1
|
Wang X, Zhang C, Zhang Y, Wang S, Thomas R, Yang X. NK Cells Modulate Dendritic Cell (DC) Signaling Pathways and DC Recruitment in Chlamydial Infection. Int J Mol Sci 2025; 26:3769. [PMID: 40332391 PMCID: PMC12028113 DOI: 10.3390/ijms26083769] [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: 02/22/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Previous studies have demonstrated the significant impact of NK cells on adaptive immune responses against chlamydial infections through modulating DCs, yet the molecular mechanisms remain incompletely understood. This study investigates the role of NK cells in modulating DC signaling pathways and the recruitment of DCs during Chlamydia muridarum infection. Transcriptomic analyses revealed significant downregulation of key genes in DCs from NK-depleted mice involved in type I immunity, including IL12rb2, IL-18rap, and chemokine signaling components such as Ccl3, Ccl5, and Ccr5. Gene ontology (GO) analyses confirmed impaired chemokine-chemokine receptor interactions in DCs from NK-depleted mice. Moreover, flow cytometry analysis showed that NK-cell depletion reduced CCR5 expression on splenic and pulmonary DCs, impairing their migration toward CCL3 and CCL5. Furthermore, IFN-γ enhanced CCR5 expression on the surface of DCs, consequently promoting their migration, which was blocked by anti-IFN-γ antibodies. In vitro migration assays showed that treatment of DCs with IFN-γ increased their responsiveness to CCL3 and CCL5, the ligands of CCR5. Collectively, this study provides new insights into the indispensable role of NK cells in orchestrating DC signaling and the recruitment of DCs during chlamydial infection.
Collapse
Affiliation(s)
- Xinting Wang
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (X.W.)
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Chunyan Zhang
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (X.W.)
| | - Yongci Zhang
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (X.W.)
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Shuhe Wang
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (X.W.)
| | - Rony Thomas
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (X.W.)
| | - Xi Yang
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (X.W.)
| |
Collapse
|
2
|
Yount KS, Darville T. Immunity to Sexually Transmitted Bacterial Infections of the Female Genital Tract: Toward Effective Vaccines. Vaccines (Basel) 2024; 12:863. [PMID: 39203989 PMCID: PMC11359697 DOI: 10.3390/vaccines12080863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Sexually transmitted infections (STIs) caused by bacterial pathogens Chlamydia trachomatis, Neisseria gonorrhoeae, and Treponema pallidum present significant public health challenges. These infections profoundly impact reproductive health, leading to pelvic inflammatory disease, infertility, and increased susceptibility to other infections. Prevention measures, including antibiotic treatments, are limited by the often-asymptomatic nature of these infections, the need for repetitive and continual screening of sexually active persons, antibiotic resistance for gonorrhea, and shortages of penicillin for syphilis. While vaccines exist for viral STIs like human papillomavirus (HPV) and hepatitis B virus (HBV), there are no vaccines available for bacterial STIs. This review examines the immune responses in the female genital tract to these bacterial pathogens and the implications for developing effective vaccines against bacterial STIs.
Collapse
Affiliation(s)
| | - Toni Darville
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
3
|
Mayall JR, Horvat JC, Mangan NE, Chevalier A, McCarthy H, Hampsey D, Donovan C, Brown AC, Matthews AY, de Weerd NA, de Geus ED, Starkey MR, Kim RY, Daly K, Goggins BJ, Keely S, Maltby S, Baldwin R, Foster PS, Boyle MJ, Tanwar PS, Huntington ND, Hertzog PJ, Hansbro PM. Interferon-epsilon is a novel regulator of NK cell responses in the uterus. EMBO Mol Med 2024; 16:267-293. [PMID: 38263527 PMCID: PMC10897320 DOI: 10.1038/s44321-023-00018-6] [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: 10/22/2021] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
The uterus is a unique mucosal site where immune responses are balanced to be permissive of a fetus, yet protective against infections. Regulation of natural killer (NK) cell responses in the uterus during infection is critical, yet no studies have identified uterine-specific factors that control NK cell responses in this immune-privileged site. We show that the constitutive expression of IFNε in the uterus plays a crucial role in promoting the accumulation, activation, and IFNγ production of NK cells in uterine tissue during Chlamydia infection. Uterine epithelial IFNε primes NK cell responses indirectly by increasing IL-15 production by local immune cells and directly by promoting the accumulation of a pre-pro-like NK cell progenitor population and activation of NK cells in the uterus. These findings demonstrate the unique features of this uterine-specific type I IFN and the mechanisms that underpin its major role in orchestrating innate immune cell protection against uterine infection.
Collapse
Affiliation(s)
- Jemma R Mayall
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Jay C Horvat
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Niamh E Mangan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Anne Chevalier
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Huw McCarthy
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Daniel Hampsey
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Chantal Donovan
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2000, Australia
| | - Alexandra C Brown
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Antony Y Matthews
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Nicole A de Weerd
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Eveline D de Geus
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Malcolm R Starkey
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
- Immunology and Pathology, Central Clinical School, Monash University, Clayton, VIC, 3168, Australia
| | - Richard Y Kim
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2000, Australia
| | - Katie Daly
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Bridie J Goggins
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Simon Keely
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Steven Maltby
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Rennay Baldwin
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Paul S Foster
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Michael J Boyle
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
- Immunology and Infectious Diseases Unit, John Hunter Hospital, Newcastle, NSW, 2305, Australia
| | - Pradeep S Tanwar
- Gynecology Oncology Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Nicholas D Huntington
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3168, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Philip M Hansbro
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia.
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2000, Australia.
| |
Collapse
|
4
|
Peng Y, Qiao S, Wang H, Shekhar S, Wang S, Yang J, Fan Y, Yang X. Enhancement of Macrophage Immunity against Chlamydial Infection by Natural Killer T Cells. Cells 2024; 13:133. [PMID: 38247825 PMCID: PMC10813948 DOI: 10.3390/cells13020133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Lung macrophage (LM) is vital in host defence against bacterial infections. However, the influence of other innate immune cells on its function, including the polarisation of different subpopulations, remains poorly understood. This study examined the polarisation of LM subpopulations (monocytes/undifferentiated macrophages (Mo/Mφ), interstitial macrophages (IM), and alveolar macrophages (AM)). We further assessed the effect of invariant natural killer T cells (iNKT) on LM polarisation in a protective function against Chlamydia muridarum, an obligate intracellular bacterium, and respiratory tract infection. We found a preferentially increased local Mo/Mφ and IMs with a significant shift to a type-1 macrophage (M1) phenotype and higher expression of iNOS and TNF-α. Interestingly, during the same infection, the alteration of macrophage subpopulations and the shift towards M1 was much less in iNKT KO mice. More importantly, functional testing by adoptively transferring LMs isolated from iNKT KO mice (iNKT KO-Mφ) conferred less protection than those isolated from wild-type mice (WT-Mφ). Further analyses showed significantly reduced gene expression of the JAK/STAT signalling pathway molecules in iNKT KO-Mφ. The data show an important role of iNKT in promoting LM polarisation to the M1 direction, which is functionally relevant to host defence against a human intracellular bacterial infection. The alteration of JAK/STAT signalling molecule gene expression in iNKT KO-Mφ suggests the modulating effect of iNKT is likely through the JAK/STAT pathway.
Collapse
Affiliation(s)
- Ying Peng
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Sai Qiao
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Hong Wang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Medical Microbiology, School of Medicine, Shandong University, Jinan 250100, China
| | - Sudhanshu Shekhar
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Shuhe Wang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jie Yang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Yijun Fan
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Xi Yang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
5
|
Zeng J, Yang S, Sun R, Tuo Y, Tan L, Zhang H, Zhang Y, Che X, Lu T, Zhang X, Bai H. A Pathogenic Role for FcγRI in the Immune Response against Chlamydial Respiratory Infection. Microorganisms 2022; 11:microorganisms11010039. [PMID: 36677333 PMCID: PMC9862693 DOI: 10.3390/microorganisms11010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/11/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
FcγRI is an important cell surface receptor reported to be involved in multiple immune responses, although it has not yet been extensively studied in intracellular bacterial infections. Here, using a mouse model of C. muridarum respiratory infection, we were able to determine how FcγRI regulates the host resistance against chlamydial invasion. According to our findings, the chlamydial loads and pulmonary pathology were both reduced in FcγRI deficient (Fcgr1-/-) animals. Being infected, monocytes, macrophages, neutrophils, DCs, CD4+/CD8+ T cells, and effector Th1 subsets displayed increased FcγRI expression patterns. Altered infiltration of these cells in the lungs of Fcgr1-/- mice further demonstrated the regulation of FcγRI in the immune system and identified Th1 cells and macrophages as its target cell populations. As expected, we observed that the Th1 response was augmented in Fcgr1-/- mice, while the pro-inflammatory M1 macrophage polarization was constrained. These findings might indicate FcγRI as a potential regulator for host immunity and inflammatory response during chlamydial infection.
