1
|
Niu Q, Wang M, Liu XS. The evolving landscape of IL-10, IL-22 and IL-26 in pleurisy especially in tuberculous pleurisy. Respir Res 2024; 25:275. [PMID: 39003443 PMCID: PMC11245850 DOI: 10.1186/s12931-024-02896-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/29/2024] [Indexed: 07/15/2024] Open
Abstract
Pleurisy can be categorized as primary or secondary, arising from immunological, tumorous, or microbial conditions. It often results in lung structure damage and the development of various respiratory issues. Among the different types, tuberculous pleurisy has emerged as a prominent focus for both clinical and scientific investigations. The IL-10 family, known for its anti-inflammatory properties in the human immune system, is increasingly being studied for its involvement in the pathogenesis of pleurisy. This review aims to present a detailed overview of the intricate role of IL-10 family members (specifically IL-10, IL-22, and IL-26) in human and animal pleuritic diseases or relevant animal models. These insights could serve as valuable guidance and references for further studies on pleurisy and potential therapeutic strategies.
Collapse
Affiliation(s)
- Qian Niu
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathology, Baoji Gaoxin Hospital, Baoji, 721000, China
| | - Xian-Sheng Liu
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Martinez-Martinez YB, Huante MB, Chauhan S, Naqvi KF, Bharaj P, Endsley JJ. Helper T cell bias following tuberculosis chemotherapy identifies opportunities for therapeutic vaccination to prevent relapse. NPJ Vaccines 2023; 8:165. [PMID: 37898618 PMCID: PMC10613213 DOI: 10.1038/s41541-023-00761-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023] Open
Abstract
Therapeutic vaccines have promise as adjunctive treatment for tuberculosis (TB) or as preventives against TB relapse. An important development challenge is the limited understanding of T helper (Th) cell roles during these stages of disease. A murine model of TB relapse was used to identify changes in Th populations and cytokine microenvironment. Active TB promoted expansion of Th1, Th2, Th17, and Th22 cells and cytokines in the lung. Following drug therapy, pulmonary Th17 and Th22 cells contracted, Th1 cells remained elevated, while Th cells producing IL-4 or IL-10 expanded. At relapse, Th22 cells failed to re-expand in the lung despite a moderate re-expansion of Th1 and Th17 cells and an increase in Th cytokine polyfunctionality. The dynamics of Th populations further differed by tissue compartment and disease presentation. These outcomes identify immune bias by Th subpopulations during TB relapse as candidate mechanisms for pathogenesis and targets for therapeutic vaccination.
Collapse
Affiliation(s)
- Yazmin B Martinez-Martinez
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Matthew B Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sadhana Chauhan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kubra F Naqvi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Preeti Bharaj
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
3
|
Zhang K, Chen L, Zhu C, Zhang M, Liang C. Current Knowledge of Th22 Cell and IL-22 Functions in Infectious Diseases. Pathogens 2023; 12:pathogens12020176. [PMID: 36839448 PMCID: PMC9965464 DOI: 10.3390/pathogens12020176] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
T helper 22 (Th22) cells, a newly defined CD4+ T-cell lineage, are characterized by their distinct cytokine profile, which primarily consists of IL-13, IL-22 and TNF-α. Th22 cells express a wide spectrum of chemokine receptors, such as CCR4, CCR6 and CCR10. The main effector molecule secreted by Th22 cells is IL-22, a member of the IL-10 family, which acts by binding to IL-22R and triggering a complex downstream signaling system. Th22 cells and IL-22 have been found to play variable roles in human immunity. In preventing the progression of infections such as HIV and influenza, Th22/IL-22 exhibited protective anti-inflammatory characteristics, and their deleterious proinflammatory activities have been demonstrated to exacerbate other illnesses, including hepatitis B and Helicobacter pylori infection. Herein, we review the current understanding of Th22 cells, including their definition, differentiation and mechanisms, and the effect of Th22/IL-22 on human infectious diseases. According to studies on Th22 cells, Th22/IL-22 may be a promising therapeutic target and an effective treatment strategy for various infections.
Collapse
Affiliation(s)
- Kunyu Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- The Second Clinical Medical College, Anhui Medical University, Hefei 230032, China
| | - Lei Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
| | - Chenyu Zhu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- The Second Clinical Medical College, Anhui Medical University, Hefei 230032, China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- Correspondence: (M.Z.); (C.L.); Tel./Fax: +86-55162922034 (M.Z.); +86-55162922034 (C.L.)
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- Correspondence: (M.Z.); (C.L.); Tel./Fax: +86-55162922034 (M.Z.); +86-55162922034 (C.L.)
| |
Collapse
|
4
|
Ritter K, Behrends J, Rückerl D, Hölscher A, Volz J, Prinz I, Hölscher C. High-Dose Mycobacterium tuberculosis H37rv Infection in IL-17A- and IL-17A/F-Deficient Mice. Cells 2022; 11:cells11182875. [PMID: 36139450 PMCID: PMC9496946 DOI: 10.3390/cells11182875] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/02/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
During experimental tuberculosis (TB), interleukin (IL)-17A appears to be involved in the formation of lung granulomas, possibly through the attraction of neutrophils to the sites of infection. However, the protective impact of cytokine appears to depend on the degree of its induction. Hence, robust production of IL-17A in mice infected with the hypervirulent isolate Mycobacterium tuberculosis (Mtb) HN878 mediates protection, while the cytokine is dispensable for protective immune responses against low-dose infection with the less virulent strain H37rv. Here, we show that after experimental infection with high doses of Mtb H37rv, IL-17A-deficient (−/−) mice exhibited high susceptibility to the infection, which was mediated by the strong accumulation of neutrophils in the infected lung tissue. Accordingly, we observed nearly unrestricted bacterial replication within the neutrophils, indicating that they may serve as a survival niche for Mtb. By use of IL-17A/IL-17F-double-deficient mice, we demonstrated that the susceptibility in the absence of IL-17A is mediated by a compensatory expression of IL-17F, which, however, appeared not to be dependent on neutrophils. Together, our results illustrate the compensatory potential of the Th17-secreted cytokines IL-17A and IL-17F in the context of experimental TB and once again emphasize the detrimental effect of excessive neutrophil infiltration in response to Mtb.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Center Borstel, D-23845 Borstel, Germany
| | - Jochen Behrends
- Fluorescence Cytometry Core Unit, Research Center Borstel, D-23845 Borstel, Germany
| | - Dominik Rückerl
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
| | | | - Johanna Volz
- Infection Immunology, Research Center Borstel, D-23845 Borstel, Germany
| | - Immo Prinz
- Center for Molecular Neurobiology Hamburg, Eppendorf University Medical Center, D-20246 Hamburg, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Center Borstel, D-23845 Borstel, Germany
- Thematic Translational Unit Tuberculosis, German Center for Infection Research (DZIF), D-38124 Braunschweig, Germany
- Correspondence:
| |
Collapse
|
5
|
Makatsa MS, Omondi FMA, Bunjun R, Wilkinson RJ, Riou C, Burgers WA. Characterization of Mycobacterium tuberculosis-Specific Th22 Cells and the Effect of Tuberculosis Disease and HIV Coinfection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:446-455. [PMID: 35777848 PMCID: PMC9339498 DOI: 10.4049/jimmunol.2200140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/26/2022] [Indexed: 02/03/2023]
Abstract
The development of a highly effective tuberculosis (TB) vaccine is likely dependent on our understanding of what constitutes a protective immune response to TB. Accumulating evidence suggests that CD4+ T cells producing IL-22, a distinct subset termed "Th22" cells, may contribute to protective immunity to TB. Thus, we characterized Mycobacterium tuberculosis-specific Th22 (and Th1 and Th17) cells in 72 people with latent TB infection or TB disease, with and without HIV-1 infection. We investigated the functional properties (IFN-γ, IL-22, and IL-17 production), memory differentiation (CD45RA, CD27, and CCR7), and activation profile (HLA-DR) of M. tuberculosis-specific CD4+ T cells. In HIV-uninfected individuals with latent TB infection, we detected abundant circulating IFN-γ-producing CD4+ T cells (median, 0.93%) and IL-22-producing CD4+ T cells (median, 0.46%) in response to M. tuberculosis The frequency of IL-17-producing CD4+ T cells was much lower, at a median of 0.06%. Consistent with previous studies, IL-22 was produced by a distinct subset of CD4+ T cells and not coexpressed with IL-17. M. tuberculosis-specific IL-22 responses were markedly reduced (median, 0.08%) in individuals with TB disease and HIV coinfection compared with IFN-γ responses. M. tuberculosis-specific Th22 cells exhibited a distinct memory and activation phenotype compared with Th1 and Th17 cells. Furthermore, M. tuberculosis-specific IL-22 was produced by conventional CD4+ T cells that required TCR engagement. In conclusion, we confirm that Th22 cells are a component of the human immune response to TB. Depletion of M. tuberculosis-specific Th22 cells during HIV coinfection may contribute to increased risk of TB disease.
Collapse
Affiliation(s)
- Mohau S Makatsa
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - F Millicent A Omondi
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rubina Bunjun
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Imperial College London, London, U.K.; and
- Francis Crick Institute Mill Hill laboratory, London, U.K
| | - Catherine Riou
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Wendy A Burgers
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa;
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
6
|
Brandenburg J, Heyckendorf J, Marwitz F, Zehethofer N, Linnemann L, Gisch N, Karaköse H, Reimann M, Kranzer K, Kalsdorf B, Sanchez-Carballo P, Weinkauf M, Scholz V, Malm S, Homolka S, Gaede KI, Herzmann C, Schaible UE, Hölscher C, Reiling N, Schwudke D. Tuberculostearic Acid-Containing Phosphatidylinositols as Markers of Bacterial Burden in Tuberculosis. ACS Infect Dis 2022; 8:1303-1315. [PMID: 35763439 PMCID: PMC9274766 DOI: 10.1021/acsinfecdis.2c00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
One-fourth of the
global human population is estimated to be infected
with strains of the Mycobacterium tuberculosis complex (MTBC), the causative agent of tuberculosis (TB). Using
lipidomic approaches, we show that tuberculostearic acid (TSA)-containing
phosphatidylinositols (PIs) are molecular markers for infection with
clinically relevant MTBC strains and signify bacterial burden. For
the most abundant lipid marker, detection limits of ∼102 colony forming units (CFUs) and ∼103 CFUs
for bacterial and cell culture systems were determined, respectively.
