Bacterial lipoproteins are an often-underappreciated class of microbe-associated molecular patterns with potent immunomodulatory activity. We previously reported that vaccination of BALB/c mice with Mycoplasma pneumoniae (Mp) lipid-associated membrane proteins (LAMPs) resulted in lipoprotein-dependent vaccine enhanced disease after challenge with virulent Mp, though the immune responses underpinning this phenomenon remain poorly understood. Herein, we report that lipoprotein-induced VED in a mouse model is associated with elevated inflammatory cytokines TNF-α, IL-1β, IL-6, IL-17A, and KC in lung lavage fluid and with suppurative pneumonia marked by exuberant neutrophilia in the pulmonary parenchyma. Whole-lung-digest flow cytometry and RNAScope analysis identified multiple cellular sources for IL-17A, and the numbers of IL-17A producing cells were increased in LAMPs-vaccinated/Mp-challenged animals compared to controls. Specific IL-17A or neutrophil depletion reduced disease severity in our VED model—indicating that Mp lipoproteins induce VED in an IL-17A-dependent manner and through exuberant neutrophil recruitment. IL-17A neutralization reduced levels of TNF-α, IL-1β, IL-6, and KC, indicating that IL-17A preceded other inflammatory cytokines. Surprisingly, we found that IL-17A neutralization impaired bacterial clearance, while neutrophil depletion improved it—indicating that, while IL-17A appears to confer both maladaptive and protective responses, neutrophils play an entirely maladaptive role in VED. Given that lipoproteins are found in virtually all bacteria, the potential for lipoprotein-mediated maladaptive inflammatory responses should be taken into consideration when developing vaccines against bacterial pathogens.
Collapse
|
24
|
Abstract
The interleukin-23 [IL-23] cytokine, derived predominantly from macrophages and dendritic cells in response to microbial stimulation, has emerged as a critical promoter of chronic intestinal inflammation. Genome-wide association studies linking variants in IL23R to disease protection, bolstered by experimental evidence from colitis models, and the successful application of therapies against the IL-12/IL-23 shared p40 subunit in the treatment of inflammatory bowel disease [IBD] all provide compelling evidence of a crucial role for IL-23 in disease pathogenesis. Moreover, targeting the p19 subunit specific for IL-23 has shown considerable promise in recent phase 2 studies in IBD. The relative importance of the diverse immunological pathways downstream of IL-23 in propagating mucosal inflammation in the gut, however, remains contentious. Here we review current understanding of IL-23 biology and explore its pleiotropic effects on T cells, and innate lymphoid, myeloid and intestinal epithelial cells in the context of the pathogenesis of IBD. We furthermore discuss these pathways in the light of recent evidence from clinical trials and indicate emerging targets amenable to therapeutic intervention and translation into clinical practice.
Collapse
Affiliation(s)
- Gavin W Sewell
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Arthur Kaser
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| |
Collapse
|
25
|
Omokanye A, Ong LC, Lebrero-Fernandez C, Bernasconi V, Schön K, Strömberg A, Bemark M, Saelens X, Czarnewski P, Lycke N. Clonotypic analysis of protective influenza M2e-specific lung resident Th17 memory cells reveals extensive functional diversity. Mucosal Immunol 2022; 15:717-729. [PMID: 35260804 PMCID: PMC8903128 DOI: 10.1038/s41385-022-00497-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/31/2022] [Accepted: 02/06/2022] [Indexed: 02/06/2023]
Abstract
The fate of tissue-resident memory CD4 T cells (Trm) has been incompletely investigated. Here we show that intranasal, but not parenteral, immunization with CTA1-3M2e-DD stimulated M2e-specific Th17 Trm cells, which conferred strong protection against influenza virus infection in the lung. These cells rapidly expanded upon infection and effectively restricted virus replication as determined by CD4 T cell depletion studies. Single-cell RNAseq transcriptomic and TCR VDJ-analysis of M2e-tetramer-sorted CD4 T cells on day 3 and 8 post infection revealed complete Th17-lineage dominance (no Th1 or Tregs) with extensive functional diversity and expression of gene markers signifying mature resident Trm cells (Cd69, Nfkbid, Brd2, FosB). Unexpectedly, the same TCR clonotype hosted cells with different Th17 subcluster functions (IL-17, IL-22), regulatory and cytotoxic cells, suggesting a tissue and context-dependent differentiation of reactivated Th17 Trm cells. A gene set enrichment analysis demonstrated up-regulation of regulatory genes (Lag3, Tigit, Ctla4, Pdcd1) in M2e-specific Trm cells on day 8, indicating a tissue damage preventing function. Thus, contrary to current thinking, lung M2e-specific Th17 Trm cells are sufficient for controlling infection and for protecting against tissue injury. These findings will have strong implications for vaccine development against respiratory virus infections and influenza virus infections, in particular.
Collapse
Affiliation(s)
- Ajibola Omokanye
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Li Ching Ong
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Cristina Lebrero-Fernandez
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Valentina Bernasconi
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anneli Strömberg
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mats Bemark
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Xavier Saelens
- grid.5342.00000 0001 2069 7798VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paulo Czarnewski
- grid.10548.380000 0004 1936 9377Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Nils Lycke
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
26
|
Ma X, Hu J, Wang C, Gu Y, Cao S, Huang X, Wen Y, Zhao Q, Wu R, Zuo Z, Yu S, Shen L, Zhong Z, Peng G, Ling S. Innate and mild Th17 cutaneous immune responses elicited by subcutaneous infection of immunocompetent mice with Cladosporium cladosporioides. Microb Pathog 2021; 163:105384. [PMID: 34974124 DOI: 10.1016/j.micpath.2021.105384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 10/19/2020] [Accepted: 12/28/2021] [Indexed: 01/02/2023]
Abstract
Cladosporium cladosporioides is a dematiaceous hyphomycete that is pathogenic in the superficial and deep tissues of both immunodeficient and immunocompetent humans and animals. Our aim was to evaluate the antifungal immune responses elicited by C. cladosporioides in immunocompetent mice. Hence, we subcutaneously injected suspensions of C. cladosporioides spores into immunocompetent mice to investigate the anti-fungal immune responses in the skin. We collected skin tissue samples for histopathological examination, immunofluorescence staining, and quantitative real-time polymerase chain reaction analysis. We observed subcutaneous abscesses in mice after subcutaneous injection of C. cladosporioides. A large number of inflammatory cells, including dendritic cells, macrophages, and neutrophils, infiltrated the focal abscess, with comparatively few infiltrating inflammatory cells in the epidermal and dermal layers of the skin. We detected the expression of CD54 in the abscesses and the skin. Gene expression of the pattern recognition receptors Dectin-1 and TLR-2 was higher in infected mice than in controls. Gene expression of the cytokines IL-6, IL-1β, and IL-17A also increased after infection, suggesting that the Th17 signaling pathway may be involved in the anti-fungal response. Although the pathogenicity of C. cladosporioides in healthy mice was weak after subcutaneous infection, resulting in few serious pathological phenomena, it appears that innate and Th17 immune responses play important roles in the cutaneous host response to C. cladosporioides. These findings lay a foundation for further study of the pathogenic mechanism and treatment of C. cladosporioides infection.
