1
|
Machura E, Krakowczyk H, Kleszyk M, Swiętochowska E, Grzywna-Rozenek E, Rusek M, Góra A, Chrobak E, Pukas-Bochenek A, Szczepanska M. Serum Levels of Selected Cytokines and Chemokines and IgG4 in Children With Recurrent Respiratory Tract Infections. J Immunol Res 2024; 2024:5170588. [PMID: 39431236 PMCID: PMC11490343 DOI: 10.1155/2024/5170588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Background: Respiratory tract infections are a common health problem. Cytokines/chemokines play a critical role in the regulation of the immune system. Their defective production may predispose to recurrent respiratory tract infections (RRIs), and an excessive immune response may lead to chronic inflammation and cause damage to the respiratory tract. Another biomarker of respiratory infections may be immunoglobulin-IgG4. Its meaning has still been little explored. We wanted to assess the suitability of the levels of biomarkers tested: interleukin (IL)-17A, IL-18, IL-23, normal T cells expressed and secreted (RANTES), and induced protein (IP)-10, as well as immunoglobilun G4 (IgG4) to predict recurrent infections. Methods: The study group (SG) included a total of 130 children (68 girls, 62 boys) between 3 and 17 years of age with RRI. The control group (CG) included 86 healthy children with no symptoms of inflammatory or allergic diseases (44 girls and 42 boys) of the same age. Blood samples were collected in fasting state and then serum samples were frozen and stored until biomarker assay. Results: Serum RANTES, IL-18, IL-23, and IgG4 concentration were higher in all children with recurrent infections vs. those in the CG (p < 0001). Serum levels of IL-17A and IP-10 were also significantly higher in the SG than in the CG, but only in the youngest children. Among the six serum markers, RANTES demonstrated the highest area under the receiver operating characteristic curve (area under curve) value (0.998, 95% confidence interval [CI]: 0.98-1.0, p < 0.001) for the diagnosis of RRIs, followed by IL-23 (0.99, 95% CI 0.966-0,999, p < 0.001) and IL-18 (0.957, 95% CI 0.921-0.980, p < 0.001). Conclusions: RANTES, IL-23, and IL-18 could be strong predictors of respiratory infections recurrence in children.
Collapse
Affiliation(s)
- Edyta Machura
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Helena Krakowczyk
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Magdalena Kleszyk
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Elzbieta Swiętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Ewa Grzywna-Rozenek
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Malgorzata Rusek
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Anna Góra
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Ewelina Chrobak
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Anna Pukas-Bochenek
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| | - Maria Szczepanska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
2
|
Duan C, Abudureheman T, Wang S, Suo J, Yu Y, Shi F, Liu X, Salama DB, Srivastav RK, Gupta N, Suo X. Expression of IL-1β in transgenic Eimeria necatrix enhances the immunogenicity of parasites and promotes mucosal immunity against coccidiosis. Front Immunol 2024; 15:1435702. [PMID: 39221251 PMCID: PMC11361970 DOI: 10.3389/fimmu.2024.1435702] [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: 05/20/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Anticoccidial vaccines comprising living oocysts of Eimeria tenella, Eimeria necatrix, Eimeria maxima, and Eimeria acervulina are used to control coccidiosis. This study explored the potential of IL-1β to act as a molecular adjuvant for enhancing the immunogenicity of Eimeria necatrix and mucosal immunity. We engineered E. necatrix to express a functional chIL-1β (EnIL-1β) and immunized chickens with oocysts of the wild type (EnWT) and tranegenic (EnIL-1β) strains, respectively. The chickens were then challenged with EnWT oocysts to examine the immunogenicity-enhancing potential of chIL-1β. As expected, the oocyst output of EnIL-1β-immunized chickens was significantly reduced compared to those immunized using EnWT. No difference in body weight gain and lesion scores of EnIL-1β and EnWT groups was observed. The parasite load in the small intestine and caeca showed that the invasion and replication of EnIL-1β was not affected. However, the markers of immunogenicity and mucosal barrier, Claudin-1 and avian β-defensin-1, were elevated in EnIL-1β-infected chickens. Ectopic expression of chIL-1β in E. necatrix thus appears to improve its immunogenicity and mucosal immunity, without increasing pathogenicity. Our findings support chIL-1β as a candidate for development of effective live-oocyst-based anticoccidial vaccines.
Collapse
Affiliation(s)
- Chunhui Duan
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Tumalisi Abudureheman
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Si Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingxia Suo
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ying Yu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Fangyun Shi
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xianyong Liu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dina B Salama
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Intracellular Parasite Education And Research Labs (iPEARL), Department of Biological Sciences, Birla Institute of Technology and Science, Pilani (BITS-P), Hyderabad, India
- Parasitology and Animal Disease Department, Veterinary Research Institute, National Research Centre, Giza, Egypt
| | - Ratnesh Kumar Srivastav
- Intracellular Parasite Education And Research Labs (iPEARL), Department of Biological Sciences, Birla Institute of Technology and Science, Pilani (BITS-P), Hyderabad, India
| | - Nishith Gupta
- Intracellular Parasite Education And Research Labs (iPEARL), Department of Biological Sciences, Birla Institute of Technology and Science, Pilani (BITS-P), Hyderabad, India
- Department of Molecular Parasitology, Institute of Biology, Faculty of Life Sciences, Humboldt University, Berlin, Germany
| | - Xun Suo
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Bhowmick S, Gupta S, Mondal S, Mallick AI. Activation of Antiviral Host Responses against Avian Influenza Virus and Remodeling of Gut Microbiota by rLAB Vector Expressing rIL-17A in Chickens. ACS Infect Dis 2024; 10:3026-3041. [PMID: 38970488 DOI: 10.1021/acsinfecdis.4c00377] [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: 07/08/2024]
Abstract
Low-pathogenic avian influenza virus (LPAIV) remains the most common subtype of type-A influenza virus that causes moderate to severe infection in poultry with significant zoonotic and pandemic potential. Due to high mutability, increasing drug resistance, and limited vaccine availability, the conventional means to prevent intra- or interspecies transmission of AIV is highly challenging. As an alternative to control AIV infections, cytokine-based approaches to augment antiviral host defense have gained significant attention. However, the selective application of cytokines is critical since unregulated expression of cytokines, particularly proinflammatory ones, can cause substantial tissue damage during acute phases of immune responses. Moreover, depending on the type of cytokine and its impact on intestinal microbiota, outcomes of cytokine-gut microflora interaction can have a critical effect on overall host defense against AIV infections. Our recent study demonstrated some prominent roles of chicken IL-17A (ChIL-17A) in regulating antiviral host responses against AIV infection, however, in an in vitro model. For more detailed insights into ChIL-17A function, in the present study, we investigated whether ChIL-17A-meditated elevated antiviral host responses can translate into effective immune protection against AIV infection in an in vivo system. Moreover, considering the role of gut health in fostering innate or local host responses, we further studied the contributory relationships between gut microbiota and host immunity against AIV infection in chickens. For this, we employed a recombinant lactic acid-producing bacterial (LAB) vector, Lactococcus lactis, expressing ChIL-17A and analyzed the in vivo functionality in chickens against an LPAIV (A/H9N2) infection. Our study delineates that mucosal delivery of rL. lactis expressing ChIL-17A triggers proinflammatory signaling cascades and can drive a positive shift in phylum Firmicutes, along with a marked decline in phylum Actinobacteriota and Proteobacteria, favoring effective antiviral host responses against AIV infection in chickens. We propose that ChIL-17A-mediated selective expansion of beneficial gut microbiota might form a healthy microbial community that augments the effective immune protection against AIV infections in chickens.
Collapse
Affiliation(s)
- Sucharita Bhowmick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Subhadeep Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Samiran Mondal
- Department of Veterinary Pathology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| |
Collapse
|
4
|
Giannuzzi D, Capra E, Bisutti V, Vanzin A, Marsan PA, Cecchinato A, Pegolo S. Methylome-wide analysis of milk somatic cells upon subclinical mastitis in dairy cattle. J Dairy Sci 2024; 107:1805-1820. [PMID: 37939836 DOI: 10.3168/jds.2023-23821] [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: 05/31/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023]
Abstract
Better understanding of the molecular mechanisms behind bovine mastitis is fundamental for improving the management of this disease, which continues to be of major concern for the dairy industry, especially in its subclinical form. Disease severity and progression depend on numerous aspects, such as livestock genetics, and the interaction between the causative agent, the host, and the environment. In this context, epigenetic mechanisms have proven to have a role in controlling the response of the animal to inflammation. Therefore, in this study we aimed to explore genome-wide DNA methylation of milk somatic cells (SC) in healthy cows (n = 15) and cows affected by naturally occurring subclinical mastitis by Streptococcus agalactiae (n = 12) and Prototheca spp. (n = 11), to better understand the role of SC methylome in the host response to disease. Differentially methylated regions (DMR) were evaluated comparing: (1) Strep. agalactiae-infected versus healthy; (2) Prototheca-infected versus healthy, and (3) mastitis versus healthy and (4) Strep. agalactiae-infected versus Prototheca-infected. The functional analysis was performed at 2 levels. To begin with, we extracted differentially methylated genes (DMG) from promoter DMR, which were analyzed using the Cytoscape ClueGO plug-in. Coupled with this DMG-driven approach, all the genes associated with promoter-methylated regions were fed to the Pathifier algorithm. From the DMR analysis, we identified 1,081 hypermethylated and 361 hypomethylated promoter regions in Strep. agalactiae-infected animals, while 1,514 hypermethylated and 358 hypomethylated promoter regions were identified in Prototheca-infected animals, when compared with the healthy controls. When considering infected animals as a whole group (regardless of the pathogen), we found 1,576 hypermethylated and 460 hypomethylated promoter regions. Both pathogens were associated with methylation differences in genes involved in pathways related to meiosis, reproduction and tissue remodeling. Exploring the whole methylome, in subclinically infected cows we observed a strong deregulation of immune-related pathways, such as nuclear factor kB and toll-like receptors signaling pathways, and of energy-related pathways such as the tricarboxylic acid cycle and unsaturated fatty acid biosynthesis. In conclusion, no evident pathogen-specific SC methylome signature was detected in the present study. Overall, we observed a clear regulation of host immune response driven by DNA methylation upon subclinical mastitis. Further studies on a larger cohort of animals are needed to validate our results and to possibly identify a unique SC methylome that signifies pathogen-specific alterations.
Collapse
Affiliation(s)
- D Giannuzzi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| | - E Capra
- Institute of Agricultural Biology and Biotechnology, National Research Council (IBBA CNR), 26900, Lodi, Italy
| | - V Bisutti
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy.
| | - A Vanzin
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| | - P Ajmone Marsan
- Department of Animal Science, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, 29122, Piacenza, Italy
| | - A Cecchinato
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| | - S Pegolo
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| |
Collapse
|
5
|
Eshwar V, Kamath A. Assessment of safety profile of secukinumab in real-world scenario using United States food and drug administration adverse event reporting system database. Sci Rep 2024; 14:1222. [PMID: 38216608 PMCID: PMC10786882 DOI: 10.1038/s41598-023-50013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 12/14/2023] [Indexed: 01/14/2024] Open
Abstract
Secukinumab is an anti-IL-17 monoclonal antibody approved for treating psoriasis and various arthritides. A comprehensive evaluation of its safety, especially in a real-world setting, is necessary. This study aimed to describe the adverse events (AE) associated with secukinumab use using the United States Food and Drug Administration Adverse Event Reporting System (FAERS) database. FAERS data files containing AE reports from 2015 to 2021 were downloaded for data mining. Primary or secondary suspect medications indicated for psoriasis were identified and analyzed. Medical dictionary for regulatory activities (MedDRA version 24.1) was used to analyze the AE terms. To detect potential safety signals of AE from secukinumab use, disproportionality analysis was used. A total of 365,590 adverse event reports were identified; of these, 44,761 reports involved the use of secukinumab. Safety signals were identified for ocular infections and gastrointestinal adverse events at the standardised MedDRA query level. Safety signals for oral candidiasis, oral herpes, conjunctivitis, eye infections, and ulcerative colitis were identified at the preferred term level. The findings of our study are consistent with those of earlier studies, such as the increased risk of infections and inflammatory bowel disease. However, our study also identified additional safety signals that need to be further evaluated.