Collapse
|
6
|
Garcinuño S, Gil-Etayo FJ, Mancebo E, López-Nevado M, Lalueza A, Díaz-Simón R, Pleguezuelo DE, Serrano M, Cabrera-Marante O, Allende LM, Paz-Artal E, Serrano A. Effective Natural Killer Cell Degranulation Is an Essential Key in COVID-19 Evolution. Int J Mol Sci 2022; 23:ijms23126577. [PMID: 35743021 PMCID: PMC9224310 DOI: 10.3390/ijms23126577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/05/2023] Open
Abstract
NK degranulation plays an important role in the cytotoxic activity of innate immunity in the clearance of intracellular infections and is an important factor in the outcome of the disease. This work has studied NK degranulation and innate immunological profiles and functionalities in COVID-19 patients and its association with the severity of the disease. A prospective observational study with 99 COVID-19 patients was conducted. Patients were grouped according to hospital requirements and severity. Innate immune cell subpopulations and functionalities were analyzed. The profile and functionality of innate immune cells differ between healthy controls and severe patients; CD56dim NK cells increased and MAIT cells and NK degranulation rates decreased in the COVID-19 subjects. Higher degranulation rates were observed in the non-severe patients and in the healthy controls compared to the severe patients. Benign forms of the disease had a higher granzymeA/granzymeB ratio than complex forms. In a multivariate analysis, the degranulation capacity resulted in a protective factor against severe forms of the disease (OR: 0.86), whereas the permanent expression of NKG2D in NKT cells was an independent risk factor (OR: 3.81; AUC: 0.84). In conclusion, a prompt and efficient degranulation functionality in the early stages of infection could be used as a tool to identify patients who will have a better evolution.
Collapse
Affiliation(s)
- Sara Garcinuño
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
| | - Francisco Javier Gil-Etayo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Esther Mancebo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Marta López-Nevado
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
| | - Antonio Lalueza
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Raquel Díaz-Simón
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Daniel Enrique Pleguezuelo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Manuel Serrano
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Oscar Cabrera-Marante
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Luis M. Allende
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Antonio Serrano
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-652-085-293
| |
Collapse
|
7
|
Zhao L, Yang X. Cross Talk Between Natural Killer T and Dendritic Cells and Its Impact on T Cell Responses in Infections. Front Immunol 2022; 13:837767. [PMID: 35185930 PMCID: PMC8850912 DOI: 10.3389/fimmu.2022.837767] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 11/16/2022] Open
Abstract
Both innate and adaptive immunity is vital for host defense against infections. Dendritic cells (DCs) are critical for initiating and modulating adaptive immunity, especially for T-cell responses. Natural killer T (NKT) cells are a small population of innate-like T cells distributed in multiple organs. Many studies have suggested that the cross-talk between these two immune cells is critical for immunobiology and host defense mechanisms. Not only can DCs influence the activation/function of NKT cells, but NKT cells can feedback on DCs also, thus modulating the phenotype and function of DCs and DC subsets. This functional feedback of NKT cells on DCs, especially the preferential promoting effect on CD8α+ and CD103+ DC subsets in lymphoid and non-lymphoid tissues, significantly impacts the systemic and local adaptive CD4 and CD8 T cell responses in infections. This review focuses on the two-way interaction between NKT cells and DCs, emphasizing the importance of NKT cell feedback on DCs in bridging innate and adaptive immune responses for host defense purposes.
Collapse
Affiliation(s)
- Lei Zhao
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xi Yang
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
8
|
Zhao L, Li J, Zhou X, Pan Q, Zhao W, Yang X, Wang H. Natural Killer Cells Regulate Pulmonary Macrophages Polarization in Host Defense Against Chlamydial Respiratory Infection. Front Cell Infect Microbiol 2022; 11:775663. [PMID: 35059323 PMCID: PMC8764407 DOI: 10.3389/fcimb.2021.775663] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
NK cells and pulmonary macrophages both are important components of innate immunity. The interaction between NK cells and pulmonary macrophages during chlamydial infection is poorly understood. In this study, we explored the effect of NK cells on regulation of pulmonary macrophage function during chlamydial respiratory infection. We found that NK depletion led to polarization of pulmonary macrophages from M1 to M2 phenotype, and it is related to reduced miR-155 expression in lung macrophage. Using adoptive transfer approach, we found that the recipients receiving lung macrophages isolated from C. muridarum-infected NK-cell-depleted mice exhibited an increased bacterial load and severe inflammation in the lung upon chlamydial challenge infection when compared with the recipients of lung macrophages from infected isotype control antibody treated mice. Herein, the effects of NK cells on macrophage polarization were examined in vitro. We found that NK cells from chlamydial-infected mice (iNK) significantly induced M1 polarization compared to that from uninfected mice (uNK). Inhibition of miR-155 expression in macrophages reduced M1 polarization induced by iNK, while miR-155 over-expression enhanced it. Furthermore, neutralization of IFN-γ in the coculture system decreased the expression of miR-155 by macrophages, and resulted in weakened M1 polarization. The data indicates that NK cells promote M1 polarization through up-regulation of miR-155 in macrophages by producing IFN-γ during chlamydial infection, and NK-regulated macrophage polarization is functionally relevant to host defense against the infection.
Collapse
Affiliation(s)
- Lei Zhao
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Li
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaoqing Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qianqian Pan
- Department of Respiratory, Laiwu Central Hospital, Jinan, China
| | - Weiming Zhao
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xi Yang
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Hong Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
9
|
Richardson S, Medhavi F, Tanner T, Lundy S, Omosun Y, Igietseme JU, Carroll D, Eko FO. Cellular Basis for the Enhanced Efficacy of the Fms-Like Tyrosine Kinase 3 Ligand (FL) Adjuvanted VCG-Based Chlamydia abortus Vaccine. Front Immunol 2021; 12:698737. [PMID: 34249004 PMCID: PMC8264281 DOI: 10.3389/fimmu.2021.698737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
Efficacious vaccines are needed to control genital chlamydial diseases in humans and the veterinary industry. We previously reported a C. abortus (Cab) vaccine comprising recombinant Vibrio cholerae ghosts (rVCG) expressing the conserved and immunogenic N-terminal region of the Cab polymorphic membrane protein D (rVCG-Pmp18.1) protein that protected mice against intravaginal challenge. In this study, we investigated the immunomodulatory effect of the hematopoietic progenitor activator cytokine, Fms-like tyrosine kinase 3-ligand (FL) when co-administered with the rVCG-Pmp18.1 vaccine as a strategy to enhance the protective efficacy and the potential mechanism of immunomodulation. Groups of female C57BL/6J mice were immunized and boosted twice intranasally (IN) with rVCG-PmpD18.1 with and without FL or purified rPmp18.1 or rVCG-gD2 (antigen control) or PBS (medium) per mouse. The results revealed that co-administration of the vaccine with FL enhanced antigen-specific cellular and humoral immune responses and protected against live Cab genital infection. Comparative analysis of immune cell phenotypes infiltrating mucosal and systemic immune inductive tissue sites following immunization revealed that co-administration of rVCG-Pmp18.1 with FL significantly enhanced the number of macrophages, dendritic and NK cells, γδ and NK T cells in the spleen (systemic) and iliac lymph nodes (ILN) draining the genital tract (mucosal) tissues compared to rVCG-Pmp18.1 alone. Furthermore, FL enhanced monocyte infiltration in the ILN, while CD19+ B cells and CD4+ T cells were enhanced in the spleen. These results indicate that the immunomodulatory effect of FL is associated with its ability to mobilize innate immune cells and subsequent activation of robust antigen-specific immune effectors in mucosal and systemic lymphoid tissues.
Collapse
Affiliation(s)
- Shakyra Richardson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Fnu Medhavi
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Tayhlor Tanner
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Stephanie Lundy
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Joseph U. Igietseme
- National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Darin Carroll
- National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
10
|
Niu W, Xu Y, Zha X, Zeng J, Qiao S, Yang S, Zhang H, Tan L, Sun L, Pang G, Liu T, Zhao H, Zheng N, Zhang Y, Bai H. IL-21/IL-21R Signaling Aggravated Respiratory Inflammation Induced by Intracellular Bacteria through Regulation of CD4 + T Cell Subset Responses. THE JOURNAL OF IMMUNOLOGY 2021; 206:1586-1596. [PMID: 33608454 DOI: 10.4049/jimmunol.2001107] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/19/2021] [Indexed: 12/16/2022]
Abstract
The IL-21/IL-21R interaction plays an important role in a variety of immune diseases; however, the roles and mechanisms in intracellular bacterial infection are not fully understood. In this study, we explored the effect of IL-21/IL-21R on chlamydial respiratory tract infection using a chlamydial respiratory infection model. The results showed that the mRNA expression of IL-21 and IL-21R was increased in Chlamydia muridarum-infected mice, which suggested that IL-21 and IL-21R were involved in host defense against C. muridarum lung infection. IL-21R-/- mice exhibited less body weight loss, a lower bacterial burden, and milder pathological changes in the lungs than wild-type (WT) mice during C. muridarum lung infection. The absolute number and activity of CD4+ T cells and the strength of Th1/Th17 responses in IL-21R-/- mice were significantly higher than those in WT mice after C. muridarum lung infection, but the Th2 response was weaker. Consistently, IL-21R-/- mice showed higher mRNA expression of Th1 transcription factors (T-bet/STAT4), IL-12p40, a Th17 transcription factor (STAT3), and IL-23. The mRNA expression of Th2 transcription factors (GATA3/STAT6), IL-4, IL-10, and TGF-β in IL-21R-/- mice was significantly lower than that in WT mice. Furthermore, the administration of recombinant mouse IL-21 aggravated chlamydial lung infection in C57BL/6 mice and reduced Th1 and Th17 responses following C. muridarum lung infection. These findings demonstrate that IL-21/IL-21R may aggravate chlamydial lung infection by inhibiting Th1 and Th17 responses.
