1
|
Guo F, Song Y, Dong S, Wei J, Li B, Xu T, Wang H. Characterization and anti-tuberculosis effects of γδ T cells expanded and activated by Mycobacterium tuberculosis heat-resistant antigen. Virulence 2025; 16:2462092. [PMID: 39921673 PMCID: PMC11810100 DOI: 10.1080/21505594.2025.2462092] [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: 08/09/2024] [Revised: 12/01/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (Mtb) that poses a severe threat to human health. A variety of highly immunogenic tuberculosis proteins have been used as targets in vaccine development to mitigate the spread of TB. Although Th1-type immunity has long been considered a crucial part of resistance to Mtb, γδ T cells, the predominant source of IL-17, are not negligible in controlling the early stages of TB infection. In addition to classical phosphoantigens, Mycobacterium tuberculosis heat-resistant antigens (HAg), a complex containing 564 proteins obtained from live tuberculosis bacteria after heat treatment at 121 °C for 20 min, have been confirmed to be highly effective γδ T cell stimulators as well. Several studies have demonstrated that HAg-activated γδ T cells can participate in TB immunity by secreting multiple cytokines against Mtb or by interacting with other innate immune cells. In this review, we present a possible mechanism of HAg stimulation of γδ T cells and the role of HAg-activated γδ T cells in anti-TB immunity. We also highlight the limitations of studies on HAg activation of γδ T cells and suggest further research directions on the relationship between HAg and γδ T cells.
Collapse
Affiliation(s)
- Fangzheng Guo
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Yamin Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Sihang Dong
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Jing Wei
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Baiqing Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Tao Xu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Hongtao Wang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| |
Collapse
|
2
|
Franzoni G, Signorelli F, Mazzone P, Donniacuo A, De Matteis G, Grandoni F, Schiavo L, Zinellu S, Dei Giudici S, Bezos J, De Carlo E, Galiero G, Napolitano F, Martucciello A. Cytokines as potential biomarkers for the diagnosis of Mycobacterium bovis infection in Mediterranean buffaloes ( Bubalus bubalis). Front Vet Sci 2024; 11:1512571. [PMID: 39776597 PMCID: PMC11703857 DOI: 10.3389/fvets.2024.1512571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Mycobacterium bovis (M. bovis) is the primary agent of bovine tuberculosis (TB) in Mediterranean buffalo, which has a negative economic impact on buffalo herds. Improving TB diagnostic performance in this species represents a key step to eradicate efficiently this disease. We have recently shown the utility of the IFN-γ assay in the diagnosis of M. bovis infection in Mediterranean buffaloes (Bubalus bubalis), but other cytokines might be useful immunological biomarkers of this infection. We therefore investigated the utility of key immune cytokines as diagnostic biomarkers of M. bovis infection in this species. Thirty-six Italian Mediterranean buffaloes were used in this study: healthy animals (N = 11), infected (IFN-γ test positive, no post-mortem lesions, no M. bovis detection; N = 14), and affected (IFN-γ test positive, visible post-mortem lesions; N = 11). Heparin blood samples were stimulated with bovine purified protein derivative (PPD-B), alongside controls, and 18-24 h later plasma were collected. Levels of 14 key cytokines were measured: IFN-γ, IL-17, IL-4, IL-10, TNF, IL-1α, IL-1β, IL-6, IP-10, MIP-1α, MIP-1β, MCP-1, IL-36Ra, and VEGF-A. We observed that both infected and affected animals released higher levels of IFN-γ, IL-17, IL-10, TNF, IL-1α, IL-6, MIP-1β, in response to PPD-B compared to healthy subjects. Mycobacterium bovis infected animals released also higher levels of IL-1β and IP-10 in response to PPD-B compared to healthy subjects, whereas only tendencies were detected in affected animals. Affected animals only released MIP-1α in response to PPD-B compared to healthy subjects and in this group higher values of PPD-B specific TNF was also observed. Finally, canonical discriminant analysis (CDA) was used to generate predictive cytokine profiles by groups. Our data suggest that IL-10, TNF, IL-1α, IL-6, MIP-1β could be useful biomarkers of TB in Mediterranean Buffalo and can improve the TB diagnostic performance in this specie.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Federica Signorelli
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Piera Mazzone
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, Perugia, Italy
| | - Anna Donniacuo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Giovanna De Matteis
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Francesco Grandoni
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Lorena Schiavo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Javier Bezos
- VISAVET Health Surveillance Centre, Complutense University of Madrid, Madrid, Spain
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad Complutense de Madrid, Madrid, Spain
| | - Esterina De Carlo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Giorgio Galiero
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Francesco Napolitano
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Alessandra Martucciello
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| |
Collapse
|
3
|
Yang Z, Zhang D, Jiang Z, Peng J, Wei H. The formidable guardian: Type 3 immunity in the intestine of pigs. Virulence 2024; 15:2424325. [PMID: 39497434 PMCID: PMC11552283 DOI: 10.1080/21505594.2024.2424325] [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: 03/14/2024] [Revised: 09/19/2024] [Accepted: 10/11/2024] [Indexed: 11/09/2024] Open
Abstract
Well-intestinal health is crucial for better growth performance in pigs. Type 3 immunity, which is one of the three types of immune responses in mammals, plays a vital role in maintaining intestinal homoeostasis. Therefore, we initially introduce the type 3 immune cells in the intestine of pigs, including their distribution, development, and function. We then discuss the type 3 immune response under infection, encompassing bacterial, fungal, and viral infections. It also covers two major stresses in pigs: heat stress and weaning stress. Lastly, we discuss the effects of various nutrients and feed additives on the regulation of the type 3 immune response in pigs under infection. This review aims to contribute to the understanding of the interaction between infection and type 3 immunity in pigs and to illustrate how various nutrients modulate the type 3 immune response in pigs under diverse infections.
Collapse
Affiliation(s)
- Zhipeng Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Dou Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhoudan Jiang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
4
|
Kerro Dego O, Vidlund J. Staphylococcal mastitis in dairy cows. Front Vet Sci 2024; 11:1356259. [PMID: 38863450 PMCID: PMC11165426 DOI: 10.3389/fvets.2024.1356259] [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: 01/17/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Bovine mastitis is one of the most common diseases of dairy cattle. Even though different infectious microorganisms and mechanical injury can cause mastitis, bacteria are the most common cause of mastitis in dairy cows. Staphylococci, streptococci, and coliforms are the most frequently diagnosed etiological agents of mastitis in dairy cows. Staphylococci that cause mastitis are broadly divided into Staphylococcus aureus and non-aureus staphylococci (NAS). NAS is mainly comprised of coagulase-negative Staphylococcus species (CNS) and some coagulase-positive and coagulase-variable staphylococci. Current staphylococcal mastitis control measures are ineffective, and dependence on antimicrobial drugs is not sustainable because of the low cure rate with antimicrobial treatment and the development of resistance. Non-antimicrobial effective and sustainable control tools are critically needed. This review describes the current status of S. aureus and NAS mastitis in dairy cows and flags areas of knowledge gaps.
Collapse
Affiliation(s)
- Oudessa Kerro Dego
- Department of Animal Science, University of Tennessee, Knoxville, TN, United States
| | - Jessica Vidlund
- Department of Animal Science, University of Tennessee, Knoxville, TN, United States
- East Tennessee AgResearch and Education Center-Little River Animal and Environmental Unit, University of Tennessee, Walland, TN, United States
| |
Collapse
|
5
|
Application of Multiparametric Flow Cytometry Panels to Study Lymphocyte Subpopulations in Tuberculin-Positive Cattle. Vet Sci 2023; 10:vetsci10030197. [PMID: 36977236 PMCID: PMC10057637 DOI: 10.3390/vetsci10030197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Flow cytometry (FC) is widely used in microbiology, immunology, hematology, and oncology. In the veterinary field, FC enabled the study of the immune response in cattle infected with different pathogens, as well as vaccine testing. However, few fluorochrome-conjugated antibodies recognize bovine antigens, limiting the possible benefits of FC and the implementation of multiparametric analysis for more complex studies. Two cytometry panels with five colors each were designed and implemented for the study and identification of populations and subpopulations of T cells derived from the peripheral blood mononuclear cells of dairy heifers. Both panels detected differences in T cell subpopulations between heifers positively and negatively tested for tuberculin; they detected overexpression of CD25+ and CD45RO+ in tuberculin-positive heifers after stimulation with a culture filtrate protein extract (CFPE) from Mycobacterium bovis (M. bovis). We identified subpopulations of T cells from peripheral blood mononuclear cells using two multicolor panels. These panels could be used to analyze total bovine blood in immunopathogenic studies and vaccine development. The same strategy could be implemented in other species of veterinary interest.
Collapse
|
6
|
Li J, Fu X, Sun L, Xue X, Liu H, Zhang F. Case Report: Lepromatous Leprosy and Psoriasis: An Uncommon Coincidence. Am J Trop Med Hyg 2023; 108:317-319. [PMID: 36572011 PMCID: PMC9896323 DOI: 10.4269/ajtmh.22-0324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 12/27/2022] Open
Abstract
Leprosy, a chronic infectious disease, and psoriasis, an inflammatory disorder, are distinct entities. Epidemiology data show that these two diseases are almost mutually exclusive, with only a few reported cases of their coexistence. Here, we present the case of a patient manifesting intermingled psoriatic and leprosy lesions diagnosed as borderline lepromatous leprosy and plaque psoriasis. Of note, Mycobacterium leprae bacilli were detected not only in the two types of lesions but also in normal-appearing skin and blood.