Collapse
Affiliation(s)
- Vishnu Eshwar
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
6
|
Nejabat M, Heydari M, Motamedifar M, Foroozanfar Z, Fard SA, Hashempour A, Nazari N, Rezaei E, Heydari Z. Association of polymorphism of IL-17A, IL-17F, and IL-6 with Toxoplasma gondii infection susceptibility in HIV/AIDS patients in Shiraz, southern Iran. Immun Inflamm Dis 2024; 12:e1117. [PMID: 38270309 PMCID: PMC10777877 DOI: 10.1002/iid3.1117] [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: 09/08/2023] [Revised: 10/31/2023] [Accepted: 12/09/2023] [Indexed: 01/26/2024] Open
Abstract
INTRODUCTION Toxoplasma gondii infection is considered as one of the most important opportunistic infections and cause of death in HIV patients. METHODS In this cross-sectional study, 334 HIV positive patients were included. The molecular test was performed by the restriction fragment length polymorphism-polymerase chain reaction method. Allelic frequency, haplotype analyses, and linkage disequilibrium were calculated. The odds ratio was calculated. The linear regression model was used to analysis of interleukin (IL)-17A, IL-17F, and IL-6 single-nucleotide polymorphism genotypes in HIV patients with and without toxoplasmosis. RESULTS In total, 95 tested'patients (28.4%) were positive for toxoplasmosis. The risk of toxoplasma infection in the current study did not correlate with IL-17 and IL-6 polymorphism and the risk of contracting toxoplasma was also not significantly correlated in this study. There was no association between the frequency of alleles and the risk of toxoplasma infection in IL-17 haplotype analysis. CONCLUSION The findings of this study revealed that there were significant differences in the serum levels of IL-6 and IL-17A, but not IL-17F, between the case and control groups in various genetic models. However, these polymorphisms did not show a significant relationship with toxoplasma infection in HIV-positive patients. This study represents the first investigation in Iran to explore the role of IL-6 and IL-17 polymorphisms in toxoplasma infection among HIV-positive patients.
Collapse
Affiliation(s)
- Maryam Nejabat
- HIV/AIDS Research Center, Institute of HealthShiraz University of Medical SciencesShirazIran
| | - Mohammadreza Heydari
- HIV/AIDS Research Center, Institute of HealthShiraz University of Medical SciencesShirazIran
| | - Mohammad Motamedifar
- HIV/AIDS Research Center, Institute of HealthShiraz University of Medical SciencesShirazIran
- Department of Bacteriology and Virology, Shiraz Medical SchoolShiraz University of Medical SciencesShirazFarsIran
| | - Zohre Foroozanfar
- HIV/AIDS Research Center, Institute of HealthShiraz University of Medical SciencesShirazIran
| | - Saeid Amirizadeh Fard
- Virology Section, Diagnostic Laboratory Sciences and Technology Research CenterSchool of Paramedical SciencesShirazIran
| | - Ava Hashempour
- HIV/AIDS Research Center, Institute of HealthShiraz University of Medical SciencesShirazIran
| | - Nazani Nazari
- Department of Immunology, Shiraz Medical SchoolShiraz University of Medical SciencesShirazIran
| | - Esmaeil Rezaei
- Department of Biochemistry, Faculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Zahra Heydari
- Department of Cell and Molecular Biology, Faculty of Life Sciences and BiotechnologyShahid Behesti UniversityTehranIran
| |
Collapse
|
7
|
Casaux ML, Neto WS, Schild CO, Costa RA, Macías-Rioseco M, Caffarena RD, Silveira CS, Aráoz V, Díaz BD, Giannitti F, Fraga M. Epidemiological and clinicopathological findings in 15 fatal outbreaks of salmonellosis in dairy calves and virulence genes in the causative Salmonella enterica Typhimurium and Dublin strains. Braz J Microbiol 2023; 54:475-490. [PMID: 36602750 PMCID: PMC9943839 DOI: 10.1007/s42770-022-00898-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
Salmonella enterica is a major food-borne pathogen that affects cattle-rearing systems worldwide. Little information is available on the epidemiology and pathology of salmonellosis and the virulence genes (VGs) carried by Salmonella in spontaneous outbreaks in cattle. We describe epidemiological findings in 15 fatal outbreaks of salmonellosis in Uruguayan dairy farms and the age, clinical signs, and pathology in 20 affected calves. We also describe the serotypes and frequencies of 17 VGs in the causative Salmonella strains and explore their associations with epidemiological, clinical, and pathological findings. Salmonella Typhimurium and Dublin were identified in 11/15 and 4/15 outbreaks, respectively. The most frequent reason for consultation was digestive disease (8 outbreaks caused by S. Typhimurium), followed by sudden death (4 outbreaks, 3 caused by S. Dublin). Morbidity, mortality, and lethality ranged 4.8-100%, 3.8-78.9%, and 10-100%, without significant differences between serotypes. Diarrhea, the most common clinical sign (14 cases), was associated with the Typhimurium serotype (OR = 26.95), especially in ≤ 30-day-old calves with fibrinous enteritis as the main autopsy finding. The Dublin serotype affected ≥ 50-day-old calves and was associated with fibrinosuppurative splenitis (p = 0.01) and tubulointerstitial nephritis (OR = 48.95). The chances of the Dublin serotype increased significantly with age. There was low variability of VG across serotypes. The pefA gene was associated with the Typhimurium serotype (OR = 21.95), macroscopic enteritis (p = 0.03), and microscopic fibrinosuppurative splenitis (p = 0.04). Understanding the epidemiology, pathology, and virulence of S. enterica at the farm level is key to delineating prevention and control strategies to mitigate its impact on animal and human health.
Collapse
Affiliation(s)
- M L Casaux
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
| | - W Santiago Neto
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
| | - C O Schild
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
| | - R A Costa
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
| | - M Macías-Rioseco
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
- California Animal Health and Food Safety (CAHFS) Laboratory, Tulare Branch, University of California at Davis, Tulare, CA, USA
| | - R D Caffarena
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
- Unidad Académica Salud de los Rumiantes, Facultad de Veterinaria, Universidad de la República, Montevideo, Uruguay
| | - C S Silveira
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
| | - V Aráoz
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
| | - B Doncel Díaz
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
- Laboratorio de Patología Veterinaria Facultad de Medicina Veterinaria y de Zootecnia, Universidad Nacional de Colombia, Bogotá, Colombia
| | - F Giannitti
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay
| | - M Fraga
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Estación Experimental La Estanzuela, Ruta 50, Km 11, El Semillero, Uruguay.
| |
Collapse
|
8
|
Inflammation-mediated tissue damage in pulmonary tuberculosis and host-directed therapeutic strategies. Semin Immunol 2023; 65:101672. [PMID: 36469987 DOI: 10.1016/j.smim.2022.101672] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 12/04/2022]
Abstract
Treatment of tuberculosis (TB) involves the administration of anti-mycobacterial drugs for several months. The emergence of drug-resistant strains of Mycobacterium tuberculosis (Mtb, the causative agent) together with increased disease severity in people with co-morbidities such as diabetes mellitus and HIV have hampered efforts to reduce case fatality. In severe disease, TB pathology is largely attributable to over-exuberant host immune responses targeted at controlling bacterial replication. Non-resolving inflammation driven by host pro-inflammatory mediators in response to high bacterial load leads to pulmonary pathology including cavitation and fibrosis. The need to improve clinical outcomes and reduce treatment times has led to a two-pronged approach involving the development of novel antimicrobials as well as host-directed therapies (HDT) that favourably modulate immune responses to Mtb. HDT strategies incorporate aspects of immune modulation aimed at downregulating non-productive inflammatory responses and augmenting antimicrobial effector mechanisms to minimise pulmonary pathology and accelerate symptom resolution. HDT in combination with existing antimycobacterial agents offers a potentially promising strategy to improve the long-term outcome for TB patients. In this review, we describe components of the host immune response that contribute to inflammation and tissue damage in pulmonary TB, including cytokines, matrix metalloproteinases, lipid mediators, and neutrophil extracellular traps. We then proceed to review HDT directed at these pathways.
Collapse
|
9
|
Ali S, Majid S, Ali MN, Banday MZ, Taing S, Wani S, Almuqbil M, Alshehri S, Shamim K, Rehman MU. Immunogenetic Role of IL17A Polymorphism in the Pathogenesis of Recurrent Miscarriage. J Clin Med 2022; 11:jcm11247448. [PMID: 36556060 PMCID: PMC9785316 DOI: 10.3390/jcm11247448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Interleukin-17A (IL17A) is a proinflammatory cytokine and is assumed to play an important role in fetal rejection. In order to evaluate the potential role of IL17A polymorphism in the pathogenesis of recurrent miscarriage (RM), serum IL17A levels were estimated by ELISA. Single-nucleotide polymorphism was assessed by polymerase chain reaction-restriction fragment-length polymorphism (PCR-RFLP) using gene-specific primers and the EcoNI restriction enzyme. Serum IL17A levels were nonsignificantly (p > 0.5) low in RM patients compared with the control group. IL17A gene amplification by PCR yielded the undigested product of 815 bp, and its digestion with EcoNI enzyme produced 815, 529, 286, and 270 bp fragments for the GG genotype; 529, 286, and 270 bp fragments for the GA genotype; and 529 and 286 bp fragments for the AA genotype. The genotype frequency between the RM and control groups exhibited a significant difference (p = 0.001), whereas no significant difference was observed between allele frequencies in the two groups (p = 0.0954). These data suggest that the IL17A gene polymorphism exhibits no significant effect on IL17A gene expression. However, it significantly decreases and increases RM risk in the homozygous and recessive models, suggesting its potential pregnancy-protecting and -harming roles in the AA and GA + GG genotypes, respectively.
Collapse
Affiliation(s)
- Shafat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, Srinagar 190006, J&K, India
- Department of Biochemistry, Government Medical College, Srinagar 190010, J&K, India
- Multidisciplinary Research Unit, Government Medical College, Srinagar 190010, J&K, India
- Department of Obstetrics and Gynaecology, Government Medical College-Associated Lalla Ded Hospital, Srinagar 190008, J&K, India
| | - Sabhiya Majid
- Department of Biochemistry, Government Medical College, Srinagar 190010, J&K, India
- Multidisciplinary Research Unit, Government Medical College, Srinagar 190010, J&K, India
- Correspondence: (S.M.); (M.N.A.); (M.U.R.)
| | - Md. Niamat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, Srinagar 190006, J&K, India
- Correspondence: (S.M.); (M.N.A.); (M.U.R.)
| | - Mujeeb Zafar Banday
- Department of Biochemistry, Government Medical College, Srinagar 190010, J&K, India
| | - Shahnaz Taing
- Department of Obstetrics and Gynaecology, Government Medical College-Associated Lalla Ded Hospital, Srinagar 190008, J&K, India
| | - Saima Wani
- Department of Obstetrics and Gynaecology, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Soura, Srinagar 190011, J&K, India
| | - Mansour Almuqbil
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Kashif Shamim
- National Centre for Natural Products Research, University of Mississippi, Oxford, MS 38677, USA
| | - Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence: (S.M.); (M.N.A.); (M.U.R.)
| |
Collapse
|
10
|
Eshwar V, Kamath A, Shastry R, Shenoy AK, Kamath P. A Review of the Safety of Interleukin-17A Inhibitor Secukinumab. Pharmaceuticals (Basel) 2022; 15:1365. [PMID: 36355537 PMCID: PMC9695424 DOI: 10.3390/ph15111365] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Secukinumab is an anti-interleukin (IL)-17A IgG1-κ monoclonal antibody approved for psoriasis, psoriatic arthritis, and ankylosing spondylitis. Its efficacy is well documented, but the complete safety profile of secukinumab, especially on long-term use, needs to be studied. IL-17 inhibitors increase the risk of infections, especially respiratory tract infections and candidiasis, and inflammatory bowel disease; the causal relationships are well described. However, evidence regarding the other adverse events is scarce, and causal associations between the adverse events and the biologic remain unresolved. This review aims to present a narrative perspective on the safety of secukinumab and identify some key areas where the safety of secukinumab may potentially be useful in understanding the scope of secukinumab therapy and making informed clinical decisions.