Collapse
Affiliation(s)
- Wenhao Niu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yueyue Xu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Xiaoyu Zha
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Jiajia Zeng
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Sai Qiao
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Shuaini Yang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Hong Zhang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Lu Tan
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Lida Sun
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Gaoju Pang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Tengli Liu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Huili Zhao
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Ningbo Zheng
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yongci Zhang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Hong Bai
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| |
Collapse
|
11
|
D Helble J, N Starnbach M. T cell responses to Chlamydia. Pathog Dis 2021; 79:6164867. [PMID: 33693620 DOI: 10.1093/femspd/ftab014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/07/2021] [Indexed: 12/27/2022] Open
Abstract
Chlamydia trachomatis is the most commonly reported sexually transmitted infection in the United States. The high prevalence of infection and lack of a vaccine indicate a critical knowledge gap surrounding the host's response to infection and how to effectively generate protective immunity. The immune response to C. trachomatis is complex, with cells of the adaptive immune system playing a crucial role in bacterial clearance. Here, we discuss the CD4+ and CD8+ T cell response to Chlamydia, the importance of antigen specificity and the role of memory T cells during the recall response. Ultimately, a deeper understanding of protective immune responses is necessary to develop a vaccine that prevents the inflammatory diseases associated with Chlamydia infection.
Collapse
Affiliation(s)
- Jennifer D Helble
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
12
|
Wang X, Zhang C, Wang S, Rashu R, Thomas R, Yang J, Yang X. SND1 promotes Th1/17 immunity against chlamydial lung infection through enhancing dendritic cell function. PLoS Pathog 2021; 17:e1009295. [PMID: 33635920 PMCID: PMC7946287 DOI: 10.1371/journal.ppat.1009295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 03/10/2021] [Accepted: 01/07/2021] [Indexed: 12/31/2022] Open
Abstract
To date, no reports have linked the multifunctional protein, staphylococcal nuclease domain-containing protein 1 (SND1), to host defense against intracellular infections. In this study, we investigated the role and mechanisms of SND1, by using SND1 knockout (SND1-/-) mice, in host defense against the lung infection of Chlamydia muridarum, an obligate intracellular bacterium. Our data showed that SND1-/- mice exhibited significantly greater body weight loss, higher organism growth, and more severe pathological changes compared with wild-type mice following the infection. Further analysis showed significantly reduced Chlamydia-specific Th1/17 immune responses in SND1-/- mice after infection. Interestingly, the dendritic cells (DCs) isolated from SND1-/- mice showed lower costimulatory molecules expression and IL-12 production, but higher IL-10 production compared with those from wild-type control mice. In the DC-T cell co-culture system, DCs isolated from SND1-/- infected mice showed significantly reduced ability to promote Chlamydia-specific IFN-γ producing Th1 cells but enhanced capacity to induce CD4+T cells into Foxp3+ Treg cells. Adoptive transfer of DCs isolated from SND1-/- mice, unlike those from wild-type control mice, failed to protect the recipients against challenge infection. These findings provide in vivo evidence that SND1 plays an important role in host defense against intracellular bacterial infection, and suggest that SND1 can promote Th1/17 immunity and inhibit the expansion of Treg cells through modulation of the function of DCs.
Collapse
Affiliation(s)
- Xinting Wang
- Department of Immunology, University of Manitoba, Winnipeg, Canada
- Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Key Laboratory of Cellular and Molecular Immunology in Tianjin, Department of Biochemistry and Molecular Biology, Excellent Talent Project, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chunyan Zhang
- Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Shuhe Wang
- Department of Immunology, University of Manitoba, Winnipeg, Canada
| | | | - Rony Thomas
- Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Jie Yang
- Department of Immunology, University of Manitoba, Winnipeg, Canada
- * E-mail: (JY); (XY)
| | - Xi Yang
- Department of Immunology, University of Manitoba, Winnipeg, Canada
- * E-mail: (JY); (XY)
| |
Collapse
|
13
|
Coxiella burnetii-Infected NK Cells Release Infectious Bacteria by Degranulation. Infect Immun 2020; 88:IAI.00172-20. [PMID: 32817330 DOI: 10.1128/iai.00172-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/10/2020] [Indexed: 01/24/2023] Open
Abstract
Natural killer (NK) cells are critically involved in the early immune response against various intracellular pathogens, including Coxiella burnetii and Chlamydia psittaci Chlamydia-infected NK cells functionally mature, induce cellular immunity, and protect themselves by killing the bacteria in secreted granules. Here, we report that infected NK cells do not allow intracellular multiday growth of Coxiella, as is usually observed in other host cell types. C. burnetii-infected NK cells display maturation and gamma interferon (IFN-γ) secretion, as well as the release of Coxiella-containing lytic granules. Thus, NK cells possess a potent program to restrain and expel different types of invading bacteria via degranulation. Strikingly, though, in contrast to Chlamydia, expulsed Coxiella organisms largely retain their infectivity and, hence, escape the cell-autonomous self-defense mechanism in NK cells.
Collapse
|
14
|
Li J, Zheng J, Wang M, Zhang Y, Jiang Y, Zhang X, Guo P. [Inhibition of CD96 enhances interferon-γ secretion by natural killer cells to alleviate lung injury in mice with pulmonary Chlamydia muridarum infection]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:930-935. [PMID: 32895152 DOI: 10.12122/j.issn.1673-4254.2020.07.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To assess the effect of neutralizing CD96 on natural killer (NK) cell functions in mice with pulmonary Chlamydia muridarum infection and explore the possible mechanism. METHODS Male BALB/c mice were randomly divided into infection group (Cm group), anti-CD96 treatment group (anti-CD96 group) and control group (n=5). In the former two groups, C. muridarum was inoculated via intranasal administration to establish mouse models of pulmonary C. muridarum infection, and the mice in the control group received intranasal administration of the inhalation buffer. In anti-CD96 group, the mice were injected with anti-CD96 antibody intraperitoneally at the dose of 250 μg every 3 days after the infection; the mice in Cm group received intraperitoneal injections of saline. The body weight of the mice was recorded daily. The mice were sacrificed 5 days after C. muridarum infection, and CD96 expression was detected by quantitative real-time PCR and Western blotting. HE staining and pathological scores were used to evaluate pneumonia of the mice. The inclusion body forming units (IFUs) were detected in the lung tissue homogenates to assess lung tissue chlamydia load. Flow cytometry and ELISA were used to assess the capacity of the lung NK cells to produce interferon-γ (IFN-γ) and regulate macrophages and Th1 cells. RESULTS C. muridarum infection inhibited CD96 expression in NK cells of the mice. Compared with those in Cm group, the mice in antiCD96 mice showed significantly milder lung inflammation (P < 0.05) and reduced chlamydia load in the lung tissue (P < 0.05). Neutralizing CD96 with anti-CD96 significantly enhanced IFN-γ secretion by the NK cells (P < 0.05) and augmented the immunoregulatory effect of the NK cells shown by enhanced responses of the lung macrophages (P < 0.05) and Th1 cells (P < 0.05). CONCLUSIONS Inhibition of CD96 alleviates pneumonia in C. muridarum-infected mice possibly by enhancing IFN-γ secretion by NK cells and augmenting the immunoregulatory effect of the NK cells on innate and adaptive immunity.
Collapse
Affiliation(s)
- Jing Li
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Jing Zheng
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Minda Wang
- Anhui Provincial Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
| | - Yan Zhang
- Anhui Provincial Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
| | - Yifan Jiang
- Anhui Provincial Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
| | - Xiaofeng Zhang
- Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Pu Guo
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
15
|
Zhao L, Wang H, Thomas R, Gao X, Bai H, Shekhar S, Wang S, Yang J, Zhao W, Yang X. NK cells modulate T cell responses via interaction with dendritic cells in Chlamydophila pneumoniae infection. Cell Immunol 2020; 353:104132. [DOI: 10.1016/j.cellimm.2020.104132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 12/27/2022]
|
16
|
Wang H, Li J, Dong X, Zhou X, Zhao L, Wang X, Rashu R, Zhao W, Yang X. NK Cells Contribute to Protective Memory T Cell Mediated Immunity to Chlamydia muridarum Infection. Front Cell Infect Microbiol 2020; 10:296. [PMID: 32626664 PMCID: PMC7311576 DOI: 10.3389/fcimb.2020.00296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/19/2020] [Indexed: 12/23/2022] Open
Abstract
We previously reported that NK cells can promote type 1 T cell immune response that is essential for protection to a primary infection of Chlamydia muridarum. In this study, we have investigated the contribution of NK cells to memory T cells associated immunity during chlamydial infection. We have found that NK cell depletion led to impaired production of IFN-γ by memory T cells upon re-stimulation with chlamydial antigens in vitro. Mice with depleted NK cells also exhibited reduced type 1 T cell recall responses, with increased production of IL-4 from CD4+ T cells and a lower level of Chlamydia-specific IgG2a titers compared to control mice. In addition, Tregs response was significantly increased in mice with NK cell depletion. Moreover, NK cell-depleted mice showed an increased bacterial loads and more severe inflammatory pathological changes than control mice. These findings indicate that NK cells contribute to protective memory T cell associated immunity to chlamydial re-infection through modulating the cytokine pattern of T cell and inhibition of Tregs response.
Collapse
Affiliation(s)
- Hong Wang
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jing Li
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaojing Dong
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xaoqing Zhou
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Lei Zhao
- Institute of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Wang
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Rasheduzzaman Rashu
- Department of Immunology and Department of Medical Microbiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Weiming Zhao
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xi Yang
- Department of Immunology and Department of Medical Microbiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
17
|
Effect of Time of Day of Infection on Chlamydia Infectivity and Pathogenesis. Sci Rep 2019; 9:11405. [PMID: 31388084 PMCID: PMC6684580 DOI: 10.1038/s41598-019-47878-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 07/09/2019] [Indexed: 01/11/2023] Open
Abstract
Genital chlamydia infection in women causes complications such as pelvic inflammatory disease and tubal factor infertility, but it is unclear why some women are more susceptible than others. Possible factors, such as time of day of chlamydia infection on chlamydial pathogenesis has not been determined. We hypothesised that infections during the day, will cause increased complications compared to infections at night. Mice placed under normal 12:12 light: dark (LD) cycle were infected intravaginally with Chlamydia muridarum either at zeitgeber time 3, ZT3 and ZT15. Infectivity was monitored by periodic vaginal swabs and chlamydiae isolation. Blood and vaginal washes were collected for host immunologic response assessments. The reproductive tracts of the mice were examined histopathologically, and fertility was determined by embryo enumeration after mating. Mice infected at ZT3 shed significantly more C. muridarum than mice infected at ZT15. This correlated with the increased genital tract pathology observed in mice infected at ZT3. Mice infected at ZT3 were less fertile than mice infected at ZT15. The results suggest that the time of day of infection influences chlamydial pathogenesis, it indicates a possible association between complications from chlamydia infection and host circadian clock, which may lead to a better understanding of chlamydial pathogenesis.