Collapse
Affiliation(s)
- Jinghui Li
- Shandong Provincial Hospital for Skin Diseases, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Provincial Hospital of Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xi’an Fu
- Shandong Provincial Hospital of Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Lele Sun
- Shandong Provincial Hospital of Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaotong Xue
- Shandong Provincial Hospital of Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hong Liu
- Shandong Provincial Hospital for Skin Diseases, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Provincial Hospital of Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Furen Zhang
- Shandong Provincial Hospital for Skin Diseases, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Provincial Hospital of Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
7
|
Giannou AD, Lücke J, Kleinschmidt D, Shiri AM, Steglich B, Nawrocki M, Zhang T, Zazara DE, Kempski J, Zhao L, Giannou O, Agalioti T, Brockmann L, Bertram F, Sabihi M, Böttcher M, Ewald F, Schulze K, von Felden J, Machicote A, Maroulis IC, Arck PC, Grass JK, Mercanoglu B, Reeh M, Wolter S, Tachezy M, Seese H, Theodorakopoulou M, Lykoudis PM, Heumann A, Uzunoglu FG, Ghadban T, Mann O, Izbicki JR, Li J, Duprée A, Melling N, Gagliani N, Huber S. A Critical Role of the IL-22-IL-22 Binding Protein Axis in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14246019. [PMID: 36551508 PMCID: PMC9775560 DOI: 10.3390/cancers14246019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the five most common cancer entities worldwide and leads to hundred-thousands of deaths every year. Despite some groundbreaking therapeutical revelations during the last years, the overall prognosis remains poor. Although the immune system fights malignant transformations with a robust anti-tumor response, certain immune mediators have also been shown to promote cancer development. For example, interleukin (IL)-22 has been associated with HCC progression and worsened prognosis in multiple studies. However, the underlying mechanisms of the pathological role of IL-22-signaling as well as the role of its natural antagonist IL-22 binding protein (IL-22BP) in HCC remain elusive. Here, we corroborate the pathogenic role of IL-22 in HCC by taking advantage of two mouse models. Moreover, we observed a protective role of IL-22BP during liver carcinogenesis. While IL-22 was mainly produced by CD4+ T cells in HCC, IL-22BP was abundantly expressed by neutrophils during liver carcinogenesis. Hepatocytes could be identified as a major target of this pathological IL-22-signaling. Moreover, abrogation of IL-22 signaling in hepatocytes in IL22ra1flox/flox × AlbCre+ mice reduced STEAP4 expression-a known oncogene-in HCC in vivo. Likewise, STEAP4 expression correlated with IL22 levels in human HCC samples, but not in healthy liver specimens. In conclusion, these data encourage the development of therapeutical approaches that target the IL-22-IL-22BP axis in HCC.
Collapse
Affiliation(s)
- Anastasios D. Giannou
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Correspondence: (A.D.G.); (S.H.); Tel.: +49-40-7410-20980 (A.D.G.); +49-40-7410-53910 (S.H.)
| | - Jöran Lücke
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Dörte Kleinschmidt
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ahmad Mustafa Shiri
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Babett Steglich
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mikolaj Nawrocki
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tao Zhang
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Dimitra E. Zazara
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Laboratory for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jan Kempski
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- The Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lilan Zhao
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Olympia Giannou
- Department of Computer Engineering & Informatics, University of Patras, 26500 Patras, Greece
| | - Theodora Agalioti
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Leonie Brockmann
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Franziska Bertram
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Morsal Sabihi
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marius Böttcher
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Florian Ewald
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kornelius Schulze
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Johann von Felden
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andres Machicote
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ioannis C. Maroulis
- Department of Surgery, University of Patras Medical School, 26500 Patras, Greece
| | - Petra C. Arck
- Laboratory for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Julia-Kristin Grass
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Baris Mercanoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Matthias Reeh
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Wolter
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hannes Seese
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Myrto Theodorakopoulou
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Panagis M. Lykoudis
- 3rd Department of Surgery, National & Kapodistrian University of Athens, 11527 Athens, Greece
- Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK
| | - Asmus Heumann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Faik G. Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tarik Ghadban
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Oliver Mann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jun Li
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anna Duprée
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nicola Gagliani
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Correspondence: (A.D.G.); (S.H.); Tel.: +49-40-7410-20980 (A.D.G.); +49-40-7410-53910 (S.H.)
| |
Collapse
|
8
|
Wherry TLT, Stabel JR. Bovine Immunity and Vitamin D 3: An Emerging Association in Johne's Disease. Microorganisms 2022; 10:1865. [PMID: 36144467 PMCID: PMC9500906 DOI: 10.3390/microorganisms10091865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP) is an environmentally hardy pathogen of ruminants that plagues the dairy industry. Hallmark clinical symptoms include granulomatous enteritis, watery diarrhea, and significant loss of body condition. Transition from subclinical to clinical infection is a dynamic process led by MAP which resides in host macrophages. Clinical stage disease is accompanied by dysfunctional immune responses and a reduction in circulating vitamin D3. The immunomodulatory role of vitamin D3 in infectious disease has been well established in humans, particularly in Mycobacterium tuberculosis infection. However, significant species differences exist between the immune system of humans and bovines, including effects induced by vitamin D3. This fact highlights the need for continued study of the relationship between vitamin D3 and bovine immunity, especially during different stages of paratuberculosis.
Collapse
Affiliation(s)
- Taylor L. T. Wherry
- Department of Veterinary Pathology, Iowa State University, Ames, IA 50011, USA
- Infectious Bacterial Diseases Research Unit, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), National Animal Disease Center, Ames, IA 50010, USA
| | - Judith R. Stabel
- Infectious Bacterial Diseases Research Unit, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), National Animal Disease Center, Ames, IA 50010, USA
| |
Collapse
|
9
|
Jiao X, Li K, Geng M, Li K, Liang W, Zhang J, Zhang Q, Gao H, Wei X, Yang J. Activated T cells are the cellular source of IL-22 that enhances proliferation and survival of lymphocytes in Nile tilapia. FISH & SHELLFISH IMMUNOLOGY 2022; 128:216-227. [PMID: 35934242 DOI: 10.1016/j.fsi.2022.07.079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
As a pleiotropic cytokine mainly secreted by CD4+ T cells, interleukin (IL)-22 plays an important role in immune regulation and infection elimination. Despite IL-22 homologues have been identified in non-mammal, whether and how IL-22 participates in the adaptive immune response of early vertebrates have not been fully addressed. In this study, we identified an evolutionarily conserved IL-22 from Nile tilapia Oreochromis niloticus (defined as OnIL-22), proved by its properties regarding sequence, gene structure, functional domain, tertiary structure and phylogeny. IL-22 was broadly expressed in lymphoid-related tissues of tilapia, and with relatively higher levels in skin, gill, intestine and liver. The expression of OnIL-22 in spleen lymphocytes was markedly induced at the adaptive immune stage after Streptococcus agalactiae infection. Moreover, once lymphocytes were activated by PMA plus ionomycin or T-cell specific mitogen PHA in vitro, OnIL-22 expression was obviously up-regulated at both mRNA and protein levels. These results thus suggest that activated T cells produce IL-22 to take part in the adaptive immune response of tilapia. Furthermore, treatment of lymphocytes with recombinant OnIL-22 increased the expression of genes related to proliferation and survival, and further promoted the proliferation and reduced the apoptosis of lymphocytes during bacterial infection or T-cell activation. These cellular effects of IL-22 seem to be associated with JAK1/STAT3 axis downstream of IL-22, because IL-22 application not only elevated the mRNA expression of JAK1 and STAT3, but also enhanced their phosphorylation in lymphocytes. Altogether, we suggest that activated T cells produce IL-22 to promote lymphocyte proliferation and survival probability via JAK1/STAT3 signaling pathway, thus participating in adaptive immune response of Nile tilapia. Our study therefore provides helpful perspective for understanding the function and mechanism of adaptive immune system in teleost.
Collapse
Affiliation(s)
- Xinying Jiao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ming Geng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kunming Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wei Liang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Haiyou Gao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
10
|
Khalid H, van Hooij A, Connelley TK, Geluk A, Hope JC. Protein Levels of Pro-Inflammatory Cytokines and Chemokines as Biomarkers of Mycobacterium bovis Infection and BCG Vaccination in Cattle. Pathogens 2022; 11:pathogens11070738. [PMID: 35889984 PMCID: PMC9320177 DOI: 10.3390/pathogens11070738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 12/10/2022] Open
Abstract
Bovine tuberculosis (bTB), caused by Mycobacterium bovis, is a globally prevalent infectious disease with significant animal welfare and economic impact. Difficulties in implementing test-and-slaughter measures in low- and middle-income countries (LMICs) and the underperformance of the current diagnostics establish a clear need to develop improved diagnostics. Adaptive immunity biomarkers other than IFNγ could be useful as suggested by various gene expression studies; however, a comprehensive assessment at the protein level is lacking. Here, we screened a range of chemokines and cytokines for their potential as biomarkers in samples from M. bovis experimentally challenged or naive animals. Although serum concentrations for most proteins were low, the pro-inflammatory markers, IL-2, CXCL-9, IP-10 and CCL4, in addition to IFNγ, were found to be significantly elevated in bovine tuberculin (PPDb)-stimulated whole blood supernatants. Further assessment of these molecules in BCG-vaccinated with or without subsequent M. bovis challenge or naive animals revealed that PPDb-specific IL-2 and IP-10, in addition to IFNγ, could discriminate naive and BCG-vaccinated from M. bovis challenged animals. Moreover, these proteins, along with CCL4, showed DIVA potential, i.e., enabling differentiation of M. bovis-infected animals from BCG-vaccinated animals. Combined analysis of cytokines and chemokines could also accurately identify M. bovis infection with strong correlations observed between PPDb-specific IFNγ, IL-2 and IP-10 levels. This provides proof of concept for utilizing multiple biomarker signatures for discrimination of animals with respect to M. bovis infection or BCG vaccination status.
