1
|
Sachen KL, Hammaker D, Sarabia I, Stoveken B, Hartman J, Leppard KL, Manieri NA, Bao P, Greving C, Lacy ER, DuPrie M, Wertheimer J, Deming JD, Brown J, Hart A, Li H(H, Freeman TC, Keyes B, Kohler K, White I, Karpowich N, Steele R, Elloso MM, Fakharzadeh S, Goyal K, Lavie F, Abreu MT, Allez M, Atreya R, Bissonnette R, Eyerich K, Krueger JG, McGonagle D, McInnes IB, Ritchlin C, Fourie AM. Guselkumab binding to CD64 + IL-23-producing myeloid cells enhances potency for neutralizing IL-23 signaling. Front Immunol 2025; 16:1532852. [PMID: 40145093 PMCID: PMC11937023 DOI: 10.3389/fimmu.2025.1532852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
IL-23 is implicated in the pathogenesis of immune-mediated inflammatory diseases, and myeloid cells that express Fc gamma receptor 1 (FcγRI or CD64) on their surface have been recently identified as a primary source of IL-23 in inflamed tissue. Our complementary analyses of transcriptomic datasets from psoriasis and IBD showed increased expression of CD64 and IL-23 transcripts in inflamed tissue, and greater abundance of cell types with co-expression of CD64 and IL-23. These findings led us to explore potential implications of CD64 binding on the function of IL-23-targeting monoclonal antibodies (mAbs). Guselkumab and risankizumab are mAbs that target the IL-23p19 subunit. Guselkumab has a native Fc domain while risankizumab contains mutations that diminish binding to FcγRs. In flow cytometry assays, guselkumab, but not risankizumab, showed Fc-mediated binding to CD64 on IFNγ-primed monocytes. Guselkumab bound CD64 on IL-23-producing inflammatory monocytes and simultaneously captured IL-23 secreted from these cells. Guselkumab binding to CD64 did not induce cytokine production. In live-cell confocal imaging of CD64+ macrophages, guselkumab, but not risankizumab, mediated IL-23 internalization to low-pH intracellular compartments. Guselkumab and risankizumab demonstrated similar potency for inhibition of IL-23 signaling in cellular assays with exogenous addition of IL-23. However, in a co-culture of IL-23-producing CD64+ THP-1 cells with an IL-23-responsive reporter cell line, guselkumab demonstrated Fc-dependent enhanced potency compared to risankizumab for inhibiting IL-23 signaling. These in vitro data highlight the potential for guselkumab binding to CD64 in inflamed tissue to contribute to the potent neutralization of IL-23 at its cellular source.
Collapse
Affiliation(s)
| | | | | | | | - John Hartman
- Johnson & Johnson, Spring House, PA, United States
| | | | | | - Phuc Bao
- Johnson & Johnson, San Diego, CA, United States
| | | | | | | | | | | | | | - Amy Hart
- Johnson & Johnson, Spring House, PA, United States
| | | | | | - Brice Keyes
- Johnson & Johnson, San Diego, CA, United States
| | | | - Ian White
- Johnson & Johnson, Spring House, PA, United States
| | | | - Ruth Steele
- Johnson & Johnson, Spring House, PA, United States
| | | | | | | | | | - Maria T. Abreu
- University of Miami, Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Matthieu Allez
- Hôpital Saint-Louis, Université Paris Cité, Paris, France
| | - Raja Atreya
- Department of Medicine 1, Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Kilian Eyerich
- Medical Center, University of Freiburg, Freiburg, Germany
- Department of Medicine – Division of Dermatology and Venereology, Karolinska Institute, Stockholm, Sweden
| | - James G. Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, United States
| | - Dennis McGonagle
- Leeds Biomedical Research Centre, University of Leeds, Leeds, United Kingdom
| | - Iain B. McInnes
- College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christopher Ritchlin
- Center for Musculoskeletal Research, Allergy, Immunology, and Rheumatology Division, University of Rochester, Rochester, NY, United States
| | | |
Collapse
|
2
|
Geyer CE, Newling M, Sritharan L, Griffith GR, Chen HJ, Baeten DLP, den Dunnen J. C-Reactive Protein Controls IL-23 Production by Human Monocytes. Int J Mol Sci 2021; 22:ijms222111638. [PMID: 34769069 PMCID: PMC8583945 DOI: 10.3390/ijms222111638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023] Open
Abstract
C-reactive protein (CRP) is an acute-phase protein in humans that is produced in high quantities by the liver upon infection and under inflammatory conditions. Although CRP is commonly used as a marker of inflammation, CRP can also directly contribute to inflammation by eliciting pro-inflammatory cytokine production by immune cells. Since CRP is highly elevated in serum under inflammatory conditions, we have studied the CRP-induced cytokine profile of human monocytes, one of the main innate immune cell populations in blood. We identified that CRP is relatively unique in its capacity to induce production of the pro-inflammatory cytokine IL-23, which was in stark contrast to a wide panel of pattern recognition receptor (PRR) ligands. We show that CRP-induced IL-23 production was mediated at the level of gene transcription, since CRP particularly promoted gene transcription of IL23A (encoding IL-23p19) instead of IL12A (encoding IL-12p35), while PRR ligands induce the opposite response. Interestingly, when CRP stimulation was combined with PRR ligand stimulation, as for example, occurs in the context of sepsis, IL-23 production by monocytes was strongly reduced. Combined, these data identify CRP as a unique individual ligand to induce IL-23 production by monocytes, which may contribute to shaping systemic immune responses under inflammatory conditions.