We developed a targeted lipid assay, which can be performed within
a day including sample preparation—roughly 30-fold faster than
in conventional methods based on bacterial culture. This indirect
and culture-free detection approach allowed us to determine pathogen
loads in infected murine macrophages, human neutrophils, and murine
lung tissue. These marker lipids inferred from mycobacterial PIs were
found in higher levels in peripheral blood mononuclear cells of TB
patients compared to healthy individuals. Moreover, in a small cohort
of drug-susceptible TB patients, elevated levels of these molecular
markers were detected at the start of therapy and declined upon successful
anti-TB treatment. Thus, the concentration of TSA-containing PIs can
be used as a correlate for the mycobacterial burden in experimental
models and in vitro systems and may prospectively also provide a clinically
relevant tool to monitor TB severity.
Collapse
Affiliation(s)
- Julius Brandenburg
- Division of Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Jan Heyckendorf
- Division of Clinical Infectious Disease, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Clinical Tuberculosis Center, 23845 Borstel, Germany
| | - Franziska Marwitz
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, 23845 Borstel, Germany
| | - Nicole Zehethofer
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, 23845 Borstel, Germany
| | - Lara Linnemann
- Division of Cellular Microbiology, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Hande Karaköse
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, 23845 Borstel, Germany
| | - Maja Reimann
- Division of Clinical Infectious Disease, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Clinical Tuberculosis Center, 23845 Borstel, Germany
| | - Katharina Kranzer
- National Reference Center for Mycobacteria, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Barbara Kalsdorf
- Division of Clinical Infectious Disease, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Clinical Tuberculosis Center, 23845 Borstel, Germany
| | - Patricia Sanchez-Carballo
- Division of Clinical Infectious Disease, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Clinical Tuberculosis Center, 23845 Borstel, Germany
| | - Michael Weinkauf
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Verena Scholz
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Sven Malm
- Division of Molecular and Experimental Mycobacteriology, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Susanne Homolka
- Division of Molecular and Experimental Mycobacteriology, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Karoline I Gaede
- BioMaterialBank Nord, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Lung Research (DZL), Airway Research Center North (ARCN), Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Christian Herzmann
- Center for Clinical Studies, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Ulrich E Schaible
- German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, 23845 Borstel, Germany.,Division of Cellular Microbiology, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Christoph Hölscher
- German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, 23845 Borstel, Germany.,Division of Infection Immunology, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Norbert Reiling
- Division of Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, 23845 Borstel, Germany
| | - Dominik Schwudke
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany.,German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, 23845 Borstel, Germany.,German Center for Lung Research (DZL), Airway Research Center North (ARCN), Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| |
Collapse
|
7
|
Bunjun R, Omondi FMA, Makatsa MS, Keeton R, Wendoh JM, Müller TL, Prentice CSL, Wilkinson RJ, Riou C, Burgers WA. Th22 Cells Are a Major Contributor to the Mycobacterial CD4 + T Cell Response and Are Depleted During HIV Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1239-1249. [PMID: 34389623 PMCID: PMC8387408 DOI: 10.4049/jimmunol.1900984] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 07/03/2021] [Indexed: 12/13/2022]
Abstract
HIV-1 infection substantially increases the risk of developing tuberculosis (TB). Mechanisms such as defects in the Th1 response to Mycobacterium tuberculosis in HIV-infected persons have been widely reported. However, Th1-independent mechanisms also contribute to protection against TB. To identify a broader spectrum of defects in TB immunity during HIV infection, we examined IL-17A and IL-22 production in response to mycobacterial Ags in peripheral blood of persons with latent TB infection and HIV coinfection. Upon stimulating with mycobacterial Ags, we observed a distinct CD4+ Th lineage producing IL-22 in the absence of IL-17A and IFN-γ. Mycobacteria-specific Th22 cells were present at high frequencies in blood and contributed up to 50% to the CD4+ T cell response to mycobacteria, comparable in magnitude to the IFN-γ Th1 response (median 0.91% and 0.55%, respectively). Phenotypic characterization of Th22 cells revealed that their memory differentiation was similar to M. tuberculosis-specific Th1 cells (i.e., predominantly early differentiated CD45RO+CD27+ phenotype). Moreover, CCR6 and CXCR3 expression profiles of Th22 cells were similar to Th17 cells, whereas their CCR4 and CCR10 expression patterns displayed an intermediate phenotype between Th1 and Th17 cells. Strikingly, mycobacterial IL-22 responses were 3-fold lower in HIV-infected persons compared with uninfected persons, and the magnitude of responses correlated inversely with HIV viral load. These data provide important insights into mycobacteria-specific Th subsets in humans and suggest a potential role for IL-22 in protection against TB during HIV infection. Further studies are needed to fully elucidate the role of IL-22 in protective TB immunity.
Collapse
Affiliation(s)
- Rubina Bunjun
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Fidilia M A Omondi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mohau S Makatsa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Roanne Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Jerome M Wendoh
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Tracey L Müller
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Caryn S L Prentice
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Robert J Wilkinson
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Imperial College London, London, United Kingdom; and
- The Francis Crick Institute, London, United Kingdom
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa;
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
8
|
Ritter K, Sodenkamp JC, Hölscher A, Behrends J, Hölscher C. IL-6 is not Absolutely Essential for the Development of a TH17 Immune Response after an Aerosol Infection with Mycobacterium Tuberculosis H37rv. Cells 2020; 10:cells10010009. [PMID: 33375150 PMCID: PMC7822128 DOI: 10.3390/cells10010009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022] Open
Abstract
Anti-inflammatory treatment of chronic inflammatory diseases often increases susceptibility to infectious diseases such as tuberculosis (TB). Since numerous chronic inflammatory and autoimmune diseases are mediated by interleukin (IL)-6-induced T helper (TH) 17 cells, a TH17-directed anti-inflammatory therapy may be preferable to an IL-12-dependent TH1 inhibition in order to avoid reactivation of latent infections. To assess, however, the risk of inhibition of IL-6-dependent TH17-mediated inflammation, we examined the TH17 immune response and the course of experimental TB in IL-6- and T-cell-specific gp130-deficient mice. Our study revealed that the absence of IL-6 or gp130 on T cells has only a minor effect on the development of antigen-specific TH1 and TH17 cells. Importantly, these gene-deficient mice were as capable as wild type mice to control mycobacterial infection. Together, in contrast to its key function for TH17 development in other inflammatory diseases, IL-6 plays an inferior role for the generation of TH17 immune responses during experimental TB.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.C.S.); (A.H.)
| | - Jan Christian Sodenkamp
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.C.S.); (A.H.)
| | - Alexandra Hölscher
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.C.S.); (A.H.)
| | - Jochen Behrends
- Core Facility Fluorescence Cytometry, Research Centre Borstel, D-23845 Borstel, Germany;
| | - Christoph Hölscher
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.C.S.); (A.H.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, D-23845 Borstel, Germany
- Correspondence:
| |
Collapse
|
9
|
Schuurbiers MMF, Bruno M, Zweijpfenning SMH, Magis-Escurra C, Boeree M, Netea MG, van Ingen J, van de Veerdonk F, Hoefsloot W. Immune defects in patients with pulmonary Mycobacterium abscessus disease without cystic fibrosis. ERJ Open Res 2020; 6:00590-2020. [PMID: 33263065 PMCID: PMC7682720 DOI: 10.1183/23120541.00590-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of Mycobacterium abscessus infections in non-cystic fibrosis (CF) patients has increased in recent years. In this study, we investigate whether immune defects explain the apparent susceptibility to this opportunistic infection in non-CF patients. We performed stimulations of peripheral blood mononuclear cells and whole blood from 13 patients with M. abscessus pulmonary disease and 13 healthy controls to investigate their cytokine production after 24 h and 7 days. Patients were predominantly women (54%) with a mean age of 59 years; 62% had nodular bronchiectatic disease. Many patients had predisposing pulmonary diseases, such as COPD (46%), and asthma (23%). Patients with COPD showed an impaired interleukin (IL)-6 response to M. abscessus and a reduced IL-17 response to Candida, together with a M. abscessus-specific enhanced IL-22 production. Patients without COPD showed higher levels of interleukin-1 receptor antagonist (IL-1Ra), an anti-inflammatory molecule. Within the non-COPD patients, those with bronchiectasis showed defective interferon (IFN)-γ production in response to Candida albicans. In conclusion, susceptibility to M. abscessus is likely determined by a combination of immunological defects and predisposing pulmonary disease. The main defect in the innate immune response was a shift of the ratio of IL-1β to IL-1Ra, which decreased the bioactivity of this pathway in the adaptive immune response. In the adaptive immune response there was defective IL-17 and IFN-γ production. Patients with COPD and bronchiectasis showed different cytokine defects. It is therefore crucial to interpret the immunological results within the clinical background of the patients tested. Measurement of defects in both the innate and adaptive immune responses in patients with M. abscessus pulmonary disease show that susceptibility to M. abscessus is determined by a combination of immunological defects and predisposing pulmonary diseasehttps://bit.ly/2DtbycY
Collapse
Affiliation(s)
- Milou M F Schuurbiers
- Radboud University Medical Centre, University Centre of Chronic Diseases Dekkerswald, Dept of Pulmonary Diseases, Nijmegen, The Netherlands.,These authors contributed equally
| | - Mariolina Bruno
- Dept of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,These authors contributed equally
| | - Sanne M H Zweijpfenning
- Radboud University Medical Centre, University Centre of Chronic Diseases Dekkerswald, Dept of Pulmonary Diseases, Nijmegen, The Netherlands.,These authors contributed equally
| | - Cecile Magis-Escurra
- Radboud University Medical Centre, University Centre of Chronic Diseases Dekkerswald, Dept of Pulmonary Diseases, Nijmegen, The Netherlands
| | - Martin Boeree
- Radboud University Medical Centre, University Centre of Chronic Diseases Dekkerswald, Dept of Pulmonary Diseases, Nijmegen, The Netherlands
| | - Mihai G Netea
- Dept of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Dept for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Jakko van Ingen
- Radboud University Medical Centre, Dept of Medical Microbiology, Nijmegen, The Netherlands
| | - Frank van de Veerdonk
- Dept of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wouter Hoefsloot
- Radboud University Medical Centre, University Centre of Chronic Diseases Dekkerswald, Dept of Pulmonary Diseases, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Personalized Approach as a Basis for the Future Diagnosis of Tuberculosis (Literature Review). ACTA BIOMEDICA SCIENTIFICA 2019. [DOI: 10.29413/abs.2019-4.3.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The global spread of tuberculosis remains one of actual problems of public health despite of introduction of public health safety programs. Early, rapid and accurate identification of M. tuberculosis and determination of drug susceptibility are essential for treatment and management of this disease. Delay in delivering results prolongs potentially inappropriate antituberculosis therapy, contributing to emergence of drug resistance, reducing treatment options and increasing treatment duration and associated costs, resulting in increased mortality and morbidity. Faster, more comprehensive diagnostics will enable earlier use of the most appropriate drug regimen, thus improving patient outcomes and reducing overall healthcare costs. The treatment of infection based on the using of massive antimicrobial therapy with analysis of bacterial strains resistance to first line drugs (FLD) isoniazid (INH), rifampin (RIF), pyrazinamide (PZA), ethambutol (EMB) and streptomycin (SM). However, the public health practitioners pay no attention to functional activity of human immune system genes. The interaction of bacterial genomes and immune system genes plays the major role in infection progress. There is growing evidence that, together with human and environmental factors, Mycobacterium tuberculosis complex strain diversity contributes to the variable outcome of infection and disease in human TB. We suppose that the future of diagnosis and treatment of tuberculosis lies in the field of personal medicine with comprehensive analysis of host and pathogen genes.