Collapse
Affiliation(s)
- Hamza Khalid
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK;
- Center for Inflammation Research, The Queen’s Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
- Correspondence: (H.K.); (J.C.H.)
| | - Anouk van Hooij
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.v.H.); (A.G.)
| | - Timothy K. Connelley
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK;
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.v.H.); (A.G.)
| | - Jayne C. Hope
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK;
- Correspondence: (H.K.); (J.C.H.)
| |
Collapse
|
11
|
Elsayed MSAE, Salah A, Elbadee AA, Roshdy T. Real-time PCR using atpE, conventional PCR targeting different regions of difference, and flow cytometry for confirmation of Mycobacterium bovis in buffaloes and cattle from the Delta area of Egypt. BMC Microbiol 2022; 22:154. [PMID: 35689185 PMCID: PMC9188198 DOI: 10.1186/s12866-022-02568-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
Background Mycobacterium bovis notoriously causes detrimental infections in bovines and humans. In this study, 1500 buffaloes and 2200 cattle were tested by single intradermal comparative cervical tuberculin test and compared with the detection rates of M. bovis isolation, real-time and simplex PCR, and flow Cytometry. Results The tuberculin test is the reference test in Egypt, the positive rate was 54/3700 (1.5%) composed of 18/1500 (1.2%) buffaloes and 36/2200 (1.6%) cattle which were mandatorily slaughtered under the Egyptian legislation, after postmortem examination the non-visible-lesion proportion was 39/54 (72.2%) which surpassed the visible-lesion rate 15/54 (27.8%) with (p < 0.0001). The samples from each case were pooled into one sample representing the case, and the isolation rate of M. bovis was 25/54 (46.3%). Real-time PCR using atpE was positive for mycobacteria on the genus level in 18/18 (100%) and 5/5 (100%) of tissue samples and isolates, respectively; simplex PCR detected M. bovis in 44/54 (81.5%) and 25/25 (100%) of tissue samples and isolates, respectively. Flow Cytometry evaluation of the CD4+, CD8+, WC1+δγ, and CD2+ cell phenotypes showed increased counts in the tuberculin-positive cases compared with negative cases (p < 0.0001), and these phenotypes in the tuberculin-positive cases increased after antigen stimulation than in the negative cases (p < 0.0001). Detection rates of PCR techniques and flow Cytometry exceeded that of bacterial isolation (p < 0.0001) and exhibited a strong correlation. Conclusions The skin test suffers from interference from non-tuberculous mycobacteria able to cause false-positive reactions in cattle and other species. Real-time PCR using atpE, conventional PCR targeting RDs, and flow Cytometry are rapid and accurate methods that correlate with the isolation and can be promising for detection and confirmation of infected live and slaughtered cases.
Collapse
Affiliation(s)
- Mohamed Sabry Abd Elraheam Elsayed
- Department of Bacteriology, Mycology, and Immunology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menoufia, 32897, Egypt.
| | - Ahmed Salah
- Department of Molecular Biology, Genetic Engineering, and Biotechnology Research Institute, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Ahmed Abd Elbadee
- Animal Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Tamer Roshdy
- Department of Molecular Biology, Genetic Engineering, and Biotechnology Research Institute, University of Sadat City, Sadat City, Menoufia, Egypt
| |
Collapse
|
12
|
Le Page L, Baldwin CL, Telfer JC. γδ T cells in artiodactyls: Focus on swine. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 128:104334. [PMID: 34919982 DOI: 10.1016/j.dci.2021.104334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Vaccination is the most effective medical strategy for disease prevention but there is a need to improve livestock vaccine efficacy. Understanding the structure of the immune system of swine, which are considered a γδ T cell "high" species, and thus, particularly how to engage their γδ T cells for immune responses, may allow for development of vaccine optimization strategies. The propensity of γδ T cells to home to specific tissues, secrete pro-inflammatory and regulatory cytokines, exhibit memory or recall responses and even function as antigen-presenting cells for αβ T cells supports the concept that they have enormous potential for priming by next generation vaccine constructs to contribute to protective immunity. γδ T cells exhibit several innate-like antigen recognition properties including the ability to recognize antigen in the absence of presentation via major histocompatibility complex (MHC) molecules enabling γδ T cells to recognize an array of peptides but also non-peptide antigens in a T cell receptor-dependent manner. γδ T cell subpopulations in ruminants and swine can be distinguished based on differential expression of the hybrid co-receptor and pattern recognition receptors (PRR) known as workshop cluster 1 (WC1). Expression of various PRR and other innate-like immune receptors diversifies the antigen recognition potential of γδ T cells. Finally, γδ T cells in livestock are potent producers of critical master regulator cytokines such as interferon (IFN)-γ and interleukin (IL)-17, whose production orchestrates downstream cytokine and chemokine production by other cells, thereby shaping the immune response as a whole. Our knowledge of the biology, receptor expression and response to infectious diseases by swine γδ T cells is reviewed here.
Collapse
Affiliation(s)
- Lauren Le Page
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Cynthia L Baldwin
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Janice C Telfer
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
13
|
Palmer MV, Kanipe C, Boggiatto PM. The Bovine Tuberculoid Granuloma. Pathogens 2022; 11:61. [PMID: 35056009 PMCID: PMC8780557 DOI: 10.3390/pathogens11010061] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 02/05/2023] Open
Abstract
The bovine tuberculoid granuloma is the hallmark lesion of bovine tuberculosis (bTB) due to Mycobacterium bovis infection. The pathogenesis of bTB, and thereby the process of bovine tuberculoid granuloma development, involves the recruitment, activation, and maintenance of cells under the influence of antigen, cytokines and chemokines in affected lungs and regional lymph nodes. The granuloma is key to successful control of bTB by preventing pathogen dissemination through containment by cellular and fibrotic layers. Paradoxically, however, it may also provide a niche for bacterial replication. The morphologic and cellular characteristics of granulomas have been used to gauge disease severity in bTB pathogenesis and vaccine efficacy studies. As such, it is critical to understand the complex mechanisms behind granuloma initiation, development, and maintenance.
Collapse
Affiliation(s)
- Mitchell V. Palmer
- Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA; (C.K.); (P.M.B.)
| | | | | |
Collapse
|
14
|
Lücke J, Shiri AM, Zhang T, Kempski J, Giannou AD, Huber S. Rationalizing heptadecaphobia: T H 17 cells and associated cytokines in cancer and metastasis. FEBS J 2021; 288:6942-6971. [PMID: 33448148 DOI: 10.1111/febs.15711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/13/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the leading causes of death worldwide. When cancer patients are diagnosed with metastasis, meaning that the primary tumor has spread to at least one different site, their life expectancy decreases dramatically. In the past decade, the immune system´s role in fighting cancer and metastasis has been studied extensively. Importantly, immune cells and inflammatory reactions generate potent antitumor responses but also contribute to tumor development. However, the molecular and cellular mechanisms underlying this dichotomic interaction between the immune system and cancer are still poorly understood. Recently, a spotlight has been cast on the distinct subsets of immune cells and their derived cytokines since evidence has implicated their crucial impact on cancer development. T helper 17 cell (TH 17) cells, which express the master transcriptional factor Retinoic acid-receptor-related orphan receptor gamma t, are among these critical cell subsets and are defined by their production of type 3 cytokines, such as IL-17A, IL-17F, and IL-22. Depending on the tumor microenvironment, these cytokines can also be produced by other immune cell sources, such as T cytotoxic 17 cell, innate lymphoid cells, NKT cells, or γδ T cells. To date, a lot of data have been collected describing the divergent functions of IL-17A, IL-17F, and IL-22 in malignancies. In this comprehensive review, we discuss the role of these TH 17- and non-TH 17-derived type 3 cytokines in different tumor entities. Furthermore, we will provide a structured insight into the strict regulation and subsequent downstream mechanisms of these cytokines in cancer and metastasis.
Collapse
Affiliation(s)
- Jöran Lücke
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| | - Ahmad Mustafa Shiri
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| | - Tao Zhang
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| | - Jan Kempski
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- The Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Anastasios D Giannou
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Germany
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
15
|
Alvarez AH. Revisiting tuberculosis screening: An insight to complementary diagnosis and prospective molecular approaches for the recognition of the dormant TB infection in human and cattle hosts. Microbiol Res 2021; 252:126853. [PMID: 34536677 DOI: 10.1016/j.micres.2021.126853] [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: 06/22/2021] [Revised: 08/21/2021] [Accepted: 08/22/2021] [Indexed: 12/17/2022]
Abstract
Tuberculosis (TB) is defined as a chronic infection in both human and cattle hosts and many subclinical cases remain undetected. After the pathogen is inhaled by a host, phagocyted bacilli can persist inside macrophages surviving intracellularly. Hosts develop granulomatous lesions in the lungs or lymph nodes, limiting infection. However, bacilli become persister cells. Immunological diagnosis of TB is performed basically by routine tuberculin skin test (TST), and in some cases, by ancillary interferon-gamma release assay (IGRA). The concept of human latent TB infection (LTBI) by M. tuberculosis is recognized in cohorts without symptoms by routine clinical diagnostic tests, and nowadays IGRA tests are used to confirm LTBI with either active or latent specific antigens of M. tuberculosis. On the other hand, dormant infection in cattle by M. bovis has not been described by TST or IGRA testing as complications occur by cross-reactive immune responses to homolog antigens of environmental mycobacteria or a false-negative test by anergic states of a wained bovine immunity, evidencing the need for deciphering more specific biomarkers by new-generation platforms of analysis for detection of M. bovis dormant infection. The study and description of bovine latent TB infection (boLTBI) would permit the recognition of hidden animal infection with an increase in the sensitivity of routine tests for an accurate estimation of infected dairy cattle. Evidence of immunological and experimental analysis of LTBI should be taken into account to improve the study and the description of the still neglected boLTBI.