Collapse
Affiliation(s)
- Chiara E. Geyer
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Melissa Newling
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (M.N.); (L.S.); (D.L.P.B.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Lathees Sritharan
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (M.N.); (L.S.); (D.L.P.B.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Guillermo R. Griffith
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (G.R.G.); (H.-J.C.)
| | - Hung-Jen Chen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (G.R.G.); (H.-J.C.)
| | - Dominique L. P. Baeten
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (M.N.); (L.S.); (D.L.P.B.)
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Correspondence: ; Tel.: +31-205668043
| |
Collapse
|
3
|
Nickel Sulfate Promotes IL-17A Producing CD4+ T Cells by an IL-23-Dependent Mechanism Regulated by TLR4 and Jak-STAT Pathways. J Invest Dermatol 2017. [PMID: 28634033 DOI: 10.1016/j.jid.2017.05.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Allergic contact dermatitis, caused by nickel, is a delayed-type hypersensitivity reaction, and 14.5% of the general population may be affected in Europe. Among a wide range of cytokines, the IL-12 family has unique structural and immunological characteristics. Whereas IL-12p70 promotes T helper (Th) 1 cell polarization, IL-23 promotes Th17 cell development and both have been isolated from nickel-allergic patients. In this work, we were interested in understanding the mechanism behind nickel-induced Th17 cell development. We showed that nickel induced an early production of IL-23 in human monocyte-derived dendritic cells along with an increase in the expression of il-23p19 and il-12p40 mRNA. However, the production of a significant level of IL-12p70 required an additional signal such as IFN-γ. Moreover, nickel-treated monocyte-derived dendritic cells induced an increase in the percentage of IL-17A+ CD4+ T cells, an effect reduced by IL-23 neutralization. We then investigated the molecular mechanism of IL-23 production. Our results showed that toll-like receptor 4, p38 mitogen-activated protein kinase, and NF-κB were involved in IL-23 production induced by nickel. However, Jak-signal transducer and activator of transcription activation seems to maintain the IL-23/IL-12p70 balance by limiting IL-23 production and promoting Th1 polarization. These results indicate that nickel-induced Th17 cell development is dependent on the production of IL-23 by human monocyte-derived dendritic cells via toll-like receptor 4, p38 mitogen-activated protein kinase, NF-κB, and Jak-signal transducer and activator of transcription pathways.
Collapse
|
4
|
Heppell CW, Egan JR, Hall I. A human time dose response model for Q fever. Epidemics 2017; 21:30-38. [PMID: 28666604 PMCID: PMC5729200 DOI: 10.1016/j.epidem.2017.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/30/2017] [Accepted: 06/07/2017] [Indexed: 12/16/2022] Open
Abstract
The causative agent of Q fever, Coxiella burnetii, has the potential to be developed for use in biological warfare and it is classified as a bioterrorism threat agent by the Centers for Disease Control and Prevention (CDC) and as a category B select agent by the National Institute of Allergy and Infectious Diseases (NIAID). In this paper we focus on the in-host properties that arise when an individual inhales a dose of C. burnetii and establish a human time-dose response model. We also propagate uncertainty throughout the model allowing us to robustly estimate key properties including the infectious dose and incubation period. Using human study data conducted in the 1950's we conclude that the dose required for a 50% probability of infection is about 15 organisms, and that one inhaled organism of C. burnetti can cause infection in 5% of the exposed population. In addition, we derive a low dose incubation period of 17.6 days and an extracellular doubling time of half a day. In conclusion this paper provides a framework for detailing the parameters and approaches that would be required for risk assessments associated with exposures to C. burnetii that might cause human infection.