Collapse
|
18
|
The Important Role of Dendritic Cell (DC) in iNKT-Mediated Modulation of NK Cell Function in Chlamydia pneumoniae Lung Infection. Mediators Inflamm 2019; 2019:4742634. [PMID: 31236064 PMCID: PMC6545808 DOI: 10.1155/2019/4742634] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/29/2019] [Accepted: 04/15/2019] [Indexed: 01/30/2023] Open
Abstract
Chlamydia pneumoniae (Cpn) infection causes multiple acute and chronic human diseases. The role of DCs in host defense against Cpn infection has been well documented. The same is true for invariant natural killer T (iNKT) cells and NK cells, but the interaction among cells is largely unknown. In this study, we investigated the influence and mechanism of iNKT cell on the differentiation and function of NK cell in Cpn lung infection and the role played by DCs in this process. We found that expansion of IFN-γ-producing NK cells quickly happened after the infection, but this response was altered in iNKT knockout (KO) mice. The expression of activation markers and the production of IFN-γ by different NK subsets were significantly lower in KO mice than wild-type (WT) mice. Using in vitro DC-NK coculture and in vivo adoptive transfer approaches, we further examined the role of DCs in iNKT-mediated modulation of NK cell function. We found that NK cells expressed lower levels of activation markers and produced less IFN-γ when they were cocultured with DCs from KO mice than WT mice. More importantly, we found that the adoptive transfer of DCs from the KO mice induced less NK cell activation and IFN-γ production. The results provided evidence on the modulating effect of iNKT cell on NK cell function, particularly the critical role of DCs in this modulation process. The finding suggests the complexity of cellular interactions in Cpn lung infection, which should be considered in designing preventive and therapeutic approaches for diseases and infections.
Collapse
|
19
|
Dzopalić T, Božić-Nedeljković B, Jurišić V. Function of innate lymphoid cells in the immune-related disorders. Hum Cell 2019; 32:231-239. [DOI: 10.1007/s13577-019-00257-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/05/2019] [Indexed: 12/12/2022]
|
20
|
Qiao S, Zhang H, Zha X, Niu W, Liang J, Pang G, Tang Y, Liu T, Zhao H, Wang Y, Bai H. Endogenous IL-17A mediated neutrophil infiltration by promoting chemokines expression during chlamydial lung infection. Microb Pathog 2019; 129:106-111. [PMID: 30703475 DOI: 10.1016/j.micpath.2019.01.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 01/16/2019] [Accepted: 01/25/2019] [Indexed: 01/04/2023]
Abstract
Chlamydia is an obligate intracellular bacteria, which can infect cervix, urethra, conjunctiva, joints, lungs and so on. Neutrophils are important in host protection against microbial invasion during the early phase of infection. Here, to investigate the mechanism of IL-17A in recruiting neutrophils during Chlamydia muridarum (Cm) lung infection, we introduced IL-17A antibodies and IL-17-/- mice to confirm the effect of IL-17A on influencing neutrophil attractants expressions. From the analysis of the data, we found that showed that Cm infection could upregulate the expression of neutrophil-related chemokines such as KC, MIP-2 and IL-6, as well as adhesion molecules including ICAM-1 and VCAM-1. With blocking endogenous IL-17A, the upregulated MIP-2 and IL-6 were decreased, which induced less neutrophil recruitment in lung. Comparing to WT mice, IL-17-/- mice showed decreased infiltration of neutrophils in lung during the early phase of Cm infection, which were accordant with decreased chemokines, such as KC, MIP-2 and IL-6 expression. Whereas, the expression of adhesion molecules including ICAM and VCAM-1 in lungs were significantly increased in IL-17-/- mice comparing to WT mice during Cm lung infection. The results demonstrated that IL-17A influenced neutrophil infiltration by affecting expression of chemokines and adhesion molecules during the early phase of chlamydial lung infection.
Collapse
Affiliation(s)
- Sai Qiao
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Hong Zhang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Xiaoyu Zha
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Wenhao Niu
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Juyou Liang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Gaoju Pang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Yingying Tang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Tengli Liu
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Huili Zhao
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Yue Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China
| | - Hong Bai
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, Tianjin, PR China.
| |
Collapse
|
21
|
Chauhan P, Saha B. Metabolic regulation of infection and inflammation. Cytokine 2018; 112:1-11. [PMID: 30472107 DOI: 10.1016/j.cyto.2018.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/30/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022]
Abstract
Immunometabolic framework provides a way to understand the immune regulation via cell intrinsic metabolic fluxes and metabolites during infections, tumors, and inflammatory disorders. During these diseases, the immune cells are activated requiring more energy and moderating their metabolic functions. The two categories of metabolic alterations are therefore causally associated with energy derivation and cellular functions. Pathogens, tumors and inflammation target energy metabolism, primarily glucose uptake, glucose catabolism, gluconeogenesis for continuing lipid metabolism through mainstream pathways such as glycolysis, tricarboxylic acid cycle, mitochondrial respiration and pentose phosphate pathway. Many biosynthetic pathways such as those of cholesterol, ceramide, sphingolipids, and fatty acids are altered explaining the metabolic interface in molecular pathogenesis in various infectious and non-infectious inflammatory diseases. The emerging immune-metabolic framework also identifies the key regulatory elements such as metabolites, signalling intermediates and transcription factors. These regulatory elements play key roles in deciding the fate of an infection, tumor or autoimmune diseases. The original research articles and the review articles in this Special issue of Cytokine on "Infection, Inflammation and Immunometabolomes" highlight these aspects of metabolic reprogramming and the role of some 'metabolomic regulators' in controlling the outcome of infectious and non-infectious diseases. In this Editorial, we introduce the readers to these articles discussing the elements in immune-metabolic framework.
Collapse
Affiliation(s)
- Prashant Chauhan
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Bhaskar Saha
- Trident Academy of Creative Technology, Bhubaneswar 750019, India
| |
Collapse
|
22
|
Walwyn-Brown K, Guldevall K, Saeed M, Pende D, Önfelt B, MacDonald AS, Davis DM. Human NK Cells Lyse Th2-Polarizing Dendritic Cells via NKp30 and DNAM-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2028-2041. [PMID: 30120122 PMCID: PMC6139540 DOI: 10.4049/jimmunol.1800475] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/23/2018] [Indexed: 01/08/2023]
Abstract
Cross-talk between NK cells and dendritic cells (DCs) is important in Th1 immune responses, including antitumor immunity and responses to infections. DCs also play a crucial role in polarizing Th2 immunity, but the impact of NK cell-DC interactions in this context remains unknown. In this study, we stimulated human monocyte-derived DCs in vitro with different pathogen-associated molecules: LPS or polyinosinic-polycytidylic acid, which polarize a Th1 response, or soluble egg Ag from the helminth worm Schistosoma mansoni, a potent Th2-inducing Ag. Th2-polarizing DCs were functionally distinguishable from Th1-polarizing DCs, and both showed distinct morphology and dynamics from immature DCs. We then assessed the outcome of autologous NK cells interacting with these differently stimulated DCs. Confocal microscopy showed polarization of the NK cell microtubule organizing center and accumulation of LFA-1 at contacts between NK cells and immature or Th2-polarizing DCs but not Th1-polarizing DCs, indicative of the assembly of an activating immune synapse. Autologous NK cells lysed immature DCs but not DCs treated with LPS or polyinosinic-polycytidylic acid as reported previously. In this study, we demonstrated that NK cells also degranulated in the presence of Th2-polarizing DCs. Moreover, time-lapse live-cell microscopy showed that DCs that had internalized fluorescently labeled soluble egg Ag were efficiently lysed. Ab blockade of NK cell-activating receptors NKp30 or DNAM-1 abrogated NK cell lysis of Th2-polarizing DCs. Thus, these data indicate a previously unrecognized role of NK cell cytotoxicity and NK cell-activating receptors NKp30 and DNAM-1 in restricting the pool of DCs involved in Th2 immune responses.
Collapse
Affiliation(s)
- Katherine Walwyn-Brown
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Karolin Guldevall
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, SE-106 91 Stockholm, Sweden
| | - Mezida Saeed
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Daniela Pende
- Laboratorio Immunologia, Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Policlinico San Martino, 16132 Genova, Italy; and
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, SE-106 91 Stockholm, Sweden
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Andrew S MacDonald
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom;
| |
Collapse
|
23
|
Immunometabolism of T cells and NK cells: metabolic control of effector and regulatory function. Inflamm Res 2018; 67:813-828. [PMID: 30066126 DOI: 10.1007/s00011-018-1174-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 01/10/2023] Open
Abstract
Metabolic flux can dictate cell fate, including immune cell effector and regulatory function. The metabolic regulation of cell function is well characterized with respect to effector, memory, and regulatory T cells. This knowledge may allow for manipulation of T cell metabolic pathways that set the stage for more effective T cell therapy. Natural Killer (NK) and T-lymphocytes have complementary roles in the defense against pathogens. However, studies of NK cell metabolism are only beginning to emerge and there is comparatively little knowledge on the metabolic regulation of NK-cell activation and effector function. Given their common lymphoid lineage, effector functions and cellular memory potential our current knowledge on T cell metabolism could inform investigation of metabolic reprogramming in NK cells. In this review, we compare the current knowledge of metabolic regulation in T cell and NK cell development, activation, effector and memory function. Commonalties in glucose transport, hypoxia-inducible factors and mTOR highlight metabolic control points in both cells types. Contrasting the glycolytic and oxidative nodes of metabolic regulation in T cells versus NK cells may provide insight into the contribution of specific immune responses to disease and promote the development of immunotherapeutic approaches targeting both innate and adaptive immune responses.