Collapse
Affiliation(s)
- Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Jing Hu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chengdong Wang
- China Conservation and Research Center for the Giant Panda, Chengdu, Sichuan, 611800, China.
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Sanjie Cao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaobo Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yiping Wen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qin Zhao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Rui Wu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shumin Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Liuhong Shen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shanshan Ling
- China Conservation and Research Center for the Giant Panda, Chengdu, Sichuan, 611800, China
| |
Collapse
|
27
|
Neutrophil-Mediated Lung Injury Both via TLR2-Dependent Production of IL-1α and IL-12 p40, and TLR2-Independent CARDS Toxin after Mycoplasma pneumoniae Infection in Mice. Microbiol Spectr 2021; 9:e0158821. [PMID: 34937175 PMCID: PMC8694186 DOI: 10.1128/spectrum.01588-21] [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] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mycoplasma pneumoniae (Mp) residing extracellularly in the respiratory tract is the primary cause of bacterial community-acquired pneumonia in humans. However, the detailed pathological mechanism of Mp infection, especially inflammation in the lung, remains unclear. This study examined the role of the neutrophils in the inflammation of Mp-induced pneumonia in mice and the mechanism of neutrophil infiltration into the lungs in the Mp-induced pneumonia. We observed massive infiltration of neutrophils in the bronchoalveolar lavage fluid (BALF) and lung injury after the Mp challenge. The neutrophils were shown to contribute to lung injury in Mp pneumonia but were not involved in eliminating Mp, suggesting that neutrophils are detrimental to the host in Mp pneumonia. Mp also induced the production of inflammatory cytokines and chemokines in the BALF in a toll-like receptor 2 (TLR2)-dependent manner. Particularly, both interleukin (IL)-1α and IL-12 p40 played a crucial role in neutrophil infiltration into the BALF in a coordinated manner. Both IL-1α and IL-12 p40 were released from the alveolar macrophages depending on the TLR2 and reactive oxygen species. In addition, the community-acquired respiratory distress syndrome (CARDS) toxin from Mp were found to induce neutrophil infiltration into BALF in a TLR2-independent and IL-1α-dependent manner. Collectively, the TLR2-dependent production of both IL-1α and IL-12 p40, and CARDS toxin have been elucidated to play an important role in neutrophil infiltration into the lungs subsequently leading to the lung injury upon Mp infection in mice. These data will aid in the development of therapeutics and vaccines for Mp pneumonia. IMPORTANCE Although Mp-induced pneumonia is usually a self-limiting disease, refractory life-threatening pneumonia is often induced. In addition, the development of alternative therapeutic strategies for Mp is expected because of the emergence of antibiotic-resistant Mp. However, the lack of knowledge regarding the pathogenesis of Mp-induced pneumonia, especially inflammation upon the Mp infection, makes it tedious to design novel therapeutics and vaccines. For example, although neutrophil infiltration is widely recognized as one of the characteristics of Mp-induced pneumonia, the precise role of neutrophils in the aggravation of Mp pneumonia remains unclear. This study showed that the infiltration of neutrophils in the lungs is detrimental to the host in Mp-induced pneumonia in mice. Furthermore, the TLR2-dependent IL-1α and IL-12 p40 production, and CARDS toxin play important roles in neutrophil infiltration into the lung, following lung injury. Our findings apply to the rational design of novel therapeutics and vaccines against Mp.
Collapse
|
28
|
Pons S, Arrii E, Arnaud M, Loiselle M, Ferry J, Nouacer M, Lion J, Cohen S, Mooney N, Zafrani L. Immunomodulation of endothelial cells induced by macrolide therapy in a model of septic stimulation. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1656-1669. [PMID: 34636179 PMCID: PMC8589380 DOI: 10.1002/iid3.518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 01/18/2023]
Abstract
Objectives Sepsis is defined as the host's inflammatory response to a life‐threatening infection. The endothelium is implicated in immunoregulation during sepsis. Macrolides have been proposed to display immunomodulatory properties. The goal of this study was to analyze whether macrolides can exert immunomodulation of endothelial cells (ECs) in an experimental model of sepsis. Methods Human ECs were stimulated by proinflammatory cytokines and lipopolysaccharide before exposure to macrolides. ECs phenotypes were analyzed by flow cytometry. Cocultures of ECs and peripheral blood mononuclear cells (PBMCs) were performed to study the ECs ability to alter T‐cell viability and differentiation in the presence of macrolides. Soluble factor production was assessed. Results ECs act as non‐professional antigen presenting cells and expressed human leukocyte antigen (HLA) antigens, the adhesion molecules CD54, CD106, and the coinhibitory molecule CD274 after septic stimulation. Incubation with macrolides induced a significant decrease of HLA class I and HLA class II HLA‐DR on septic‐stimulated ECs, but did not alter either CD54, CD106, nor CD274 expression. Interleukin‐6 (IL‐6) and IL‐8 production by stimulated ECs were unaltered by incubation with macrolides, whereas Clarithromycin exposure significantly decreased IL‐6 gene expression. In cocultures of septic ECs with PBMCs, neither the proportion of CD4 + , CD8 + T nor their viability was altered by macrolides. T‐helper lymphocyte subsets Th1, Th17, and Treg polarization by stimulated ECs were unaltered by macrolides. Conclusion This study reports phenotypic and gene expression changes in septic‐stimulated ECs exposed to macrolides, without resulting in altered immunogenicity of ECs in co‐cultures with PBMCs. In vivo studies may help to further understand the impact of macrolide therapy on ECs immune homeostasis during sepsis.