Collapse
|
11
|
Masonou T, Hokey DA, Lahey T, Halliday A, Berrocal-Almanza LC, Wieland-Alter WF, Arbeit RD, Lalvani A, von Reyn CF. CD4+ T cell cytokine responses to the DAR-901 booster vaccine in BCG-primed adults: A randomized, placebo-controlled trial. PLoS One 2019; 14:e0217091. [PMID: 31120957 PMCID: PMC6532882 DOI: 10.1371/journal.pone.0217091] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/30/2019] [Indexed: 11/18/2022] Open
Abstract
Background DAR-901 is an inactivated whole cell tuberculosis booster vaccine, prepared using a new scalable, broth-grown method from the master cell bank of SRL172, a vaccine previously shown to prevent tuberculosis. This study examined whether DAR-901 (a) induces CD4+ T cell cytokine profiles previously proposed as correlates of protection and (b) has a specific vaccine-induced immunological signature compared to BCG or placebo. Methods We analysed CD4+ T cell cytokine immune responses from 10 DAR-901 recipients, 9 BCG recipients and 9 placebo recipients from the Phase I DAR-901 MDES trial. In that study, HIV-negative, IGRA-negative participants with prior BCG immunization were randomized (double-blind) to receive three intradermal injections of DAR-901 or saline placebo or two injections of saline placebo followed by an intradermal injection of BCG. Antigen-specific functional and phenotypic CD4+ T cell responses along with effector phenotype of responder cells were measured by intracellular cytokine staining. Results DAR-901 recipients exhibited increased DAR-901 antigen-specific polyfunctional or bifunctional T cell responses compared to baseline. Vaccine specific CD4+ IFNγ, IL2, TNFα and any cytokine responses peaked at 7 days post-dose 3. Th1 responses predominated, with most responder cells exhibiting a polyfunctional effector memory phenotype. BCG induced greater CD4+ T cell responses than placebo while the more modest DAR-901 responses did not differ from placebo. Neither DAR-901 nor BCG induced substantial or sustained Th17 /Th22 cytokine responses. Conclusion DAR-901, a TB booster vaccine grown from the master cell bank of SRL 172 which was shown to prevent TB, induced low magnitude polyfunctional effector memory CD4+ T cell responses. DAR-901 responses were lower than those induced by BCG, a vaccine that has been shown ineffective as a booster to prevent tuberculosis disease. These results suggest that induction of higher levels of CD4+ cytokine stimulation may not be a critical or pre-requisite characteristic for candidate TB vaccine boosters. Trial registration ClinicalTrials.gov NCT02063555.
Collapse
Affiliation(s)
- Tereza Masonou
- Tuberculosis Research Centre, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- * E-mail: (TM); (AL); (CFvR)
| | | | - Timothy Lahey
- Larner College of Medicine, University of Vermont, Burlington, VT, United States of America
| | - Alice Halliday
- Tuberculosis Research Centre, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Luis C. Berrocal-Almanza
- Tuberculosis Research Centre, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Robert D. Arbeit
- Tufts University School of Medicine, Boston, MA, United States of America
| | - Ajit Lalvani
- Tuberculosis Research Centre, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- * E-mail: (TM); (AL); (CFvR)
| | - C. Fordham von Reyn
- Geisel School of Medicine, Hanover, NH, United States of America
- * E-mail: (TM); (AL); (CFvR)
| |
Collapse
|
12
|
He J, Zhang R, Shen Y, Wan C, Zeng N, Qin J, Tian P, Chen L. Diagnostic accuracy of interleukin-22 and adenosine deaminase for tuberculous pleural effusions. Curr Res Transl Med 2018; 66:103-106. [PMID: 30217555 DOI: 10.1016/j.retram.2018.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Reliable markers for accurately diagnosing tuberculous pleural effusions (TPE) are needed. This study sought to investigate the diagnostic potential of pleural interleukin-22 (IL-22) and compare it with the performance of adenosine deaminase (ADA). METHOD This prospective study involved 49 patients with TPE and 60 patients with pleural effusion of other causes. Pleural levels of IL-22 and ADA were determined, respectively, using ELISA or an enzymatic method. A receiver operating characteristic curve was constructed and the area under the curve (AUC) was calculated to summarize the diagnostic accuracy of single markers or marker combinations. RESULTS Levels of IL-22 in pleural effusion were significantly higher in TPE patients than in other patients (322.36 ± 406.65 vs. 83.13 ± 22.15 pg/ml, P < 0.05). With a cut-off value of 97.82 pg/ml, the diagnostic sensitivity of IL-22 for TPE was 71.42%, specificity was 81.67%, and the area under the curve (AUC) was 0.83. ADA levels were also increased in TPE, and its AUC for diagnosing TPE was 0.90. The combination of IL-22 and ADA enhanced diagnostic accuracy, offering sensitivity of 83.67%, specificity of 91.67%, and an AUC of 0.93. CONCLUSION IL-22 may be useful for diagnosing TPE, and combining it with ADA may further enhance diagnostic accuracy. Our results justify more rigorous studies with larger samples to confirm the diagnostic potential of IL-22 for TPE.
Collapse
Affiliation(s)
- Junyun He
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Rui Zhang
- Department of Medical Informatics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yongchun Shen
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China; Department of Medical Informatics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Chun Wan
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ni Zeng
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jiangyue Qin
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Panwen Tian
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Lei Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
13
|
Ronacher K, Sinha R, Cestari M. IL-22: An Underestimated Player in Natural Resistance to Tuberculosis? Front Immunol 2018; 9:2209. [PMID: 30319650 PMCID: PMC6167461 DOI: 10.3389/fimmu.2018.02209] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/06/2018] [Indexed: 12/22/2022] Open
Abstract
Approximately 10% of individuals latently infected with Mycobacterium tuberculosis (Mtb) develop active tuberculosis (TB) during their lifetime. Although it is well recognized that T-helper 1 immune responses are crucial for containing latent TB infection, the full array of host factors conferring protective immunity from TB progression are not completely understood. IL-22 is produced by cells of the innate and adaptive immune system including innate lymphoid cells, and natural killer cells as well as T lymphocytes (Th1, Th17, and Th22) and binds to its cognate receptor, the IL-22R1, which is expressed on non-hematopoietic cells such as lung epithelial cells. However, recent studies suggest that Mtb induces expression of the IL-22R1 on infected macrophages and multiple studies have indicated a protective role of IL-22 in respiratory tract infections. Reduced concentrations of circulating IL-22 in active TB compared to latent TB and decreased percentages of Mtb-specific IL-22 producing T cells in TB patients compared to controls designate this cytokine as a key player in TB immunology. More recently, it has been shown that in type 2 diabetes (T2D) and TB co-morbidity serum IL-22 concentrations are further reduced compared to TB patients without co-morbidities. However, whether a causative link between low IL-22 and increased susceptibility to TB and disease severity of TB exists remains to be established. This review summarizes the contribution of IL-22, a potentially under-appreciated key player in natural resistance to TB, at the interface between the immune response to Mtb and the lung epithelium.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Disease Resistance/immunology
- Epithelial Cells/immunology
- Epithelial Cells/microbiology
- Humans
- Interleukins/immunology
- Interleukins/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Latent Tuberculosis/blood
- Latent Tuberculosis/immunology
- Latent Tuberculosis/microbiology
- Lung/cytology
- Lung/immunology
- Lung/microbiology
- Lymphocyte Activation/immunology
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/microbiology
- Mycobacterium tuberculosis/immunology
- Receptors, Interleukin/immunology
- Receptors, Interleukin/metabolism
- Respiratory Mucosa/cytology
- Respiratory Mucosa/immunology
- Respiratory Mucosa/microbiology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Tuberculosis, Pulmonary/blood
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/microbiology
- Interleukin-22
Collapse
Affiliation(s)
- Katharina Ronacher
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, SAMRC Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- Infection, Immunity and Metabolism Group, Translational Research Institute, Mater Research Institute and The University of Queensland, Brisbane, QLD, Australia
| | - Roma Sinha
- Infection, Immunity and Metabolism Group, Translational Research Institute, Mater Research Institute and The University of Queensland, Brisbane, QLD, Australia
| | - Michelle Cestari
- Infection, Immunity and Metabolism Group, Translational Research Institute, Mater Research Institute and The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
14
|
Ahmed A, Adiga V, Nayak S, Uday Kumar JAJ, Dhar C, Sahoo PN, Sundararaj BK, Souza GD, Vyakarnam A. Circulating HLA-DR+CD4+ effector memory T cells resistant to CCR5 and PD-L1 mediated suppression compromise regulatory T cell function in tuberculosis. PLoS Pathog 2018; 14:e1007289. [PMID: 30231065 PMCID: PMC6166982 DOI: 10.1371/journal.ppat.1007289] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 10/01/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Chronic T cell activation is a hallmark of pulmonary tuberculosis (PTB). The mechanisms underpinning this important phenomenon are however, poorly elucidated, though known to rely on control of T effector cells (Teff) by regulatory T cells (Treg). Our studies show that circulating natural Treg cells in adults with PTB preserve their suppressive potential but Teff cells from such subjects are resistant to Treg-mediated suppression. We found this to be due to expansion of an activated Teff subset identified by Human Leukocyte Antigen (HLA)-DR expression. Sensitivity to suppression was restored to control levels by depletion of this subset. Comparative transcriptome analysis of Teff cells that contain HLA-DR+ cells versus the fraction depleted of this population identified putative resistance mechanisms linked to IFNG, IL17A, IL22, PD-L1 and β-chemokines CCL3L3, CCL4 expression. Antibody blocking experiments confirmed HLA-DR+ Teff cells, but not the fraction depleted of HLA-DR+ effectors, to be resistant to Treg suppression mediated via CCR5 and PD-L1 associated pathways. In the presence of HLA-DR+ Teff cells, activation of NFκB downstream of CCR5 and PD-L1 was perturbed. In addition, HLA-DR+ Teff cells expressed significantly higher levels of Th1/Th17 cytokines that may regulate Treg function through a reciprocal counter-balancing relationship. Taken together, our study provides novel insight on how activated HLA-DR+CD4+ T cells may contribute to disease associated inflammation by compromising Treg-mediated suppression in PTB. An important marker of progression to PTB following Mycobacterium tuberculosis (Mtb) infection in humans is elevated frequencies of HLA-DR+CD4+ T cells, reflecting chronic T cell activation. However, the mechanisms by which activated HLA-DR+CD4+ T cells contribute to disease process is not known. We show that CD25- HLA-DR+CD4+ memory Teff from PTB patients are resistant to suppression mediated by Treg cells. An unbiased transcriptome analysis identified several key pathways that contribute to this resistance. Specifically, presence of HLA-DR+CD4+ T cells renders the effector population resistant to CCR5 and PD-L1 mediated suppression by Treg cells. In addition, the HLA-DR+CD4+ memory Teff cells express elevated levels of Th1/Th17 cytokines known to counter-regulate and dampen Treg suppression. These findings provide fresh insight to disease process in TB and identify HLA-DR+ Teff resistant to Treg suppression as a potential functional marker of disease.