Collapse
|
24
|
Lai HC, Chang CJ, Lin CS, Wu TR, Hsu YJ, Wu TS, Lu JJ, Martel J, Ojcius DM, Ku CL, Young JD, Lu CC. NK Cell-Derived IFN-γ Protects against Nontuberculous Mycobacterial Lung Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:1478-1490. [PMID: 30061197 DOI: 10.4049/jimmunol.1800123] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/05/2018] [Indexed: 01/09/2023]
Abstract
In developed countries, pulmonary nontuberculous mycobacteria (NTM) infections are more prevalent than Mycobacterium tuberculosis infections. Given the differences in the pathogenesis of NTM and M. tuberculosis infections, separate studies are needed to investigate the pathological effects of NTM pathogens. Our previous study showed that anti-IFN-γ autoantibodies are detected in NTM-infected patients. However, the role of NK cells and especially NK cell-derived IFN-γ in this context has not been studied in detail. In the current study, we show that NK1.1 cell depletion increases bacterial load and mortality in a mouse model of pulmonary NTM infection. NK1.1 cell depletion exacerbates NTM-induced pathogenesis by reducing macrophage phagocytosis, dendritic cell development, cytokine production, and lung granuloma formation. Similar pathological phenomena are observed in IFN-γ-deficient (IFN-γ-/-) mice following NTM infection, and adoptive transfer of wild-type NK cells into IFN-γ-/- mice considerably reduces NTM pathogenesis. Injection of rIFN-γ also prevents NTM-induced pathogenesis in IFN-γ-/- mice. We observed that NK cells represent the main producers of IFN-γ in the lungs and production starts as soon as 1 d postinfection. Accordingly, injection of rIFN-γ into IFN-γ-/- mice 1 d (but not 2 wk) postinfection significantly improves immunity against NTM infection. NK cells also stimulate mycobacterial killing and IL-12 production by macrophages. Our results therefore indicate that IFN-γ production by NK cells plays an important role in activating and enhancing innate and adaptive immune responses at early stages of pulmonary NTM infection.
Collapse
Affiliation(s)
- Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan 33303, Taiwan.,Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan 33303, Taiwan
| | - Chih-Jung Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Chuan-Sheng Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Tsung-Ru Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Ya-Jing Hsu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Ting-Shu Wu
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - Jang-Jih Lu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103
| | - Cheng-Lung Ku
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY 10021; and
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, Xinzhuang, New Taipei City 24205, Taiwan
| |
Collapse
|
25
|
V γ4+ T Cells: A Novel IL-17-Producing γδ T Subsets during the Early Phase of Chlamydial Airway Infection in Mice. Mediators Inflamm 2018; 2018:6265746. [PMID: 29670466 PMCID: PMC5835244 DOI: 10.1155/2018/6265746] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Our previous studies showed that γδ T cells provided immune protection against Chlamydial muridarum (Cm), an obligate intracellular strain of chlamydia trachomatis, lung infection by producing abundant IL-17. In this study, we investigated the proliferation and activation of lung γδ T cell subsets, specifically the IL-17 and IFNγ production by them following Cm lung infection. Our results found that five γδ T cell subsets, Vγ1+ T, Vγ2+ T, Vγ4+ T, Vγ5+ T, and Vγ6+ T, expressed in lungs of naïve mice, while Cm lung infection mainly induced the proliferation and activation of Vγ4+ T cells at day 3 p.i., following Vγ1+ T cells at day 7 p.i. Cytokine detection showed that Cm lung infection induced IFNγ secretion firstly by Vγ4+ T cells at very early stage (day 3) and changed to Vγ1+ T cells at midstage (day 7). Furthermore, Vγ4+ T cell is the main γδ T cell subset that secretes IL-17 at the very early stage of Cm lung infection and Vγ1+ T cell did not secrete IL-17 during the infection. These findings provide in vivo evidence that Vγ4+T cells are the major IL-17 and IFNγ-producing γδ T cell subsets at the early period of Cm lung infection.
Collapse
|
26
|
Alamri A, Soussi Gounni A, Kung SKP. View Point: Semaphorin-3E: An Emerging Modulator of Natural Killer Cell Functions? Int J Mol Sci 2017; 18:E2337. [PMID: 29113093 PMCID: PMC5713306 DOI: 10.3390/ijms18112337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/21/2017] [Accepted: 11/01/2017] [Indexed: 12/29/2022] Open
Abstract
Semaphorin-3E (Sema-3E) is a member of a large family of proteins originally identified as axon guidance cues in neural development. It is expressed in different cell types, such as immune cells, cancer cells, neural cells, and epithelial cells. Subsequently, dys-regulation of Sema-3E expression has been reported in various biological processes that range from cancers to autoimmune and allergic diseases. Recent work in our laboratories revealed a critical immunoregulatory role of Sema-3E in experimental allergic asthma. We further speculate possible immune modulatory function(s) of Sema-3E on natural killer (NK) cells.
Collapse
Affiliation(s)
- Abdulaziz Alamri
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| | - Abdelilah Soussi Gounni
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| | - Sam K P Kung
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| |
Collapse
|
27
|
Lemire P, Galbas T, Thibodeau J, Segura M. Natural Killer Cell Functions during the Innate Immune Response to Pathogenic Streptococci. Front Microbiol 2017; 8:1196. [PMID: 28706510 PMCID: PMC5489694 DOI: 10.3389/fmicb.2017.01196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/12/2017] [Indexed: 01/21/2023] Open
Abstract
Dendritic cells (DCs) and NK cells play a crucial role in the first phase of host defense against infections. Group B Streptococcus (GBS) and Streptococcus suis are encapsulated streptococci causing severe systemic inflammation, leading to septicemia and meningitis. Yet, the involvement of NK cells in the innate immune response to encapsulated bacterial infection is poorly characterized. Here, it was observed that these two streptococcal species rapidly induce the release of IFN-γ and that NK cells are the major cell type responsible for this production during the acute phase of the infection. Albeit S. suis capacity to activate NK cells was lower than that of GBS, these cells partially contribute to S. suis systemic infection; mainly through amplification of the inflammatory loop. In contrast, such a role was not observed during GBS systemic infection. IFN-γ release by NK cells required the presence of DCs, which in turn had a synergistic effect on DC cytokine production. These responses were mainly mediated by direct DC-NK cell contact and partially dependent on soluble factors. Though IL-12 and LFA-1 were shown to be critical in S. suis-mediated activation of the DC-NK cell crosstalk, different or redundant molecular pathways modulate DC-NK interactions during GBS infection. The bacterial capsular polysaccharides also differently modulated NK cell activation. Together, these results demonstrated a role of NK cells in the innate immune response against encapsulated streptococcal infections; yet the molecular pathways governing NK activation seem to differ upon the pathogen and should not be generalized when studying bacterial infections.
Collapse
Affiliation(s)
- Paul Lemire
- Laboratory of Immunology of the Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of MontrealSt-Hyacinthe, QC, Canada
| | - Tristan Galbas
- Laboratory of Molecular Immunology, Faculty of Medicine, University of MontrealMontreal, QC, Canada
| | - Jacques Thibodeau
- Laboratory of Molecular Immunology, Faculty of Medicine, University of MontrealMontreal, QC, Canada
| | - Mariela Segura
- Laboratory of Immunology of the Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of MontrealSt-Hyacinthe, QC, Canada
| |
Collapse
|
28
|
NK-DC Crosstalk in Immunity to Microbial Infection. J Immunol Res 2016; 2016:6374379. [PMID: 28097157 PMCID: PMC5206438 DOI: 10.1155/2016/6374379] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/13/2016] [Indexed: 12/11/2022] Open
Abstract
The interaction between natural killer (NK) cell and dendritic cell (DC), two important cellular components of innate immunity, started to be elucidated in the last years. The crosstalk between NK cells and DC, which leads to NK cell activation, DC maturation, or apoptosis, involves cell-cell contacts and soluble factors. This interaction either in the periphery or in the secondary lymphoid organs acts as a key player linking innate and adaptive immune responses to microbial stimuli. This review focuses on the mechanisms of NK-DC interaction and their relevance in antimicrobial responses. We specifically aim to emphasize the ability of various microbial infections to differently influence NK-DC crosstalk thereby contributing to distinct adaptive immune response.