Collapse
Affiliation(s)
- Stéphanie Pons
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France.,Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, GRC 29, AP-HP, DMU DREAM, Sorbonne University, Paris, France
| | - Eden Arrii
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Marine Arnaud
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Maud Loiselle
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Juliette Ferry
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Manel Nouacer
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Julien Lion
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Shannon Cohen
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Nuala Mooney
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Lara Zafrani
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France.,Medical Intensive Care Unit, AP-HP, Saint-Louis Teaching Hospital, Paris, France
| |
Collapse
|
29
|
Silva JRDA, Oliveira PVSDE, Nolasco P, Santana H, Rezende IS, Santos DPD, Timenetsky J, Marques LM, Figueiredo TB, Silva RAADA. Ureaplasma diversum clearance in lung mice infection is mediated by neutrophils. AN ACAD BRAS CIENC 2021; 93:e20200424. [PMID: 34431870 DOI: 10.1590/0001-3765202120200424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/22/2020] [Indexed: 11/22/2022] Open
Abstract
Pneumonia in cattle is one of the causes of morbidity rates and economic loss. The host response to lung infections caused by Ureaplasma diversum in bovines is virtually unknown. Here in the immune response was evaluated in a murine model for an experimental pulmonary infection by U. diversum. Therefore, AJ, BALB/C and C57BL/6 mice received intratracheal inoculation of U. diversum and were evaluated after 1, 2, 3, 7 and 14 days and the clinical specimens were collected. In bronchoalveolar lavages (BAL) an increase of inflammatory cells was observed. Neutrophils were the main cells recruited to the site of infection and the infiltration was coincided with the production of pro-inflammatory cytokines. We found a large amount of neutrophil in this initial period, followed by a decrease 7 and 14 days post infection, accompanied by bacterial clearance. Our results evidenced the presence of U. diversum within the neutrophil that suggests a phagocytic role of this cell in the elimination of the infection. The immune response features reported here are the initial evidence that healthy immune systems may control these microorganisms. This may be the first step to design new strategies immune based to control the infections in naturally infected hosts.
Collapse
Affiliation(s)
- Jamile R DA Silva
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil.,Universidade de São Paulo, Departamento de Microbiologia, ICB II, Laboratório de Desenvolvimento de Vacinas, Avenida Prof. Lineu Prestes, 1374, Cidade Universitária, 05508-000 São Paulo, SP, Brazil
| | - Percíllia V S DE Oliveira
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil
| | - Patricia Nolasco
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil
| | - Hugo Santana
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil
| | - Izadora S Rezende
- Centro Universitário FG - UniFG, Departamento de Farmácia, Av. Barão do Rio Branco, 459, Centro, 46430-000 Guanambi, BA, Brazil
| | - Denisar P Dos Santos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil.,Centro Universitário FG - UniFG, Departamento de Farmácia, Av. Barão do Rio Branco, 459, Centro, 46430-000 Guanambi, BA, Brazil
| | - Jorge Timenetsky
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, Av. Professor Lineu Prestes, 1374, Cidade Universitária 05508-900 São Paulo, SP, Brazil
| | - Lucas M Marques
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil.,Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, Av. Professor Lineu Prestes, 1374, Cidade Universitária 05508-900 São Paulo, SP, Brazil
| | - Tiana B Figueiredo
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil
| | - Robson A A DA Silva
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Rua Hormindo Barros, 58, Candeias, 45029-094 Vitoria da Conquista, BA, Brazil
| |
Collapse
|
30
|
Kadry NA, Porsch EA, Shen H, St Geme JW. Immunization with HMW1 and HMW2 adhesins protects against colonization by heterologous strains of nontypeable Haemophilus influenzae. Proc Natl Acad Sci U S A 2021; 118:e2019923118. [PMID: 34344825 PMCID: PMC8364133 DOI: 10.1073/pnas.2019923118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) is a common cause of localized respiratory tract disease and results in significant morbidity. The pathogenesis of NTHi disease begins with nasopharyngeal colonization, and therefore, the prevention of colonization represents a strategy to prevent disease. The NTHi HMW1 and HMW2 proteins are a family of conserved adhesins that are present in 75 to 80% of strains and have been demonstrated to play a critical role in colonization of the upper respiratory tract in rhesus macaques. In this study, we examined the vaccine potential of HMW1 and HMW2 using a mouse model of nasopharyngeal colonization. Immunization with HMW1 and HMW2 by either the subcutaneous or the intranasal route resulted in a strain-specific antibody response associated with agglutination of bacteria and restriction of bacterial adherence. Despite the specificity of the antibody response, immunization resulted in protection against colonization by both the parent NTHi strain and heterologous strains expressing distinct HMW1 and HMW2 proteins. Pretreatment with antibody against IL-17A eliminated protection against heterologous strains, indicating that heterologous protection is IL-17A dependent. This work demonstrates the vaccine potential of the HMW1 and HMW2 proteins and highlights the importance of IL-17A in protection against diverse NTHi strains.
Collapse
Affiliation(s)
- Nadia A Kadry
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Eric A Porsch
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Joseph W St Geme
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104;
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
31
|
Żabińska M, Kościelska-Kasprzak K, Krajewska J, Bartoszek D, Augustyniak-Bartosik H, Krajewska M. Immune Cells Profiling in ANCA-Associated Vasculitis Patients-Relation to Disease Activity. Cells 2021; 10:1773. [PMID: 34359942 PMCID: PMC8307495 DOI: 10.3390/cells10071773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/11/2021] [Indexed: 12/05/2022] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) are a group of necrotizing multiorgan autoimmune vasculitides that predominantly affect small blood vessels and are associated with the presence of ANCAs. The aim was to assess regulatory and effector cell populations accompanied by the suPAR biomarker level and link the so-defined immune state to the AAV disease activity. The research involved a multicomponent description of an immune state encompassing a range of B and T cell subsets such as transitional/regulatory B cells (CD19+CD24++CD38++), naïve B cells (CD19+CD24INTCD38INT), Th17 cells, T regulatory cells (CD4+CD25+FoxP3+) and cytotoxic CD4+CD28- cells by flow cytometry. The suPAR plasma level was measured by ELISA. The results indicate that AAV is associated with an increased suPAR plasma level and immune fingerprint characterized by an expansion of Th17 cells and T cells lacking the costimulatory molecule CD28, accompanied by a decrease of regulatory populations (Tregs and transitional B cells) and NK cells. Decreased numbers of regulatory T cells and transitional B cells were shown to be linked to activation of the AAV disease while the increased suPAR plasma level-to AAV-related deterioration of kidney function. The observed immune fingerprint might be a reflection of peripheral tolerance failure responsible for development and progression of ANCA-associated vasculitides.
Collapse
Affiliation(s)
- Marcelina Żabińska
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Katarzyna Kościelska-Kasprzak
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Joanna Krajewska
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland;
| | - Dorota Bartoszek
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Hanna Augustyniak-Bartosik
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| |
Collapse
|
32
|
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
|
33
|
Luo Y, Li C, Zhou Z, Gong Z, Zhu C, Lei A. Biological functions of IL-17-producing cells in mycoplasma respiratory infection. Immunology 2021; 164:223-230. [PMID: 33930194 DOI: 10.1111/imm.13346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/07/2021] [Accepted: 04/18/2021] [Indexed: 12/17/2022] Open
Abstract
Mycoplasmas are the smallest and simplest bacteria that lack a cell wall but have the capability of self-replication. Among them, Mycoplasma pneumoniae is one of the most common causes of community-acquired pneumonia. The hallmark of mycoplasma respiratory diseases is the persistence of lung inflammation that involves both innate and adaptive immune responses. In recent years, a growing body of evidence demonstrates that IL-17 plays an important role in respiratory mycoplasma infection, and associates with the pathologic outcomes of infection, such as pneumonitis and asthma. Numerous studies have shown that a variety of cells, in particular Th17 cells, in the lung can secrete IL-17 during respiratory mycoplasma infection. In this article, we review the biological functions of distinct IL-17-producing cells in mycoplasma respiratory infection with a focus on the effect of IL-17 on the outcomes of infection.