Collapse
Affiliation(s)
| | - Joseph R Egan
- University of Southampton, Hampshire SO17 1BJ, United Kingdom.
| | - Ian Hall
- Public Health England, Porton, Wiltshire SP4 0JG, United Kingdom.
| |
Collapse
|
5
|
Sugitharini V, Shahana P, Prema A, Berla Thangam E. TLR2 and TLR4 co-activation utilizes distinct signaling pathways for the production of Th1/Th2/Th17 cytokines in neonatal immune cells. Cytokine 2016; 85:191-200. [DOI: 10.1016/j.cyto.2016.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 12/30/2022]
|
6
|
Yasuda K, Ushio H. Keyhole limpet hemocyanin induces innate immunity via Syk and Erk phosphorylation. EXCLI JOURNAL 2016; 15:474-481. [PMID: 27822175 PMCID: PMC5083961 DOI: 10.17179/excli2016-488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 07/11/2016] [Indexed: 01/01/2023]
Abstract
Hemocyanin is an extracellular respiratory protein containing copper in hemolymph of invertebrates, such as Mollusk and Arthropod. Keyhole limpet hemocyanin (KLH) is one of hemocyanins and has many years of experience for vaccine developments and immunological studies in mammals including human. However, the association between KLH and the immune systems, especially the innate immune systems, remains poorly understood. The aim of this study is to clarify the direct effects of KLH on the innate immune systems. KLH activated an inflammation-related transcription factor NF-κB as much as lipopolysaccharide (LPS) in a human monocytic leukemia THP-1 reporter cell line. We have found that the KLH-induced NF-κB activation is partially involved in a spleen tyrosine kinase (Syk) pathway. We have also successfully revealed that an extracellular signal-regulated kinase (Erk), a member of mitogen-activated protein kinases, is located in an upstream of NF-κB activation induced by KLH. Furthermore, a Syk phosphorylation inhibitor partially suppressed the Erk activation in KLH-stimulated THP-1. These results suggest that both Syk and Erk associate with the KLH-induced NF-κB activation in the human monocyte.
Collapse
Affiliation(s)
- Kyoko Yasuda
- Laboratory of Marine Biochemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo
| | - Hideki Ushio
- Laboratory of Marine Biochemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo
| |
Collapse
|
7
|
Kulsantiwong P, Pudla M, Boondit J, Wikraiphat C, Dunachie SJ, Chantratita N, Utaisincharoen P. Burkholderia pseudomallei induces IL-23 production in primary human monocytes. Med Microbiol Immunol 2015; 205:255-60. [DOI: 10.1007/s00430-015-0440-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/30/2015] [Indexed: 10/22/2022]
|
8
|
Wood RM, Egan JR, Hall IM. A dose and time response Markov model for the in-host dynamics of infection with intracellular bacteria following inhalation: with application to Francisella tularensis. J R Soc Interface 2014; 11:20140119. [PMID: 24671937 PMCID: PMC4006251 DOI: 10.1098/rsif.2014.0119] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
In a novel approach, the standard birth–death process is extended to incorporate a fundamental mechanism undergone by intracellular bacteria, phagocytosis. The model accounts for stochastic interaction between bacteria and cells of the immune system and heterogeneity in susceptibility to infection of individual hosts within a population. Model output is the dose–response relation and the dose-dependent distribution of time until response, where response is the onset of symptoms. The model is thereafter parametrized with respect to the highly virulent Schu S4 strain of Francisella tularensis, in the first such study to consider a biologically plausible mathematical model for early human infection with this bacterium. Results indicate a median infectious dose of about 23 organisms, which is higher than previously thought, and an average incubation period of between 3 and 7 days depending on dose. The distribution of incubation periods is right-skewed up to about 100 organisms and symmetric for larger doses. Moreover, there are some interesting parallels to the hypotheses of some of the classical dose–response models, such as independent action (single-hit model) and individual effective dose (probit model). The findings of this study support experimental evidence and postulations from other investigations that response is, in fact, influenced by both in-host and between-host variability.