Collapse
Affiliation(s)
- Asma Ahmed
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Soumya Nayak
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | | | - Chirag Dhar
- Division of Infectious Diseases, St John’s Research Institute, Bangalore, India
| | - Pravat Nalini Sahoo
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Bharath K. Sundararaj
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - George D. Souza
- Dept. of Pulmonary Medicine & Division of Infectious Diseases, St John’s Research Institute, Bangalore, India
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Department of Infectious Diseases, King’s College London, London, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, Guy's Campus, London, United Kingdom
- * E-mail: ,
| |
Collapse
|
15
|
Xu L, Ding W, Stohl LL, Zhou XK, Azizi S, Chuang E, Lam J, Wagner JA, Granstein RD. Regulation of T helper cell responses during antigen presentation by norepinephrine-exposed endothelial cells. Immunology 2018; 154:104-121. [PMID: 29164596 PMCID: PMC5904699 DOI: 10.1111/imm.12871] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/01/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022] Open
Abstract
Dermal blood vessels and regional lymph nodes are innervated by sympathetic nerves and, under stress, sympathetic nerves release norepinephrine (NE). Exposure of primary murine dermal microvascular endothelial cells (pDMECs) to NE followed by co-culture with Langerhans cells (LCs), responsive CD4+ T-cells and antigen resulted in modulation of CD4+ T-cell responses. NE-treatment of pDMECs induced increased production of interleukin (IL)-6 and IL-17A while down-regulating interferon (IFN)-γ and IL-22 release. This effect did not require contact between pDMECs and LCs or T-cells and depended upon pDMEC production of IL-6. The presence of NE-treated pDMECs increased the proportion of CD4+ T-cells expressing intracellular IL-17A and increased IL-17A mRNA while decreasing the proportion of IFN-γ- or IL-22-expressing CD4+ T-cells and mRNA levels for those cytokines. Retinoic acid receptor-related orphan receptor gamma (ROR-γt) mRNA was significantly increased in CD4+ T-cells while T-box transcription factor (T-bet) mRNA was decreased. Intradermal administration of NE prior to hapten immunization at the injection site produced a similar bias in draining lymph node CD4+ T-cells towards IL-17A and away from IFN-γ and IL-22 production. Under stress, release of NE may have significant regulatory effects on the outcome of antigen presentation through actions on ECs with enhancement of inflammatory skin disorders involving IL-17/T helper type 17 (Th17) cells.
Collapse
Affiliation(s)
- Linghui Xu
- Department of DermatologyWeill Cornell MedicineNew YorkNYUSA
- Present address:
Department of DermatologyThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Wanhong Ding
- Department of DermatologyWeill Cornell MedicineNew YorkNYUSA
| | - Lori L. Stohl
- Department of DermatologyWeill Cornell MedicineNew YorkNYUSA
| | - Xi K. Zhou
- Health Care Policy and ResearchWeill Cornell MedicineNew YorkNYUSA
| | - Shayan Azizi
- Department of DermatologyWeill Cornell MedicineNew YorkNYUSA
| | - Ethan Chuang
- Department of DermatologyWeill Cornell MedicineNew YorkNYUSA
| | - Jimmy Lam
- Department of DermatologyWeill Cornell MedicineNew YorkNYUSA
| | - John A. Wagner
- Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
- Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| | | |
Collapse
|
16
|
Ramos-Espinosa O, Islas-Weinstein L, Peralta-Álvarez MP, López-Torres MO, Hernández-Pando R. The use of immunotherapy for the treatment of tuberculosis. Expert Rev Respir Med 2018; 12:427-440. [PMID: 29575946 DOI: 10.1080/17476348.2018.1457439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Tuberculosis (TB) is the first cause of mortality by a single infectious agent in the world, causing more than one million deaths worldwide as reported by the World Health Organization (WHO). For the optimal control of TB infection, a protective immune response that limits bacterial spread without causing damage to the host is essential. Although most healthy individuals are capable of generating protective responses, patients who suffer pulmonary TB commonly present a defective immune function. Areas covered: We intend to highlight the potential of novel immunotherapeutic strategies that enhance and promote effective immune responses. The following methodology was undertaken for establishing a literature search: the authors used PubMed to search for 'Pulmonary Tuberculosis' and keywords that denoted the novel immunotherapeutic strategies discussed in length in the text including antibodies, antimicrobial peptides, cell therapy, cytokines and gene therapy. Expert commentary: The current therapeutic regimens for this disease are complex and involve the prolonged use of multiple antibiotics with diverse side effects that lead to therapeutic failure and bacterial resistance. The standard appliance of immunotherapy and its deployment to vulnerable populations will require coordinated work and may serve as a powerful tool to combat the ensuing threat of TB.
Collapse
Affiliation(s)
- Octavio Ramos-Espinosa
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| | - León Islas-Weinstein
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| | - Marco Polo Peralta-Álvarez
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México.,b Laboratory of Immunochemistry, Department of Immunology , Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional , México City , México
| | - Manuel Othoniel López-Torres
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| | - Rogelio Hernández-Pando
- a Section of Experimental Pathology, Department of Pathology , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , México
| |
Collapse
|
17
|
Voigt C, May P, Gottschlich A, Markota A, Wenk D, Gerlach I, Voigt S, Stathopoulos GT, Arendt KAM, Heise C, Rataj F, Janssen KP, Königshoff M, Winter H, Himsl I, Thasler WE, Schnurr M, Rothenfußer S, Endres S, Kobold S. Cancer cells induce interleukin-22 production from memory CD4 + T cells via interleukin-1 to promote tumor growth. Proc Natl Acad Sci U S A 2017; 114:12994-12999. [PMID: 29150554 PMCID: PMC5724250 DOI: 10.1073/pnas.1705165114] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IL-22 has been identified as a cancer-promoting cytokine that is secreted by infiltrating immune cells in several cancer models. We hypothesized that IL-22 regulation would occur at the interface between cancer cells and immune cells. Breast and lung cancer cells of murine and human origin induced IL-22 production from memory CD4+ T cells. In the present study, we found that IL-22 production in humans is dependent on activation of the NLRP3 inflammasome with the subsequent release of IL-1β from both myeloid and T cells. IL-1 receptor signaling via the transcription factors AhR and RORγt in T cells was necessary and sufficient for IL-22 production. In these settings, IL-1 induced IL-22 production from a mixed T helper cell population comprised of Th1, Th17, and Th22 cells, which was abrogated by the addition of anakinra. We confirmed these findings in vitro and in vivo in two murine tumor models, in primary human breast and lung cancer cells, and in deposited expression data. Relevant to ongoing clinical trials in breast cancer, we demonstrate here that the IL-1 receptor antagonist anakinra abrogates IL-22 production and reduces tumor growth in a murine breast cancer model. Thus, we describe here a previously unrecognized mechanism by which cancer cells induce IL-22 production from memory CD4+ T cells via activation of the NLRP3 inflammasome and the release of IL-1β to promote tumor growth. These findings may provide the basis for therapeutic interventions that affect IL-22 production by targeting IL-1 activity.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- CD4-Positive T-Lymphocytes/metabolism
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Proliferation
- Culture Media, Conditioned
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Inflammasomes/metabolism
- Interleukin-1beta/physiology
- Interleukins/biosynthesis
- Interleukins/metabolism
- Leukocytes, Mononuclear/metabolism
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Neoplasm Transplantation
- Signal Transduction
- Tumor Burden
- Interleukin-22
Collapse
Affiliation(s)
- Cornelia Voigt
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Comprehensive Pneumology Center, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Institute for Lung Biology and Disease, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Helmholtz Zentrum München, 81377 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Peter May
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Adrian Gottschlich
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Anamarija Markota
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Daniel Wenk
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Inga Gerlach
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | | | - Georgios T Stathopoulos
- Comprehensive Pneumology Center, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Institute for Lung Biology and Disease, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, University of Patras, Rio, Achaia, 26504 Greece
- Faculty of Medicine, University of Patras, Rio, Achaia, 26504 Greece
| | - Kristina A M Arendt
- Comprehensive Pneumology Center, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Institute for Lung Biology and Disease, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Helmholtz Zentrum München, 81377 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Constanze Heise
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Felicitas Rataj
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Klaus-Peter Janssen
- Chirurgische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität, 81675 Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Institute for Lung Biology and Disease, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Helmholtz Zentrum München, 81377 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Hauke Winter
- German Center for Lung Research, 81377 Munich, Germany
- Department of Thoracic Surgery, University Hospital, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Isabelle Himsl
- Brustzentrum Klinikum Dritter Orden, 80638 Munich, Germany
| | - Wolfgang E Thasler
- Biobank, Department of General, Visceral and Transplantation Surgery, University Hospital, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Max Schnurr
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Simon Rothenfußer
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany;
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
- German Center for Lung Research, 81377 Munich, Germany
| |
Collapse
|
18
|
Treerat P, Prince O, Cruz-Lagunas A, Muñoz-Torrico M, Salazar-Lezama MÁ, Selman M, Fallert-Junecko B, Reinhardt T, Alcorn JF, Kaushal D, Zuñiga J, Rangel-Moreno J, Kolls JK, Khader SA. Novel role for IL-22 in protection during chronic Mycobacterium tuberculosis HN878 infection. Mucosal Immunol 2017; 10:1069-1081. [PMID: 28247861 PMCID: PMC5477058 DOI: 10.1038/mi.2017.15] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/23/2017] [Indexed: 02/04/2023]
Abstract
Approximately 2 billion people are infected with Mycobacterium tuberculosis (Mtb), resulting in 1.4 million deaths every year. Among Mtb-infected individuals, clinical isolates belonging to the W-Beijing lineage are increasingly prevalent, associated with drug resistance, and cause severe disease immunopathology in animal models. Therefore, it is exceedingly important to identify the immune mechanisms that mediate protection against rapidly emerging Mtb strains, such as W-Beijing lineage. IL-22 is a member of the IL-10 family of cytokines with both protective and pathological functions at mucosal surfaces. Thus far, collective data show that IL-22 deficient mice are not more susceptible to aerosolized infection with less virulent Mtb strains. Thus, in this study we addressed the functional role for the IL-22 pathway in immunity to emerging Mtb isolates, using W-Beijing lineage member, Mtb HN878 as a prototype. We show that Mtb HN878 stimulates IL-22 production in TLR2 dependent manner and IL-22 mediates protective immunity during chronic stages of Mtb HN878 infection in mice. Interestingly, IL-22-dependent pathways in both epithelial cells and macrophages mediate protective mechanisms for Mtb HN878 control. Thus, our results project a new protective role for IL-22 in emerging Mtb infections.