Collapse
Affiliation(s)
- Ying Luo
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China.,Changsha Central Hospital, University of South China, Changsha, China
| | - Cheng Li
- Changsha Central Hospital, University of South China, Changsha, China
| | - Zhou Zhou
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Zhande Gong
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Cuiming Zhu
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Aihua Lei
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation enter for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| |
Collapse
|
34
|
Zhao Y, Liu Z, Qin L, Wang T, Bai O. Insights into the mechanisms of Th17 differentiation and the Yin-Yang of Th17 cells in human diseases. Mol Immunol 2021; 134:109-117. [PMID: 33756352 DOI: 10.1016/j.molimm.2021.03.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/28/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023]
Abstract
Th17 cells are a lineage of CD4+ T helper cells with Th17-specific transcription factors RORγt and RoRα. Since its discovery in 2005, research on Th17 has been in rapid progress, and increasing cytokines or transcription factors have been uncovered in the activation and differentiation of Th17 cells. Furthermore, growing evidence proves there are two different subsets of Th17 cells, namely non-pathogenic Th17 (non-pTh17) and pathogenic Th17 (pTh17), both of which play important roles in adaptive immunity, especially in host defenses, autoimmune diseases, and cancer. In this review, we summarize and discuss the mechanisms of Th17 cells differentiation, and their roles in immunity and diseases.
Collapse
Affiliation(s)
- Yangzhi Zhao
- Department of Hematology, The First Hospital of Jilin University, Changchun, China.
| | - Zhongshan Liu
- Department of Radiation Oncology, the Second Affiliated Hospital of Jilin University, Changchun, China.
| | - Lei Qin
- Institute for Immunology, Tsinghua University, Beijing, China.
| | - Tiejun Wang
- Department of Radiation Oncology, the Second Affiliated Hospital of Jilin University, Changchun, China.
| | - Ou Bai
- Department of Hematology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
35
|
Ma X, Hu J, Yu Y, Wang C, Gu Y, Cao S, Huang X, Wen Y, Zhao Q, Wu R, Zuo Z, Deng J, Ren Z, Yu S, Shen L, Zhong Z, Peng G. Assessment of the pulmonary adaptive immune response to Cladosporium cladosporioides infection using an experimental mouse model. Sci Rep 2021; 11:909. [PMID: 33441700 PMCID: PMC7806624 DOI: 10.1038/s41598-020-79642-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/10/2020] [Indexed: 01/29/2023] Open
Abstract
Cladosporium cladosporioides causes asthma and superficial and deep infections, mostly in immunodeficient individuals and animals. This study aimed to investigate whether C. cladosporioides spores can enter the lungs through pulmonary circulation and influence pulmonary immune response. We intravenously injected mice with C. cladosporioides spore suspension and conducted several assays on the lungs. Pulmonary hemorrhage symptoms and congestion were most severe on days 1, 2, and 3 post-inoculation (PI). Extensive inflammatory cell infiltration occurred throughout the period of infection. More spores and hyphae colonizing the lungs were detected on days 1, 2, and 3 PI, and fewer spores and hyphae were observed within 21 d of infection. Numerous macrophages, dendritic cells, and neutrophils were observed on day 5 PI, along with upregulation of CD54, an intercellular adhesion molecule. Th1 and Th2 cells increased after infection; specifically, Th2 cells increased considerably on day 5 PI. These results suggest that days 2 and 5 PI represent the inflammatory peak in the lungs and that the Th2 and Th1 signaling pathways are potentially involved in pulmonary immune responses. In conclusion, the further adaptive immune responses played important roles in establishing effective pulmonary immunity against C. cladosporioides systemic infections based on innate immune responses.
Collapse
Affiliation(s)
- Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jing Hu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yan Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chengdong Wang
- China Conservation and Research Center for the Giant Panda, Chengdu, 611800, Sichuan, China.
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Sanjie Cao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaobo Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yiping Wen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qin Zhao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Rui Wu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhihua Ren
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shumin Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Liuhong Shen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| |
Collapse
|
36
|
Karan S, Choudhury D, Dixit A. Immunogenic characterization and protective efficacy of recombinant CsgA, major subunit of curli fibers, against Vibrio parahaemolyticus. Appl Microbiol Biotechnol 2021; 105:599-616. [PMID: 33404830 DOI: 10.1007/s00253-020-11038-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022]
Abstract
Vibrio parahaemolyticus is one of the major pathogens responsible for vibriosis and zoonotic infections in teleosts, marine invertebrates, and also humans through consumption of contaminated or unprocessed seafood. Emergence of resistance against current accessible antibiotics and spread to the food chain and environment necessitate the development of safe and effective subunit vaccine against this bacterium. Many bacteria including V. parahaemolyticus produce extracellular curli fibrils, heteropolymeric filaments of major and minor subunit, which have been implicated in adhesion, biofilm formation, and virulence. Adhesins are the primary contact points with the host which help in establishing infection and colonization. CsgA, an adhesin, is the major structural component of the curli fiber that forms homopolymers of several hundred units. Due to their exposure on the cell surface, the curli fibers are recognized by the host's immune system, would generate high immune response, and therefore can serve as effective vaccine candidate. In the present study, we describe characterization of the csgA gene, and preparation of recombinant soluble CsgA of V. parahaemolyticus (rVpCsgA), and evaluation of its vaccine potential. Immunization of BALB/c mice with the rVpCsgA mounted a strong immune response. Cellular immune assays such as antibody isotyping, in vitro splenocyte proliferation analysis, and cytokine profiling revealed mixed T-helper cell immune response. The anti-rVpCsgA antiserum was agglutination positive and specifically cross-reacted with the curli CsgA present on the outer membrane of V. parahaemolyticus cells, thus demonstrating its neutralization potential. One hundred percent survival of the immunized mice upon challenge with the lethal dosage of the bacterium established that the rVpCsgA could serve as an effective vaccine against the bacterium. KEY POINTS: • Recombinant histidine-tagged CsgA of V. parahaemolyticus, rVpCsgA, was purified. • The rVpCsgA immunization produced mixed immune response and agglutinating antibodies. • Immunization with the rVpCsgA protected mice against V. parahaemolyticus challenge.