Collapse
Affiliation(s)
- R M Wood
- Bioterrorism and Emerging Disease Analysis, Microbial Risk Assessment and Behavioural Science, Public Health England, , Porton Down SP4 0JG, UK
| | | | | |
Collapse
|
9
|
Gillette DD, Curry HM, Cremer T, Ravneberg D, Fatehchand K, Shah PA, Wewers MD, Schlesinger LS, Butchar JP, Tridandapani S, Gavrilin MA. Virulent Type A Francisella tularensis actively suppresses cytokine responses in human monocytes. Front Cell Infect Microbiol 2014; 4:45. [PMID: 24783062 PMCID: PMC3988375 DOI: 10.3389/fcimb.2014.00045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/27/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Human monocyte inflammatory responses differ between virulent and attenuated Francisella infection. RESULTS A mixed infection model showed that the virulent F. tularensis Schu S4 can attenuate inflammatory cytokine responses to the less virulent F. novicida in human monocytes. CONCLUSION F. tularensis dampens inflammatory response by an active process. SIGNIFICANCE This suppression may contribute to enhanced pathogenicity of F. tularensis. Francisella tularensis is a Gram-negative facultative bacterium that can cause the disease tularemia, even upon exposure to low numbers of bacteria. One critical characteristic of Francisella is its ability to dampen or subvert the host immune response. Previous work has shown that monocytes infected with highly virulent F. tularensis subsp. tularensis strain Schu S4 responded with a general pattern of quantitatively reduced pro-inflammatory signaling pathway genes and cytokine production in comparison to those infected with the less virulent related F. novicida. However, it has been unclear whether the virulent Schu S4 was merely evading or actively suppressing monocyte responses. By using mixed infection assays with F. tularensis and F. novicida, we show that F. tularensis actively suppresses monocyte pro-inflammatory responses. Additional experiments show that this suppression occurs in a dose-dependent manner and is dependent upon the viability of F. tularensis. Importantly, F. tularensis was able to suppress pro-inflammatory responses to earlier infections with F. novicida. These results lend support that F. tularensis actively dampens human monocyte responses and this likely contributes to its enhanced pathogenicity.
Collapse
Affiliation(s)
- Devyn D Gillette
- Integrated Biomedical Graduate Program, The Ohio State University Columbus, OH, USA
| | - Heather M Curry
- Department of Microbial Infection and Immunity, The Ohio State University Columbus, OH, USA ; Center for Microbial Interface Biology, The Ohio State University Columbus, OH, USA
| | - Thomas Cremer
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - David Ravneberg
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - Kavin Fatehchand
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - Prexy A Shah
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - Mark D Wewers
- Center for Microbial Interface Biology, The Ohio State University Columbus, OH, USA ; Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - Larry S Schlesinger
- Department of Microbial Infection and Immunity, The Ohio State University Columbus, OH, USA ; Center for Microbial Interface Biology, The Ohio State University Columbus, OH, USA
| | - Jonathan P Butchar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - Susheela Tridandapani
- Center for Microbial Interface Biology, The Ohio State University Columbus, OH, USA ; Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - Mikhail A Gavrilin
- Center for Microbial Interface Biology, The Ohio State University Columbus, OH, USA ; Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| |
Collapse
|
10
|
Costa MM, Saraceni PR, Forn-Cuní G, Dios S, Romero A, Figueras A, Novoa B. IL-22 is a key player in the regulation of inflammation in fish and involves innate immune cells and PI3K signaling. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:746-55. [PMID: 23999050 DOI: 10.1016/j.dci.2013.08.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/20/2013] [Accepted: 08/24/2013] [Indexed: 06/02/2023]
Abstract
IL-22 plays a role in various disorders in mammals, including mucosal-associated infections and inflammatory diseases. No functional IL-22 studies have been conducted on non-mammals to date. In this study, recombinant IL-22 (rIL-22) from turbot was produced to investigate its effects as a bioactive molecule. The expression of several pro-inflammatory cytokines was increased after rIL-22 treatment and reduced by pre-treatment with a JAK/STAT inhibitor. The involvement of the PI3K pathway in IL-22 induction was demonstrated. rIL-22 reduced the mortality in Aeromonas salmonicida-infected turbot, while higher Aeromonas hydrophila- or LPS-induced mortality was observed when IL-22 was blocked in zebrafish embryos. IL-22 knockdown increased pro-inflammatory cytokine expression in bacteria-stimulated fish. In zebrafish, IL-22 expression was detected primarily in the myeloid innate linage. It was found during early developmental stages when the adaptive immune response is not yet functional and in rag1(-)/(-) fish that lack an adaptive immune system. Our results clarify the conserved role of IL-22 in lower vertebrates. We suggest for the first time that IL-22 constitutes a key regulator of inflammatory homeostasis even in distant species such as teleosts, which diverged from mammals more than 350 million years ago.