Collapse
Affiliation(s)
- Angel H Alvarez
- Centro de Investigación y Asistencia en Tecnología y diseño del Estado de Jalisco A.C. (CIATEJ), Consejo Nacional de Ciencia y Tecnología (CONACYT), Av. Normalistas 800 C.P. 44270, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
16
|
Lücke J, Sabihi M, Zhang T, Bauditz LF, Shiri AM, Giannou AD, Huber S. The good and the bad about separation anxiety: roles of IL-22 and IL-22BP in liver pathologies. Semin Immunopathol 2021; 43:591-607. [PMID: 33851257 PMCID: PMC8443499 DOI: 10.1007/s00281-021-00854-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022]
Abstract
The human liver fulfills several vital tasks daily and possesses an impressive ability to self-regenerate. However, the capacity of this self-healing process can be exhausted by a variety of different liver diseases, such as alcoholic liver damage, viral hepatitis, or hepatocellular carcinoma. Over time, all these diseases generally lead to progressive liver failure that can become fatal if left untreated. Thus, a great effort has been directed towards the development of innovative therapies. The most recently discovered therapies often involve modifying the patient's immune system to enhance a beneficial immune response. Current data suggest that, among others, the cytokine IL-22 might be a promising therapeutical candidate. IL-22 and its endogenous antagonist, IL-22BP, have been under thorough scientific investigation for nearly 20 years. While IL-22 is mainly produced by TH22 cells, ILC3s, NKT cells, or γδ T cells, sources of IL-22BP include dendritic cells, eosinophils, and CD4+ cells. In many settings, IL-22 was shown to promote regenerative potential and, thus, could protect tissues from pathogens and damage. However, the effects of IL-22 during carcinogenesis are more ambiguous and depend on the tumor entity and microenvironment. In line with its capabilities of neutralizing IL-22 in vivo, IL-22BP possesses often, but not always, an inverse expression pattern compared to its ligand. In this comprehensive review, we will summarize past and current findings regarding the roles of IL-22 and IL-22BP in liver diseases with a particular focus on the leading causes of advanced liver failure, namely, liver infections, liver damage, and liver malignancies.
Collapse
Affiliation(s)
- Jöran Lücke
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Morsal Sabihi
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tao Zhang
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lennart Fynn Bauditz
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ahmad Mustafa Shiri
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Anastasios D Giannou
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
17
|
Blanco FC, Gravisaco MJ, Bigi MM, García EA, Marquez C, McNeil M, Jackson M, Bigi F. Identifying Bacterial and Host Factors Involved in the Interaction of Mycobacterium bovis with the Bovine Innate Immune Cells. Front Immunol 2021; 12:674643. [PMID: 34335572 PMCID: PMC8319915 DOI: 10.3389/fimmu.2021.674643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis is an important animal and zoonotic disease caused by Mycobacterium bovis. The innate immune response is the first line of defense against pathogens and is also crucial for the development of an efficient adaptive immune response. In this study we used an in vitro co-culture model of antigen presenting cells (APC) and autologous lymphocytes derived from peripheral blood mononuclear cells to identify the cell populations and immune mediators that participate in the development of an efficient innate response capable of controlling the intracellular replication of M. bovis. After M. bovis infection, bovine immune cell cultures displayed upregulated levels of iNOS, IL-22 and IFN-γ and the induction of the innate immune response was dependent on the presence of differentiated APC. Among the analyzed M. bovis isolates, only a live virulent M. bovis isolate induced an efficient innate immune response, which was increased upon stimulation of cell co-cultures with the M. bovis culture supernatant. Moreover, we demonstrated that an allelic variation of the early secreted protein ESAT-6 (ESAT6 T63A) expressed in the virulent strain is involved in this increased innate immune response. These results highlight the relevance of the compounds secreted by live M. bovis as well as the variability among the assessed M. bovis strains to induce an efficient innate immune response.
Collapse
Affiliation(s)
- Federico Carlos Blanco
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
- (Consejo Nacional de Investigaciones Científicas y Tecnológicas) National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - María José Gravisaco
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
| | - María Mercedes Bigi
- (Facultad de Agronomía, Universidad de Buenos Aires) School of Agronomy, University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Elizabeth Andrea García
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
| | - Cecilia Marquez
- High Technology Analytical Centre, Laboratory, Buenos Aires, Argentina
| | - Mike McNeil
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mary Jackson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Fabiana Bigi
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
- (Consejo Nacional de Investigaciones Científicas y Tecnológicas) National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
18
|
Jiang Q, Yang G, Xiao F, Xie J, Wang S, Lu L, Cui D. Role of Th22 Cells in the Pathogenesis of Autoimmune Diseases. Front Immunol 2021; 12:688066. [PMID: 34295334 PMCID: PMC8290841 DOI: 10.3389/fimmu.2021.688066] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Upon antigenic stimulation, naïve CD4+T cells differentiate into different subsets and secrete various cytokines to exert biological effects. Th22 cells, a newly identified CD4+T cell subset,are distinct from the Th1, Th2 and Th17 subsets. Th22 cells secrete certain cytokines such as IL-22, IL-13 and TNF-α, but not others, such as IL-17, IL-4, or interferon-γ (IFN-γ), and they express chemokine receptors CCR4, CCR6 and CCR10. Th22 cells were initially found to play a role in skin inflammatory diseases, but recent studies have demonstrated their involvement in the development of various autoimmune diseases. Here, we review research advances in the origin, characteristics and effector mechanisms of Th22 cells, with an emphasis on the role of Th22 cells and their main effector cytokine IL-22 in the pathogenesis of autoimmune diseases. The findings presented here may facilitate the development of new therapeutic strategies for targeting these diseases.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Blood Transfusion, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Guocan Yang
- Department of Blood Transfusion, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Fan Xiao
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong; Chongqing International Institute for Immunology, Chongqing, China
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong; Chongqing International Institute for Immunology, Chongqing, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
19
|
Temporal dynamics of intradermal cytokine response to tuberculin in Mycobacterium bovis BCG-vaccinated cattle using sampling microneedles. Sci Rep 2021; 11:7074. [PMID: 33782422 PMCID: PMC8007627 DOI: 10.1038/s41598-021-86398-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/15/2021] [Indexed: 11/08/2022] Open
Abstract
Bovine tuberculosis (bTB) is a disease of livestock with severe and worldwide economic, animal welfare and zoonotic consequences. Application of test-and-slaughter-based control polices reliant on tuberculin skin testing has been the mainstay of bTB control in cattle. However, little is known about the temporal development of the bovine tuberculin skin test response at the dermal sites of antigen injection. To fill this knowledge gap, we applied minimally-invasive sampling microneedles (SMNs) for intradermal sampling of interstitial fluid at the tuberculin skin test sites in Mycobacterium bovis BCG-vaccinated calves and determined the temporal dynamics of a panel of 15 cytokines and chemokines in situ and in the peripheral blood. The results reveal an orchestrated and coordinated cytokine and local chemokine response, identified IL-1RA as a potential soluble biomarker of a positive tuberculin skin response, and confirmed the utility of IFN-γ and IP-10 for bTB detection in blood-based assays. Together, the results highlight the utility of SMNs to identify novel biomarkers and provide mechanistic insights on the intradermal cytokine and chemokine responses associated with the tuberculin skin test in BCG-sensitized cattle.
Collapse
|
20
|
Smith K, Kleynhans L, Warren RM, Goosen WJ, Miller MA. Cell-Mediated Immunological Biomarkers and Their Diagnostic Application in Livestock and Wildlife Infected With Mycobacterium bovis. Front Immunol 2021; 12:639605. [PMID: 33746980 PMCID: PMC7969648 DOI: 10.3389/fimmu.2021.639605] [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: 12/09/2020] [Accepted: 02/08/2021] [Indexed: 01/06/2023] Open
Abstract
Mycobacterium bovis has the largest host range of the Mycobacterium tuberculosis complex and infects domestic animal species, wildlife, and humans. The presence of global wildlife maintenance hosts complicates bovine tuberculosis (bTB) control efforts and further threatens livestock and wildlife-related industries. Thus, it is imperative that early and accurate detection of M. bovis in all affected animal species is achieved. Further, an improved understanding of the complex species-specific host immune responses to M. bovis could enable the development of diagnostic tests that not only identify infected animals but distinguish between infection and active disease. The primary bTB screening standard worldwide remains the tuberculin skin test (TST) that presents several test performance and logistical limitations. Hence additional tests are used, most commonly an interferon-gamma (IFN-γ) release assay (IGRA) that, similar to the TST, measures a cell-mediated immune (CMI) response to M. bovis. There are various cytokines and chemokines, in addition to IFN-γ, involved in the CMI component of host adaptive immunity. Due to the dominance of CMI-based responses to mycobacterial infection, cytokine and chemokine biomarkers have become a focus for diagnostic tests in livestock and wildlife. Therefore, this review describes the current understanding of host immune responses to M. bovis as it pertains to the development of diagnostic tools using CMI-based biomarkers in both gene expression and protein release assays, and their limitations. Although the study of CMI biomarkers has advanced fundamental understanding of the complex host-M. bovis interplay and bTB progression, resulting in development of several promising diagnostic assays, most of this research remains limited to cattle. Considering differences in host susceptibility, transmission and immune responses, and the wide variety of M. bovis-affected animal species, knowledge gaps continue to pose some of the biggest challenges to the improvement of M. bovis and bTB diagnosis.