Collapse
Affiliation(s)
- Sweta Karan
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru Universitys, New Delhi, 110067, India
| | - Devapriya Choudhury
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru Universitys, New Delhi, 110067, India.
| | - Aparna Dixit
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru Universitys, New Delhi, 110067, India.
| |
Collapse
|
37
|
Bruellman R, Llorente C. A Perspective Of Intestinal Immune-Microbiome Interactions In Alcohol-Associated Liver Disease. Int J Biol Sci 2021; 17:307-327. [PMID: 33390852 PMCID: PMC7757023 DOI: 10.7150/ijbs.53589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Uncovering the intricacies of the gut microbiome and how it interacts with the host immune system has opened up pathways in the search for the treatment of disease conditions. Alcohol-associated liver disease is a major cause of death worldwide. Research has shed light on the breakdown of the protective gut barriers, translocation of gut microbes to the liver and inflammatory immune response to microbes all contributing to alcohol-associated liver disease. This knowledge has opened up avenues for alternative therapies to alleviate alcohol-associated liver disease based on the interaction of the commensal gut microbiome as a key player in the regulation of the immune response. This review describes the relevance of the intestinal immune system, the gut microbiota, and specialized and non-specialized intestinal cells in the regulation of intestinal homeostasis. It also reflects how these components are altered during alcohol-associated liver disease and discusses new approaches for potential future therapies in alcohol-associated liver disease.
Collapse
Affiliation(s)
- Ryan Bruellman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
38
|
Pavlenko D, Funahashi H, Sakai K, Hashimoto T, Lozada T, Yosipovitch G, Akiyama T. IL-23 modulates histamine-evoked itch and responses of pruriceptors in mice. Exp Dermatol 2020; 29:1209-1215. [PMID: 33010057 DOI: 10.1111/exd.14206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2023]
Abstract
Accumulating evidence has highlighted the essential roles of cytokines in itch processing. Although IL-23 and Th17 cytokines are elevated in inflammatory skin disorders, their role in itch is unknown. Here, we investigated the role of IL-23 and IL-17A in itch response using an in vitro calcium imaging of mouse dorsal root ganglion (DRG) neurons and an in vivo behaviour test. Calcium imaging studies revealed that a few DRG neurons (~5%) responded to either IL-23 or IL-17A. Pretreatment cells with IL-23 significantly reduced calcium responses to histamine and capsaicin but not chloroquine. Behaviour experiments showed neither IL-23 nor IL-17A evoked scratching. IL-23 significantly decreased histamine-evoked scratching without affecting chloroquine-evoked scratching. There was no difference in scratching between IL-17A- and vehicle-treated groups. These results indicate that IL-23 might play a role in regulating histaminergic itch via modulation of TRPV1 activity.
Collapse
Affiliation(s)
- Darya Pavlenko
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hideki Funahashi
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Psychiatry, University of Miyazaki, Kiyotake, Japan
| | - Kent Sakai
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Takashi Hashimoto
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Taisa Lozada
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gil Yosipovitch
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tasuku Akiyama
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
39
|
Wang Z, Bao H, Liu Y, Wang Y, Qin J, Yang L. Interleukin-23 derived from CD16 + monocytes drives IL-17 secretion by TLR4 pathway in children with mycoplasma pneumoniae pneumonia. Life Sci 2020; 258:118149. [PMID: 32726660 DOI: 10.1016/j.lfs.2020.118149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/10/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
AIMS The study aimed to investigate whether IL-23 is amplified in monocyte subsets of MP pneumonia and to determine its relevant pathway. MATERIALS AND METHODS We firstly analyze the IL-23p19 expression in monocyte subgroups in MP pneumonia patients and healthy controls subjects by using flow cytometry. Then, we also analyzed the percentage of IL-17+γδT cells and Th17 cells in patients with MP pneumonia and controls subjects. At the same time, the relation between IL-23 and IL-17 were also assessed. Furthermore, we constructed the recombinant community-acquired respiratory distress syndrome (CARDS) toxin and intend to stimulate peripheral blood mononuclear cells and RAW264.7 cells in vitro. IL-23p19 was detected by flow cytometry and the mRNA levels were measured by real-time PCR. Finally, TLR4 pathway was also investigated by TAK242 inhibitor. KEY FINDINGS It turned out that the expression of IL-23p19 was increased in CD14brightCD16+ monocyte of MP pneumonia patients than controls subjects. The patients with MP pneumonia had significantly higher the percentage of IL-17+γδT cells and Th17 cells than controls subjects. Interestingly, the levels of IL-23 were positively related to IL-17 in MP pneumonia patients. CD16+ monocytes and RAW264.7 cells, respectively can be induced by CARDS toxin to secrete IL-23 by TLR4 pathway in vitro. SIGNIFICANCE These results indicated that IL-23-IL-17+γδT/Th17 axis may play a role in the pathogenesis of MP pneumonia, whereas IL-23 derived from CD16+ monocytes was expanded in MP pneumonia by TLR4 pathway.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Pediatrics, Tianjin Nankai Hospital, Tianjin, China; Nankai Clinical School, Tianjin Medical University, Tianjin, China
| | - Huijing Bao
- Integrative Medical Diagnosis Laboratory, Tianjin Nankai Hospital, Tianjin, China
| | - Yan Liu
- Department of Pediatrics, Tianjin Nankai Hospital, Tianjin, China; Nankai Clinical School, Tianjin Medical University, Tianjin, China
| | - Yushui Wang
- Department of Pediatrics, Tianjin Nankai Hospital, Tianjin, China; Nankai Clinical School, Tianjin Medical University, Tianjin, China
| | - Junfang Qin
- Medical School of Nankai University, Nankai University, Tianjin, China
| | - Lei Yang
- Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, China.
| |
Collapse
|
40
|
Orlov M, Wander PL, Morrell ED, Mikacenic C, Wurfel MM. A Case for Targeting Th17 Cells and IL-17A in SARS-CoV-2 Infections. THE JOURNAL OF IMMUNOLOGY 2020; 205:892-898. [PMID: 32651218 DOI: 10.4049/jimmunol.2000554] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022]
Abstract
SARS-CoV-2, the virus causing COVID-19, has infected millions and has caused hundreds of thousands of fatalities. Risk factors for critical illness from SARS-CoV-2 infection include male gender, obesity, diabetes, and age >65. The mechanisms underlying the susceptibility to critical illness are poorly understood. Of interest, these comorbidities have previously been associated with increased signaling of Th17 cells. Th17 cells secrete IL-17A and are important for clearing extracellular pathogens, but inappropriate signaling has been linked to acute respiratory distress syndrome. Currently there are few treatment options for SARS-CoV-2 infections. This review describes evidence linking risk factors for critical illness in COVID-19 with increased Th17 cell activation and IL-17 signaling that may lead to increased likelihood for lung injury and respiratory failure. These findings provide a basis for testing the potential use of therapies directed at modulation of Th17 cells and IL-17A signaling in the treatment of COVID-19.