Collapse
Affiliation(s)
- Maria M Costa
- Instituto de Investigaciones Marinas (IIM), CSIC, Eduardo Cabello 6, 36208 Vigo, Spain.
| | | | | | | | | | | | | |
Collapse
|
11
|
Slight SR, Monin L, Gopal R, Avery L, Davis M, Cleveland H, Oury TD, Rangel-Moreno J, Khader SA. IL-10 restrains IL-17 to limit lung pathology characteristics following pulmonary infection with Francisella tularensis live vaccine strain. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1397-1404. [PMID: 24007881 DOI: 10.1016/j.ajpath.2013.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 07/17/2013] [Accepted: 07/22/2013] [Indexed: 12/29/2022]
Abstract
IL-10 production during intracellular bacterial infections is generally thought to be detrimental because of its role in suppressing protective T-helper cell 1 (Th1) responses. Francisella tularensis is a facultative intracellular bacterium that activates both Th1 and Th17 protective immune responses. Herein, we report that IL-10-deficient mice (Il10(-/-)), despite having increased Th1 and Th17 responses, exhibit increased mortality after pulmonary infection with F. tularensis live vaccine strain. We demonstrate that the increased mortality observed in Il10(-/-)-infected mice is due to exacerbated IL-17 production that causes increased neutrophil recruitment and associated lung pathology. Thus, although IL-17 is required for protective immunity against pulmonary infection with F. tularensis live vaccine strain, its production is tightly regulated by IL-10 to generate efficient induction of protective immunity without mediating pathology. These data suggest a critical role for IL-10 in maintaining the delicate balance between host immunity and pathology during pulmonary infection with F. tularensis live vaccine strain.
Collapse
Affiliation(s)
- Samantha R Slight
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Leticia Monin
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Radha Gopal
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lyndsay Avery
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marci Davis
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hillary Cleveland
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Shabaana A Khader
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
12
|
Blahoianu MA, Rahimi AAR, Gajanayaka N, Kozlowski M, Angel JB, Kumar A. Engagement of CD14 sensitizes primary monocytes to IFN-γ to produce IL-12/23p40 and IL-23 through p38 mitogen-activated protein kinase and independent of the janus kinase/signal transducers and activators of transcription signaling. J Interferon Cytokine Res 2013; 33:434-45. [PMID: 23679818 DOI: 10.1089/jir.2012.0058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Interferon (IFN)-γ is a potent stimulator of the IL-12 family Th1 cytokines, including IL-12/23p40 and IL-23, responsible for coordinating the innate and adaptive immune responses. Our results show that IFN-γ induced the production of IL-12/23p40 and IL-23p19 mRNA as well as IL-12p40 and IL-23 proteins in primary human monocytes isolated by positive selection through anti-CD14 microbeads. These results were confirmed by IFN-γ stimulation of CD14-activated monocytes resulting in IL-12/23p40 and IL-23 production. We investigated the signaling pathways governing the regulation of IL-23 and its subunits IL-23p40 and IL-23p19 following IFN-γ stimulation. We observed a differential regulation of IL-23p19, IL-12/23p40, and IL-23 following IFN-γ stimulation. IFN-γ-induced IL-23 and IL-12/23p40 expression was positively regulated by the p38 mitogen-activated protein kinases (MAPKs), independent of the Janus kinase (Jak)/signal transducers and activators of transcription (STAT) signaling. In contrast, IL-12 and IL-23 were negatively regulated by the Jak/STAT, phosphatidylinositol 3-kinase (PI3K), and the c-Jun-N-terminal kinase (JNK) MAPKs in IFN-γ-stimulated monocytes. Overall, our results suggest for the first time a differential positive regulation of IL-12p40 and IL-23 by p38 MAPKs independent of the Jak/STAT pathways and negative regulation by the Jak/STAT, JNK, and PI3K pathways in CD14-activated primary human monocytes stimulated with IFN-γ.