Collapse
Affiliation(s)
| | - Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | | | | | | |
Collapse
|
11
|
Mucosal immunization with a delta-inulin adjuvanted recombinant spike vaccine elicits lung-resident immune memory and protects mice against SARS-CoV-2. Mucosal Immunol 2022; 15:1405-1415. [PMID: 36411332 PMCID: PMC9676795 DOI: 10.1038/s41385-022-00578-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/12/2022] [Accepted: 10/09/2022] [Indexed: 11/22/2022]
Abstract
Multiple SARS-CoV-2 vaccine candidates have been approved for use and have had a major impact on the COVID-19 pandemic. There remains, however, a significant need for vaccines that are safe, easily transportable and protective against infection, as well as disease. Mucosal vaccination is favored for its ability to induce immune memory at the site of infection, making it appealing for SARS-CoV-2 vaccine strategies. In this study we performed in-depth analysis of the immune responses in mice to a subunit recombinant spike protein vaccine formulated with the delta-inulin adjuvant Advax when administered intratracheally (IT), versus intramuscular delivery (IM). Both routes produced robust neutralizing antibody titers (NAb) and generated sterilizing immunity against SARS-CoV-2. IT delivery, however, produced significantly higher systemic and lung-local NAb that resisted waning up to six months post vaccination, and only IT delivery generated inducible bronchus-associated lymphoid tissue (iBALT), a site of lymphocyte antigen presentation and proliferation. This was coupled with robust and long-lasting lung tissue-resident memory CD4+ and CD8+ T cells that were not observed in IM-vaccinated mice. This study provides a detailed view of the lung-resident cellular response to IT vaccination against SARS-CoV-2 and demonstrates the importance of delivery site selection in the development of vaccine candidates.
Collapse
|
12
|
Akter S, Chauhan KS, Dunlap MD, Choreño-Parra JA, Lu L, Esaulova E, Zúñiga J, Artyomov MN, Kaushal D, Khader SA. Mycobacterium tuberculosis infection drives a type I IFN signature in lung lymphocytes. Cell Rep 2022; 39:110983. [PMID: 35732116 PMCID: PMC9616001 DOI: 10.1016/j.celrep.2022.110983] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) infects 25% of the world's population and causes tuberculosis (TB), which is a leading cause of death globally. A clear understanding of the dynamics of immune response at the cellular level is crucial to design better strategies to control TB. We use the single-cell RNA sequencing approach on lung lymphocytes derived from healthy and Mtb-infected mice. Our results show the enrichment of the type I IFN signature among the lymphoid cell clusters, as well as heat shock responses in natural killer (NK) cells from Mtb-infected mice lungs. We identify Ly6A as a lymphoid cell activation marker and validate its upregulation in activated lymphoid cells following infection. The cross-analysis of the type I IFN signature in human TB-infected peripheral blood samples further validates our results. These findings contribute toward understanding and characterizing the transcriptional parameters at a single-cell depth in a highly relevant and reproducible mouse model of TB.
Collapse
Affiliation(s)
- Sadia Akter
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA,These authors contributed equally
| | - Kuldeep S. Chauhan
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA,These authors contributed equally
| | - Micah D. Dunlap
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - José Alberto Choreño-Parra
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA,Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City 14080, Mexico,Laboratorio de Inmunoquímica I, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Lan Lu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ekaterina Esaulova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joaquin Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City 14080, Mexico,Laboratorio de Inmunoquímica I, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Maxim N. Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA,Lead contact,Correspondence: (D.K.), (S.A.K.) https://doi.org/10.1016/j.celrep.2022.110983
| |
Collapse
|
13
|
Autoimmunity in human CE: Correlative with the fertility status of the CE cyst. Helminthologia 2022; 59:1-17. [PMID: 35601761 PMCID: PMC9075880 DOI: 10.2478/helm-2022-0011] [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: 09/11/2020] [Accepted: 01/27/2022] [Indexed: 11/20/2022] Open
Abstract
Cystic echinococcosis is speculated to exert several immune-evasion strategies involving autoimmune-phenomena. We evaluated the hypothesizes that the prevalence of autoantibodies increases in the sera of CE patients that may evidence the association between the parasite and autoimmune diseases. Sera from 63 subjects at distinct types of CE cyst fertility were investigated for antinuclear antibodies (ANA), and anti-CCP antibodies. Plasma levels and cellular production of IL-17A cytokine were specifically defined as being assumed to prime for autoimmunity. Healthy-controls were age and gender-matched to test sera. ANA expressions inside the surgically removed metacestode and adventitial layer were also assayed. Out of 63 patients, 35 % had fertile highly viable cysts (group-1), 41 % had fertile low viable cysts (group-2) and 24 % had non-fertile cysts (group-3). A four-fold increase in ANA sera-levels was detected in group-1 compared with their controls (p-value 0.001) while anti-CCP levels were of insignificant differences. In group-2 and group-3, no significant differences were detected between ANA and anti-CCP sera-levels in CE patients and their controls. IL-17A sera-levels in group-1 and group- 2 were significantly higher than their healthy-controls while being of insignificant differences in group-3, p-value= 0.300. No association was detected between sera-levels of IL-17A and ANA as well as anti-CCP antibodies. Interestingly, relative IL-17A cellular expression associated positive ANA deposition in the parasite cells and adventitial layer. Collectively, based on the parasite fertility, IL-17A and ANA seemed to be involved in the host immune defenses against CE. There is no association between CE and anti-CCP antibodies.
Collapse
|
14
|
El Saftawy EA, Abdelmoktader A, Sabry MM, Alghandour SM. Histological and immunological insights to hydatid disease in camels. Vet Parasitol Reg Stud Reports 2021; 26:100635. [PMID: 34879946 DOI: 10.1016/j.vprsr.2021.100635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 08/31/2021] [Accepted: 09/06/2021] [Indexed: 11/15/2022]
Abstract
PURPOSE To investigate the immuno-histological evidences in viable and non-viable hydatid cysts obtained from naturally infected camels. METHODS A cohort study (February 2018-December 2019), a total of 15 hydatidosis-infected camels from slaughter houses in Cairo were involved. Specimens were investigated for parasite viability, liver histological changes, IL-17A cytokine immunohistochemical expressions in the adventitial layer, and the anti-nuclear antibodies (ANAs) immunofluorescent expression in the metacestode's structures. Real-Time Quantitative -Morphocytometry and SPSS were utilized. RESULTS Multi-focal lesions and high viability were found in 60% of the cases. Overall accumulation of collagen associated the parasite establishment that involved infiltrations of mononuclear cells with significantly increased IL-17A expression. Interestingly, the ANAs appeared to have a role in the immune-defense against the metacestode showing different patterns. ANAs production correlated with IL-17A expression and the viability of the parasite. CONCLUSION IL-17A responses in hydatidosis is associated with collagen deposition and ANA production as a sort of anti-parasite immunity in a viability dependent manner.
Collapse
Affiliation(s)
- Enas A El Saftawy
- Medical Parasitology Department, Faculty of Medicine, Cairo University, Cairo, Egypt; Medical Parasitology Department, Faculty of Medicine, Armed Forces College of Medicine, Cairo, Egypt.
| | - Abdelrahman Abdelmoktader
- Medical Microbiology and Immunology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Marwa Mohamed Sabry
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
15
|
Sahu U, Khare P. Role of interleukin-17 in human papillomavirus infection and associated malignancies. Microb Pathog 2021; 161:105294. [PMID: 34798279 DOI: 10.1016/j.micpath.2021.105294] [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: 07/23/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023]
Abstract
Human papillomavirus infection is among the leading viral infections in the world, causing severe mortality and morbidity. The virus mainly targets the female genital tract-cervix, vulva, anus but it is also reported to infect the lungs and oropharyngeal region of the body. The host immune response plays a vital role in the persistence of viral infection. Interleukin 17 (IL-17) is mainly secreted by Th17 cells and mediates the immune response that enhances the disease severity in HPV infection. IL-17 is reported to promote lesions and tumour progression by creating a hyperinflammatory condition leading to cancer. The current minireview summarizes the pathogenic role of IL-17 in HPV infection and HPV-induced malignancies. Further study on IL-17 associated pathology of HPV infection would be useful in developing therapeutic measures.
Collapse
Affiliation(s)
- Utkarsha Sahu
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Prashant Khare
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India.
| |
Collapse
|
16
|
Leyva-Castillo JM, Das M, Kane J, Strakosha M, Singh S, Wong DSH, Horswill AR, Karasuyama H, Brombacher F, Miller LS, Geha RS. Basophil-derived IL-4 promotes cutaneous Staphylococcus aureus infection. JCI Insight 2021; 6:149953. [PMID: 34747366 PMCID: PMC8663570 DOI: 10.1172/jci.insight.149953] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022] Open
Abstract
Superficial cutaneous Staphylococcus aureus (S. aureus) infection in humans can lead to soft tissue infection, an important cause of morbidity and mortality. IL-17A production by skin TCRγδ+ cells in response to IL-1 and IL-23 produced by epithelial and immune cells is important for restraining S. aureus skin infection. How S. aureus evades this cutaneous innate immune response to establish infection is not clear. Here we show that mechanical injury of mouse skin by tape stripping predisposed mice to superficial skin infection with S. aureus. Topical application of S. aureus to tape-stripped skin caused cutaneous influx of basophils and increased Il4 expression. This basophil-derived IL-4 inhibited cutaneous IL-17A production by TCRγδ+ cells and promoted S. aureus infection of tape-stripped skin. We demonstrate that IL-4 acted on multiple checkpoints that suppress the cutaneous IL-17A response. It reduced Il1 and Il23 expression by keratinocytes, inhibited IL-1+IL-23-driven IL-17A production by TCRγδ+ cells, and impaired IL-17A-driven induction of neutrophil-attracting chemokines by keratinocytes. IL-4 receptor blockade is shown to promote Il17a expression and enhance bacterial clearance in tape-stripped mouse skin exposed to S. aureus, suggesting that it could serve as a therapeutic approach to prevent skin and soft tissue infection.
Collapse
Affiliation(s)
- Juan-Manuel Leyva-Castillo
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Mrinmoy Das
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Kane
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Strakosha
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sonal Singh
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Sen Hoi Wong
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology Cape Town Component and Health Science Faculty, University of Cape Town, Cape Town, South Africa
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Yao YE, Qin CC, Yang CM, Huang TX. γδT17/γδTreg cell subsets: a new paradigm for asthma treatment. J Asthma 2021; 59:2028-2038. [PMID: 34634976 DOI: 10.1080/02770903.2021.1980585] [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] [Indexed: 12/24/2022]
Abstract
Bronchial asthma (abbreviated as asthma), is a heterogeneous disease characterized by chronic airway inflammation and airway hyperresponsiveness. The main characteristics of asthma include variable reversible airflow limitation and airway remodeling. The pathogenesis of asthma is still unclear. Th1/Th2 imbalance, Th1 deficiency and Th2 hyperfunction are classic pathophysiological mechanisms of asthma. Some studies have shown that the imbalance of the Th1/Th2 cellular immune model and Th17/Treg imbalance play a key role in the occurrence and development of asthma; however, these imbalances do not fully explain the disease. In recent years, studies have shown that γδT and γδT17 cells are involved in the pathogenesis of asthma. γδTreg has a potential immunosuppressive function, but its regulatory mechanisms have not been fully elucidated. In this paper, we reviewed the role of γδT17/γδTreg cells in bronchial asthma, including the mechanisms of their development and activation. Here we propose that γδT17/Treg cell subsets contribute to the occurrence and development of asthma, constituting a novel potential target for asthma treatment.