Collapse
Affiliation(s)
- Katrin Smith
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Léanie Kleynhans
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Robin M Warren
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Wynand J Goosen
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Michele A Miller
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
21
|
Yap YA, McLeod KH, McKenzie CI, Gavin PG, Davalos-Salas M, Richards JL, Moore RJ, Lockett TJ, Clarke JM, Eng VV, Pearson JS, Hamilton-Williams EE, Mackay CR, Mariño E. An acetate-yielding diet imprints an immune and anti-microbial programme against enteric infection. Clin Transl Immunology 2021; 10:e1233. [PMID: 33489123 PMCID: PMC7809703 DOI: 10.1002/cti2.1233] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 11/16/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Objectives During gastrointestinal infection, dysbiosis can result in decreased production of microbially derived short‐chain fatty acids (SCFAs). In response to the presence of intestinal pathogens, we examined whether an engineered acetate‐ or butyrate‐releasing diet can rectify the deficiency of SCFAs and lead to the resolution of enteric infection. Methods We tested whether a high acetate‐ or butyrate‐producing diet (HAMSA or HAMSB, respectively) condition Citrobacterrodentium infection in mice and assess its impact on host‐microbiota interactions. We analysed the adaptive and innate immune responses, changes in gut microbiome function, epithelial barrier function and the molecular mechanism via metabolite sensing G protein‐coupled receptor 43 (GPR43) and IL‐22 expression. Results HAMSA diet rectified the deficiency in acetate production and protected against enteric infection. Increased SCFAs affect the expression of pathogen virulence genes. HAMSA diet promoted compositional and functional changes in the gut microbiota during infection similar to healthy microbiota from non‐infected mice. Bacterial changes were evidenced by the production of proteins involved in acetate utilisation, starch and sugar degradation, amino acid biosynthesis, carbohydrate transport and metabolism. HAMSA diet also induced changes in host proteins critical in glycolysis, wound healing such as GPX1 and epithelial architecture such as EZR1 and PFN1. Dietary acetate assisted in rapid epithelial repair, as shown by increased colonic Muc‐2, Il‐22, and anti‐microbial peptides. We found that acetate increased numbers of colonic IL‐22 producing TCRαβ+CD8αβ+ and TCRγδ+CD8αα+ intraepithelial lymphocytes expressing GPR43. Conclusion HAMSA diet may be an effective therapeutic approach for fighting inflammation and enteric infections and offer a safe alternative that may impact on human health.
Collapse
Affiliation(s)
- Yu Anne Yap
- Department of Biochemistry and Molecular Biology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia
| | - Keiran H McLeod
- Department of Biochemistry and Molecular Biology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia
| | - Craig I McKenzie
- Department of Biochemistry and Molecular Biology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia
| | - Patrick G Gavin
- The University of Queensland Diamantina Institute The University of Queensland Brisbane QLD Australia
| | - Mercedes Davalos-Salas
- Department of Biochemistry and Molecular Biology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia
| | - James L Richards
- Department of Biochemistry and Molecular Biology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia
| | - Robert J Moore
- Department of Microbiology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia.,School of Science RMIT University Bundoora VIC Australia
| | | | | | - Vik Ven Eng
- Department of Microbiology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia.,Centre for Innate Immunity and Infectious Diseases Hudson Institute of Medical Research Clayton, Melbourne VIC Australia
| | - Jaclyn S Pearson
- Department of Microbiology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia.,Centre for Innate Immunity and Infectious Diseases Hudson Institute of Medical Research Clayton, Melbourne VIC Australia.,Department of Molecular and Translational Research Monash University Clayton, Melbourne VIC Australia
| | - Emma E Hamilton-Williams
- The University of Queensland Diamantina Institute The University of Queensland Brisbane QLD Australia
| | - Charles R Mackay
- Department of Microbiology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia
| | - Eliana Mariño
- Department of Biochemistry and Molecular Biology Infection and Immunity Program Biomedicine Discovery Institute Monash University Clayton, Melbourne VIC Australia
| |
Collapse
|
22
|
Xuan X, Zhang L, Tian C, Wu T, Ye H, Cao J, Chen F, Liang Y, Yang H, Huang C. Interleukin-22 and connective tissue diseases: emerging role in pathogenesis and therapy. Cell Biosci 2021; 11:2. [PMID: 33407883 PMCID: PMC7788945 DOI: 10.1186/s13578-020-00504-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 11/27/2020] [Indexed: 01/14/2023] Open
Abstract
Interleukin-22 (IL-22), a member of the IL-10 family of cytokines, is produced by a number of immune cells involved in the immune microenvironment of the body. IL-22 plays its pivotal roles by binding to the IL-22 receptor complex (IL-22R) and subsequently activating the IL-22R downstream signalling pathway. It has recently been reported that IL-22 also contributes to the pathogenesis of many connective tissue diseases (CTDs). In this review, we will discuss the role of IL-22 in several CTDs, such as system lupus erythematosus, rheumatoid arthritis, Sjögren’s syndrome, systemic sclerosis and dermatomyositis, suggesting that IL-22 may be a potential therapeutic target in CTDs.
Collapse
Affiliation(s)
- Xiuyun Xuan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Lin Zhang
- Department of Gerontology, Jinan City People's Hospital, Jinan, 271199, Shandong, China
| | - Chunxia Tian
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ting Wu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Haihua Ye
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Juanmei Cao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Fangqi Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yan Liang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Huilan Yang
- Department of Dermatology, General Hospital of Southern Theatre Command, Guangzhou, 510000, China.
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
23
|
Gupta SK, Parlane NA, Luo D, Rehm BHA, Heiser A, Buddle BM, Wedlock DN. Self-assembled particulate vaccine elicits strong immune responses and reduces Mycobacterium avium subsp. paratuberculosis infection in mice. Sci Rep 2020; 10:22289. [PMID: 33339863 PMCID: PMC7749150 DOI: 10.1038/s41598-020-79407-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/08/2020] [Indexed: 01/14/2023] Open
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP) causes chronic progressive granulomatous enteritis leading to diarrhoea, weight loss, and eventual death in ruminants. Commercially available vaccines provide only partial protection against MAP infection and can compromise the use of bovine tuberculosis diagnostic tests. Here, we report the development of a protein-particle-based vaccine containing MAP antigens Ag85A202-347-SOD1-72-Ag85B173-330-74F1-148+669-786 as a fusion ('MAP fusion protein particle'). The fusion antigen displayed on protein particles was identified using mass spectrometry. Surface exposure and accessibility of the fusion antigen was confirmed by flow cytometry and ELISA. The MAP fusion protein particle vaccine induced strong antigen-specific T-cell immune responses in mice, as indicated by increased cytokine (IFN-γ and IL-17A) and costimulatory signals (CD40 and CD86) in these animals. Following MAP-challenge, a significant reduction in bacterial burden was observed in multiple organs of the mice vaccinated with the MAP fusion protein particle vaccine compared with the PBS group. The reduction in severity of MAP infection conferred by the MAP fusion protein particle vaccine was similar to that of Silirum and recombinant protein vaccines. Overall, the results provide evidence that MAP antigens can be engineered as a protein particulate vaccine capable of inducing immunity against MAP infection. This utility offers an attractive platform for production of low-cost particulate vaccines against other intracellular pathogens.
Collapse
Affiliation(s)
- Sandeep K Gupta
- Grasslands Research Centre, AgResearch, Hopkirk Research Institute, Private Bag 11008, Palmerston North, 4442, New Zealand.
| | - Natalie A Parlane
- Grasslands Research Centre, AgResearch, Hopkirk Research Institute, Private Bag 11008, Palmerston North, 4442, New Zealand
| | - Dongwen Luo
- Bioinformatics and Statistics, AgResearch, Palmerston North, New Zealand
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
- Menzies Health Institute Queensland (MHIQ), Griffith University (Gold Coast Campus), Southport, Australia
| | - Axel Heiser
- Grasslands Research Centre, AgResearch, Hopkirk Research Institute, Private Bag 11008, Palmerston North, 4442, New Zealand
| | - Bryce M Buddle
- Grasslands Research Centre, AgResearch, Hopkirk Research Institute, Private Bag 11008, Palmerston North, 4442, New Zealand
| | - D Neil Wedlock
- Grasslands Research Centre, AgResearch, Hopkirk Research Institute, Private Bag 11008, Palmerston North, 4442, New Zealand
| |
Collapse
|
24
|
Benedictus L, Steinbach S, Holder T, Bakker D, Vrettou C, Morrison WI, Vordermeier M, Connelley T. Hydrophobic Mycobacterial Antigens Elicit Polyfunctional T Cells in Mycobacterium bovis Immunized Cattle: Association With Protection Against Challenge? Front Immunol 2020; 11:588180. [PMID: 33281817 PMCID: PMC7688591 DOI: 10.3389/fimmu.2020.588180] [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: 07/28/2020] [Accepted: 10/20/2020] [Indexed: 01/09/2023] Open
Abstract
Bovine tuberculosis (bTB), caused by Mycobacterium bovis, is a chronic disease of cattle with a detrimental impact on food quality and production. Research on bTB vaccines has predominantly been focused on proteinaceous antigens. However, mycobacteria have a thick and intricate lipid outer layer and lipids as well as lipopeptides are important for immune-evasion and virulence. In humans, lipid extracts of M. tuberculosis have been shown to elicit immune responses effective against M. tuberculosis in vitro. Chloroform-methanol extraction (CME) was applied to M. bovis BCG to obtain a hydrophobic antigen extract (CMEbcg) containing lipids and lipopeptides. CMEbcg stimulated IFN-γ+IL-2+ and IL-17A+IL-22+ polyfunctional T cells and elicited T cell responses with a Th1 and Th17 cytokine release profile in both M. bovis BCG vaccinated and M. bovis challenged calves. Lipopeptides were shown to be the immunodominant antigens in CMEbcg, stimulating CD4 T cells via MHC class II. CMEbcg expanded T cells killed CMEbcg loaded monocytes and the CMEbcg-specific CD3 T cell proliferative response following M. bovis BCG vaccination was the best predictor for reduced pathology following challenge with M. bovis. Although the high predictive value of CMEbcg-specific immune responses does not confirm a causal relationship with protection against M. bovis challenge, when taking into account the in vitro antimycobacterial phenotype of CMEbcg-specific T cells (e.g. Th1/Th17 cytokine profile), it is indicative that CMEbcg-specific immune responses could play a functional role in immunity against M. bovis. Based on these findings we conclude that lipopeptides of M. bovis are potential novel subunit vaccine candidates and that further studies into the functional characterization of lipopeptide-specific immune responses together with their role in protection against bovine tuberculosis are warranted.