Collapse
Affiliation(s)
- Maria A Blahoianu
- Department of Pathology and Laboratory Medicine, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada.
| | | | | | | | | | | |
Collapse
|
13
|
Interleukin-17 protects against the Francisella tularensis live vaccine strain but not against a virulent F. tularensis type A strain. Infect Immun 2013; 81:3099-105. [PMID: 23774604 DOI: 10.1128/iai.00203-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Francisella tularensis is a highly infectious intracellular bacterium that causes the zoonotic infection tularemia. While much literature exists on the host response to F. tularensis infection, the vast majority of work has been conducted using attenuated strains of Francisella that do not cause disease in humans. However, emerging data indicate that the protective immune response against attenuated F. tularensis versus F. tularensis type A differs. Several groups have recently reported that interleukin-17 (IL-17) confers protection against the live vaccine strain (LVS) of Francisella. While we too have found that IL-17Rα(-/-) mice are more susceptible to F. tularensis LVS infection, our studies, using a virulent type A strain of F. tularensis (SchuS4), indicate that IL-17Rα(-/-) mice display organ burdens and pulmonary gamma interferon (IFN-γ) responses similar to those of wild-type mice following infection. In addition, oral LVS vaccination conferred equivalent protection against pulmonary challenge with SchuS4 in both IL-17Rα(-/-) and wild-type mice. While IFN-γ was found to be critically important for survival in a convalescent model of SchuS4 infection, IL-17 neutralization from either wild-type or IFN-γ(-/-) mice had no effect on morbidity or mortality in this model. IL-17 protein levels were also higher in the lungs of mice infected with the LVS rather than F. tularensis type A, while IL-23p19 mRNA expression was found to be caspase-1 dependent in macrophages infected with LVS but not SchuS4. Collectively, these results demonstrate that IL-17 is dispensable for host immunity to type A F. tularensis infection, and that induced and protective immunity differs between attenuated and virulent strains of F. tularensis.
Collapse
|
14
|
Yannam GR, Gutti T, Poluektova LY. IL-23 in infections, inflammation, autoimmunity and cancer: possible role in HIV-1 and AIDS. J Neuroimmune Pharmacol 2011; 7:95-112. [PMID: 21947740 DOI: 10.1007/s11481-011-9315-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 09/11/2011] [Indexed: 12/30/2022]
Abstract
The growing family of interleukin (IL)-12-like cytokines produced by activated macrophages and dendritic cells became the important players in the control of infections, development of inflammation, autoimmunity and cancer. However, the role of one of them-heterodimer IL-23, which consists of IL12p40 and the unique p19 subunit in HIV-1 infection pathogenesis and progression to AIDS, represent special interest. We overviewed findings of IL-23 involvement in control of peripheral bacterial pathogens and opportunistic infection, central nervous system (CNS) viral infections and autoimmune disorders, and tumorogenesis, which potentially could be applicable to HIV-1 and AIDS.
Collapse
Affiliation(s)
- Govardhana Rao Yannam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | |
Collapse
|
15
|
Slight SR, Lin Y, Messmer M, Khader SA. Francisella tularensis LVS-induced Interleukin-12 p40 cytokine production mediates dendritic cell migration through IL-12 Receptor β1. Cytokine 2011; 55:372-9. [PMID: 21669537 PMCID: PMC3148298 DOI: 10.1016/j.cyto.2011.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 04/20/2011] [Accepted: 05/18/2011] [Indexed: 12/22/2022]
Abstract
Three cytokines use the IL-12p40 cytokine subunit namely: IL-12p70 (IL-12-comprised of IL-12p40 and IL-12p35), IL-23 (comprised of the IL-12p40 and IL-23p19 subunits) and homodimeric IL-12p40 (IL-12(p40)(2)). Following activation, immature dendritic cells (DCs) upregulate the chemokine receptor Chemokine-C-Receptor 7 (CCR7), and migrate in response to homeostatic chemokines such as chemokine (C-C motif) ligand 19 (CCL19). Induction of the cytokine IL-12p40 in response to pathogen-exposure, likely in its homodimeric form, is one of the primary events that mediates migration of DCs in response to CCL19. Here we show that following exposure to Francisella tularensis Live Vaccine Strain (LVS), DCs produce IL-12p40 and promote the migration of DCs to the chemokine CCL19 in an IL-12Rβ1- and IL-12p(40)(2)-dependent manner. Induction of IL-12p40 and resulting chemokine responsiveness in DCs is TLR2-dependent and coincides with the uptake of F. tularensis LVS and activation of DCs. Importantly, we show that IL-12Rβ1 signaling is required for DC migration from the lung to the draining lymph node following F. tularensis LVS exposure and coincides with accumulation of IL-12p40 expressing DCs in the draining lymph nodes. Together, these findings illustrate that IL-12p40 is induced rapidly in response to F. tularensis LVS and is required for DC migration through an IL-12Rβ1-IL-12(p40)(2) dependent mechanism.