Collapse
Affiliation(s)
- Yi-En Yao
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Cai-Cheng Qin
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chao-Mian Yang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tian-Xia Huang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
18
|
Pre-treatment with chicken IL-17A secreted by bioengineered LAB vector protects chicken embryo fibroblasts against Influenza Type A Virus (IAV) infection. Mol Immunol 2021; 140:106-119. [PMID: 34678620 DOI: 10.1016/j.molimm.2021.10.003] [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: 05/28/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 01/01/2023]
Abstract
The recent advances in our understanding of the host factors in orchestrating qualitatively different immune responses against influenza Type A virus (IAV) have changed the perception of conventional approaches for controlling avian influenza virus (AIV) infection in chickens. Given that infection-induced pathogenicity and replication of influenza virus largely rely on regulating host immune responses, immunoregulatory cytokine profiles often determine the disease outcomes. However, in contrast to the function of other inflammatory cytokines, interleukin-17A (IL-17A) has been described as a 'double-edged sword', indicating that in addition to antiviral host responses, IL-17A has a distinct role in promoting viral infection. Therefore, in the present study, we investigated the chicken IL-17A mediated antiviral immune effects on IAVs infection in primary chicken embryo fibroblasts cells (CEFs). To this end, we first bioengineered a food-grade Lactic Acid Producing Bacteria (LAB), Lactococcus lactis (L. lactis), secreting bioactive recombinant chicken IL-17A (sChIL-17A). Next, the functionality of sChIL-17A was confirmed by transcriptional upregulation of several genes associated with antiviral host responses, including granulocyte-monocyte colony-stimulating factor (GM-CSF) (CSF3 in the chickens), interleukin-6 (IL-6), interferon-α (IFN-α), -β and -γ genes in primary CEFs cells. Consistent with our hypothesis that such a pro-inflammatory state may translate to immunoprotection against IAVs infection, we observed that sChIL-17A pre-treatment could significantly limit the viral replication and protect the primary CEFs cells against two heterotypic IAVs such as A/turkey/Wisconsin/1/1966(H9N2) and A/PR/8/1934(H1N1). Together, the data presented in this work suggest that exogenous application of sChIL-17A secreted by modified LAB vector may represent an alternative strategy for improving antiviral immunity against avian influenza virus infection in chickens.
Collapse
|
19
|
Asad M, Sabur A, Kamran M, Shadab M, Das S, Ali N. Effector functions of Th17 cells are regulated by IL-35 and TGF-β in visceral leishmaniasis. FASEB J 2021; 35:e21755. [PMID: 34383962 DOI: 10.1096/fj.202002356rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 06/05/2021] [Accepted: 06/10/2021] [Indexed: 11/11/2022]
Abstract
Visceral leishmaniasis (VL) is a debilitating human pathogenesis in which the body's immune functions are severely compromised. Various subsets of T cells, including Th17 cells are important regulators of immune responses observed in various pathologies. The role of Th17 cells and its correlation with immuno-regulatory cytokines are however not well understood in human VL. Herein we studied how IL-17 is associated with the progression of Leishmania donovani infection using murine model of VL. We found induction of a strong IL-17 response at the early phase of infection which progressively reduced to basal level during chronic VL. The mechanistic study of this behavior was found to be linked with the role of regulatory T cells (CD4+ CD25+ T cells) that suppresses the proliferation of the Th17 cell population. Moreover, TGF-β and IL-35 derived from CD4+ CD25+ T cells are the key mediators for the downregulation of IL-17 during chronic VL. Thus, this study points to an antagonistic effect of Tregs and Th17 cells that can be used for designing better therapeutic and preventive strategies against leishmaniasis.
Collapse
Affiliation(s)
- Mohammad Asad
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India.,Department of Medicine, Albert Einstein College of Medicine/ Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Abdus Sabur
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India.,Raiganj Surendranath Mahavidyalaya, Affiliated to University of Gour Banga, Uttar Dinajpur, Raiganj, West Bengal, 733134, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Md Shadab
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India.,Department of Pediatrics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Sonali Das
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
20
|
Kridin K, Schonmann Y, Tzur Bitan D, Damiani G, Peretz A, Weinstein O, Cohen AD. Coronavirus Disease 2019 (COVID-19)-Associated Hospitalization and Mortality in Patients with Psoriasis: A Population-Based Study. Am J Clin Dermatol 2021; 22:709-718. [PMID: 34060006 PMCID: PMC8166379 DOI: 10.1007/s40257-021-00605-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2021] [Indexed: 12/16/2022]
Abstract
Background The impact of immune-related conditions on the outcomes of coronavirus disease 2019 (COVID-19) is poorly understood. Determinants of COVID-19 outcomes among patients with psoriasis are yet to be established. Objective Th objective of this study was to characterize a large cohort of patients with psoriasis with COVID-19 and to identify predictors of COVID-19-associated hospitalization and mortality. Methods A population-based nested case-control study was performed using the computerized database of Clalit Health Services, Israel. Multivariable logistic regression was used to estimate odds ratios (ORs) and 95% confidence (CIs) of predictors for COVID-19-associated hospitalization and mortality. Results The study population included 3151 patients with psoriasis who tested positive for COVID-19. Subclinical COVID-19 infection occurred in 2818 (89.4%) of the patients while 122 (3.9%), 71 (2.3%), 123 (3.9%), and 16 (0.5%) of the patients experienced a mild, moderate, severe, and critical disease, respectively. Overall, 332 (10.5%) patients were hospitalized and 50 (1.6%) patients died because of COVID-19 complications. Intake of methotrexate independently predicted COVID-19-associated hospitalization (adjusted OR 2.30; 95% CI 1.11–4.78; p = 0.025). Use of biologic agents was not associated with COVID-19-associated hospitalization (OR 0.75; 95% CI 0.32–1.73; p = 0.491) or mortality (OR 0.85; 95% CI 0.12–6.21; p = 0.870). Older age, the presence of comorbid cardiovascular diseases, metabolic syndrome, chronic obstructive pulmonary disease, and chronic renal failure independently predicted both COVID-19-associated hospitalization and mortality. Conclusions The use of oral methotrexate was associated with an increased odds of COVID-associated hospitalization, whereas the use of biologic drugs was not associated with worse outcomes of COVID-19 among patients with psoriasis. Supplementary Information The online version contains supplementary material available at 10.1007/s40257-021-00605-8.
Collapse
Affiliation(s)
- Khalaf Kridin
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | | | - Dana Tzur Bitan
- Department of Behavioral Sciences, Ariel University, Ariel, Israel
| | - Giovanni Damiani
- Clinical Dermatology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | | | - Orly Weinstein
- Clalit Health Services, Tel-Aviv, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Ben-Gurion Ave, Beer Sheva, Israel
| | - Arnon D Cohen
- Clalit Health Services, Tel-Aviv, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Ben-Gurion Ave, Beer Sheva, Israel
| |
Collapse
|
21
|
Wu Q, Cui D, Chao X, Chen P, Liu J, Wang Y, Su T, Li M, Xu R, Zhu Y, Zhang Y. Transcriptome Analysis Identifies Strategies Targeting Immune Response-Related Pathways to Control Enterotoxigenic Escherichia coli Infection in Porcine Intestinal Epithelial Cells. Front Vet Sci 2021; 8:677897. [PMID: 34447800 PMCID: PMC8383179 DOI: 10.3389/fvets.2021.677897] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/05/2021] [Indexed: 11/13/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important cause of post-weaning diarrhea (PWD) worldwide, resulting in huge economic losses to the swine industry worldwide. In this study, to understand the pathogenesis, the transcriptomic analysis was performed to explore the biological processes (BP) in porcine intestinal epithelial J2 cells infected with an emerging ETEC strain isolated from weaned pigs with diarrhea. Under the criteria of |fold change| (FC) ≥ 2 and P < 0.05 with false discovery rate < 0.05, a total of 131 referenced and 19 novel differentially expressed genes (DEGs) were identified after ETEC infection, including 96 upregulated DEGs and 54 downregulated DEGs. The Gene Ontology (GO) analysis of DEGs showed that ETEC evoked BP specifically involved in response to lipopolysaccharide (LPS) and negative regulation of intracellular signal transduction. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that immune response-related pathways were mainly enriched in J2 cells after ETEC infection, in which tumor necrosis factor (TNF), interleukin 17, and mitogen-activated protein kinase (MAPK) signaling pathways possessed the highest rich factor, followed by nucleotide-binding and oligomerization domain-like receptor (NLRs), C-type lectin receptor (CLR), cytokine–cytokine receptor interaction, and Toll-like receptor (TLR), and nuclear factor kappa-B (NF-κB) signaling pathways. Furthermore, 30 of 131 referenced DEGs, especially the nuclear transcription factor AP-1 and NF-κB, participate in the immune response to infection through an integral signal cascade and can be target molecules for prevention and control of enteric ETEC infection by probiotic Lactobacillus reuteri. Our data provide a comprehensive insight into the immune response of porcine intestinal epithelial cells (IECs) to ETEC infection and advance the identification of targets for prevention and control of ETEC-related PWD.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.,Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Defeng Cui
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.,Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Xinyu Chao
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Peng Chen
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Jiaxuan Liu
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Yiding Wang
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Tongjian Su
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Meng Li
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Ruyu Xu
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Yaohong Zhu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yonghong Zhang
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.,Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
22
|
Jia YH, Liu JQ, Wang YC, Wang HT, Tao K, Zheng Z, Hu DH. [Research advances on the regulation of interleukin-17 signal transduction and the implication of interleukin-17 in sepsis]. ZHONGHUA SHAO SHANG ZA ZHI = ZHONGHUA SHAOSHANG ZAZHI = CHINESE JOURNAL OF BURNS 2021; 37:675-680. [PMID: 34304410 PMCID: PMC11917318 DOI: 10.3760/cma.j.cn501120-20200515-00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Sepsis remains a leading cause of death in critical patients. Both excessive inflammatory response and long-term immunosuppression can lead to the death of sepsis patients. As a key pro-inflammatory cytokine, interleukin-17 (IL-17) plays an important role in the body's inflammatory response and immune system. The signal transduction of IL-17 is a key link in maintaining the body's health and participating in the onset and development of sepsis. This review mainly summarizes and discusses the regulation of IL-17 signal transduction and pathogenic and protective role of IL-17 in sepsis.
Collapse
Affiliation(s)
- Y H Jia
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - J Q Liu
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - Y C Wang
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - H T Wang
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - K Tao
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - Z Zheng
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - D H Hu
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| |
Collapse
|
23
|
Abstract
Gammaherpesviruses establish lifelong infections in a majority of humans and are associated with B cell lymphomas. IL-17A is a host cytokine that plays a well-established role in the clearance of bacterial and fungal infections; however, the role of IL-17A in viral infections is poorly understood. Gammaherpesviruses establish lifelong infection and are associated with a variety of cancers, including B cell lymphomas. These viruses manipulate the B cell differentiation process to establish lifelong infection in memory B cells. Specifically, gammaherpesviruses infect naive B cells and promote entry of both infected and uninfected naive B cells into germinal centers, where the virus usurps rapid proliferation of germinal center B cells to exponentially increase its cellular latent reservoir. In addition to facilitating the establishment of latent infection, germinal center B cells are thought to be the target of viral transformation. In this study, we have uncovered a novel proviral role of host interleukin 17A (IL-17A), a well-established antibacterial and antifungal factor. Loss of IL-17A signaling attenuated the establishment of chronic gammaherpesvirus infection and gammaherpesvirus-driven germinal center response in a route of inoculation-dependent manner. Further, IL-17A treatment directly supported gammaherpesvirus reactivation and de novo lytic infection. This study is the first demonstration of a multifaceted proviral role of IL-17 signaling.
Collapse
|
24
|
Ergun T, Seckin D, Demir G, Direskeneli H. Secukinumab and infectious adverse effects: A real-life experience of 63 psoriasis patients. Australas J Dermatol 2021; 62:e423-e426. [PMID: 33788261 DOI: 10.1111/ajd.13588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/31/2021] [Accepted: 02/15/2021] [Indexed: 01/07/2023]
Affiliation(s)
- Tulin Ergun
- Department of Dermatology, Marmara University School of Medicine, Istanbul, Turkey
| | - Dilek Seckin
- Department of Dermatology, Marmara University School of Medicine, Istanbul, Turkey
| | - Gizem Demir
- Department of Dermatology, Marmara University School of Medicine, Istanbul, Turkey
| | - Haner Direskeneli
- Department of Rheumatology, Marmara University School of Medicine, Istanbul, Turkey
| |
Collapse
|
25
|
Yiu ZZN, Parisi R, Lunt M, Warren RB, Griffiths CEM, Langan SM, Ashcroft DM. Risk of hospitalization and death due to infection in people with psoriasis: a population-based cohort study using the Clinical Practice Research Datalink. Br J Dermatol 2021; 184:78-86. [PMID: 32222069 DOI: 10.1111/bjd.19052] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Psoriasis is associated with risk factors for serious infections, but the independent relationship between psoriasis and serious infection is as yet unclear. OBJECTIVES To determine whether people with psoriasis have a higher risk of hospitalization due to any infection, respiratory infections, soft-tissue and skin infections, or a higher risk of death due to infection. METHODS We conducted a cohort study of people (≥ 18 years) with psoriasis using the UK Clinical Practice Research Datalink (CPRD GOLD) linked to Hospital Episode Statistics (HES) and Office for National Statistics (ONS) mortality records between 1 April 2003 and 31 December 2016, and matched with up to six comparators on age, sex and general practice. Hospitalization was ascertained from HES records; death was ascertained from ONS mortality records. Stratified Cox proportional hazard models were estimated, with stepwise adjustment in different models for potential confounders or mediators between psoriasis and serious infection. RESULTS There were 69 315 people with psoriasis and 338 620 comparators who were followed up for a median (interquartile range) of 4·9 (5·9) and 5·1 (6·3) years, respectively. People with psoriasis had a higher incidence rate of serious infection [20·5 per 1000 person-years, 95% confidence interval (CI) 20·0-21·0, n = 7631] compared with those without psoriasis (16·1 per 1000 person-years, 95% CI 15·9-16·3, n = 30 761). The fully adjusted hazard ratio for the association between psoriasis and serious infection was 1·36 (95% CI 1·31-1·40), with similar results across the other outcomes. CONCLUSIONS Psoriasis is associated with a small increase in the risk of serious infection. Further research is needed to understand how psoriasis predisposes to a higher risk of infection.