Collapse
Affiliation(s)
- Puthayalai Treerat
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Oliver Prince
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Alfredo Cruz-Lagunas
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Marcela Muñoz-Torrico
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | | | - Moises Selman
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Beth Fallert-Junecko
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Todd Reinhardt
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F. Alcorn
- Division of Pulmonology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Deepak Kaushal
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Centre, Covington, LA, USA
| | - Joaquin Zuñiga
- Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Centre, Rochester, NY, USA
| | - Jay K. Kolls
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
19
|
Yokosawa M, Kondo Y, Tahara M, Iizuka-Koga M, Segawa S, Kaneko S, Tsuboi H, Yoh K, Takahashi S, Matsumoto I, Sumida T. T-bet over-expression regulates aryl hydrocarbon receptor-mediated T helper type 17 differentiation through an interferon (IFN)γ-independent pathway. Clin Exp Immunol 2017; 188:22-35. [PMID: 27936495 DOI: 10.1111/cei.12912] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2016] [Indexed: 01/27/2023] Open
Abstract
Various transcription factors are also known to enhance or suppress T helper type 17 (Th17) differentiation. We have shown previously that the development of collagen-induced arthritis was suppressed in T-bet transgenic (T-bet Tg) mice, and T-bet seemed to suppress Th17 differentiation through an interferon (IFN)-γ-independent pathway, although the precise mechanism remains to be clarified. The present study was designed to investigate further the mechanisms involved in the regulation of Th17 differentiation by T-bet over-expression, and we found the new relationship between T-bet and aryl hydrocarbon receptor (AHR). Both T-bet Tg mice and IFN-γ-/- -over-expressing T-bet (T-bet Tg/IFN-γ-/- ) mice showed inhibition of retinoic acid-related orphan receptor (ROR)γt expression and IL-17 production by CD4+ T cells cultured under conditions that promote Th-17 differentiation, and decreased IL-6 receptor (IL-6R) expression and signal transducer and activator of transcription-3 (STAT-3) phosphorylation in CD4+ T cells. The mRNA expression of ahr and rorc were suppressed in CD4+ T cells cultured under Th-17 conditions from T-bet Tg mice and T-bet Tg/IFN-γ-/- mice. CD4+ T cells of wild-type (WT) and IFN-γ-/- mice transduced with T-bet-expressing retrovirus also showed inhibition of IL-17 production, whereas T-bet transduction had no effect on IL-6R expression and STAT-3 phosphorylation. Interestingly, the mRNA expression of ahr and rorc were suppressed in CD4+ T cells with T-bet transduction cultured under Th17 conditions. The enhancement of interleukin (IL)-17 production from CD4+ T cells by the addition of AHR ligand with Th17 conditions was cancelled by T-bet over-expression. Our findings suggest that T-bet over-expression-induced suppression of Th17 differentiation is mediated through IFN-γ-independent AHR suppression.
Collapse
Affiliation(s)
- M Yokosawa
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - Y Kondo
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - M Tahara
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - M Iizuka-Koga
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - S Segawa
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - S Kaneko
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - H Tsuboi
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - K Yoh
- Division of Nephrology, Department of Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan.,Department of Anatomy and Embryology, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - S Takahashi
- Department of Anatomy and Embryology, University of Tsukuba, Tsukuba city, Ibaraki, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba city, Ibaraki, Japan.,Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba city, Ibaraki, Japan.,Laboratory Animal Resource Center (LARC), University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - I Matsumoto
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| | - T Sumida
- Division of Rheumatology, Department of Internal Medicine, University of Tsukuba, Tsukuba city, Ibaraki, Japan
| |
Collapse
|
20
|
Guo M, Xian QY, Rao Y, Zhang J, Wang Y, Huang ZX, Wang X, Bao R, Zhou L, Liu JB, Tang ZJ, Guo DY, Qin C, Li JL, Ho WZ. SIV Infection Facilitates Mycobacterium tuberculosis Infection of Rhesus Macaques. Front Microbiol 2017; 7:2174. [PMID: 28133458 PMCID: PMC5233680 DOI: 10.3389/fmicb.2016.02174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/29/2016] [Indexed: 01/14/2023] Open
Abstract
Tuberculosis (TB) is a common opportunistic infection and the leading cause of death for human immunodeficiency virus (HIV)-infected patients. Thus, it is necessary to understand the pathogenetic interactions between M.tb and HIV infection. In this study, we examined M.tb and/or simian immunodeficiency virus (SIV) infection of Chinese rhesus macaques. While there was little evidence that M.tb enhanced SIV infection of macaques, SIV could facilitate M.tb infection as demonstrated by X-rays, pathological and microbiological findings. Chest X-rays showed that co-infected animals had disseminated lesions in both left and right lungs, while M.tb mono-infected animals displayed the lesions only in right lungs. Necropsy of co-infected animals revealed a disseminated M.tb infection not only in the lungs but also in the extrapulmonary organs including spleen, pancreas, liver, kidney, and heart. The bacterial counts in the lungs, the bronchial lymph nodes, and the extrapulmonary organs of co-infected animals were significantly higher than those of M.tb mono-infected animals. The mechanistic studies demonstrated that two of three co-infected animals had lower levels of M.tb specific IFN-γ and IL-22 in PBMCs than M.tb mono-infected animals. These findings suggest that Chinese rhesus macaque is a suitable and alternative non-human primate model for SIV/M.tb coinfection studies. The impairment of the specific anti-TB immunity is likely to be a contributor of SIV-mediated enhancement M.tb infection.
Collapse
Affiliation(s)
- Ming Guo
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Qiao-Yang Xian
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Yan Rao
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Jing Zhang
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Yong Wang
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Zhi-Xiang Huang
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Xin Wang
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Rong Bao
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Li Zhou
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Jin-Biao Liu
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Zhi-Jiao Tang
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - De-Yin Guo
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei, China
| | - Chuan Qin
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College Beijing, China
| | - Jie-Liang Li
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine Philadelphia, PA, USA
| | - Wen-Zhe Ho
- School of Basic Medical Sciences, Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan UniversityWuhan, Hubei, China; Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of MedicinePhiladelphia, PA, USA
| |
Collapse
|
21
|
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
|
22
|
Domingo-Gonzalez R, Prince O, Cooper A, Khader SA. Cytokines and Chemokines in Mycobacterium tuberculosis Infection. Microbiol Spectr 2016; 4:10.1128/microbiolspec.TBTB2-0018-2016. [PMID: 27763255 PMCID: PMC5205539 DOI: 10.1128/microbiolspec.tbtb2-0018-2016] [Citation(s) in RCA: 266] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Indexed: 02/06/2023] Open
Abstract
Chemokines and cytokines are critical for initiating and coordinating the organized and sequential recruitment and activation of cells into Mycobacterium tuberculosis-infected lungs. Correct mononuclear cellular recruitment and localization are essential to ensure control of bacterial growth without the development of diffuse and damaging granulocytic inflammation. An important block to our understanding of TB pathogenesis lies in dissecting the critical aspects of the cytokine/chemokine interplay in light of the conditional role these molecules play throughout infection and disease development. Much of the data highlighted in this review appears at first glance to be contradictory, but it is the balance between the cytokines and chemokines that is critical, and the "goldilocks" (not too much and not too little) phenomenon is paramount in any discussion of the role of these molecules in TB. Determination of how the key chemokines/cytokines and their receptors are balanced and how the loss of that balance can promote disease is vital to understanding TB pathogenesis and to identifying novel therapies for effective eradication of this disease.