Collapse
|
29
|
Ji Y, Wen J, Veldhuisen B, Haer-Wigman L, Wang Z, Lodén-van Straaten M, Wei L, Luo G, Fu Y, van der Schoot CE. Validation of the multiplex ligation-dependent probe amplification assay and its application on the distribution study of the major alleles of 17 blood group systems in Chinese donors from Guangzhou. Transfusion 2016; 57:423-432. [DOI: 10.1111/trf.13940] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/26/2016] [Accepted: 10/02/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Yanli Ji
- Institute of Clinical Blood Transfusion, Guangzhou Blood Center; Guangzhou People's Republic of China
- Sanquin Research and Landsteiner Laboratory; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| | - Jizhi Wen
- Institute of Clinical Blood Transfusion, Guangzhou Blood Center; Guangzhou People's Republic of China
| | - Barbera Veldhuisen
- Sanquin Research and Landsteiner Laboratory; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| | - Lonneke Haer-Wigman
- Sanquin Research and Landsteiner Laboratory; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| | - Zhen Wang
- Institute of Clinical Blood Transfusion, Guangzhou Blood Center; Guangzhou People's Republic of China
| | | | - Ling Wei
- Institute of Clinical Blood Transfusion, Guangzhou Blood Center; Guangzhou People's Republic of China
| | - Guangping Luo
- Institute of Clinical Blood Transfusion, Guangzhou Blood Center; Guangzhou People's Republic of China
| | - Yongshui Fu
- Institute of Clinical Blood Transfusion, Guangzhou Blood Center; Guangzhou People's Republic of China
| | - C. Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
30
|
Fernández-Ruiz M, Silva J, López-Medrano F, Allende L, San Juan R, Cambra F, Justo I, Paz-Artal E, Jiménez C, Aguado J. Post-transplant monitoring of NK cell counts as a simple approach to predict the occurrence of opportunistic infection in liver transplant recipients. Transpl Infect Dis 2016; 18:552-65. [DOI: 10.1111/tid.12564] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/03/2016] [Accepted: 03/29/2016] [Indexed: 01/02/2023]
Affiliation(s)
- M. Fernández-Ruiz
- Unit of Infectious Diseases; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - J.T. Silva
- Unit of Infectious Diseases; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - F. López-Medrano
- Unit of Infectious Diseases; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - L.M. Allende
- Department of Immunology; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - R. San Juan
- Unit of Infectious Diseases; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - F. Cambra
- Department of Abdominal Organ Transplantation and General and Digestive Surgery; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - I. Justo
- Department of Abdominal Organ Transplantation and General and Digestive Surgery; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - E. Paz-Artal
- Department of Immunology; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - C. Jiménez
- Department of Abdominal Organ Transplantation and General and Digestive Surgery; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| | - J.M. Aguado
- Unit of Infectious Diseases; Hospital Universitario “12 de Octubre”; Instituto de Investigación Hospital “12 de Octubre” (i+12); School of Medicine; Universidad Complutense; Madrid Spain
| |
Collapse
|
31
|
Genital Chlamydia trachomatis: understanding the roles of innate and adaptive immunity in vaccine research. Clin Microbiol Rev 2016; 27:346-70. [PMID: 24696438 DOI: 10.1128/cmr.00105-13] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted disease worldwide, and despite significant advances in chlamydial research, a prophylactic vaccine has yet to be developed. This Gram-negative obligate intracellular bacterium, which often causes asymptomatic infection, may cause pelvic inflammatory disease (PID), ectopic pregnancies, scarring of the fallopian tubes, miscarriage, and infertility when left untreated. In the genital tract, Chlamydia trachomatis infects primarily epithelial cells and requires Th1 immunity for optimal clearance. This review first focuses on the immune cells important in a chlamydial infection. Second, we summarize the research and challenges associated with developing a chlamydial vaccine that elicits a protective Th1-mediated immune response without inducing adverse immunopathologies.
Collapse
|
32
|
Wei S, Sun A, Ding S, Meng Q, Wang H, Duan S, Tian J, Chen Y, Liu X, Li Y, Liao Y. Mur (MNS 10) screening with a novel loop-mediated isothermal amplification assay in Zhongshan, China. Transfus Med 2016; 26:215-9. [DOI: 10.1111/tme.12305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/10/2016] [Accepted: 03/26/2016] [Indexed: 11/30/2022]
Affiliation(s)
- S.S. Wei
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - A.N. Sun
- Zhongshan Blood Center; Zhongshan China
| | - S.H. Ding
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - Q.L. Meng
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - H.M. Wang
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - S.B. Duan
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - J.J. Tian
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - Y.Z. Chen
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - X.M. Liu
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - Y. Li
- CAS Key Lab of Bio-Medical Diagnostics; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences; Suzhou China
| | - Y.T. Liao
- Zhongshan Blood Center; Zhongshan China
| |
Collapse
|
33
|
Li J, Dong X, Zhao L, Wang X, Wang Y, Yang X, Wang H, Zhao W. Natural killer cells regulate Th1/Treg and Th17/Treg balance in chlamydial lung infection. J Cell Mol Med 2016; 20:1339-51. [PMID: 27028780 PMCID: PMC4929289 DOI: 10.1111/jcmm.12821] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/30/2016] [Indexed: 12/20/2022] Open
Abstract
Natural killer (NK) cell is an important component in innate immunity, playing a critical role in bridging innate and adaptive immunity by modulating the function of other immune cells including T cells. In this study, we focused on the role of NK cells in regulating Th1/Treg and Th17/Treg balance during chlamydial lung infection. We found that NK cell‐depleted mice showed decreased Th1 and Th17 cells, which was correlated with reduced interferon‐γ, interleukin (IL)‐12, IL‐17 and IL‐22 production as well as T‐bet and receptor‐related orphan receptor gamma t expression compared with mice treated with the isotype control antibody. In contrast, NK cell depletion significantly increased Treg in cell number and related transcription factor (Foxp3) expression. The opposite trends of changes of Th1/Th17 and Treg led to significant reduction in the Th1/Treg and Th17/Treg ratios. The data implicate that NK cells play an important role in host defence against chlamydial lung infection, mainly through maintaining Th1/Treg and Th17/Treg balance.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xiaojing Dong
- Department of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Lei Zhao
- Institute of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiao Wang
- Department of Pathology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Yan Wang
- Department of Pathology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xi Yang
- Department of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China.,Department of Immunology and Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Hong Wang
- Department of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Weiming Zhao
- Department of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| |
Collapse
|
34
|
Shekhar S, Peng Y, Gao X, Joyee AG, Wang S, Bai H, Zhao L, Yang J, Yang X. NK cells modulate the lung dendritic cell-mediated Th1/Th17 immunity during intracellular bacterial infection. Eur J Immunol 2015. [PMID: 26222048 DOI: 10.1002/eji.201445390] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The impact of the interaction between NK cells and lung dendritic cells (LDCs) on the outcome of respiratory infections is poorly understood. In this study, we investigated the effect and mechanism of NK cells on the function of LDCs during intracellular bacterial lung infection of Chlamydia muridarum in mice. We found that the naive mice receiving LDCs from C. muridarum-infected NK-cell-depleted mice (NK-LDCs) showed more serious body weight loss, bacterial burden, and pathology upon chlamydial challenge when compared with the recipients of LDCs from infected sham-treated mice (NK+LDCs). Cytokine analysis of the local tissues of the former compared with the latter exhibited lower levels of Th1 (IFN-γ) and Th17 (IL-17), but higher levels of Th2 (IL-4), cytokines. Consistently, NK-LDCs were less efficient in directing C. muridarum-specific Th1 and Th17 responses than NK+LDCs when cocultured with CD4(+) T cells. In NK cell/LDC coculture experiments, the blockade of NKG2D receptor reduced the production of IL-12p70, IL-6, and IL-23 by LDCs. The neutralization of IFN-γ in the culture decreased the production of IL-12p70 by LDCs, whereas the blockade of TNF-α resulted in diminished IL-6 production. Our findings demonstrate that NK cells modulate LDC function to elicit Th1/Th17 immunity during intracellular bacterial infection.
Collapse
Affiliation(s)
- Sudhanshu Shekhar
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ying Peng
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xiaoling Gao
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Antony G Joyee
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shuhe Wang
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hong Bai
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lei Zhao
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jie Yang
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xi Yang
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
35
|
Chang MC, Chen YL, Chiang YC, Chen TC, Tang YC, Chen CA, Sun WZ, Cheng WF. Mesothelin-specific cell-based vaccine generates antigen-specific immunity and potent antitumor effects by combining with IL-12 immunomodulator. Gene Ther 2015; 23:38-49. [PMID: 26262583 DOI: 10.1038/gt.2015.85] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 06/12/2015] [Accepted: 06/23/2015] [Indexed: 12/30/2022]
Abstract
Ovarian cancer is a gynecologic malignancy with a high mortality rate. In the present study, we developed a novel cell-based vaccine, Meso-VAX, to generate mesothelin antigen-specific immune responses and immunotherapy against ovarian cancer. Mesothelin, a secreted protein anchored at the cell membrane, has recently been identified as a potential new tumor antigen for ovarian cancer. In this study, mice vaccinated with Meso-VAX and adeno-associated virus (AAV)-IL-12 exhibited dramatic increases in the number of mesothelin-specific CD4(+) helper and CD8(+) cytotoxic T-cell precursors, higher titers of anti-mesothelin Abs and in vitro tumor killing activity, and all of these mice were tumor-free after 60 days of tumor challenge. In addition, a significant reduction in peritoneal tumors and longer survival were noted in the mice vaccinated with Meso-VAX combined with AAV-IL-12. CD4(+) helper and CD8(+) cytotoxic T lymphocytes were essential for the antitumor effect generated by Meso-VAX combined with AAV-IL-12. The post-vaccination sera of the mice vaccinated with Meso-VAX and AAV-IL-12 also showed mesothelin-specific complement-dependent cell-mediated cytotoxicity. Our results suggest that a Meso-VAX cell-based vaccine combined with AAV-IL-12 can generate antigen-specific immunological responses and antitumor effects on ovarian cancer.