Collapse
Affiliation(s)
- Z Z N Yiu
- Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - R Parisi
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M Lunt
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - R B Warren
- Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - C E M Griffiths
- Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - S M Langan
- Faculty of Epidemiology, and Population Health, London School of Hygiene and Tropical Medicine, London, UK.,Health Data Research UK
| | - D M Ashcroft
- Centre for Pharmacoepidemiology and Drug Safety, School of Health Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
26
|
Silvane L, Celias DP, Romagnoli PA, Maletto BA, Sanchez Vallecillo MF, Chiapello LS, Palma SD, Allemandi DA, Sanabria REF, Pruzzo CI, Motrán CC, Cervi L. A Vaccine Based on Kunitz-Type Molecule Confers Protection Against Fasciola hepatica Challenge by Inducing IFN-γ and Antibody Immune Responses Through IL-17A Production. Front Immunol 2020; 11:2087. [PMID: 33193292 PMCID: PMC7641617 DOI: 10.3389/fimmu.2020.02087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/31/2020] [Indexed: 01/03/2023] Open
Abstract
Fasciola hepatica is helminth parasite found around the world that causes fasciolosis, a chronic disease affecting mainly cattle, sheep, and occasionally humans. Triclabendazole is the drug of choice to treat this parasite. However, the continuous use of this drug has led to the development of parasite resistance and, consequently, the limitation of its effectiveness. Hence, vaccination appears as an attractive option to develop. In this work, we evaluated the potential of F. hepatica Kunitz-type molecule (FhKTM) as an antigen formulated with a liquid crystal nanostructure formed by self-assembly of 6-O-ascorbyl palmitate ester (Coa-ASC16) and the synthetic oligodeoxynucleotide containing unmethylated cytosine-guanine motifs (CpG-ODN) during an experimental model of fasciolosis in mice, and we further dissected the immune response associated with host protection. Our results showed that immunization of mice with FhKTM/CpG-ODN/Coa-ASC16 induces protection against F. hepatica challenge by preventing liver damage and improving survival after F. hepatica infection. FhKTM/CpG-ODN/Coa-ASC16-immunized mice elicited potent IFN-γ and IL-17A with high levels of antigen-specific IgG1, IgG2a, and IgA serum antibodies. Strikingly, IL-17A blockade during infection decreased IgG2a and IgA antibody levels as well as IFN-γ production, leading to an increase in mortality of vaccinated mice. The present study highlights the potential of a new vaccine formulation to improve control and help the eradication of F. hepatica infection, with potential applications for natural hosts such as cattle and sheep.
Collapse
Affiliation(s)
- Leonardo Silvane
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Daiana Pamela Celias
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Pablo Alberto Romagnoli
- Centro de Investigación en Medicina Traslacional Severo Amuchastegui (CIMETSA), Córdoba, Argentina.,Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Belkys Angélica Maletto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - María Fernanda Sanchez Vallecillo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Silvina Chiapello
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Santiago Daniel Palma
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Unidad de Investigación y desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Daniel Alberto Allemandi
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Unidad de Investigación y desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Rodrigo Eduardo Fabrizio Sanabria
- Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, La Plata, Argentina.,Instituto Tecnológico Chascomús (INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín (CONICET/UNSAM), Chascomús, Argentina
| | - César Iván Pruzzo
- Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, La Plata, Argentina
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| |
Collapse
|
27
|
Counoupas C, Ferrell KC, Ashhurst A, Bhattacharyya ND, Nagalingam G, Stewart EL, Feng CG, Petrovsky N, Britton WJ, Triccas JA. Mucosal delivery of a multistage subunit vaccine promotes development of lung-resident memory T cells and affords interleukin-17-dependent protection against pulmonary tuberculosis. NPJ Vaccines 2020; 5:105. [PMID: 33298977 PMCID: PMC7665186 DOI: 10.1038/s41541-020-00255-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/02/2020] [Indexed: 11/29/2022] Open
Abstract
The development of effective vaccines against bacterial lung infections requires the induction of protective, pathogen-specific immune responses without deleterious inflammation within the pulmonary environment. Here, we made use of a polysaccharide-adjuvanted vaccine approach to elicit resident pulmonary T cells to protect against aerosol Mycobacterium tuberculosis infection. Intratracheal administration of the multistage fusion protein CysVac2 and the delta-inulin adjuvant Advax™ (formulated with a TLR9 agonist) provided superior protection against aerosol M. tuberculosis infection in mice, compared to parenteral delivery. Surprisingly, removal of the TLR9 agonist did not impact vaccine protection despite a reduction in cytokine-secreting T cell subsets, particularly CD4+IFN-γ+IL-2+TNF+ multifunctional T cells. CysVac2/Advax-mediated protection was associated with the induction of lung-resident, antigen-specific memory CD4+ T cells that expressed IL-17 and RORγT, the master transcriptional regulator of Th17 differentiation. IL-17 was identified as a key mediator of vaccine efficacy, with blocking of IL-17 during M. tuberculosis challenge reducing phagocyte influx, suppressing priming of pathogen-specific CD4+ T cells in local lymph nodes and ablating vaccine-induced protection. These findings suggest that tuberculosis vaccines such as CysVac2/Advax that are capable of eliciting Th17 lung-resident memory T cells are promising candidates for progression to human trials.
Collapse
Affiliation(s)
- Claudio Counoupas
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.
| | - Kia C Ferrell
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Anneliese Ashhurst
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Nayan D Bhattacharyya
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Gayathri Nagalingam
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Erica L Stewart
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale and Flinders University, Adelaide, Australia
| | - Carl G Feng
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale and Flinders University, Adelaide, Australia
| | - Warwick J Britton
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - James A Triccas
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.
- Charles Perkins Centre and Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
28
|
Ladda M, Lynde C, Fleming P. Severe Acute Respiratory Syndrome Coronavirus 2 and the Use of Biologics in Patients With Psoriasis. J Cutan Med Surg 2020; 24:625-632. [PMID: 32757760 DOI: 10.1177/1203475420945234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Coronavirus disease (COVID-19), a respiratory disease caused by a novel coronavirus designated severe acute respiratory syndrome coronavirus 2, has rapidly spread worldwide and has been recognized as a pandemic by the World Health Organization. Patients with altered immunologic function are at higher risk of acquiring COVID-19. In patients with psoriasis, inhibition of select pro-inflammatory cytokines through the use of biologic agents has been shown to be an effective treatment option. Pro-inflammatory cytokines have key immunomodulatory effects and are known to be involved in the hosts' immune response to a variety of viral infections. Though little is currently known about the role of inflammatory cytokines in COVID-19, early reports have shown patients with severe disease to have elevated serum levels of select inflammatory cytokines such as tumor necrosis factor alpha. This review will summarize key information that is currently known about COVID-19, the role of select cytokines in viral defense, and important considerations for patients with psoriasis using biologic agents during this pandemic. Currently, there is insufficient evidence to discontinue biologic therapy in patients with psoriasis who have not tested positive for COVID-19. The decision to pause biologic therapy should be considered on a case-by-case basis in patients in higher risk populations, and should take into account individual risk and benefit. Until more is known about the impact of biologic therapy on COVID-19 outcomes, we recommend patients with psoriasis who test positive for COVID-19 be instructed to discontinue or postpone biologic treatment until they have recovered from infection.
Collapse
Affiliation(s)
- Matthew Ladda
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Charles Lynde
- Lynde Institute for Dermatology, Markham, ON, Canada
- 210484 Division of Dermatology, Department of Medicine, University of Toronto, Toronto ON, Canada
| | - Patrick Fleming
- Lynde Institute for Dermatology, Markham, ON, Canada
- 210484 Division of Dermatology, Department of Medicine, University of Toronto, Toronto ON, Canada
| |
Collapse
|
29
|
Segueni N, Jacobs M, Ryffel B. Innate type 1 immune response, but not IL-17 cells control tuberculosis infection. Biomed J 2020; 44:165-171. [PMID: 32798210 PMCID: PMC8178558 DOI: 10.1016/j.bj.2020.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/16/2020] [Accepted: 06/29/2020] [Indexed: 01/22/2023] Open
Abstract
The role of the innate immune response and host resistance to Mycobacterium tuberculosis infection (TB) is reviewed. Based on our data and the abundant literature, an early type 1 immune response is critical for infection control, while ILC3 and Th17 cells seem to be dispensable. Indeed, in M. tuberculosis infected mice, transcriptomic levels of Il17, Il17ra, Il22 and Il23a were not significantly modified as compared to controls, suggesting a limited role of IL-17 and IL-22 pathways in TB infection control. Neutralization of IL-17A or IL-17F did not affect infection control either. Ongoing clinical studies with IL-17 neutralizing antibodies show high efficacy in patients with psoriasis without increased incidence of TB infection or reactivation. Therefore, both experimental studies in mice and clinical trials in human patients suggest no risk of TB infection or reactivation by therapeutic IL-17 antibodies, unlike by TNF.
Collapse
Affiliation(s)
- Noria Segueni
- Molecular and Experimental Immunology and Neurogenetics, UMR 7355, INEM, CNRS-University of Orleans, Orleans, France
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, South Africa; National Health Laboratory Service, Johannesburg, South Africa; Immunology of Infectious Disease Research Unit, University of Cape Town, South Africa
| | - Bernhard Ryffel
- Molecular and Experimental Immunology and Neurogenetics, UMR 7355, INEM, CNRS-University of Orleans, Orleans, France.
| |
Collapse
|
30
|
Graham JB, Swarts JL, Menachery VD, Gralinski LE, Schäfer A, Plante KS, Morrison CR, Voss KM, Green R, Choonoo G, Jeng S, Miller DR, Mooney MA, McWeeney SK, Ferris MT, Pardo-Manuel de Villena F, Gale M, Heise MT, Baric RS, Lund JM. Immune Predictors of Mortality After Ribonucleic Acid Virus Infection. J Infect Dis 2020; 221:882-889. [PMID: 31621854 PMCID: PMC7107456 DOI: 10.1093/infdis/jiz531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Virus infections result in a range of clinical outcomes for the host, from asymptomatic to severe or even lethal disease. Despite global efforts to prevent and treat virus infections to limit morbidity and mortality, the continued emergence and re-emergence of new outbreaks as well as common infections such as influenza persist as a health threat. Challenges to the prevention of severe disease after virus infection include both a paucity of protective vaccines as well as the early identification of individuals with the highest risk that may require supportive treatment. METHODS We completed a screen of mice from the Collaborative Cross (CC) that we infected with influenza, severe acute respiratory syndrome-coronavirus, and West Nile virus. RESULTS The CC mice exhibited a range of disease manifestations upon infections, and we used this natural variation to identify strains with mortality after infection and strains exhibiting no mortality. We then used comprehensive preinfection immunophenotyping to identify global baseline immune correlates of protection from mortality to virus infection. CONCLUSIONS These data suggest that immune phenotypes might be leveraged to identify humans at highest risk of adverse clinical outcomes upon infection, who may most benefit from intensive clinical interventions, in addition to providing insight for rational vaccine design.