Collapse
Affiliation(s)
- Marika Orlov
- Hospitalist and Specialty Medicine, Department of Veterans Affairs, Puget Sound, Seattle, WA 98108;
| | - Pandora L Wander
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, WA 98195; and
| | - Eric D Morrell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104
| | - Carmen Mikacenic
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104
| |
Collapse
|
41
|
Tamiya S, Yoshikawa E, Ogura M, Kuroda E, Suzuki K, Yoshioka Y. Vaccination using inactivated Mycoplasma pneumoniae induces detrimental infiltration of neutrophils after subsequent infection in mice. Vaccine 2020; 38:4979-4987. [PMID: 32536549 DOI: 10.1016/j.vaccine.2020.05.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/18/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022]
Abstract
Mycoplasma pneumoniae (Mp) is one of the most common causes of community-acquired pneumonia. Given the emergence and high rates of antibiotic-resistant Mp strains, vaccines that prevent the pneumonia and secondary complications due to Mp infection are urgently needed. Although several studies have shown the protective efficacy of Mp vaccines in human clinical trials, some reports suggest that vaccination against Mp exacerbates disease upon subsequent Mp challenge. Therefore, to develop optimal vaccines against Mp, understanding the immune responses that contribute to post-vaccination exacerbation of inflammation is crucial. Here we examined whether Mp vaccination might exacerbate pneumonia after subsequent Mp infection in mice. We found that vaccination with inactivated Mp plus aluminum salts as an adjuvant induced Mp-specific IgG, Th1 cells, and Th17 cells. Toll-like receptor 2 signaling contributed to the induction of an Mp-specific IgG response and was necessary for Mp-specific Th17-cell-but not Th1-cell-responses in vaccinated mice. In addition, vaccination with inactivated Mp plus aluminum salts suppressed the number of Mp organisms in the bronchoalveolar lavage fluid, indicating that vaccination can reduce Mp infection. However, the numbers of total immune cells and neutrophils in bronchoalveolar lavage fluid after Mp challenge did not differ between vaccinated mice and non-vaccinated control mice. Furthermore, depletion of CD4+ T cells prior to Mp challenge decreased pulmonary neutrophil infiltration in vaccinated mice, suggesting that Th1 or Th17 cells (or both) are responsible for the vaccination-induced neutrophil infiltration. These results suggest that, despite reducing Mp infection, vaccination of mice by using inactivated Mp fails to suppress inflammation, such as neutrophil infiltration into the lung, after subsequent Mp infection.
Collapse
Affiliation(s)
- Shigeyuki Tamiya
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Eisuke Yoshikawa
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Monami Ogura
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Etsushi Kuroda
- Department of Immunology and Medical Zoology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan; Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, NIBIOHN, Ibaraki, Osaka, Japan
| | - Koichiro Suzuki
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan; BIKEN Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
42
|
Cammayo PLT, Fernandez-Colorado CP, Flores RA, Roy A, Kim S, Lillehoj HS, Kim WH, Min W. IL-17A treatment influences murine susceptibility to experimental Riemerella anatipestifer infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 106:103633. [PMID: 31991165 DOI: 10.1016/j.dci.2020.103633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 06/10/2023]
Abstract
Riemerella anatipestifer causes infectious disease and considerable economic loss in the duck industry worldwide. Our previous studies demonstrated an association between proinflammatory cytokine interleukin (IL)-17A and R. anatipestifer infection. Here, we provide evidence for IL-17A involvement in R. anatipestifer infection using a mouse model. Mice showed higher resistance to R. anatipestifer infection than ducks, with median lethal doses (LD50) of 3.5 × 1010 and 5 × 107 colony-forming units (CFU), respectively. Twenty-four hours after infection, mice with a sub-lethal dose (3.5 × 109 CFU) exhibited levels of IL-17A and IL-23 expression similar to uninfected mice. Thus, we hypothesized that exogenous IL-17A or IL-23 administration affects susceptibility of mice to R. anatipestifer. Mice pretreated with IL-17A or IL-23 prior to sub-lethal dose infection of R. anatipestifer exhibited increased bacterial burden and spleen weights compared to untreated infected mice, confirming the involvement of IL-17A in susceptibility to R. anatipestifer infection in vivo.
Collapse
Affiliation(s)
- Paula Leona T Cammayo
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Cherry P Fernandez-Colorado
- Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of the Philippines Los Banos, College, Laguna, 4031, Philippines
| | - Rochelle A Flores
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Anindita Roy
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Suk Kim
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, 20705, USA
| | - Woo H Kim
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, 20705, USA
| | - Wongi Min
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
43
|
Interleukin-17 mediates lung injury by promoting neutrophil accumulation during the development of contagious caprine pleuropneumonia. Vet Microbiol 2020; 243:108651. [PMID: 32273025 DOI: 10.1016/j.vetmic.2020.108651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/05/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
Contagious caprine pleuropneumonia (CCPP) is a highly contagious infectious disease of goats caused by Mycoplasma capricolum subspecies capripneumoniae (Mccp). CCPP outbreaks usually result in high morbidity and mortality of the affected goats, making this disease a major cause of economic losses to goat producers globally. However, the pathogenesis of CCPP remains unclear. Here, we show that IL-17-driven neutrophil accumulation is involved in the lung damage in CCPP goats. During CCPP development, intense inflammatory infiltrates could be observed in the injured lungs. Specifically, neutrophils were observed to be present within the alveoli. Increased IL-17 release drove the excessive influx of neutrophils into the lung, as IL-17 effectively stimulated the production of neutrophil chemoattractants from lung epithelial cells following Mccp infection. Our data highlight a critical role of IL-17-driven neutrophil accumulation in the pathogenesis of CCPP and suggest that IL-17 may potentially be a useful immunotherapeutic target for the treatment of CCPP.
Collapse
|
44
|
Th17/IL-17 Axis Regulated by Airway Microbes Get Involved in the Development of Asthma. Curr Allergy Asthma Rep 2020; 20:11. [PMID: 32172346 DOI: 10.1007/s11882-020-00903-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Bronchial asthma is a common respiratory disease induced by immune imbalance, characterized by chronic non-specific airway inflammation and airway hyperresponsiveness (AHR). Many factors induce asthma, among which respiratory infection is the important cause. In this review, we discuss how respiratory microbes participate in the occurrence and progression of asthma via Th17/IL-17 axis. RECENT FINDINGS Pathogenesis of asthma has been considered as closely related to the imbalance in number and function of Th1/Th2 in the CD4+ T lymphocyte subsets. Recent studies have shown that Th17 cell and its secretory IL-17 also play an important role in AHR. Respiratory virus, bacteria, fungi, and other respiratory microbial infections can directly or indirectly induce the differentiation of Th17 cell and the production of related cytokines to induce AHR. Respiratory microbial infection can affect the TH17/IL-17A axis through a variety of mechanisms, thereby promoting the occurrence and development of asthma, and these specific mechanisms may provide new effective therapeutic ideas for asthma.