Collapse
Affiliation(s)
| | | | - Michelle Messmer
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Shabaana A. Khader
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| |
Collapse
|
16
|
Wang Q, Franks HA, Porte J, El Refaee M, Shah S, Crooks J, Patel PM, Jackson AM. Novel approach for interleukin-23 up-regulation in human dendritic cells and the impact on T helper type 17 generation. Immunology 2011; 134:60-72. [PMID: 21718315 PMCID: PMC3173695 DOI: 10.1111/j.1365-2567.2011.03467.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 05/25/2011] [Accepted: 05/31/2011] [Indexed: 12/15/2022] Open
Abstract
Interleukin-23 (IL-23) is important for T helper type 17 (Th17) responses and strategies to regulate IL-23 in human dendritic cells (DC) are limited. This study describes a novel means to control IL-23 secretion by conditioning DC with a phosphatidyl inositol 3-kinase inhibitor Wortmannin (WM). Treatment of monocyte-derived DC with WM increased Toll-like receptor (TLR) -dependent IL-23 secretion 10-fold and IL-12p70 twofold, but IL-27 was unaffected. The effect of WM was restricted to TLR3/4 pathways, did not occur through TLR2, TLR7/8 or Dectin-1, and was characterized by increased p19, p35 and p40 transcription. These responses were not solely dependent on phosphatidyl inositol 3-kinase as the alternative inhibitor LY294002 did not modulate IL-23 production. The normal patterns of activation of mitogen-activated protein kinase pathways were unaffected by WM-conditioning but IL-23 secretion required p38, ERK and JNK pathways. Importantly, this effect was manifest in populations of blood DC. Conditioning freshly isolated myeloid DC with WM before TLR3 or TLR4 triggering resulted in high levels of IL-23 secretion and an absence of IL-12p70. These WM-conditioned myeloid DC were highly effective at priming Th17 responses from naive CD4(+) T cells. Our findings provide a novel means to generate IL-23-rich environments and Th17 responses and suggest as yet unidentified regulatory factors, identification of which will provide new approaches to control IL-23-dependent immunity in infectious disease, autoimmunity and malignancy.
Collapse
Affiliation(s)
- Qunwei Wang
- Academic Unit of Clinical Oncology, University of Nottingham, Nottingham, UK
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Bauler TJ, Chase JC, Bosio CM. IFN-β mediates suppression of IL-12p40 in human dendritic cells following infection with virulent Francisella tularensis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1845-55. [PMID: 21753150 PMCID: PMC3150225 DOI: 10.4049/jimmunol.1100377] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Active suppression of inflammation is a strategy used by many viral and bacterial pathogens, including virulent strains of the bacterium Francisella tularensis, to enable colonization and infection in susceptible hosts. In this study, we demonstrated that virulent F. tularensis strain SchuS4 selectively inhibits production of IL-12p40 in primary human cells via induction of IFN-β. In contrast to the attenuated live vaccine strain, infection of human dendritic cells with virulent SchuS4 failed to induce production of many cytokines associated with inflammation (e.g., TNF-α and IL-12p40). Furthermore, SchuS4 actively suppressed secretion of these cytokines. Assessment of changes in the expression of host genes associated with suppression of inflammatory responses revealed that SchuS4, but not live vaccine strain, induced IFN-β following infection of human dendritic cells. Phagocytosis of SchuS4 and endosomal acidification were required for induction of IFN-β. Further, using a defined mutant of SchuS4, we demonstrated that the presence of bacteria in the cytosol was required, but not sufficient, for induction of IFN-β. Surprisingly, unlike previous reports, induction of IFN-β by F. tularensis was not required for activation of the inflammasome, was not associated with exacerbation of inflammatory responses, and did not control SchuS4 replication when added exogenously. Rather, IFN-β selectively suppressed the ability of SchuS4-infected dendritic cells to produce IL-12p40. Together, these data demonstrated a novel mechanism by which virulent bacteria, in contrast to attenuated strains, modulate human cells to cause disease.