Collapse
Affiliation(s)
- Jessica B Graham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jessica L Swarts
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Vineet D Menachery
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Microbiology and Immunology, University of Texas Medical Center, Galveston, Texas, USA
| | - Lisa E Gralinski
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kenneth S Plante
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Clayton R Morrison
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kathleen M Voss
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Richard Green
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Gabrielle Choonoo
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Sophia Jeng
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Darla R Miller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael A Mooney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA.,OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Shannon K McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA.,OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA.,Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Mark T Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer M Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
31
|
The risk of respiratory tract infections and symptoms in psoriasis patients treated with interleukin 17 pathway-inhibiting biologics: A meta-estimate of pivotal trials relevant to decision making during the COVID-19 pandemic. J Am Acad Dermatol 2020; 83:677-679. [PMID: 32416207 PMCID: PMC7235584 DOI: 10.1016/j.jaad.2020.05.035] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/05/2020] [Accepted: 05/09/2020] [Indexed: 11/20/2022]
|
32
|
Comparison of Sheep, Goats, and Calves as Infection Models for Mycobacterium avium subsp. paratuberculosis. Vet Immunol Immunopathol 2020; 225:110060. [PMID: 32413513 DOI: 10.1016/j.vetimm.2020.110060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 01/03/2023]
Abstract
Animal infection models to study Mycobacterium avium subsp. paratuberculosis (MAP) infection are useful for evaluating the efficacy of vaccines and other therapeutics for the prevention or treatment of infection. The goal of the present study was to compare smaller ruminants, sheep and goats, with calves as infection models. Neonatal sheep, goats, and calves (n = 4) received 109 cfu of a cattle isolate of MAP in milk replacer on days 0, 3 and 6 in a 12-month study and sampled monthly thereafter. Results demonstrated a robust antigen-specific IFN-γ response at 90 days post-inoculation for sheep and goats, with lower responses noted for calves. By 360 days, IFN-γ responses were 50 and 82% higher for calves than for goats and sheep, respectively. Although MAP-specific antibody responses were first observed in sheep at 90 days, calves had higher antibody responses throughout the remainder of the study. Following pass-through shedding on day 7, fecal shedding was fairly negligible across treatments but remained higher for calves throughout the study. Colonization of tissues was variable within treatment group and was higher for calves and sheep for the majority of tissues. Upon antigen stimulation of PBMCs, higher populations of CD4 + T cells cells and lower populations of γδ TCR + and NK cells were observed for goats and calves compared to sheep. Relative gene expression of IL-4, IL-12, and IL-17 in PBMCs was higher in goats, corresponding to lower tissue colonization with MAP. These data suggest that ruminant species are fairly comparable as infection models for MAP, but discrete differences in host responses to MAP infection exist between species.
Collapse
|
33
|
Amezcua Vesely MC, Rodríguez C, Gruppi A, Acosta Rodríguez EV. Interleukin-17 mediated immunity during infections with Trypanosoma cruzi and other protozoans. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165706. [PMID: 31987839 PMCID: PMC7071987 DOI: 10.1016/j.bbadis.2020.165706] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/05/2019] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
Host resistance during infection with Trypanosoma cruzi, and other protozoans, is dependent on a balanced immune response. Robust immunity against these pathogens requires of the concerted action of many innate and adaptive cell populations including macrophages, neutrophils, dendritic cells, CD4+, and CD8+ T cells and B cells among others. Indeed, during most protozoan infections only a balanced production of inflammatory (TH1) and anti-inflammatory (TH2/regulatory) cytokines will allow the control of parasite spreading without compromising host tissue integrity. The description of TH17 cells, a novel effector helper T cell lineage that produced IL-17 as signature cytokine, prompted the revision of our knowledge about the mechanisms that mediate protection and immunopathology during protozoan infections. In this manuscript we discuss the general features of IL-17 mediated immune responses as well as the cellular sources, effector mechanisms and overall role of IL-17 in the immune response to T. cruzi and other protozoan infections.
Collapse
Affiliation(s)
- María Carolina Amezcua Vesely
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Constanza Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Eva Virginia Acosta Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
34
|
Chen J, Vitetta L. The Role of Butyrate in Attenuating Pathobiont-Induced Hyperinflammation. Immune Netw 2020; 20:e15. [PMID: 32395367 PMCID: PMC7192831 DOI: 10.4110/in.2020.20.e15] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/09/2020] [Accepted: 01/19/2020] [Indexed: 12/17/2022] Open
Abstract
An excessive hyperinflammatory response-caused septic shock is a major medical problem that is associated with pathogenic bacterial infections leading to high mortality rates. The intestinal microbiota and the associated elaborated metabolites such as short chain fatty acid butyrate have been shown to relieve pathogenic bacterial-caused acute inflammation. Butyrate can down-regulate inflammation by inhibiting the growth of pathobionts, increasing mucosal barrier integrity, encouraging obligate anaerobic bacterial dominance and decreasing oxygen availability in the gut. Butyrate can also decrease excessive inflammation through modulation of immune cells such as increasing functionalities of M2 macrophages and regulatory T cells and inhibiting infiltration by neutrophils. Therefore, various approaches can be used to increase butyrate to relieve pathogenic bacterial-caused hyperinflammation. In this review we summarize the roles of butyrate in attenuating pathogenic bacterial-caused hyperinflammatory responses and discuss the associated plausible mechanisms.
Collapse
Affiliation(s)
| | - Luis Vitetta
- Medlab Clinical Ltd, Sydney 2015, Australia
- The University of Sydney, Faculty of Medicine and Health, Sydney 2006, Australia
| |
Collapse
|
35
|
Identification and Regulation of Interleukin-17 (IL-17) Family Ligands in the Teleost Fish European Sea Bass. Int J Mol Sci 2020; 21:ijms21072439. [PMID: 32244562 PMCID: PMC7178287 DOI: 10.3390/ijms21072439] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
Interleukin-17 (IL-17) cytokine comprises a family of six ligands in mammals with proinflammatory functions, having an important role in autoimmune disorders and against bacterial, viral, and fungal pathogens. While IL-17A and IL-17F ligands are mainly produced by Th cells (Th17 cells), the rest of the ligands are expressed by other immune and non-immune cells and have different functions. The identification of IL-17 ligands in fish has revealed the presence of six members, counterparts to mammalian ones, and a teleost-specific form, the fish IL-17N. However, tissue distribution, the regulation of gene expression, and scarce bioactivity assays point to similar functions compared to mammalian ones, though this yet to be investigated and confirmed. Thus, we have identified seven IL-17 ligands in the teleost European sea bass (Dicentrarchus labrax), for the first time, corresponding to IL-17A/F1, IL-17A/F2, IL-17A/F3, IL-17C1, IL-17C2, IL-17D, and IL-17N, according to the predicted protein sequences and phylogenetic analysis. They are constitutively and widely transcribed in sea bass tissues, with some of them being mainly expressed in the thymus, brain or intestine. Upon in vitro stimulation of head-kidney leucocytes, the mRNA levels of all sea bass IL-17 ligands were up-regulated by phytohemagglutinin treatment, a well-known T cell mitogen, suggesting a major expression in T lymphocytes. By contrast, the infection of sea bass juveniles with nodavirus (NNV), a very pathogenic virus for this fish species, resulted in the up-regulation of the transcription of IL-17C1 in the head-kidney and of IL-17C1 and IL-17D in the brain, the target tissue for NNV replication. By contrast, NNV infection led to a down-regulated transcription of IL-17A/F1, IL-17A/F2, IL-17C1, IL-17C2, and IL-17D in the head-kidney and of IL-17A/F1 and IL-17A/F3 in the brain. The data are discussed accordingly with the IL-17 ligand expression and the immune response under the different situations tested.
Collapse
|
36
|
Prawiro SR, Poeranto S, Amalia A, Widyani EL, Indraswari G, Soraya M, Dwi Pradipto SR, Prasetya A, Hidayat GR, Alitha Putri SN. Generating mucosal and systemic immune response following vaccination of vibrio cholerae adhesion molecule against shigella flexneri infection. Indian J Med Microbiol 2020; 38:37-45. [PMID: 32719207 DOI: 10.4103/ijmm.ijmm_19_411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction Previous studies have shown 37.8 kDa pili subunit protein of Vibrio cholerae and 49.8 kDa pili subunit protein of Shigella flexneri can act as an adhesion molecule to initiate infection. These molecules also have the ability to agglutinate blood. The present study assessed mucosal and systemic immunity following vaccination using 37.8 kDa V. cholerae and protection against S. flexneri. Subjects and Methods Haemagglutination test was performed after purification of V. cholerae protein, followed by an anti-haemagglutination test. The intestinal weight and colony count were used to validate the protective effect on balb/c mice which were divided into the naive group, Shigella-positive control group, Vibrio-positive control group, V. cholerae infected-Vibrio-vaccinated group and S. flexneri-infected-Vibrio-vaccinated group. Th17, Treg, interleukin (IL) IL-17A, β-defensin and secretory-immunoglobulin A (s-IgA) were also measured to determine the systemic and mucosal immunity after vaccination. Results The haemagglutination and anti-haemagglutination tests showed that the 37.8 kDa protein could inhibit 49.8 kDa of the S. flexneri pili subunit. Decreased intestinal weight and colony count of vaccinated group compared to naive group also support cross reaction findings. Vaccination also generates higher level of Th17, Treg, IL-17A, β-defensin and s-IgA significantly. Conclusions 37.8 kDa subunit pili can act as a homologous vaccine candidate to prevent V. cholerae and S. flexneri infection.
Collapse
Affiliation(s)
- Sumarno Reto Prawiro
- Department of Clinical Microbiology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Sri Poeranto
- Department of Clinical Parasitology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Aisyah Amalia
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Elsa Larissa Widyani
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Genitri Indraswari
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Merika Soraya
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Septha Rully Dwi Pradipto
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Adrian Prasetya
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Guntur Rizal Hidayat
- Bachelor Medical Program, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | | |
Collapse
|
37
|
Wu CY, Chiu HY, Tsai TF. The seroconversion rate of QuantiFERON-TB Gold In-Tube test in psoriatic patients receiving secukinumab and ixekizumab, the anti-interleukin-17A monoclonal antibodies. PLoS One 2019; 14:e0225112. [PMID: 31881026 PMCID: PMC6934285 DOI: 10.1371/journal.pone.0225112] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND For psoriatic patients receiving biologics, the concern of tuberculosis (TB) infection exists. Although the TB risk of anti-interleukin (IL)-17A agents is generally considered very low, more real-world data are needed to support the safety. OBJECTIVES This study aims to provide the real-world experience of using serial QuantiFERON-TB Gold In-Tube (QFT-GIT) test among patients treated with secukinumab or ixekizumab in Taiwan, an intermediate TB burden country, for the detection of latent TB infection (LTBI) reactivation or newly acquired TB infection. METHODS This retrospective review evaluated 100 consecutive patients with psoriasis receiving anti-IL-17A therapies who were checked with at least twice QFT-GIT between 2016 and 2019 in National Taiwan University Hospital, Taipei and Hsin-Chu, Taiwan. RESULTS Among the 100 patients, the baseline QFT-GIT results were negative in 81.0% (81/100), positive in 18.0% (18/100), and indeterminate in 1.0% (1/100) of patients. The overall outcomes in patients receiving at least 6 months of cumulative exposure to anti-IL-17A agents were persistently seronegative in 80 patients (80.0%), persistently seropositive in 14 patients (14.0%), seroconversion in 1 patient (1.0%), seroreversion in 3 patients (3.0%), and others in 2 patients (2.0%). In patients with at least 11 months of cumulative exposure, the seroconversion rate was 1.3% (1/79). The only case with seroconversion had a positive QFT-GIT result previously. No case of TB reactivation or newly acquired TB infection was identified during the follow-up. CONCLUSIONS In patients treated with anti-IL-17A monoclonal antibodies for psoriasis, routine serial repeat QFT-GIT testing was associated with lower seroconversion rate compared to real-world data of tumor necrosis factor-α inhibitors and anti-IL-12/23 antibody in Taiwan and in pivotal studies. Because clinical TB symptoms and signs are often preceded by QFT-GIF seroconversion, this result further supports the safety of anti-IL-17A agents in patients with psoriasis for LTBI.