Collapse
|
45
|
Hoang HH, Wang PC, Chen SC. The protective efficacy of recombinant hypoxic response protein 1 of Nocardia seriolae in largemouth bass (Micropterus salmoides). Vaccine 2020; 38:2925-2936. [PMID: 32115295 DOI: 10.1016/j.vaccine.2020.02.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022]
Abstract
Nocardia seriolae has become one of the major pathogens affecting the aquaculture industry and causes Nocardiosis, a highly devastating disease of marine and freshwater fish that leads to severe economic losses. Therefore, research efforts towards developing efficacious vaccines to control this disease are of high importance. In this study, the hypoxic response protein 1 (HRP1) cloned into pET32a vector was expressed, and produced in Escherichia coli strain BL21 (DE3). The antigenicity of purified recombinant TRX-tagged HRP (rHRP1) was analysed by western blotting using largemouth bass anti-N. seriolae sera. The results showed that largemouth bass anti-N. seriolae sera could specifically detect a 33 kDa rHRP1 protein. Further, the vaccine efficacy of rHRP1 was evaluated in a largemouth bass fish model by calculating the relative percent survival (RPS). rHRP1 incurred an RPS of 73.33% as compared to the control group. Immunological analysis showed that rHRP1 could produce significantly higher serum concentrations of anti-N. seriolae antibodies and serum lysozyme activity as compared to the control groups. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis showed that rHRP1 significantly enhanced the expression of immune-related genes, such as IL-12p40, IL-8, IL-1β, TNFα, IFNγ, NKEF, MHCIα, MHCIIα, CD4-1, CD8α, IgM, NF-κβ, STAT3, IRF4, RORα, and CCL20. These results indicate that rHRP1 may be a promising vaccine candidate against nocardiosis.
Collapse
Affiliation(s)
- Huy Hoa Hoang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, No. 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan.
| | - Pei-Chi Wang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, No. 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan; Southern Taiwan Fish Disease Centre, National Pingtung University of Science and Technology, No. 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan.
| | - Shih-Chu Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, No. 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan; Southern Taiwan Fish Disease Centre, National Pingtung University of Science and Technology, No. 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan; International Degree Program of Ornamental Fish Science and Technology, International College, National Pingtung University of Science and Technology, No. 1, Shuefu Road, Neipu, Pingtung 91201, Taiwan; Research Centre for Animal Biologics, National Pingtung University of Science and Technology, Pingtung, Taiwan.
| |
Collapse
|
46
|
Wang W, Deng G, Zhang G, Yu Z, Yang F, Chen J, Cai Y, Werz O, Chen X. Genetic polymorphism rs8193036 of IL17A is associated with increased susceptibility to pulmonary tuberculosis in Chinese Han population. Cytokine 2019; 127:154956. [PMID: 31864094 DOI: 10.1016/j.cyto.2019.154956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
Th17 cells play a key role in immunity against Mycobacterium tuberculosis, our previous research showed that reduced Th17 responses were associated with the severe outcome of Mtb infection. The associations between IL17A polymorphisms and susceptibility of TB has been reported, but the results are inconsistent and the underlying mechanisms is unknown. In this study, we identified a genetic variation (rs8193036) in the promoter region of IL17A is associated with susceptibility to TB. The minor allele T frequency of rs8193036 was significantly different between patients with active TB (29.7%) and healthy controls (32.3%) (OR = 0.81; 95%CI, 0.71-0.93; P = 0.0026). Peripheral blood mononuclear cells from individuals carrying rs8193036CC genotypes produced significantly lower amount of IL17A upon CD3/28 stimulation compared to the individuals carrying rs8193036TT genotypes. Functional assay by reporter luciferase activity and EMSA demonstrated that rs8193036C exhibited significantly lower promotor transcription activities. In conclusion, our study confirmed that IL17A (rs8193036) is a functional SNP that could regulate gene expression though influencing transcription factor binding activity.
Collapse
Affiliation(s)
- Wenfei Wang
- Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China; Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, Jena, Germany
| | - Guofang Deng
- Guangdong Key Laboratory for Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection & Immunity, Shenzhen Third People's Hospital, Shenzhen, China
| | - Guoliang Zhang
- Guangdong Key Laboratory for Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection & Immunity, Shenzhen Third People's Hospital, Shenzhen, China
| | - Ziqi Yu
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fan Yang
- Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jianyong Chen
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yi Cai
- Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, Jena, Germany.
| | - Xinchun Chen
- Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China; Guangdong Key Laboratory for Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection & Immunity, Shenzhen Third People's Hospital, Shenzhen, China.
| |
Collapse
|
47
|
Ebaid H, Abdel-Salam B, Alhazza I, Al-Tamimi J, Hassan I, Rady A, Mashaly A, Mahmoud A, Sammour R. Samsum ant venom modulates the immune response and redox status at the acute toxic dose in vivo. J Venom Anim Toxins Incl Trop Dis 2019; 25:e20190020. [PMID: 31839800 PMCID: PMC6892565 DOI: 10.1590/1678-9199-jvatitd-2019-0020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/05/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Ant venoms express surface molecules that participate in antigen presentation involving pro- and anti-inflammatory cytokines. This work aims to investigate the expression of MHC-II, CD80 and CD86 on the polymorphonuclear cells (PMNs) in rats injected with samsum ant venom (SAV). METHODS Rats were divided into three groups - control, SAV-treated (intraperitoneal route, 600 μg/kg), and SAV-treated (subcutaneous route, 600 μg/kg). After five doses, animals were euthanized and samples collected for analysis. RESULTS The subcutaneous SAV-trated rats presented decreased levels of glutathione with increased cholesterol and triglyceride levels. Intraperitoneal SAV-treated animals displayed significantly reduced concentrations of both IFN-γ and IL-17 in comparison with the control group. However, intraperitoneal and subcutaneous SAV-treated rats were able to upregulate the expressions of MHC-II, CD80 and CD86 on PMNs in comparison with the control respectively. The histological examination showed severe lymphocyte depletion in the splenic white pulp of the intraperitoneal SAV-injected rats. CONCLUSION Stimulation of PMNs by SAV leads to upregulation of MHC-II, CD 80, and CD 86, which plays critical roles in antigen presentation and consequently proliferation of T-cells. Subcutaneous route was more efficient than intraperitoneal by elevating MHC-II, CD80 and CD86 expression, disturbing oxidative stability and increasing lipogram concentration.