Collapse
Affiliation(s)
- Lindert Benedictus
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - Sabine Steinbach
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Thomas Holder
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Douwe Bakker
- Independent Researcher and Technical Consultant, Lelystad, Netherlands
| | - Christina Vrettou
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - W Ivan Morrison
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - Martin Vordermeier
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom.,Centre for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, University of Aberystwyth, Aberystwyth, United Kingdom
| | - Timothy Connelley
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| |
Collapse
|
25
|
Rainard P, Cunha P, Martins RP, Gilbert FB, Germon P, Foucras G. Type 3 immunity: a perspective for the defense of the mammary gland against infections. Vet Res 2020; 51:129. [PMID: 33059767 PMCID: PMC7559147 DOI: 10.1186/s13567-020-00852-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/22/2020] [Indexed: 12/23/2022] Open
Abstract
Type 3 immunity encompasses innate and adaptive immune responses mediated by cells that produce the signature cytokines IL-17A and IL-17F. This class of effector immunity is particularly adept at controlling infections by pyogenic extracellular bacteria at epithelial barriers. Since mastitis results from infections by bacteria such as streptococci, staphylococci and coliform bacteria that cause neutrophilic inflammation, type 3 immunity can be expected to be mobilized at the mammary gland. In effect, the main defenses of this organ are provided by epithelial cells and neutrophils, which are the main terminal effectors of type 3 immunity. In addition to theoretical grounds, there is observational and experimental evidence that supports a role for type 3 immunity in the mammary gland, such as the production of IL-17A, IL-17F, and IL-22 in milk and mammary tissue during infection, although their respective sources remain to be fully identified. Moreover, mouse mastitis models have shown a positive effect of IL-17A on the course of mastitis. A lot remains to be uncovered before we can safely harness type 3 immunity to reinforce mammary gland defenses through innate immune training or vaccination. However, this is a promising way to find new means of improving mammary gland defenses against infection.
Collapse
Affiliation(s)
- Pascal Rainard
- ISP, INRAE, Université de Tours, UMR1282, Tours, Nouzilly, France.
| | - Patricia Cunha
- ISP, INRAE, Université de Tours, UMR1282, Tours, Nouzilly, France
| | | | | | - Pierre Germon
- ISP, INRAE, Université de Tours, UMR1282, Tours, Nouzilly, France
| | - Gilles Foucras
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| |
Collapse
|
26
|
Early dynamics of innate immunity during pulmonary tuberculosis. Immunol Lett 2020; 221:56-60. [DOI: 10.1016/j.imlet.2020.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 01/22/2023]
|
27
|
Che Y, Su Z, Xia L. Effects of IL-22 on cardiovascular diseases. Int Immunopharmacol 2020; 81:106277. [PMID: 32062077 DOI: 10.1016/j.intimp.2020.106277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/29/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022]
Abstract
Interleukin-22 (IL-22), which belongs to the IL-10 family, is an alpha helix cytokine specifically produced by many lymphocytes, such as Th1, Th17, Th22, ILCs, CD4+ and CD8+ T cells. In recent years, more and more studies have demonstrated that IL-22 has an interesting relationship with various cardiovascular diseases, including myocarditis, myocardial infarction, atherosclerosis, and other cardiovascular diseases, and IL-22 signal may play a dual role in cardiovascular diseases. Here, we summarize the recent progress on the source, function, regulation of IL-22 and the effects of IL-22 signal in cardiovascular diseases. The study of IL-22 will suggest more specific strategies to maneuver these functions for the effective treatment of cardiovascular diseases and future clinical treatment.
Collapse
Affiliation(s)
- Yang Che
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, China; Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
28
|
DeKuiper JL, Cooperider HE, Lubben N, Ancel CM, Coussens PM. Mycobacterium avium Subspecies paratuberculosis Drives an Innate Th17-Like T Cell Response Regardless of the Presence of Antigen-Presenting Cells. Front Vet Sci 2020; 7:108. [PMID: 32258066 PMCID: PMC7089878 DOI: 10.3389/fvets.2020.00108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/12/2020] [Indexed: 01/05/2023] Open
Abstract
The gastrointestinal disease of ruminants is clinically known as Johne's disease (JD) and is caused by Mycobacterium avium subspecies paratuberculosis (MAP). An accumulative effect by insensitive diagnostic tools, a long subclinical stage of infection, and lack of effective vaccines have made the control of JD difficult. Currently lacking in the model systems of JD are undefined correlates of protection and the sources of inflammation due to JD. As an alternative to commonly studied immune responses, such as the Th1/Th2 paradigm, a non-classical Th17 immune response to MAP has been suggested. Indeed MAP antigens induce mRNAs encoding the Th17-associated cytokines IL-17A, IL-17F, IL-22, IL-23, IL-27, and IFNγ in CD3+ T cell cultures as determined by RT-qPCR. Although not as robust as when cultured with monocyte-derived macrophages (MDMs), MAP is able to stimulate the upregulation of these cytokines from sorted CD3+ T cells in the absence of antigen-presenting cells (APCs). CD4+ and CD8+ T cells are the main contributors of IL-17A and IL-22 in the absence of APCs. However, MAP-stimulated MDMs are the main contributor of IL-23. In vivo, JD+ cows have more circulating IL-23 than JD– cows, suggesting that this proinflammatory cytokine may be important in the etiology of JD. Our data in this study continue to suggest that Th17-like cells and associated cytokines may indeed play an important role in the immune responses to MAP infection and the development or control of JD.
Collapse
Affiliation(s)
- Justin L DeKuiper
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Hannah E Cooperider
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Noah Lubben
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Caitlin M Ancel
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Paul M Coussens
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
29
|
Machado VS, Silva TH. Adaptive immunity in the postpartum uterus: Potential use of vaccines to control metritis. Theriogenology 2020; 150:201-209. [PMID: 31983466 DOI: 10.1016/j.theriogenology.2020.01.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/21/2022]
Abstract
After parturition, dairy cows rely on an effective innate immune response, through the actions of neutrophils, macrophages, and antimicrobial peptides, to clear the uterus from pathogenic bacteria, such as E. coli, Bacteroides spp, F. necrophorum and T. pyogenes. However, the role of adaptive immunity in the postpartum uterus is less understood. In this review, we explore concepts of mucosal adaptive immunity and discuss recent findings regarding the efficacy of vaccines to reduce metritis in dairy cows. Areas of lymphocytic aggregates are seen throughout the bovine reproductive tract after parturition, but it is unknown if their development is influenced by previous exposure to pathogens or other intrinsic factors. Through the actions of Treg cells and γδ T cells, the uterus is an immune-tolerant environment during pregnancy. After parturition, the dynamics in the endometrial and circulating lymphocytic populations differ among cows that develop uterine diseases and healthy counterparts. However, the functionality of those cells has not yet been determined. It has been hypothesized that cows that fail to switch their uterine environment from an anti-inflammatory state prior to parturition to a pro-inflammatory state after calving are more susceptible to uterine infections. Given the nature of metritis related pathogens and the importance of innate immunity to uterine defense mechanisms, we speculate that an adaptive immunity biased towards a Th1/Th17 cellular response will provide best protection against uterine infections. Few studies have evaluated the efficacy of immunization in reducing the incidence of metritis in dairy cows revealing inconsistent findings.
Collapse
Affiliation(s)
- V S Machado
- Department of Veterinary Sciences, College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX, 79409, United States.
| | - T H Silva
- Department of Veterinary Sciences, College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX, 79409, United States; Department of Animal Science, School of Animal Science and Food Engineering, University of Sao Paulo, Pirassununga, SP, 13635-900, Brazil
| |
Collapse
|
30
|
Palmer MV, Thacker TC, Rabideau MM, Jones GJ, Kanipe C, Vordermeier HM, Ray Waters W. Biomarkers of cell-mediated immunity to bovine tuberculosis. Vet Immunol Immunopathol 2019; 220:109988. [PMID: 31846797 DOI: 10.1016/j.vetimm.2019.109988] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/15/2019] [Accepted: 11/28/2019] [Indexed: 10/25/2022]
Abstract
Whole blood based assays, particularly interferon gamma (IFN-γ) release assays (IGRAs), are used for the diagnosis of both bovine and human tuberculosis (TB). The aim of the current study was to evaluate a panel of cytokines and chemokines for potential use as diagnostic readouts indicative of Mycobacterium bovis (M. bovis) infection in cattle. A gene expression assay was used to determine the kinetics of the response to M. bovis purified protein derivative and a fusion protein consisting of ESAT-6, CFP10, and Rv3615c upon aerosol infection with ∼104 cfu of M. bovis. The panel of biomarkers included: IFN-γ, CXCL9, CXCL10, CCL2, CCL3, TNF-α, IL-1α, IL-1β, IL-1Ra, IL-22, IL-21 and IL-13. Protein levels of IFN-γ, CXCL9, and CXCL10 were determined by ELISA. Findings suggest that CXCL9, CXCL10, IL-21, IL-13, and several acute phase cytokines may be worth pursuing as diagnostic biomarkers of M. bovis infection in cattle.