Collapse
Affiliation(s)
- Chen-Yu Wu
- Department of Dermatology, Cathay General Hospital, Taipei, Taiwan
| | - Hsien-Yi Chiu
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Dermatology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
38
|
Incorporation of dynamic segmented neutrophil-to-monocyte ratio with leukocyte count for sepsis risk stratification. Sci Rep 2019; 9:19756. [PMID: 31875017 PMCID: PMC6930327 DOI: 10.1038/s41598-019-56368-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
The association between sepsis and segmented neutrophil-to-monocyte (SeMo) ratio is unclear. We postulated that an increase in dynamic SeMo ratio measurement can be applied in risk stratification. This retrospective study included 727 consecutive sepsis patients in medical intensive care units (ICUs), including a subpopulation of 153 patients. According to the leukocyte (white blood cell, WBC) count on day 3 (normal range, between 4,000/µL and 12,000/µL) and delta SeMo (value of SeMo ratio on day 3 minus value of SeMo ratio on day 1; normal delta SeMo, <7), patients were grouped into 3 (delta SeMo & WBC tool). The survival lines separated significantly with hazard ratios of 1.854 (1.342–2.560) for the delta SeMo or WBC abnormal group and 2.860 (1.849–4.439) for the delta SeMo and WBC abnormal group compared to the delta SeMo and WBC normal group. Delta SeMo & WBC tool and delta sequential organ failure assessment (SOFA) tool performed better than the other tools (delta SeMo, delta WBC, day 3 WBC, and day 1 WBC). Severity in delta SeMo & WBC tool and delta SeMo tool reflected the immune dysfunction score, cytokine expression, and human leukocyte antigen D-related monocyte expression on day 1 and day 3. There was correspondence between delta SOFA and delta WBC and between delta SeMo and delta cytokine expression. Incorporation of dynamic SeMo ratio with WBC count provides risk stratification for sepsis patients admitted in the ICU.
Collapse
|
39
|
Solaymani-Mohammadi S, Eckmann L, Singer SM. Interleukin (IL)-21 in Inflammation and Immunity During Parasitic Diseases. Front Cell Infect Microbiol 2019; 9:401. [PMID: 31867283 PMCID: PMC6904299 DOI: 10.3389/fcimb.2019.00401] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/11/2019] [Indexed: 12/30/2022] Open
Abstract
Parasitic diseases cause significant morbidity and mortality in the developing and underdeveloped countries. No efficacious vaccines are available against most parasitic diseases and there is a critical need for developing novel vaccine strategies for care. IL-21 is a pleiotropic cytokine whose functions in protection and immunopathology during parasitic diseases have been explored in limited ways. IL-21 and its cognate receptor, IL-21R, are highly expressed in parasitized organs of infected humans as well in murine models of the human parasitic diseases. Prior studies have indicated the ability of the IL-21/IL-21R signaling axis to regulate the effector functions (e.g., cytokine production) of T cell subsets by enhancing the expression of T-bet and STAT4 in human T cells, resulting in an augmented production of IFN-γ. Mice deficient for either IL-21 (Il21−/−) or IL-21R (Il21r−/−) showed significantly reduced inflammatory responses following parasitic infections as compared with their WT counterparts. Targeting the IL-21/IL-21R signaling axis may provide a novel approach for the development of new therapeutic agents for the prevention of parasite-induced immunopathology and tissue destruction.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, United States
| |
Collapse
|
40
|
Yan K, Yang J, Qian Q, Xu D, Liu H, Wei L, Li M, Xu W. Pathogenic Role of an IL-23/γδT17/Neutrophil Axis in Coxsackievirus B3-Induced Pancreatitis. THE JOURNAL OF IMMUNOLOGY 2019; 203:3301-3312. [PMID: 31748346 DOI: 10.4049/jimmunol.1900787] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/12/2019] [Indexed: 12/16/2022]
Abstract
Coxsackievirus B is a common cause of viral myocarditis and pancreatitis. IL-17A is intensively involved in the pathogenesis of viral myocarditis. Whether IL-17A plays a role in Coxsackievirus B-induced pancreatitis, characterized by acinar cell destruction and immune infiltration, remains largely unknown. We found a significant, but transient, increase of IL-17A expression and γδT influx in the pancreas of C57BL/6J mice within 3 d following CVB3 infection. The pancreatic IL-17A was mainly produced by Vγ4 γδ T cells, to a lesser extent by CD4+ Th17 cells. IL-17A-/- and TCRδ-/- mice both reduced their susceptibility to CVB3 infection and pancreatitis severity when compared with the wild-type mice, without altering viral load. mAb depletion of Vγ4γδ T cells significantly improved mice survival and pancreatic pathology via decreasing Th17 expansion and neutrophil influx into the pancreas compared with isotype-treated mice. Transfer of Vγ4γδ T cells from wild-type, but not IL-17-/-, mice reconstituted TCRδ-/- mice to produce IL-17 and develop pancreatitis to the level of wild-type mice during CVB3 infection, indicating γδ T IL-17A is required for the onset of viral pancreatitis. IL-23 was robustly induced in the pancreas within the first day of infection. Administration of exogenous rIL-23 to mice increased CVB3 pancreatitis through in vivo expansion of IL-17+γδT17 cells at 12 h postinfection. Our findings reveal a key pathogenic role for early-activated γδT17 cells in viral pancreatitis via promoting neutrophil infiltration and Th17 induction. This IL-23/γδT17/neutrophil axis is critically involved in the onset of CVB3 pancreatitis and represents a potential treating target for the disease.
Collapse
Affiliation(s)
- Kepeng Yan
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Jie Yang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Qian Qian
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Dan Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Hui Liu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lin Wei
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| |
Collapse
|
41
|
Soni B, Singh S. Cytokine Milieu in Infectious Disease: A Sword or a Boon? J Interferon Cytokine Res 2019; 40:24-32. [PMID: 31553263 DOI: 10.1089/jir.2019.0089] [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] [Indexed: 12/19/2022] Open
Abstract
Cytokines have a myriad role in an infectious disease, whether being pathogenic, bacterial, or viral. All proinflammatory and anti-inflammatory cytokine biological function are dependent on its concentration, followed by combination with the other cytokines and the stage of the disease. Plasticity in switching off from one phenotype to the other of these regulatory mediators in congruence with the traditional concept of inhibitory and stimulatory effects on immune system is dealt with. This review highlights the dual functionality of some of these cytokines and cytokine-based immunotherapy.
Collapse
Affiliation(s)
- Bhavnita Soni
- Department of Pathogenesis and Cellular Response, National Centre for Cell Science, Pune, India
| | - Shailza Singh
- Department of Pathogenesis and Cellular Response, National Centre for Cell Science, Pune, India
| |
Collapse
|
42
|
Ribero S, Licciardello M, Quaglino P, Dapavo P. Efficacy and Safety of Secukinumab in Patients with Plaque Psoriasis and Latent Tuberculosis. Case Rep Dermatol 2019; 11:23-28. [PMID: 31662735 PMCID: PMC6816124 DOI: 10.1159/000501989] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 12/30/2022] Open
Abstract
Upon the association of biologic treatments with reactivation of latent tuberculosis infection (LTBI), screening for Mycobacterium tuberculosisinfection and anti-tuberculosis chemoprophylaxis in positive patients are required prior to biologic drug administration. Nevertheless, the risk of infection relapses associated with biologic drugs seems to be different. No cases of reactivation of LTBI have been observed in secukinumab-treated subjects, in contrast with clinical reports on the risk associated with anti-tumor necrosis factor Α-based therapy. Twelve patients with moderate to severe plaque psoriasis eligible for systemic treatment and found to have LTBI received secukinumab without previous chemoprophylaxis initiation because of clinical contraindication for 10 cases and refusal by 2 patients. None of them had tuberculosis reactivation.
Collapse
Affiliation(s)
- Simone Ribero
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| | - Matteo Licciardello
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| | - Pietro Quaglino
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| | - Paolo Dapavo
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| |
Collapse
|
43
|
Luo Y, Xu J, Zhang C, Jiang C, Ma Y, He H, Wu Y, Devriendt B, Cox E, Zhang H. Toll-like receptor 5-mediated IL-17C expression in intestinal epithelial cells enhances epithelial host defense against F4 + ETEC infection. Vet Res 2019; 50:48. [PMID: 31221216 PMCID: PMC6584996 DOI: 10.1186/s13567-019-0665-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are an important cause of post-weaning diarrhea (PWD) in piglets. The IL-17 cytokine family is well known to play important roles in the host defense against bacterial infections at the mucosa. Previously, we reported the potential role of IL-17A in clearing an ETEC infection in piglets. IL-17C, another member of the IL-17 family, is highly expressed in the intestinal epithelium, however, its role during an ETEC infection is still unclear. In this study, we demonstrate that F4+ ETEC induce IL-17C mRNA and protein expression in intestinal tissues as well as in porcine intestinal epithelial cells (IPEC-J2). This IL-17C production is largely dependent on TLR5 signaling in IPEC-J2 cells. Both F4+ ETEC infection and exogenous IL-17C increased the expression of antimicrobial peptides and tight junction proteins, such as porcine beta-defensin (pBD)-2, claudin-1, claudin-2 and occludin in IPEC-J2 cells. Taken together, our data demonstrate that TLR5-mediated IL-17C expression in intestinal epithelial cells enhances mucosal host defense responses in a unique autocrine/paracrine manner in the intestinal epithelium against ETEC infection.
Collapse
Affiliation(s)
- Yu Luo
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China.
| | - Jia Xu
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Chaoying Zhang
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Chunyan Jiang
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Yanfeng Ma
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Haijian He
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Yuan Wu
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Hongbin Zhang
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| |
Collapse
|
44
|
Hui Z, Liu Z, He A, Chen Y, Zhang P, Lei B, Yao H, Yu Y, Liang R, Li Z, Zhang W. Visfatin promotes the malignancy of human acute myeloid leukemia cells via regulation of IL-17. Eur J Pharmacol 2019; 853:103-110. [PMID: 30876976 DOI: 10.1016/j.ejphar.2019.03.016] [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: 11/18/2018] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 11/29/2022]
Abstract
Understanding the mechanisms of malignancy in acute myeloid leukemia (AML) cell is important for the targeted treatment and drug development. We found that visfatin, a 52-kDa adipokine, can positively regulate the proliferation of AML cells. Targeted inhibition of visfatin via its specific siRNAs or inhibitor can suppress the proliferation of AML cells. Further, knockdown of visfatin can increase the doxorubicin (Dox) and cisplatin (CDDP) sensitivity of AML cells. Among the tested six cytokines, si-visfatin can decrease the expression of interleukin-17 (IL-17). Over expression of IL-17 can reverse si-visfatin suppressed cell proliferation and increased Dox sensitivity. The upregulation of IL-17 was also involved in visfatin induced activation of PI3K/Akt signals in AML cells. The inhibitor of PI3K/Akt can synergistically suppress the proliferation of HL60 cells which were transfected with si-visfatin. Knockdown of visfatin can increase the expression of miR-135a, which can bind to the 3'UTR of IL-17 and decrease its expression. The inhibitor of miR-135a can attenuate si-visfatin suppressed expression of IL-17 and proliferation of AML cells. Collectively, our data suggested that visfatin can increase the malignancy of AML cells via regulation of miR-135a/IL-17/PI3K/Akt signals.