Collapse
Affiliation(s)
| | - Oliver Prince
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63130
| | - Andrea Cooper
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester LE1 7RH, United Kingdom
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
23
|
Niegowska M, Rapini N, Biet F, Piccinini S, Bay S, Lidano R, Manca Bitti ML, Sechi LA. Seroreactivity against Specific L5P Antigen from Mycobacterium avium subsp. paratuberculosis in Children at Risk for T1D. PLoS One 2016; 11:e0157962. [PMID: 27336739 PMCID: PMC4919038 DOI: 10.1371/journal.pone.0157962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/07/2016] [Indexed: 01/14/2023] Open
Abstract
Aims/Hypothesis Although numerous environmental agents have been investigated over the years as possible triggers of type 1 diabetes (T1D), its causes remain unclear. We have already demonstrated an increased prevalence of antibodies against peptides derived from Mycobacterium avuim subsp. paratuberculosis (MAP) homologous to human zinc transporter 8 protein (ZnT8) and proinsulin in Italian subjects at risk for or affected by T1D. In this study, we compared titers of the previously detected antibodies with seroreactivity to MAP lipopentapetide (L5P) that recently emerged as a strong immunogenic component able to specifically distinguish MAP from other mycobacteria. Methods Plasma of 32 children and youth at risk for T1D including follow-up samples and 42 age-matched healthy controls (HC) recruited at the Tor Vergata University Hospital in Rome was analyzed by indirect ELISA for the presence of antibodies against MAP-derived epitopes MAP3865c133–141, MAP3865c125-133, MAP2404c70-85 and MAP1,4αgbp157-173 along with their ZnT8 and proinsulin homologs. The data were analyzed through two-tailed Mann-Whitney U test and relation between variables was determined by principal component analysis. Results Responses to L5P were not detectable in subjects whose initial seroreactivity to MAP peptides and their human homologs was lost in follow-up samples, whereas anti-L5P antibodies appeared constantly in individuals with a stable immunity against MAP antigens. The overall coincidence in positivity to L5P and the four MAP epitopes both in children at risk for T1D and HC exceeded 90%. Conclusions MAP-derived homologs may cross-react with ZnT8 and proinsulin peptides inducing immune responses at a young age in subjects predisposed for T1D. Thus, L5P may have a diagnostic value to immediately indicate the presence of anti-MAP seroreactivity when evaluation of a more complex antibody status is not required. Almost complete coincidence in responses to both types of antigens lends support to the involvement of MAP in T1D.
Collapse
Affiliation(s)
- Magdalena Niegowska
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Novella Rapini
- Pediatric Diabetology Unit, Policlinico di Tor Vergata, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Frank Biet
- UMR1282, Infectiologie et Santé Publique (ISP-311), INRA Centre Val de Loire, F-37380, Nouzilly, France
| | - Simona Piccinini
- Pediatric Diabetology Unit, Policlinico di Tor Vergata, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Sylvie Bay
- Institut Pasteur, Unité de Chimie des Biomolécules, Département de Biologie Structurale et Chimie, Paris, France
| | - Roberta Lidano
- Pediatric Diabetology Unit, Policlinico di Tor Vergata, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Maria Luisa Manca Bitti
- Pediatric Diabetology Unit, Policlinico di Tor Vergata, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Leonardo A. Sechi
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- * E-mail:
| |
Collapse
|
24
|
Trentini MM, de Oliveira FM, Kipnis A, Junqueira-Kipnis AP. The Role of Neutrophils in the Induction of Specific Th1 and Th17 during Vaccination against Tuberculosis. Front Microbiol 2016; 7:898. [PMID: 27375607 PMCID: PMC4901074 DOI: 10.3389/fmicb.2016.00898] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/26/2016] [Indexed: 11/15/2022] Open
Abstract
Mycobacterium tuberculosis causes tuberculosis (TB), a disease that killed more than 1.5 million people worldwide in 2014, and the Bacillus Calmette Guérin (BCG) vaccine is the only currently available vaccine against TB. However, it does not protect adults. Th1 and Th17 cells are crucial for TB control, as well as the neutrophils that are directly involved in DC trafficking to the draining lymph nodes and the activation of T lymphocytes during infection. Although several studies have shown the importance of neutrophils during M. tuberculosis infection, none have shown its role in the development of a specific response to a vaccine. The vaccine mc2-CMX was shown to protect mice against M. tuberculosis challenge, mainly due to specific Th1 and Th17 cells. This study evaluated the importance of neutrophils in the generation of the Th1- and Th17-specific responses elicited by this vaccine. The vaccine injection induced a neutrophil rich lesion with a necrotic central area. The IL-17 KO mice did not generate vaccine-specific Th1 cells. The vaccinated IL-22 KO mice exhibited Th1- and Th17-specific responses. Neutrophil depletion during vaccination abrogated the induction of Th1-specific responses and prohibited the bacterial load reduction observed in the vaccinated animals. The results show, for the first time, the role of neutrophils in the generation of specific Th1 and Th17 cells in response to a tuberculosis vaccine.
Collapse
Affiliation(s)
- Monalisa M Trentini
- Laboratory of Immunopathology of Infectious Disease, Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Disease and Public Health, Federal University of Goiás Goiânia, Brazil
| | - Fábio M de Oliveira
- Laboratory of Immunopathology of Infectious Disease, Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Disease and Public Health, Federal University of Goiás Goiânia, Brazil
| | - André Kipnis
- Laboratory of Immunopathology of Infectious Disease, Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Disease and Public Health, Federal University of Goiás Goiânia, Brazil
| | - Ana P Junqueira-Kipnis
- Laboratory of Immunopathology of Infectious Disease, Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Disease and Public Health, Federal University of Goiás Goiânia, Brazil
| |
Collapse
|
25
|
Korb VC, Phulukdaree A, Lalloo UG, Chuturgoon AA, Moodley D. TB/HIV pleurisy reduces Th17 lymphocyte proportion independent of the cytokine microenvironment. Tuberculosis (Edinb) 2016; 99:92-99. [PMID: 27450010 DOI: 10.1016/j.tube.2016.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/01/2016] [Indexed: 12/29/2022]
Abstract
T-helper (Th) 17 cells are a pro-inflammatory subset of CD4(+) effector T-cells critical in mucosal immunity. Imbalances in Th17 cell proportion have been implicated in the pathogenesis of several diseases; however, this has not been adequately explored in tuberculosis (TB) and human immunodeficiency virus (HIV) co-infection. Since Th17 cells are predominantly mucosally associated, we assessed Th17 proportion and associated microenvironment in pleural effusions from patients co-infected with TB/HIV. Our results show that TB(+)HIV(+) pleurisy results in significantly reduced frequency of CD4(+)IL-17(+)RORC(+)STAT3(+) Th17 cells compared to TB(-)HIV(-)ex vivo (p = 0.0054) and was confirmed in conditioned media studies in vitro (p = 0.0001). This was not associated with alterations in Th17 polarising cytokines IL-6, IL-21 and IL-23 or changes in Th17 signature cytokines IL-17A and F. However, the mRNA expression of Th17 signalling molecules, IL-6 (p = 0.0022), IL-6R (p = 0.0247), IL-1β (p = 0.0022) and signal transducer and activator (STAT) 3 (p = 0.0022) were significantly upregulated. Notably, TB(+)HIV(+) pleural fluid contained significantly higher concentrations of IL-1β (p = 0.0008), IL-22 (p = 0.0115), IL-31 (p = 0.0210), TNF-α (p = 0.0251) and IFN-γ (p = 0.0026) than TB(-)HIV(-) pleural fluid ex vivo. Taken together, this suggests a reduced portion of Th17 lymphocytes in TB/HIV pleurisy is independent of locally mediated cytokine polarisation.
Collapse
Affiliation(s)
- Vanessa C Korb
- Discipline of Medical Biochemistry and Chemical Pathology, University of KwaZulu-Natal, Howard College Campus, King George V Avenue, Durban 4041, South Africa.
| | - Alisa Phulukdaree
- Discipline of Medical Biochemistry and Chemical Pathology, University of KwaZulu-Natal, Howard College Campus, King George V Avenue, Durban 4041, South Africa.
| | - Umesh G Lalloo
- Discipline of Medical Biochemistry and Chemical Pathology, University of KwaZulu-Natal, Howard College Campus, King George V Avenue, Durban 4041, South Africa.
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry and Chemical Pathology, University of KwaZulu-Natal, Howard College Campus, King George V Avenue, Durban 4041, South Africa.
| | - Devapregasan Moodley
- Discipline of Medical Biochemistry and Chemical Pathology, University of KwaZulu-Natal, Howard College Campus, King George V Avenue, Durban 4041, South Africa.
| |
Collapse
|
26
|
Abdalla AE, Lambert N, Duan X, Xie J. Interleukin-10 Family and Tuberculosis: An Old Story Renewed. Int J Biol Sci 2016; 12:710-7. [PMID: 27194948 PMCID: PMC4870714 DOI: 10.7150/ijbs.13881] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/15/2016] [Indexed: 02/06/2023] Open
Abstract
The interleukin-10 (IL-10) family of cytokines consists of six immune mediators, namely IL-10, IL-19, IL-20, IL-22, IL-24 and IL-26. IL-10, IL-22, IL-24 and IL-26 are critical for the regulation of host defense against Mycobacterium tuberculosis infections. Specifically, IL-10 and IL-26 can suppress the antimycobacterial immunity and promote the survival of pathogen, while IL-22 and IL-24 can generate protective responses and inhibit the intracellular growth of pathogen. Knowledge about the new players in tuberculosis immunology, namely IL-10 family, can inform novel immunity-based countermeasures and host directed therapies against tuberculosis.
Collapse
Affiliation(s)
- Abualgasim Elgaili Abdalla
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
- 2. Department of Clinical Microbiology, College of Medical Laboratory Sciences, Omdurman Islamic University, Omdurman, Khartoum, Sudan
| | - Nzungize Lambert
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Xiangke Duan
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Jianping Xie
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| |
Collapse
|
27
|
Bellemore SM, Nikoopour E, Krougly O, Lee‐Chan E, Fouser LA, Singh B. Pathogenic T helper type 17 cells contribute to type 1 diabetes independently of interleukin-22. Clin Exp Immunol 2016; 183:380-8. [PMID: 26496462 PMCID: PMC4750601 DOI: 10.1111/cei.12735] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2015] [Indexed: 12/12/2022] Open
Abstract
We have shown that pathogenic T helper type 17 (Th17) cells differentiated from naive CD4(+) T cells of BDC2·5 T cell receptor transgenic non-obese diabetic (NOD) mice by interleukin (IL)-23 plus IL-6 produce IL-17, IL-22 and induce type 1 diabetes (T1D). Neutralizing interferon (IFN)-γ during the polarization process leads to a significant increase in IL-22 production by these Th17 cells. We also isolated IL-22-producing Th17 cells from the pancreas of wild-type diabetic NOD mice. IL-27 also blocked IL-22 production from diabetogenic Th17 cells. To determine the functional role of IL-22 produced by pathogenic Th17 cells in T1D we neutralized IL-22 in vivo by using anti-IL-22 monoclonal antibody. We found that blocking IL-22 did not alter significantly adoptive transfer of disease by pathogenic Th17 cells. Therefore, IL-22 is not required for T1D pathogenesis. The IL-22Rα receptor for IL-22 however, increased in the pancreas of NOD mice during disease progression and based upon our and other studies we suggest that IL-22 may have a regenerative and protective role in the pancreatic islets.