Collapse
Affiliation(s)
- Mitchell V Palmer
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA.
| | - Tyler C Thacker
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| | - Meaghan M Rabideau
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| | - Gareth J Jones
- TB Immunology and Vaccinology, Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surry UK
| | - Carly Kanipe
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA; Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - H Martin Vordermeier
- TB Immunology and Vaccinology, Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surry UK
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| |
Collapse
|
31
|
Cunha P, Vern YL, Gitton C, Germon P, Foucras G, Rainard P. Expansion, isolation and first characterization of bovine Th17 lymphocytes. Sci Rep 2019; 9:16115. [PMID: 31695097 PMCID: PMC6834651 DOI: 10.1038/s41598-019-52562-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin 17A-producing T helper cells (Th17) are CD4+ T cells that are crucial to immunity to extracellular bacteria. The roles of these cells in the bovine species are poorly defined, because the characterization of bovine Th17 cells lags behind for want of straightforward cultivation and isolation procedures. We have developed procedures to differentiate, expand, and isolate bovine Th17 cells from circulating CD4+ T cells of adult cows. Using polyclonal stimulation with antibodies to CD3 and CD28, we expanded IL-17A-positive CD4+ T cells in a serum-free cell culture medium supplemented with TGF-β1, IL-6 and IL-2. Populations of CD4+ T cells producing IL-17A or IFN-γ or both cytokines were obtained. Isolation of IL-17A-secreting CD4+ T cells was performed by labelling surface IL-17A, followed by flow cytometry cell sorting. The sorted Th17 cells were restimulated and could be expanded for several weeks. These cells were further characterized by cytokine profiling at transcriptomic and protein levels. They produced high amounts of IL-17A and IL-17F, and moderate amounts of IL-22 and IFN-γ. The techniques developed will be useful to characterize the phenotypic and functional properties of bovine Th17 cells.
Collapse
Affiliation(s)
- Patricia Cunha
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France
| | - Yves Le Vern
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France
| | | | - Pierre Germon
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France
| | - Gilles Foucras
- IHAP, INRA, ENVT, Université de Toulouse, Toulouse, France
| | - Pascal Rainard
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France.
| |
Collapse
|
32
|
Abstract
Gamma delta (γδ) T cells constitute a major lymphocyte population in peripheral blood and epithelial surfaces. They play nonredundant roles in host defense against diverse pathogens. Although γδ T cells share functional features with other cells of the immune system, their distinct methods of antigen recognition, rapid response, and tissue tropism make them a unique effector population. This review considers the current state of our knowledge on γδ T cell biology in ruminants and the important roles played by this nonconventional T cell population in protection against several infectious diseases of veterinary and zoonotic importance.
Collapse
|
33
|
DeKuiper JL, Coussens PM. Mycobacterium avium sp. paratuberculosis (MAP) induces IL-17a production in bovine peripheral blood mononuclear cells (PBMCs) and enhances IL-23R expression in-vivo and in-vitro. Vet Immunol Immunopathol 2019; 218:109952. [PMID: 31593889 DOI: 10.1016/j.vetimm.2019.109952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/31/2022]
Abstract
Johne's disease (JD) is a chronic inflammatory gastrointestinal disease of ruminants caused by Mycobacterium avium subspecies paratuberculosis (MAP). Control of JD is difficult largely due to insensitive diagnostic tools, a long subclinical stage of infection, and lack of effective vaccines. Correlates of protection are lacking in model systems of JD and the sources of inflammation due to JD are not well characterized. Commonly studied immune responses, such as the Th1/Th2 paradigm, do not adequately explain host responses to MAP. A potential role for non-classical immune responses to MAP, such as that mediated by Th17 cells, has been suggested. Indeed, MAP antigens induce mRNAs encoding the cytokines IL-23 and IL-17a in bovine peripheral blood mononuclear cells (PBMCs). IL-23 and IL-17a production have both been associated with Th17-like immune responses. Th17 cells are also defined by surface expression of the IL-23 receptor (IL-23R). To determine the relative prevalence of potential Th17 cells in PBMCs from MAP test positive and MAP test negative cows, PBMCs were isolated and analyzed by immunostaining and flow cytometry. Fresh PBMCs from MAP test positive cows (n = 12) contained a significantly higher proportion of IL-23R positive cells in populations of CD4+, CD8+, and Yδ + T cells than in cells from MAP test negative cows (n = 12; p < 0.05). Treatment with MAP antigens increased the percentage of all T cell subsets with surface expression of IL-23R when compared to untreated (n = 12; p < 0.05) cells. ELISA results for IL-17a secretion revealed a higher concentration of IL-17a secreted from PBMCs treated with MAP antigen (n = 20) than from PBMCs not treated with MAP antigens (n = 20) (p < 0.001), regardless of the JD test status of source cows. Also, we observed a moderate negative correlation between JD diagnostic scores for JD + cows and plasma IL-17a concentration (n = 42; r = -0.437; p-value < 0.004). Plasma with low and mid JD- scores (n = 31; n = 9; 0.1 ≤ X < 0.3) had significantly more IL-17a when compared to plasma with high JD- scores (n = 10; 0.3 ≤ X < 0.46; p-values < 0.05). Similarly, plasma with low JD + score values (0.55 ≤ X < 1.0; n = 9) had significantly more IL-17a when compared to plasma with high JD + score values (X ≥ 2.0; n = 21; p < 0.05). Overall, plasma from JD + cows (0.55 < X ≤ 2.86; n = 41) had significantly less IL-17a than plasma from JD- cows (0 < X ≤ 0.46; n = 70). Our data suggests that Th17-like cells may indeed play a role in early immune responses to MAP infection and development or control of JD.
Collapse
Affiliation(s)
- Justin L DeKuiper
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Paul M Coussens
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
34
|
Guan R, Xu W, Yuan L, Wang Y, Cui X, Hu S. Immunomodulatory effect of thymopentin on lymphocytes from supramammary lymph nodes of dairy cows. Immunol Lett 2019; 216:1-8. [PMID: 31520655 DOI: 10.1016/j.imlet.2019.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/29/2019] [Accepted: 09/10/2019] [Indexed: 11/20/2022]
Abstract
Previous study showed that injection of thymopentin (TP 5) in the area of supramammary lymph nodes (SMLN) had therapeutic effect on the intramammary infection (IMI) in cows. This study was to explore the underlying mechanisms by investigating the immunomodulatory effect of TP 5 on SMLN lymphocytes. Lymphocyte proliferation, cell cycle distribution and cytokine mRNA expression were determined by MTT, FCM and RT-qPCR, respectively. Laser scanning confocal microscope (LSCM) was used to observe the binding between TP 5 and SMLN lymphocytes. Moreover, RNA-sequencing (RNA-seq) was performed to observe the difference between the lymphocytes with and without TP 5 treatment. The results showed that TP 5 significantly promoted lymphocyte proliferation, accelerated cell cycle progression, and enhanced mRNA expression of IL-17A and IL-17F. Laser scanning confocal microscopic analysis revealed the binding of TP 5 to the surface of SMLN lymphocytes. A total of 1094 genes were identified as differentially expressed genes (DEGs) using RNA-seq with 692 up- and 402 down-regulated genes. 48 significantly enriched GO terms were identified by RNA-seq. In KEGG analysis, 1/3 of DEGs were enriched in the immune system pathway, including IL-17 signaling pathway, cytokine-cytokine receptor interaction, Th1 and Th2 cell differentiation, T cell receptor signaling pathway, Th17 cell differentiation. Among them, IL-17 signaling pathway was the most prominent. This study suggested that the therapeutic benefit of TP 5 in the treatment of bovine mastitis might be attributed to its immunomodulatory activity in SMLN lymphocytes.