Collapse
Affiliation(s)
- Hossam Ebaid
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| | - Bahaa Abdel-Salam
- Department of Biology, College of Science and Humanities in
El-Quwiaya, 11961, Shaqra University, Saudi Arabia
| | - Ibrahim Alhazza
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| | - Jameel Al-Tamimi
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| | - Iftekhar Hassan
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| | - Ahmed Rady
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| | - Ashraf Mashaly
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| | - Ahmed Mahmoud
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| | - Reda Sammour
- Department of Zoology, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
| |
Collapse
|
48
|
Transcriptome sequencing analysis of porcine MDM response to FSL-1 stimulation. Microb Pathog 2019; 138:103830. [PMID: 31689475 DOI: 10.1016/j.micpath.2019.103830] [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] [Received: 04/04/2019] [Revised: 09/18/2019] [Accepted: 10/30/2019] [Indexed: 12/18/2022]
Abstract
Mycoplasma infection can cause many diseases in pigs, resulting in great economic losses in pork production. Innate immune responses are thought to play critical roles in the pathogenesis of mycoplasma disease. However, the molecular events involved in immune responses remain to be determined. Hence, the object of this study was to use RNA-Seq to investigate the gene expression profiles of the innate immune response mediated by FSL-1 in pig monocyte-derived macrophages (MDMs). The results revealed that 1442 genes were differentially expressed in the FSL-1 group compared with the control groups, of which 777 genes were upregulated and 665 genes were downregulated. KEGG pathway analysis showed that the upregulated genes were mainly involved in innate immune-related pathways including the TNF signaling pathway, cytokine-cytokine receptor interaction, Toll-like receptor signaling pathway, Jak-STAT signaling pathway, chemokine signaling pathway, NOD-like receptor signaling pathway and NF-kappa B signaling pathway. The downregulated genes were only involved in the cGMP-PKG signaling pathway and glycerophospholipid metabolism. Our results showed that FSL-1 stimulation activated the TLR2 signaling pathway and resulted in diverse inflammatory responses. FSL-1 induced the transcription of numerous protein-coding genes involved in a complex network of innate immune-related pathways. We speculate that TNF, IL1B, IL6, NFKB1, NFKBIA, CXCL2, CXCL8, CXCL10, CCL2, CCL4 and CCL5 were the most likely hub genes that play important roles in the above pathways. This study identified the differentially expressed genes and their related signaling pathways, contributing to the comprehensive understanding of the mechanisms underlying host-pathogen interactions during mycoplasma infection and providing a reference model for further studies.
Collapse
|
49
|
Santostefano M, Herzyk D, Montgomery D, Wolf J. Nonclinical safety of tildrakizumab, a humanized anti-IL-23p19 monoclonal antibody, in nonhuman primates. Regul Toxicol Pharmacol 2019; 108:104476. [PMID: 31536773 DOI: 10.1016/j.yrtph.2019.104476] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/18/2019] [Accepted: 09/13/2019] [Indexed: 01/18/2023]
Abstract
Tildrakizumab (also known as MK-3222), is a high-affinity, humanized, immunoglobin G1κ monoclonal antibody targeting the p19 subunit of interleukin-23 recently approved for the treatment of moderate to severe plaque psoriasis in the US, Europe, and Australia. The safety profile of tildrakizumab was characterized in nonclinical studies using a pharmacologically relevant cynomolgus monkey model. In repeat-dose toxicity studies, cynomolgus monkeys were chronically treated with subcutaneous (SC) injections of 100 mg/kg of tildrakizumab every 2 weeks up to 9 months. Tildrakizumab was well tolerated, with no toxicological findings (including assessment of reproductive organs; hormonal effects; and cardiovascular, respiratory, and central nervous system function) at systemic exposures approximately 90 times higher than the recommended human dose of 100 mg. An embryofetal developmental study conducted in pregnant monkeys revealed no treatment-related effects to the developing fetus following SC administration of tildrakizumab 100 mg/kg. In a pre- and postnatal development study, 2 neonatal deaths due to potential viral infection at 100 mg/kg were considered of uncertain relationship to the treatment based on a lack of historical data on the occurrence of viral infection in neonate cynomolgus monkeys. The results of this comprehensive nonclinical safety program support the safe use of tildrakizumab.
Collapse
Affiliation(s)
- Michael Santostefano
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA, 02115-5727, United States.
| | - Danuta Herzyk
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, 19486, United States.
| | - Diana Montgomery
- Pharmacokinetics, Predictive and Clinical Immunogenicity, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, 19486, United States.
| | - Jayanthi Wolf
- Global Regulatory Affairs, Merck & Co., Inc., 351 N. Sumneytown Pike, North Wales, PA, 19454-2505, United States.
| |
Collapse
|
50
|
Chao J, Han X, Liu K, Li Q, Peng Q, Lu S, Zhu X, Hu G, Dong Y, Hu C, Chen Y, Chen J, Khan FA, Chen H, Guo AA. Calves Infected with Virulent and Attenuated Mycoplasma bovis Strains Have Upregulated Th17 Inflammatory and Th1 Protective Responses, Respectively. Genes (Basel) 2019; 10:genes10090656. [PMID: 31466385 PMCID: PMC6770603 DOI: 10.3390/genes10090656] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/14/2019] [Accepted: 08/27/2019] [Indexed: 02/08/2023] Open
Abstract
Mycoplasma bovis is a critical bovine pathogen, but its pathogenesis remains poorly understood. Here, the virulent HB0801 (P1) and attenuated HB0801-P150 (P150) strains of M. bovis were used to explore the potential pathogenesis and effect of induced immunity from calves’ differential transcriptomes post infection. Nine one-month-old male calves were infected with P1, P150, or mock-infected with medium and euthanized at 60 days post-infection. Calves in P1 group exhibited other clinical signs and pathological changes compared to the other two groups. Transcriptome profiles of peripheral blood mononuclear cells revealed seven and 10 hub differentially expressed genes (DEGs) in P1 and P150 groups compared with mock-infected group, respectively. Then, P1-induced pathogenesis was predicted to be associated with enhanced Th17, and P150-induced immunity with Th1 response and expression of ubiquitination-associated enzymes. Association analysis showed that 14 and 11 DEGs were positively and negatively correlated with pathological changes, respectively. Furthermore, up-regulated expression in molecules critical to differentiation of pathogenic Th17 cells in lung and peripheral blood mononuclear cells in P1 group was validated at RNA and protein levels. The results confirmed virulent and attenuated strains might be associated with biased differentiation of pro-inflammatory pathogenic Th17 and Th1 subsets respectively.
Collapse
Affiliation(s)
- Jin Chao
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoxiao Han
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Kai Liu
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingni Li
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | | | - Siyi Lu
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xifang Zhu
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Guyue Hu
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yaqi Dong
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Changmin Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yingyu Chen
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianguo Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Farhan Anwar Khan
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - And Aizhen Guo
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China.
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|