Collapse
Affiliation(s)
- M-C Chang
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Anesthesiology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Y-L Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan
| | - Y-C Chiang
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - T-C Chen
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Y-C Tang
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - C-A Chen
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - W-Z Sun
- Department of Anesthesiology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - W-F Cheng
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Oncology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
36
|
Mo Z, Li H, Huang L, Jiao W. Prevalence and specificity of RBC alloantibodies in the general hospitalised population in Guangxi. Transfus Med 2015; 25:313-9. [PMID: 26135712 DOI: 10.1111/tme.12222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 05/21/2015] [Accepted: 06/11/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Z. Mo
- Department of Blood Transfusion; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning China
| | - H. Li
- Department of Blood Transfusion; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning China
| | - L. Huang
- Department of Clinical Laboratory; First Affiliated Hospital of Guangxi Medical University; Nanning China
| | - W. Jiao
- Department of Blood Transfusion; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning China
| |
Collapse
|
37
|
Eberl G, Colonna M, Di Santo JP, McKenzie ANJ. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science 2015; 348:aaa6566. [PMID: 25999512 DOI: 10.1126/science.aaa6566] [Citation(s) in RCA: 646] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Innate lymphoid cells (ILCs) are a growing family of immune cells that mirror the phenotypes and functions of T cells. However, in contrast to T cells, ILCs do not express acquired antigen receptors or undergo clonal selection and expansion when stimulated. Instead, ILCs react promptly to signals from infected or injured tissues and produce an array of secreted proteins termed cytokines that direct the developing immune response into one that is adapted to the original insult. The complex cross-talk between microenvironment, ILCs, and adaptive immunity remains to be fully deciphered. Only by understanding these complex regulatory networks can the power of ILCs be controlled or unleashed in order to regulate or enhance immune responses in disease prevention and therapy.
Collapse
Affiliation(s)
- Gérard Eberl
- Institut Pasteur, Microenvironment and Immunity Unit, 75724 Paris, France.
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, INSERM U668, 75724 Paris, France
| | - Andrew N J McKenzie
- Medical Research Council (MRC) Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| |
Collapse
|
38
|
Langers I, Renoux V, Reschner A, Touzé A, Coursaget P, Boniver J, Koch J, Delvenne P, Jacobs N. Natural killer and dendritic cells collaborate in the immune response induced by the vaccine against uterine cervical cancer. Eur J Immunol 2014; 44:3585-95. [PMID: 25229656 DOI: 10.1002/eji.201444594] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 08/14/2014] [Accepted: 09/11/2014] [Indexed: 11/09/2022]
Abstract
Virus-like particles (VLPs) of human papillomavirus (HPV) are used as a vaccine against HPV-induced cancer, and recently we have shown that these VLPs are able to activate natural killer (NK) cells. Since NK cells collaborate with dendritic cells (DCs) to induce an immune response against viral infections and tumors, we studied the impact of this crosstalk in the context of HPV vaccination. NK cells in the presence of HPV-VLPs enhanced DC-maturation as shown by an upregulation of CD86 and HLA-DR and an increased production of IL-12p70, but not of the immunosuppressive cytokine IL-10. This activation was bidirectional. Indeed, in the presence of HPV-VLPs, DCs further activated NK cells by inducing the upregulation of cell surface activation markers (CD69 and HLA-DR). The function of NK cells was also improved as shown by an increase in IFN-γ secretion and cytotoxic activity against an HPV(+) cell line. This crosstalk between NK cells and DCs needed CD40 interaction and IL-12p70 secretion, whereas NKG2D was not implicated. Our results provide insight into how VLPs interact with innate immune cells and how NK cells and DCs play a role in the immune response induced by this vaccine agent.
Collapse
Affiliation(s)
- Inge Langers
- Cellular and Molecular Immunology, GIGA-Research, University of Liège, Liège, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sarmiento E, Navarro J, Fernandez-Yañez J, Palomo J, Muñoz P, Carbone J. Evaluation of an immunological score to assess the risk of severe infection in heart recipients. Transpl Infect Dis 2014; 16:802-12. [PMID: 25179534 DOI: 10.1111/tid.12284] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 03/14/2014] [Accepted: 06/18/2014] [Indexed: 01/27/2023]
Abstract
BACKGROUND We previously reported how specific humoral and cellular immunological markers that are readily available in clinical practice can be used to identify heart transplant recipients (HTR) at risk of developing severe infections. In this study, we perform an extended analysis to identify immunological profiles that could prove to be superior to individual markers in assessing the risk of infection early after heart transplantation. METHODS In a prospective follow-up study, we evaluated 100 HTR at 1 week after transplantation. Laboratory tests included determination of immunoglobulin (Ig) levels (IgG, IgA, IgM), complement factors (C3 and C4), and lymphocyte subsets (CD3+, CD4+, CD8+ T cells, B cells, and natural killer [NK] cells). The prevalence of infection during the first 3 months was registered at scheduled visits after transplantation. Severe infections were defined as all infections requiring hospitalization and intravenous antimicrobial therapy. RESULTS During follow-up, 33 patients (33%) developed severe infections. The individual risk factors of severe infection, according to the Cox regression analysis, were as follows: IgG <600 mg/dL (hazard ratio [HR], 2.41; 95% confidence interval [CI], 1.21-4.78; P = 0.012), C3 <80 mg/dL (HR, 4.65; 95% CI, 2.31-9.38; P < 0.0001), C4 <18 mg/dL (HR 2.30, 95% CI, 1.15-4.59; P = 0.018), NK count <30 cells/μL (HR 4.07, 95% CI, 1.76-9.38; P = 0.001), and CD4 count <350 cells/μL (HR, 3.04; 95% CI, 1.47-6.28; P = 0.0027). An immunological score was created. HRs were used to determine the number of points assigned to each of the 5 previously mentioned individual risk factors. The score was obtained from the sum of these factors. In the multivariate Cox regression analysis, the immunological score was useful for identifying patients at risk of infection and was the only variable that maintained a significant association with the development of infection, after adjustment for the 5 individual factors. CONCLUSION Patients with an immunological score ≥13 were at the highest risk of severe infections (HR, 9.29; 95% CI, 4.57-18.90; P < 0.0001). This score remained significantly associated with the risk of severe infection after adjustment for clinical risk factors of infection. An immunological score was useful for identifying HTR at risk of developing severe infections. If this score is validated in multicenter studies, it could be easily introduced into clinical practice.
Collapse
Affiliation(s)
- E Sarmiento
- Clinical Immunology Department, University Hospital Gregorio Marañon, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
40
|
Gao X, Zhao L, Wang S, Yang J, Yang X. Enhanced inducible costimulator ligand (ICOS-L) expression on dendritic cells in interleukin-10 deficiency and its impact on T-cell subsets in respiratory tract infection. Mol Med 2013; 19:346-56. [PMID: 24100657 DOI: 10.2119/molmed.2013.00035] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 09/25/2013] [Indexed: 11/06/2022] Open
Abstract
An association between inducible costimulator ligand (ICOS-L) expression and interleukin (IL)-10 production by dendritic cells (DCs) has been commonly found in infectious disease. DCs with higher ICOS-L expression and IL-10 production are reportedly more efficient in inducing regulatory T cells (Tregs). Here we use the Chlamydia muridarum (Cm) lung infection model in IL-10 knockout (KO) mice to test the relationship between IL-10 production and ICOS-L expression by DCs. We examined ICOS-L expression, the development of T-cell subsets, including Treg, Th17 and Th1 cell, in the background of IL-10 deficiency and its relationship with ICOS-L/ICOS signaling after infection. Surprisingly, we found that the IL-10 KO mice exhibited significantly higher ICOS-L expression by DCs. Moreover, IL-10 KO mice showed lower Tregs but higher Th17 and Th1 responses, but only the Th17 response depended on ICOS signaling. Consistently, most of the Th17 cells were ICOS⁺, whereas most of the Th1 cells were ICOS⁻ in the infected mice. Furthermore, neutralization of IL-17 in IL-10 KO mice significantly exacerbated lung infection. The data suggest that ICOS-L expression on DC may be negatively regulated by IL-10 and that ICOS-L expression on DC in the presence or absence of IL-10 costimulation may promote Treg or Th17 response, without significant impact on Th1.
Collapse
Affiliation(s)
- Xiaoling Gao
- Laboratory for Infection and Immunity, Departments of Medical Microbiology and Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lei Zhao
- Laboratory for Infection and Immunity, Departments of Medical Microbiology and Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shuhe Wang
- Laboratory for Infection and Immunity, Departments of Medical Microbiology and Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jie Yang
- Laboratory for Infection and Immunity, Departments of Medical Microbiology and Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xi Yang
- Laboratory for Infection and Immunity, Departments of Medical Microbiology and Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
41
|
Reid-Yu SA, Small CLN, Coombes BK. CD3⁻NK1.1⁺ cells aid in the early induction of a Th1 response to an attaching and effacing enteric pathogen. Eur J Immunol 2013; 43:2638-49. [PMID: 23775576 DOI: 10.1002/eji.201343435] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/24/2013] [Accepted: 06/11/2013] [Indexed: 11/06/2022]
Abstract
Extracellular attaching and effacing (A/E) pathogens including pathogenic Escherichia coli colonize the host gut causing diarrhea and inflammation. Although much is known regarding the pathogenesis of A/E bacteria, there remains an incomplete understanding of host immune responses to these microbes. NK cells are an important source of IFN-γ and are essential for early innate responses to viral pathogens; however, their role during extracellular bacterial infections is still largely unexplored. We studied the host response to the murine A/E pathogen Citrobacter rodentium to investigate NK-cell function during infection. NK1.1⁺ cell depletions and analysis of colonic intestinal inflammation following Citrobacter infection demonstrated that CD3⁻NK1.1⁺ cells play an important role in the initial clearance of C. rodentium, as evidenced by higher bacterial load, intestinal pathology, and crypt hyperplasia at the peak of inflammation in depleted mice. Loss of CD3⁻NK1.1⁺ cells resulted in lower colonic IFN-γ, TNF-α, and IL-12, and a delay in homing of IFN-γ⁺CD4⁺ T cells to the gut. Loss of this response resulted in lower anti-C. rodentium IgG in NK1.1-depleted mice. These data establish that CD3⁻NK1.1⁺ cells are critical for inducing an early Th1 response involved in clearance of a pathogen that is restricted to the gastrointestinal tract.