Collapse
Affiliation(s)
- S. M. Bellemore
- Centre for Human Immunology and Department of Microbiology and Immunology and Robarts Research Institute, University of Western OntarioLondonOntarioCanada
| | - E. Nikoopour
- Centre for Human Immunology and Department of Microbiology and Immunology and Robarts Research Institute, University of Western OntarioLondonOntarioCanada
| | - O. Krougly
- Centre for Human Immunology and Department of Microbiology and Immunology and Robarts Research Institute, University of Western OntarioLondonOntarioCanada
| | - E. Lee‐Chan
- Centre for Human Immunology and Department of Microbiology and Immunology and Robarts Research Institute, University of Western OntarioLondonOntarioCanada
| | - L. A. Fouser
- Inflammation and ImmunologyBiotherapeutics Research and Development, Pfizer Inc.CambridgeMA02140USA
| | - B. Singh
- Centre for Human Immunology and Department of Microbiology and Immunology and Robarts Research Institute, University of Western OntarioLondonOntarioCanada
| |
Collapse
|
28
|
Dixon BREA, Radin JN, Piazuelo MB, Contreras DC, Algood HMS. IL-17a and IL-22 Induce Expression of Antimicrobials in Gastrointestinal Epithelial Cells and May Contribute to Epithelial Cell Defense against Helicobacter pylori. PLoS One 2016; 11:e0148514. [PMID: 26867135 PMCID: PMC4750979 DOI: 10.1371/journal.pone.0148514] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/19/2016] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori colonization of the human stomach can lead to adverse clinical outcomes including gastritis, peptic ulcers, or gastric cancer. Current data suggest that in addition to bacterial virulence factors, the magnitude and types of immune responses influence the outcome of colonization. Specifically, CD4+ T cell responses impact the pathology elicited in response to H. pylori. Because gastritis is believed to be the initiating host response to more detrimental pathological outcomes, there has been a significant interest in pro-inflammatory T cell cytokines, including the cytokines produced by T helper 17 cells. Th17 cells produce IL-17A, IL-17F, IL-21 and IL-22. While these cytokines have been linked to inflammation, IL-17A and IL-22 are also associated with anti-microbial responses and control of bacterial colonization. The goal of this research was to determine the role of IL-22 in activation of antimicrobial responses in models of H. pylori infection using human gastric epithelial cell lines and the mouse model of H. pylori infection. Our data indicate that IL-17A and IL-22 work synergistically to induce antimicrobials and chemokines such as IL-8, components of calprotectin (CP), lipocalin (LCN) and some β-defensins in both human and primary mouse gastric epithelial cells (GEC) and gastroids. Moreover, IL-22 and IL-17A-activated GECs were capable of inhibiting growth of H. pylori in vitro. While antimicrobials were activated by IL-17A and IL-22 in vitro, using a mouse model of H. pylori infection, the data herein indicate that IL-22 deficiency alone does not render mice more susceptible to infection, change their antimicrobial gene transcription, or significantly change their inflammatory response.
Collapse
Affiliation(s)
- Beverly R. E. A. Dixon
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jana N. Radin
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - M. Blanca Piazuelo
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Diana C. Contreras
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Holly M. Scott Algood
- Veterans Affairs Tennessee Valley Healthcare Services, Nashville, Tennessee, United States of America
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
29
|
Shen W, Hixon JA, McLean MH, Li WQ, Durum SK. IL-22-Expressing Murine Lymphocytes Display Plasticity and Pathogenicity in Reporter Mice. Front Immunol 2016; 6:662. [PMID: 26834739 PMCID: PMC4717188 DOI: 10.3389/fimmu.2015.00662] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 12/26/2015] [Indexed: 12/31/2022] Open
Abstract
IL-22 has multiple activities ranging from tissue repair to inflammation. To characterize the pathogenicity and plasticity of cells that produce IL-22, a novel reporter mouse strain was generated. Homeostatic IL-22 reporter expression was observed in intestinal lymphoid cells identified as CD4 T cells and ILC3 cells. In a model of inflammatory bowel disease, CD4 T cells strongly expressed the IL-22 reporter in mesenteric lymph node. To examine plasticity of IL-22(+) T cells, they were purified after generation in vitro or in vivo from inflamed colon, and then cultured under Th1, Th2, or Th17 conditions. In vitro-generated IL-22(+) CD4 T cells showed relatively durable IL-22 expression under Th1 or Th2 conditions, whereas in vivo-generated cells rapidly lost IL-22 expression under these conditions. In vitro-generated cells could not be diverted to express Th1 or Th2 cytokines despite the expression of "master regulators." In vivo-generated cells could be diverted, at very low frequency, to express Th1 or Th2 cytokines. Both in vitro- and in vivo-generated cells could be induced in vitro to express high levels of IL-17A and IL-17F, assigning them to a "Th17 biased" class. However, IL-27 potently downregulated IL-22 expression. To examine IL-22(+) T cell pathogenicity, in vitro-generated cells were transferred into Rag1(-/-) mice, retaining the modest reporter expression and inducing moderate colitis. In contrast, IL-22 expressers from colitic mice, transferred into secondary hosts, lost reporter expression, acquired high T-bet and modest IFNγ and IL-17 expression, and induced severe colitis. These findings are consistent with a model of strong polarization under optimal in vitro conditions, but a plastic state of T cells in vivo.
Collapse
Affiliation(s)
- Wei Shen
- Cancer and Inflammation Program, Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Julie A. Hixon
- Cancer and Inflammation Program, Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Mairi H. McLean
- Cancer and Inflammation Program, Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Wen Qing Li
- Cancer and Inflammation Program, Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Scott K. Durum
- Cancer and Inflammation Program, Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| |
Collapse
|
30
|
Complexity and Controversies over the Cytokine Profiles of T Helper Cell Subpopulations in Tuberculosis. J Immunol Res 2015; 2015:639107. [PMID: 26495323 PMCID: PMC4606092 DOI: 10.1155/2015/639107] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/03/2015] [Indexed: 12/24/2022] Open
Abstract
Tuberculosis (TB) is a contagious infectious disease caused by the TB-causing bacillus Mycobacterium tuberculosis and is considered a public health problem with enormous social impact. Disease progression is determined mainly by the balance between the microorganism and the host defense systems. Although the immune system controls the infection, this control does not necessarily lead to sterilization. Over recent decades, the patterns of CD4+ T cell responses have been studied with a goal of complete understanding of the immunological mechanisms involved in the maintenance of latent or active tuberculosis infection and of the clinical cure after treatment. Conflicting results have been suggested over the years, particularly in studies comparing experimental models and human disease. In recent years, in addition to Th1, Th2, and Th17 profiles, new standards of cellular immune responses, such as Th9, Th22, and IFN-γ-IL-10 double-producing Th cells, discussed here, have also been described. Additionally, many new roles and cellular sources have been described for IL-10, demonstrating a critical role for this cytokine as regulatory, rather than merely pathogenic cytokine, involved in the establishment of chronic latent infection, in the clinical cure after treatment and in keeping antibacillary effector mechanisms active to prevent immune-mediated damage.
Collapse
|
31
|
Riccomi A, Palma C. B Cells and Programmed Death-Ligand 2 Signaling Are Required for Maximal Interferon-γ Recall Response by Splenic CD4⁺ Memory T Cells of Mice Vaccinated with Mycobacterium tuberculosis Ag85B. PLoS One 2015; 10:e0137783. [PMID: 26379242 PMCID: PMC4574766 DOI: 10.1371/journal.pone.0137783] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/21/2015] [Indexed: 02/06/2023] Open
Abstract
CD4+ T cells producing interferon-γ are crucial for protection against Mycobacterium tuberculosis infection and are the cornerstone of tuberculosis vaccination and immunological diagnostic assays. Since emerging evidence indicates that B cells can modulate T cell responses to M. tuberculosis infection, we investigated the contribution of B cells in regulating interferon-γ recall response by memory Thelper1 cells specific for Ag85B, a leading candidate for tuberculosis sub-unit vaccines. We found that B cells were able to maximize the reactivation of CD4+ memory T cells and the interferon-γ response against ex vivo antigen recall in spleens of mice vaccinated with Ag85B. B cell-mediated increase of interferon-γ response was particular evident for high interferon-γ producer CD4+ memory T cells, likely because those T cells were required for triggering and amplification of B cell activation. A positive-feedback loop of mutual activation between B cells, not necessarily antigen-experienced but with integral phosphatidylinositol-3 kinase (PI3K) pathway and a peculiar interferon-γ-producing CD4highT cell subset was established. Programed death-ligand 2 (PD-L2), expressed both on B and the highly activated CD4high T cells, contributed to the increase of interferon-γ recall response through a PD1-independent pathway. In B cell-deficient mice, interferon-γ production and activation of Ag85B-specific CD4+ T cells were blunted against ex vivo antigen recall but these responses could be restored by adding B cells. On the other hand, B cells appeared to down-regulate interleukin-22 recall response. Our data point out that nature of antigen presenting cells determines quality and size of T cell cytokine recall responses. Thus, antigen presenting cells, including B cells, deserve to be considered for a better prediction of cytokine responses by peripheral memory T cells specific for M. tuberculosis antigens. We also invite to consider B cells, PD-L2 and PI3K as potential targets for therapeutic modulation of T cell cytokine responses for tuberculosis control.
Collapse
Affiliation(s)
- Antonella Riccomi
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Carla Palma
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| |
Collapse
|
32
|
Mayer-Barber KD, Barber DL. Innate and Adaptive Cellular Immune Responses to Mycobacterium tuberculosis Infection. Cold Spring Harb Perspect Med 2015; 5:a018424. [PMID: 26187873 PMCID: PMC4665043 DOI: 10.1101/cshperspect.a018424] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Host resistance to Mycobacterium tuberculosis (Mtb) infection requires the coordinated efforts of innate and adaptive immune cells. Diverse pulmonary myeloid cell populations respond to Mtb with unique contributions to both host-protective and potentially detrimental inflammation. Although multiple cell types of the adaptive immune system respond to Mtb infection, CD4 T cells are the principal antigen-specific cells responsible for containment of Mtb infection, but they can also be major contributors to disease during Mtb infection in several different settings. Here, we will discuss the role of different myeloid populations as well as the dual nature of CD4 T cells in Mtb infection with a primary focus on data generated using in vivo cellular immunological studies in experimental animal models and in humans when available.