Collapse
Affiliation(s)
- Zengqian Hui
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Zhao Liu
- Department of Surgery, Xi'an Chest Hospital, Xi'an TB & Thoracic Tumor Hospital, Xi'an, Shaanxi 710100, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yinxia Chen
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Pengyu Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Bo Lei
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Huan Yao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yong Yu
- Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Rui Liang
- Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Zhanning Li
- Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force, Xi'an, Shaanxi 710054, China
| | - Wanggang Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
45
|
Rao M, Dodoo E, Zumla A, Maeurer M. Immunometabolism and Pulmonary Infections: Implications for Protective Immune Responses and Host-Directed Therapies. Front Microbiol 2019; 10:962. [PMID: 31134013 PMCID: PMC6514247 DOI: 10.3389/fmicb.2019.00962] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
The biology and clinical efficacy of immune cells from patients with infectious diseases or cancer are associated with metabolic programming. Host immune- and stromal-cell genetic and epigenetic signatures in response to the invading pathogen shape disease pathophysiology and disease outcomes. Directly linked to the immunometabolic axis is the role of the host microbiome, which is also discussed here in the context of productive immune responses to lung infections. We also present host-directed therapies (HDT) as a clinically viable strategy to refocus dysregulated immunometabolism in patients with infectious diseases, which requires validation in early phase clinical trials as adjuncts to conventional antimicrobial therapy. These efforts are expected to be continuously supported by newly generated basic and translational research data to gain a better understanding of disease pathology while devising new molecularly defined platforms and therapeutic options to improve the treatment of patients with pulmonary infections, particularly in relation to multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ernest Dodoo
- Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| |
Collapse
|
46
|
Wu KK, Lee MP, Lee EB, Wu JJ. Risk of herpes zoster with IL-17 inhibitor therapy for psoriasis and other inflammatory conditions. J DERMATOL TREAT 2019; 31:359-365. [PMID: 30900514 DOI: 10.1080/09546634.2019.1597246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background: Psoriasis is a chronic inflammatory skin disease that has been associated with a significantly higher risk of herpes zoster (HZ). Several newer biologics such as secukinumab, ixekizumab, and brodalumab inhibit IL-17 and have been highly effective for treatment of psoriasis. However, adverse events related to the immunosuppressive properties of these biologics have been observed.Methods: This review aims to synthesize and evaluate the literature investigating the risk of HZ in patients treated with IL-17 inhibitors, with a focus on psoriasis patients. We performed searches using the PubMED database with the following search terms: 'psoriasis,' 'herpes zoster,' 'secukinumab,' 'ixekizumab,' 'brodalumab,' 'IL-17,' 'anti-IL-17,' and 'safety.' Clinical trials, cohort studies, review articles, and meta-analyses were evaluated.Results: Studies did not detect a higher risk of HZ infections in psoriasis patients treated with IL-17 inhibitors when compared to those treated with placebo or other therapies. Studies of IL-17 inhibitors for other indications including psoriatic arthritis, rheumatoid arthritis, ankylosing spondylitis, and asthma yielded similar results.Conclusion: IL-17 inhibitors do not appear to increase risk of HZ. However, IL-17 inhibitors are relatively new medications, and further long-term data may be necessary to confirm this finding. Nevertheless, HZ vaccination should be considered on a case-bycase basis prior to initiating IL-17 therapy.
Collapse
Affiliation(s)
- Kevin K Wu
- Frank H. Netter MD School of Medicine at Quinnipiac University, North Haven, CT, USA
| | | | - Erica B Lee
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Jashin J Wu
- Dermatology Research and Education Foundation, Irvine, CA, USA
| |
Collapse
|
47
|
Hou Z, Liu D, Su S, Wang L, Zhao Z, Ma Y, Li Q, Jia C, Xu J, Zhou Y, Tao J. Comparison of splenocyte microRNA expression profiles of pigs during acute and chronic toxoplasmosis. BMC Genomics 2019; 20:97. [PMID: 30700253 PMCID: PMC6354428 DOI: 10.1186/s12864-019-5458-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 01/17/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is an obligate intracellular parasite that infects humans and other warm-blooded animals. Previous quantitative proteomic analyses of infected host cells revealed that the expression of many host proteins is modulated by T. gondii infection. However, at present limited data are available on the differentially expressed miRNAs (DEMs) associated with the pathology and host immune responses induced by acute and chronic infection with T. gondii in pigs in vivo. In this study, high-throughput sequencing was used to investigate expression profiles of spleen miRNAs at 10, 25 and 50 days post-infection (DPI) in pigs infected with Chinese I genotype strain T. gondii isolated from a dead pig. RESULTS When compared to the control group, 34, 6 and 86 DEMs were found in spleens of infected pigs at 10, 25 and 50 DPI, respectively. Gene Ontology (GO) enrichment analysis of the target genes of DEMs showed that no GO terms were enriched at 25 DPI, whereas 28 and 241 GO terms, of which two and 215 were sample-specific, were significantly enriched at 10 and 50 DPI, respectively. The top 20 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the target genes of DEMs included signal transduction, immune system, metabolism and diseases. miRNA-gene network analysis revealed that the DEMs played important roles in the host immune response to T. gondii infection by modulating expression levels of cellular immunity-related cytokines and immune-related C-type lectins. CONCLUSION Our results not only showed that host miRNA expression is altered by T. gondii but also revealed differences in the regulation of key biological processes and pathways involved in host responses to acute versus chronic T. gondii infection. This will aid future research into miRNA-target interactions during T. gondii infection in pigs and the development of novel therapies against T. gondii.
Collapse
Affiliation(s)
- Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Shijie Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Lele Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Zhenxing Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yifei Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Qiaoqiao Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Chuanli Jia
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yonghua Zhou
- Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China. .,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
48
|
Naili I, Vinot J, Baudner BC, Bernalier-Donadille A, Pizza M, Desvaux M, Jubelin G, D'Oro U, Buonsanti C. Mixed mucosal-parenteral immunizations with the broadly conserved pathogenic Escherichia coli antigen SslE induce a robust mucosal and systemic immunity without affecting the murine intestinal microbiota. Vaccine 2018; 37:314-324. [PMID: 30503655 DOI: 10.1016/j.vaccine.2018.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023]
Abstract
Emergence and dissemination of multidrug resistance among pathogenic Escherichia coli have posed a serious threat to public health across developing and developed countries. In combination with a flexible repertoire of virulence mechanisms, E. coli can cause a vast range of intestinal (InPEC) and extraintestinal (ExPEC) diseases but only a very limited number of antibiotics still remains effective against this pathogen. Hence, a broad spectrum E. coli vaccine could be a promising alternative to prevent the burden of such diseases, while offering the potential for covering against several InPEC and ExPEC at once. SslE, the Secreted and Surface-associated Lipoprotein of E. coli, is a widely distributed protein among InPEC and ExPEC. SslE functions ex vivo as a mucinase capable of degrading mucins and reaching the surface of mucus-producing epithelial cells. SslE was identified by reverse vaccinology as a protective vaccine candidate against an ExPEC murine model of sepsis, and further shown to be cross-effective against other ExPEC and InPEC models of infection. In this study, we aimed to gain insight into the immune response to antigen SslE and identify an immunization strategy suited to generate robust mucosal and systemic immune responses. We showed, by analyzing T cell and antibody responses, that mice immunized with SslE via an intranasal prime followed by two intramuscular boosts developed an enhanced overall immune response compared to either intranasal-only or intramuscular-only protocols. Importantly, we also report that this regimen of immunization did not impact the richness of the murine gut microbiota, and mice had a comparable cecal microbial composition, whether immunized with SslE or PBS. Collectively, our findings further support the use of SslE in future vaccination strategies to effectively target both InPEC and ExPEC while not perturbing the resident gut microbiota.
Collapse
Affiliation(s)
- Ilham Naili
- GSK, Siena, Italy; Université Clermont Auvergne, INRA, UMR454 MEDiS, 63000 Clermont-Ferrand, France.
| | | | | | | | | | - Mickaël Desvaux
- Université Clermont Auvergne, INRA, UMR454 MEDiS, 63000 Clermont-Ferrand, France
| | - Grégory Jubelin
- Université Clermont Auvergne, INRA, UMR454 MEDiS, 63000 Clermont-Ferrand, France
| | | | | |
Collapse
|
49
|
Müllebner A, Sassu EL, Ladinig A, Frömbling J, Miller I, Ehling-Schulz M, Hennig-Pauka I, Duvigneau JC. Actinobacillus pleuropneumoniae triggers IL-10 expression in tonsils to mediate colonisation and persistence of infection in pigs. Vet Immunol Immunopathol 2018; 205:17-23. [PMID: 30458998 DOI: 10.1016/j.vetimm.2018.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022]
Abstract
Actinobacillus pleuropneumoniae (APP) persisting in clinically healthy pigs may be the causative agent of sudden outbreaks of severe respiratory disease in swine herds. During the course of acute disease, the pathogen is eliminated from inflamed lung tissue, which is characterized by the expression of pro-inflammatory cytokines and an influx of neutrophils. However, if clearance by the porcine immune system fails, APP may switch to a persistent form. At later stages of infection, the pathogen may reside in tonsillar tissue without being eliminated by the host immune defence. To better understand the host immune response at different stages of infection, expression pattern of cytokines in tonsils and lung were recorded. In contrast to lung tissue, in which APP presence was associated with a pronounced pro-inflammatory character, APP presence in the tonsils elicited an increased IL-10 expression. In both organs of infected animals, a marked reciprocal correlation of the pro-inflammatory IL-17A and the anti-inflammatory IL-10 was found, supporting the idea that both cytokines are produced in highly associated, but reciprocal differentiated cell types, possibly APP-specific Th17 subsets. It appears that a persistent phenotype of APP triggers the anti-inflammatory immune response in tonsillar tissue in an attempt to evade the porcine immune defence.
Collapse
Affiliation(s)
- Andrea Müllebner
- Institute for Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.
| | - Elena L Sassu
- University Clinic for Swine, Department of Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Andrea Ladinig
- University Clinic for Swine, Department of Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Janna Frömbling
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Ingrid Miller
- Institute for Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Monika Ehling-Schulz
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Isabel Hennig-Pauka
- University Clinic for Swine, Department of Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - J Catharina Duvigneau
- Institute for Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
50
|
Łanocha-Arendarczyk N, Kolasa-Wołosiuk A, Wojciechowska-Koszko I, Kot K, Roszkowska P, Krasnodębska-Szponder B, Paczkowska E, Machaliński B, Łuczkowska K, Wiszniewska B, Kosik-Bogacka D. Changes in the immune system in experimental acanthamoebiasis in immunocompetent and immunosuppressed hosts. Parasit Vectors 2018; 11:517. [PMID: 30236160 PMCID: PMC6149055 DOI: 10.1186/s13071-018-3108-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/11/2018] [Indexed: 11/28/2022] Open
Abstract
Background Acanthamoebiasis is most often found in patients with immune deficiency, with infections facilitated by the intake of immunosuppressive drugs. The host immune response to Acanthamoeba spp. infection is poorly understood. Thus, in this study, we aimed to examine the course of Acanthamoeba spp. infection taking into account the host’s immunological status, including assessment of the hematological parameters, cytokine analysis, immunophenotypic changes in spleen populations, and histological spleen changes, which could help clarify some aspects of the immune response to acanthamoebiasis. In our experimental study, we used Acanthamoeba strain AM 22 isolated from the bronchoaspirate of a patient with acute myeloid leukaemia (AML) and atypical pneumonia symptoms. Results Acanthamoeba spp. affected the hematological parameters in immunocompetent and immunosuppressed mice and induced a change in spleen weight during infection. Moreover, analysis of anti-inflammatory (IL-4 and IL-10) and pro-inflammatory (IL-17A and IFN-γ) cytokines produced by splenocytes stimulated with concanavalin A demonstrated that Acanthamoeba spp. induced a selective Th1, Th2 and Th17 response at later stages of the infection in immunocompetent hosts. In the case of hosts with low immunity, Acanthamoeba elicited robust Th1 cell-mediated immunity without the participation of Th17. We observed suppression of CD8+ and CD4+ T lymphocytes and CD3+CD4-CD8- double-negative (DN) T lymphocyte populations in the beginning, and in the case of CD3+/CD4+/CD8+ double-positive (DP) T cells in the final phase of Acanthamoeba spp. infection in hosts with low immunity. Also, CD4+T lymphocytes and CD3+/CD4+ and CD3+/CD8+ lymphocyte counts during each stage of acanthamoebiasis were shown to be upregulated. Conclusions We demonstrated that analysis of the immune response and pathogenesis mechanisms of clinical isolates of Acanthamoeba spp. in an animal model not only has purely cognitive significance but above all, may help in the development of effective methods of pharmacological therapy especially in patients with low immunity.
Collapse
Affiliation(s)
- Natalia Łanocha-Arendarczyk
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Agnieszka Kolasa-Wołosiuk
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Iwona Wojciechowska-Koszko
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Karolina Kot
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Paulina Roszkowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Barbara Krasnodębska-Szponder
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Barbara Wiszniewska
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland
| | - Danuta Kosik-Bogacka
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, 70-204, Szczecin, Poland.
| |
Collapse
|