Collapse
Affiliation(s)
- Sarah A Reid-Yu
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
42
|
Gao X, Gigoux M, Yang J, Leconte J, Yang X, Suh WK. Anti-chlamydial Th17 responses are controlled by the inducible costimulator partially through phosphoinositide 3-kinase signaling. PLoS One 2012; 7:e52657. [PMID: 23285133 PMCID: PMC3527575 DOI: 10.1371/journal.pone.0052657] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 11/19/2012] [Indexed: 11/18/2022] Open
Abstract
We previously showed that mice deficient in the Inducible Costimulator ligand (ICOSL-KO) develop more severe disease and lung pathology with delayed bacterial clearance upon respiratory infection of Chlamydia muridarum. Importantly, the exacerbation of disease in ICOSL-KO mice was seen despite heightened IFN-γ/Th1 responses, the major defense mechanisms against Chlamydia. To gain insight into the mechanism of ICOS function in this model, we presently analyzed anti-Chlamydia immune responses in mice lacking the entire ICOS (ICOS-KO) versus knock-in mice expressing a mutant ICOS (ICOS-Y181F) that has selectively lost the ability to activate phosphoinositide 3-kinase (PI3K). Like ICOSL-KO mice, ICOS-KO mice showed worse disease with elevated IFN-γ/Th1 responses compared to wild-type (WT) mice. ICOS-Y181F mice developed much milder disease compared to ICOS-KO mice, yet they were still not fully protected to the WT level. This partial protection in ICOS-Y181F mice could not be explained by the magnitude of IFN-γ/Th1 responses since these mice developed a similar level of IFN-γ response compared to WT mice. It was rather IL-17/Th17 responses that reflected disease severity: IL-17/Th17 response was partially impaired in ICOS-Y181F mice compared to WT, but was substantially stronger than that of ICOS-KO mice. Consistently, we found that both polarization and expansion of Th17 cells were partially impaired in ICOS-Y181F CD4 T cells, and was further reduced in ICOS-KO CD4 T cells in vitro. Our results indicate that once the IFN-γ/Th1 response is above a threshold level, the IL-17/Th17 response becomes a limiting factor in controlling Chlamydia lung infection, and that ICOS plays an important role in promoting Th17 responses in part through the activation of PI3K.
Collapse
Affiliation(s)
- Xiaoling Gao
- Laboratory of Infection and Immunity, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mathieu Gigoux
- Immune Regulation Laboratory, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - Jie Yang
- Laboratory of Infection and Immunity, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Julien Leconte
- Immune Regulation Laboratory, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
- Département de microbiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Xi Yang
- Laboratory of Infection and Immunity, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail: (W-KS); (XY)
| | - Woong-Kyung Suh
- Immune Regulation Laboratory, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montréal, Québec, Canada
- Département de microbiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
- Département de médecine, Université de Montréal, Montréal, Québec, Canada
- * E-mail: (W-KS); (XY)
| |
Collapse
|
43
|
Siddiqui N, Hope J. Differential recruitment and activation of natural killer cell sub-populations by Mycobacterium bovis-infected dendritic cells. Eur J Immunol 2012; 43:159-69. [PMID: 23124835 DOI: 10.1002/eji.201242736] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 09/27/2012] [Accepted: 10/29/2012] [Indexed: 12/17/2022]
Abstract
Through complex interplay with APCs, subsets of NK cells play an important role in shaping adaptive immune responses. Bovine tuberculosis, caused by Mycobacterium bovis, is increasing in incidence and detailed knowledge of host-pathogen interactions in the natural host is essential to facilitate disease control. We investigated the interactions of NK-cell sub-populations and M. bovis-infected DCs to determine early innate mechanisms in the response to infection. A sub-population of NK cells (NKp46(+) CD2(-) ) selectively expressing lymphoid homing and inflammatory chemokine receptors were induced to migrate towards M. bovis-infected DCs. This migration was associated with increased expression of chemokines CCL3, 4, 5, 20 and CXCL8 by M. bovis-infected DCs. Activation of NKp46(+) CD2(-) NK cells and secretion of IFN-γ was observed, a response reliant on localised IL-12 release and direct cellular interaction. In a reciprocal manner, NKp46(+) CD2(-) cells induced an increase in the intensity of cell surface MHC class II expression on DCs. In contrast, NKp46(+) CD2(+) NK cells were unable to secrete IFN-γ and did not reciprocally affect DCs. This study provides novel evidence to demonstrate distinct effector responses between bovine NK-cell subsets during mycobacterial infection.
Collapse
|
44
|
IL-10 suppression of NK/DC crosstalk leads to poor priming of MCMV-specific CD4 T cells and prolonged MCMV persistence. PLoS Pathog 2012; 8:e1002846. [PMID: 22876184 PMCID: PMC3410900 DOI: 10.1371/journal.ppat.1002846] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 06/22/2012] [Indexed: 01/09/2023] Open
Abstract
IL-10 is an anti-inflammatory cytokine that regulates the extent of host immunity to infection by exerting suppressive effects on different cell types. Herpes viruses induce IL-10 to modulate the virus-host balance towards their own benefit, resulting in prolonged virus persistence. To define the cellular and molecular players involved in IL-10 modulation of herpes virus-specific immunity, we studied mouse cytomegalovirus (MCMV) infection. Here we demonstrate that IL-10 specifically curtails the MCMV-specific CD4 T cell response by suppressing the bidirectional crosstalk between NK cells and myeloid dendritic cells (DCs). In absence of IL-10, NK cells licensed DCs to effectively prime MCMV-specific CD4 T cells and we defined the pro-inflammatory cytokines IL-12, IFN-γ and TNF-α as well as NK cell activating receptors NKG2D and NCR-1 to regulate this bidirectional NK/DC interplay. Consequently, markedly enhanced priming of MCMV-specific CD4 T cells in Il10−/− mice led to faster control of lytic viral replication, but this came at the expense of TNF-α mediated immunopathology. Taken together, our data show that early induction of IL-10 during MCMV infection critically regulates the strength of the innate-adaptive immune cell crosstalk, thereby impacting beneficially on the ensuing virus-host balance for both the virus and the host. Cytomegalovirus (CMV) infections are very widespread in mammalian hosts. Despite the fact that CMVs are usually well controlled by the immune system, they cause persistent life-long infection and have evolved a number of strategies to effectively modulate or hide from host immunity. Since the establishment of an immunosuppressive environment favors virus persistence, IL-10 is one of the host targets that CMVs actively use to tune the virus-host balance toward their own benefit, resulting in prolonged virus persistence and hence increased chance for horizontal transmission. Here, we delineate the mechanisms of how IL-10 exerts its powerful immune-suppressing function in the context of murine cytomegalovirus (MCMV) infection. We found that IL-10 specifically restrains the priming of MCMV-specific CD4 T cell responses by suppressing dendritic cell (DC) - natural killer cell (NK) crosstalk during acute MCMV infection. Target molecules mediating this bi-directional crosstalk between DCs and NK cells were the pro-inflammatory cytokines IL-12, IFN-γ and TNF-α as well as NK cell activating receptors NKG2D and NCR-1 and all of them were markedly suppressed by IL-10. A consequence resulting from this impeded DC-NK cross-talk by IL-10, leading to poor priming of MCMV-specific CD4 T cell responses was increased lytic CMV persistence and reduced development of host tissue damage. Our study indicates that early induction of IL-10 during MCMV infection critically regulates the strength of the innate-adaptive crosstalk, thereby imparting on the ensuing virus-host balance for the benefit of both the virus and the host.
Collapse
|
45
|
Duell BL, Tan CK, Carey AJ, Wu F, Cripps AW, Ulett GC. Recent insights into microbial triggers of interleukin-10 production in the host and the impact on infectious disease pathogenesis. ACTA ACUST UNITED AC 2012; 64:295-313. [PMID: 22268692 DOI: 10.1111/j.1574-695x.2012.00931.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 01/17/2012] [Accepted: 01/17/2012] [Indexed: 02/06/2023]
Abstract
Since its initial description as a Th2-cytokine antagonistic to interferon-alpha and granulocyte-macrophage colony-stimulating factor, many studies have shown various anti-inflammatory actions of interleukin-10 (IL-10), and its role in infection as a key regulator of innate immunity. Studies have shown that IL-10 induced in response to microorganisms and their products plays a central role in shaping pathogenesis. IL-10 appears to function as both sword and shield in the response to varied groups of microorganisms in its capacity to mediate protective immunity against some organisms but increase susceptibility to other infections. The nature of IL-10 as a pleiotropic modulator of host responses to microorganisms is explained, in part, by its potent and varied effects on different immune effector cells which influence antimicrobial activity. A new understanding of how microorganisms trigger IL-10 responses is emerging, along with recent discoveries of how IL-10 produced during disease might be harnessed for better protective or therapeutic strategies. In this review, we summarize studies from the past 5 years that have reported the induction of IL-10 by different classes of pathogenic microorganisms, including protozoa, nematodes, fungi, viruses and bacteria and discuss the impact of this induction on the persistence and/or clearance of microorganisms in the host.
Collapse
Affiliation(s)
- Benjamin L Duell
- School of Medical Sciences, Centre for Medicine and Oral Health, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | | | | | | | | | | |
Collapse
|