Collapse
Affiliation(s)
- Katrin D Mayer-Barber
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Daniel L Barber
- T Lymphocyte Biology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
33
|
Pro- and anti-inflammatory cytokines in tuberculosis: A two-edged sword in TB pathogenesis. Semin Immunol 2014; 26:543-51. [DOI: 10.1016/j.smim.2014.09.011] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/28/2014] [Accepted: 09/29/2014] [Indexed: 12/19/2022]
|
34
|
Olalekan SA, Cao Y, Finnegan A. Tissue specific CD4+ T cell priming determines the requirement for interleukin-23 in experimental arthritis. Arthritis Res Ther 2014; 16:440. [PMID: 25253467 PMCID: PMC4203961 DOI: 10.1186/s13075-014-0440-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/27/2014] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory disease with striking heterogeneity in (i) clinical presentation, (ii) autoantibody profiles and (iii) responses to treatment suggesting that distinct molecular mechanisms may underlie the disease process. Proteoglycan-induced arthritis (PGIA) is induced by two pathways either by intraperitoneal (i.p.) or subcutaneous (s.c.) exposure to PG. CD4+ T cells primed by the i.p. route are T helper (Th)1 cells expressing interferon gamma (IFN-γ) whereas CD4+ T cells primed by the s.c. route are Th17 cells expressing interleukin (IL)-17. IL-23 is necessary for maintaining the phenotype of Th17 cells; however, IL-23 is inflammatory independent of IL-17. The aim of this study was to determine if PGIA induced by different routes of immunization is dependent on IL-23. METHODS BALB/c wild type (WT), IL-12p40-/- and IL-23p19-/- littermate mice were immunized with recombinant G1 (rG1) domain of human PG in adjuvant either i.p. or s.c. and development of arthritis monitored. Joint histology was assessed. CD4+ T cell cytokines in spleen, lymph node (LN), and joint were assessed by intracellular staining and cytokine enzyme-linked immunosorbent assay. RNA transcripts for cytokines and transcription factors were examined. RESULTS PGIA was suppressed in the p40-/- and p19-/- mice immunized by the s.c. route but only inhibited in p40-/- mice by the i.p. route. The joints of s.c. but not i.p. sensitized mice contained a population of CD4+ T cells expressing single positive IFN-γ and IL-17 and double positive IFN-γ/IL-17 which were dependent on IL-23 expression. The IFN-γ and IL-17 response in spleen and inguinal LN was inhibited in p19-/- mice and p40-/- mice after s.c. immunization, whereas in i.p. immunized p19-/- mice, IL-17 but not IFN-γ was reduced. Inguinal LN CD11c+ dendritic cells (DC) from s.c. immunized, but not spleen DC from i.p. immunized mice, produced IL-23, IL-1β, and IL-6 and activated T cells to produce IL-17. CONCLUSION IL-23 is necessary for the activity of Th17 after s.c. immunization and does not play a role independent of IL-17 after i.p. immunization. These data demonstrate that the molecular pathways IL-23/17 and IL-12/IFN-γ may represent subtypes of arthritis determined by the mode of induction.
Collapse
|
35
|
Heitmann L, Abad Dar M, Schreiber T, Erdmann H, Behrends J, Mckenzie ANJ, Brombacher F, Ehlers S, Hölscher C. The IL-13/IL-4Rα axis is involved in tuberculosis-associated pathology. J Pathol 2014; 234:338-50. [PMID: 24979482 PMCID: PMC4277691 DOI: 10.1002/path.4399] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 06/05/2014] [Accepted: 06/22/2014] [Indexed: 01/20/2023]
Abstract
Human tuberculosis (TB) is a leading global health threat and still constitutes a major medical challenge. However, mechanisms governing tissue pathology during post-primary TB remain elusive, partly because genetically or immunologically tractable animal models are lacking. In human TB, the demonstration of a large relative increase in interleukin (IL)-4 and IL-13 expression, which correlates with lung damage, indicates that a subversive T helper (TH)2 component in the response to Mycobacterium tuberculosis (Mtb) may undermine protective immunity and contribute to reactivation and tissue pathology. Up to now, there has been no clear evidence regarding whether IL-4/IL-13-IL-4 receptor-α (Rα)-mediated mechanisms may in fact cause reactivation and pathology. Unfortunately, the virtual absence of centrally necrotizing granulomas in experimental murine TB is associated with a poor induction of a TH2 immune response. We therefore hypothesize that, in mice, an increased production of IL-13 may lead to a pathology similar to human post-primary TB. In our study, aerosol Mtb infection of IL-13-over-expressing mice in fact resulted in pulmonary centrally necrotizing granulomas with multinucleated giant cells, a hypoxic rim and a perinecrotic collagen capsule, with an adjacent zone of lipid-rich, acid-fast bacilli-containing foamy macrophages, thus strongly resembling the pathology in human post-primary TB. Granuloma necrosis (GN) in Mtb-infected IL-13-over-expressing mice was associated with the induction of arginase-1-expressing macrophages. Indirect blockade of the endogenous arginase inhibitor l-hydroxyarginine in Mtb-infected wild-type mice resulted in a strong arginase expression and precipitated a similar pathology of GN. Together, we here introduce an experimental TB model that displays many features of centrally necrotizing granulomas in human post-primary TB and demonstrate that IL-13/IL-4Rα-dependent mechanisms leading to arginase-1 expression are involved in TB-associated tissue pathology.
Collapse
Affiliation(s)
- Lisa Heitmann
- Infection Immunology, Research Centre Borstel, Germany; Priority Research Area 'Infections', Research Centre Borstel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The adaptive immune system provides critical defense against pathogenic bacteria. Commensal bacteria have begun to receive much attention in recent years, especially in the gut where there is growing evidence of complex interactions with the adaptive immune system. In the present study, we observed that commensal skin bacteria are recognized by major populations of T cells in skin-draining lymph nodes of mice. Recombination activating gene 1 (Rag1)(-/-) mice, which lack adaptive immune cells, contained living skin-derived bacteria and bacterial sequences, especially mycobacteria, in their skin-draining lymph nodes. T cells from skin-draining lymph nodes of normal mice were shown, in vitro, to specifically recognize bacteria of several species that were grown from Rag1(-/-) lymph nodes. T cells from skin-draining lymph nodes, transferred into Rag1(-/-) mice proliferated in skin-draining lymph nodes, expressed a restricted T-cell receptor spectrotype and produced cytokines. Transfer of T cells into Rag1(-/-) mice had the effect of reducing bacterial sequences in skin-draining lymph nodes and in skin itself. Antibacterial effects of transferred T cells were dependent on IFNγ and IL-17A. These studies suggest a previously unrecognized role for T cells in controlling skin commensal bacteria and provide a mechanism to account for cutaneous infections and mycobacterial infections in T-cell-deficient patients.
Collapse
|
37
|
Kara EE, Comerford I, Fenix KA, Bastow CR, Gregor CE, McKenzie DR, McColl SR. Tailored immune responses: novel effector helper T cell subsets in protective immunity. PLoS Pathog 2014; 10:e1003905. [PMID: 24586147 PMCID: PMC3930558 DOI: 10.1371/journal.ppat.1003905] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Differentiation of naïve CD4⁺ cells into functionally distinct effector helper T cell subsets, characterised by distinct "cytokine signatures," is a cardinal strategy employed by the mammalian immune system to efficiently deal with the rapidly evolving array of pathogenic microorganisms encountered by the host. Since the T(H)1/T(H)2 paradigm was first described by Mosmann and Coffman, research in the field of helper T cell biology has grown exponentially with seven functionally unique subsets having now been described. In this review, recent insights into the molecular mechanisms that govern differentiation and function of effector helper T cell subsets will be discussed in the context of microbial infections, with a focus on how these different helper T cell subsets orchestrate immune responses tailored to combat the nature of the pathogenic threat encountered.
Collapse
Affiliation(s)
- Ervin E. Kara
- School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Iain Comerford
- School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kevin A. Fenix
- School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Cameron R. Bastow
- School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Carly E. Gregor
- School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Duncan R. McKenzie
- School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Shaun R. McColl
- School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
38
|
Mavropoulos A, Rigopoulou EI, Liaskos C, Bogdanos DP, Sakkas LI. The role of p38 MAPK in the aetiopathogenesis of psoriasis and psoriatic arthritis. Clin Dev Immunol 2013; 2013:569751. [PMID: 24151518 PMCID: PMC3787653 DOI: 10.1155/2013/569751] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 08/14/2013] [Indexed: 02/08/2023]
Abstract
The pathogenetic mechanisms responsible for the induction of immune-mediated disorders, such as psoriasis, remain not well characterized. Molecular signaling pathways are not well described in psoriasis, as well as psoriatic arthritis, which is seen in up to 40% of patients with psoriasis. Signaling pathway defects have long been hypothesized to participate in the pathology of psoriasis, yet their implication in the altered psoriatic gene expression still remains unclear. Emerging data suggest a potential pathogenic role for mitogen activated protein kinases p38 (p38 MAPK) extracellular signal-regulated kinase 1/2 (ERK1/2), and c-Jun N-terminal kinase (JNK) in the development of psoriasis. The data are still limited, though, for psoriatic arthritis. This review discusses the current data suggesting a crucial role for p38 MAPK in the pathogenesis of these disorders.
Collapse
Affiliation(s)
- Athanasios Mavropoulos
- Cellular Immunotherapy and Molecular Immunodiagnostics, Institute of Research and Technology Thessaly, 41222 Larissa, Greece
- Division of Transplantation Immunology and Mucosal Biology, Institute of Liver Studies, King's College London School of Medicine at King's College Hospital, Denmark Hill Campus, London SE5 9RS, UK
| | - Eirini I. Rigopoulou
- Department of Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Christos Liaskos
- Cellular Immunotherapy and Molecular Immunodiagnostics, Institute of Research and Technology Thessaly, 41222 Larissa, Greece
| | - Dimitrios P. Bogdanos
- Cellular Immunotherapy and Molecular Immunodiagnostics, Institute of Research and Technology Thessaly, 41222 Larissa, Greece
- Division of Transplantation Immunology and Mucosal Biology, Institute of Liver Studies, King's College London School of Medicine at King's College Hospital, Denmark Hill Campus, London SE5 9RS, UK
- Department of Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Lazaros I. Sakkas
- Department of Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
- Center of Molecular Medicine, Old Dominion University, 23529 Monarch Way, Norfolk, VA, USA
- Department of Rheumatology, Faculty of Medicine School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
| |
Collapse
|