Collapse
Affiliation(s)
- Ran Guan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Wei Xu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Lijia Yuan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Yong Wang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Xuemei Cui
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Songhua Hu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
35
|
Kathamuthu GR, Kumar NP, Moideen K, Sridhar R, Baskaran D, Babu S. Diminished type 1 and type 17 cytokine expressing - Natural killer cell frequencies in tuberculous lymphadenitis. Tuberculosis (Edinb) 2019; 118:101856. [PMID: 31430696 DOI: 10.1016/j.tube.2019.101856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/29/2019] [Accepted: 08/05/2019] [Indexed: 10/26/2022]
Abstract
Tuberculous lymphadenitis (TBL) is associated with the expansion of CD4+ and CD8+ T cells expressing Type 1 and Type 17 cytokines in the peripheral blood. However, the expression pattern of cytokine producing natural killer (NK) cells in both the peripheral blood and affected lymph nodes i.e. site of infection in TBL have not been examined. Hence, we have analyzed the baseline and mycobacterial antigen specific NK cell cytokine frequencies in whole blood of TBL and pulmonary tuberculosis (PTB) individuals. We have also examined the NK cell frequencies before and after treatment completion and in peripheral blood versus affected lymph nodes (LN) of TBL individuals. TBL is characterized by diminished frequencies of NK cells expressing Type 1 (IFNγ, TNFα), Type 17 (IL-17F) cytokines compared to PTB individuals upon antigen-specific stimulation. In contrast, TBL individuals did not exhibit any significant differences in the frequencies of NK cells expressing Type 1 and Type 17 cytokines upon completion of anti-tuberculosis treatment. LN of TBL is associated with altered frequencies of NK cells expressing Type 17 (increased IL-17F and decreased IL-22) cytokines when compared to peripheral blood. Thus, we conclude that TBL individuals are characterized by diminished frequencies of NK cells expressing Type 1/Type 17 cytokines.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India; National Institute for Research in Tuberculosis (NIRT), Chennai, India.
| | - Nathella Pavan Kumar
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | - Kadar Moideen
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | | | - Dhanaraj Baskaran
- National Institute for Research in Tuberculosis (NIRT), Chennai, India
| | - Subash Babu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India; Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
Cheng HY, Ning MX, Chen DK, Ma WT. Interactions Between the Gut Microbiota and the Host Innate Immune Response Against Pathogens. Front Immunol 2019; 10:607. [PMID: 30984184 PMCID: PMC6449424 DOI: 10.3389/fimmu.2019.00607] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/07/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian intestine is colonized by over a trillion microbes that comprise the "gut microbiota," a microbial community which has co-evolved with the host to form a mutually beneficial relationship. Accumulating evidence indicates that the gut microbiota participates in immune system maturation and also plays a central role in host defense against pathogens. Here we review some of the mechanisms employed by the gut microbiota to boost the innate immune response against pathogens present on epithelial mucosal surfaces. Antimicrobial peptide secretion, inflammasome activation and induction of host IL-22, IL-17, and IL-10 production are the most commonly observed strategies employed by the gut microbiota for host anti-pathogen defense. Taken together, the body of evidence suggests that the host gut microbiota can elicit innate immunity against pathogens.
Collapse
Affiliation(s)
- Hong-Yu Cheng
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| | - Meng-Xia Ning
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| | - De-Kun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| | - Wen-Tao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| |
Collapse
|
37
|
Elnaggar MM, Abdellrazeq GS, Dassanayake RP, Fry LM, Hulubei V, Davis WC. Characterization of αβ and γδ T cell subsets expressing IL-17A in ruminants and swine. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 85:115-124. [PMID: 29627456 DOI: 10.1016/j.dci.2018.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 06/08/2023]
Abstract
As part of our ongoing program to expand immunological reagents available for research in cattle, we developed a monoclonal antibody (mAb) to bovine interleukin-17A (IL-17A), a multifunctional cytokine centrally involved in regulating innate and adaptive immune responses. Initial comparative studies demonstrated the mAb recognizes a conserved epitope expressed on orthologues of IL-17A in sheep, goats and pigs. Comparative flow cytometric analyses of lymphocyte subsets stimulated with phorbol 12-myristate 13-acetate (PMA) and ionomycin revealed differences in expression of IL-17A by CD4, CD8, and γδ T cells across ruminants and swine species. Results in cattle showed the largest proportion of IL-17A+ cells were CD4+ followed by γδ and CD8+ T cells. Further analysis revealed the IL-17A+ γδ T cell subset was comprised of WC1.1+, WC1.2+, and WC1- subsets. Analysis of the IL-17A+ CD8+ T cell subset revealed it was comprised of αβ and γδ T cell subsets. Results in sheep and goats revealed IL-17A is expressed mainly by CD4+ and CD8+ T cells, with little expression by γδ T cells. Analysis of IL-17A+ CD8+ T cells showed the majority were CD8+ αβ in sheep, whereas they were CD8+ γδ in goats. The majority of the sheep and goat IL-17A+ γδ T cells were WC1+. Results obtained in swine showed expression of IL-17A by CD4, CD8, and γδ T cell subsets were similar to results reported in other studies. Comparison of expression of IL-17A with IFN-γ revealed subsets co-expressed IL-17A and IFN-γ in cattle, sheep, and goats. The new mAb expands opportunities for immunology research in ruminants and swine.
Collapse
Affiliation(s)
- Mahmoud M Elnaggar
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, WA, USA; Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt.
| | - Gaber S Abdellrazeq
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, WA, USA; Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt
| | - Rohana P Dassanayake
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA, USA
| | - Lindsay M Fry
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, WA, USA; Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, USA
| | - Victoria Hulubei
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, WA, USA
| | - William C Davis
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, WA, USA
| |
Collapse
|
38
|
Albarrak S, Waters W, Stabel J, Hostetter J. Evaluating the cytokine profile of the WC1+ γδ T cell subset in the ileum of cattle with the subclinical and clinical forms of MAP infection. Vet Immunol Immunopathol 2018; 201:26-31. [DOI: 10.1016/j.vetimm.2018.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 11/25/2022]
|
39
|
S. Rosenthal K. Immune monitoring of the body’s borders. AIMS ALLERGY AND IMMUNOLOGY 2018. [DOI: 10.3934/allergy.2018.3.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
40
|
Rusk RA, Palmer MV, Waters WR, McGill JL. Measuring bovine γδ T cell function at the site of Mycobacterium bovis infection. Vet Immunol Immunopathol 2017; 193-194:38-49. [PMID: 29129226 DOI: 10.1016/j.vetimm.2017.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/06/2017] [Accepted: 10/25/2017] [Indexed: 12/28/2022]
Abstract
Bovine γδ T cells are amongst the first cells to accumulate at the site of Mycobacterium bovis infection; however, their role in the developing lesion remains unclear. We utilized transcriptomics analysis, in situ hybridization, and a macrophage/γδ T cell co-culture system to elucidate the role of γδ T cells in local immunity to M. bovis infection. Transcriptomics analysis revealed that γδ T cells upregulated expression of several novel, immune-associated genes in response to stimulation with M. bovis antigen. BCG-infected macrophage/γδ T cell co-cultures confirmed the results of our RNAseq analysis, and revealed that γδ T cells from M. bovis-infected animals had a significant impact on bacterial viability. Analysis of γδ T cells within late-stage M. bovis granulomas revealed significant expression of IFN-γ and CCL2, but not IL-10, IL-22, or IL-17. Our results suggest γδ T cells influence local immunity to M. bovis through cytokine secretion and direct effects on bacterial burden.
Collapse
Affiliation(s)
- Rachel A Rusk
- Pathobiology Graduate Program, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
| | - Mitchell V Palmer
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, USDA, Ames, IA, USA
| | - W Ray Waters
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, USDA, Ames, IA, USA
| | - Jodi L McGill
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
41
|
Abstract
The interleukin-17 (IL-17) family cytokines, such as IL-17A and IL-17F, play
important protective roles in host immune response to a variety of infections
such as bacterial, fungal, parasitic, and viral. The IL-17R signaling and
downstream pathways mediate induction of proinflammatory molecules which
participate in control of these pathogens. However, the production of IL-17 can
also mediate pathology and inflammation associated with infections. In this
review, we will discuss the yin-and-yang roles of IL-17 in host immunity to
pathogens.
Collapse
Affiliation(s)
- Shibali Das
- Department of Molecular Microbiology, Washington University in St. Louis, St Louis, MO, USA
| | - Shabaana Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St Louis, MO, USA
| |
Collapse
|
42
|
Segueni N, Tritto E, Bourigault ML, Rose S, Erard F, Le Bert M, Jacobs M, Di Padova F, Stiehl DP, Moulin P, Brees D, Chibout SD, Ryffel B, Kammüller M, Quesniaux VF. Controlled Mycobacterium tuberculosis infection in mice under treatment with anti-IL-17A or IL-17F antibodies, in contrast to TNFα neutralization. Sci Rep 2016; 6:36923. [PMID: 27853279 PMCID: PMC5113257 DOI: 10.1038/srep36923] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022] Open
Abstract
Antibodies targeting IL-17A or its receptor IL-17RA show unprecedented efficacy in the treatment of autoimmune diseases such as psoriasis. These therapies, by neutralizing critical mediators of immunity, may increase susceptibility to infections. Here, we compared the effect of antibodies neutralizing IL-17A, IL-17F or TNFα on murine host responses to Mycobacterium tuberculosis infection by evaluating lung transcriptomic, microbiological and histological analyses. Coinciding with a significant increase of mycobacterial burden and pathological changes following TNFα blockade, gene array analyses of infected lungs revealed major changes of inflammatory and immune gene expression signatures 4 weeks post-infection. Specifically, gene expression associated with host-pathogen interactions, macrophage recruitment, activation and polarization, host-antimycobacterial activities, immunomodulatory responses, as well as extracellular matrix metallopeptidases, were markedly modulated by TNFα blockade. IL-17A or IL-17F neutralization elicited only mild changes of few genes without impaired host resistance four weeks after M. tuberculosis infection. Further, the absence of both IL-17RA and IL-22 pathways in genetically deficient mice did not profoundly compromise host control of M. tuberculosis over a 6-months period, ruling out potential compensation between these two pathways, while TNFα-deficient mice succumbed rapidly. These data provide experimental confirmation of the low clinical risk of mycobacterial infection under anti-IL-17A therapy, in contrast to anti-TNFα treatment.
Collapse
Affiliation(s)
- Noria Segueni
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Elaine Tritto
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Marie-Laure Bourigault
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Stéphanie Rose
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - François Erard
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Marc Le Bert
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Muazzam Jacobs
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Franco Di Padova
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Daniel P. Stiehl
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Pierre Moulin
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Dominique Brees
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Salah-Dine Chibout
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Bernhard Ryffel
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, South Africa
| | - Michael Kammüller
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Valerie F. Quesniaux
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| |
Collapse
|