Collapse
Affiliation(s)
- Timothy J. Bauler
- Immunity to Pulmonary Pathogens Section, LICP/RML/NIAID/NIH, Hamilton, MT, 59840
| | - Jennifer C. Chase
- Immunity to Pulmonary Pathogens Section, LICP/RML/NIAID/NIH, Hamilton, MT, 59840
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, LICP/RML/NIAID/NIH, Hamilton, MT, 59840
| |
Collapse
|
18
|
Al-Khodor S, Abu Kwaik Y. Triggering Ras signalling by intracellular Francisella tularensis through recruitment of PKCα and βI to the SOS2/GrB2 complex is essential for bacterial proliferation in the cytosol. Cell Microbiol 2011; 12:1604-21. [PMID: 20618341 DOI: 10.1111/j.1462-5822.2010.01494.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intracellular proliferation of Francisella tularensis is essential for manifestation of the fatal disease tularaemia, and is classified as a category A bioterrorism agent. The F. tularensis-containing phagosome (FCP) matures into a late endosome-like phagosome with limited fusion to lysosomes, followed by rapid bacterial escape into the cytosol. The Francisella pathogenicity island (FPI) encodes a type VI-like secretion system, and the FPI-encoded IglC is essential for evasion of lysosomal fusion and phagosomal escape. Many host signalling events are likely to be modulated by F. tularensis to render the cell permissive for intracellular proliferation but they are not fully understood. Here we show that within 15 min of infection, intracellular F. tularensis ssp. novicida triggers IglC-dependent temporal activation of Ras, but attached extracellular bacteria fail to trigger Ras activation, which has never been shown for other intracellular pathogens. Intracellular F. tularensis ssp. novicida triggers activation of Ras through recruitment of PKCα and PKCβI to the SOS2/GrB2 complex. Silencing of SOS2, GrB2 and PKCα and PKCβI by RNAi has no effect on evasion of lysosomal fusion and bacterial escape into the cytosol but renders the cytosol non-permissive for replication of F. tularensis ssp. novicida. Since Ras activation promotes cell survival, we show that silencing of SOS2, GrB2 and PKCα and βI is associated with rapid early activation of caspase-3 within 8 h post infection. However, silencing of SOS2, GrB2 and PKCα and βI does not affect phosphorylation of Akt or Erk, indicating that activation of the PI3K/Akt and the Erk signalling cascade are independent of the F. tularensis-triggered Ras activation. We conclude that intracellular F. tularensis ssp. novicida triggers temporal and early activation of Ras through the SOS2/GrB2/PKCα/PKCβI quaternary complex. Temporal and rapid trigger of Ras signalling by intracellular F. tularensis is essential for intracellular bacterial proliferation within the cytosol, and this is associated with downregulation of early caspase-3 activation.
Collapse
Affiliation(s)
- Souhaila Al-Khodor
- Department of Microbiology and Immunology, College of Medicine, Department of Biology, University of Louisville, Louisville, KY 40202, USA
| | | |
Collapse
|
19
|
Abstract
In recent years, studies on the intracellular pathogen Francisella tularensis have greatly intensified, generating a wealth of new information on the interaction of this organism with the immune system. Here we review the basic elements of the innate and adaptive immune responses that contribute to protective immunity against Francisella species, with special emphasis on new data that has emerged in the last 5 years. Most studies have utilized the mouse model of infection, although there has been an expansion of work on human cells and other new animal models. In mice, basic immune parameters that operate in defense against other intracellular pathogen infections, such as interferon gamma, TNF-α, and reactive nitrogen intermediates, are central for control of Francisella infection. However, new important immune mediators have been revealed, including IL-17A, Toll-like receptor 2, and the inflammasome. Further, a variety of cell types in addition to macrophages are now recognized to support Francisella growth, including epithelial cells and dendritic cells. CD4+ and CD8+ T cells are clearly important for control of primary infection and vaccine-induced protection, but new T cell subpopulations and the mechanisms employed by T cells are only beginning to be defined. A significant role for B cells and specific antibodies has been established, although their contribution varies greatly between bacterial strains of lower and higher virulence. Overall, recent data profile a pathogen that is adept at subverting host immune responses, but susceptible to many elements of the immune system's antimicrobial arsenal.
Collapse
Affiliation(s)
- Siobhán C Cowley
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration Bethesda, MD, USA
| | | |
Collapse
|
20
|
Khader SA, Gopal R. IL-17 in protective immunity to intracellular pathogens. Virulence 2010; 1:423-7. [PMID: 21178483 PMCID: PMC2953849 DOI: 10.4161/viru.1.5.12862] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 06/28/2010] [Accepted: 06/30/2010] [Indexed: 01/04/2023] Open
Abstract
The identification of a new T cell subset referred to as T helper 17 (Th17) cells and its role in protective immunity against extracellular bacterial infections is well established. In contrast, initial studies suggested that the IL-23-IL-17 pathway was not required for protection against intracellular pathogens such as mycobacterial infections. However, recent studies demonstrate that Th17-IL-23 pathway may play a crucial role in protective immunity against other intracellular pathogens by regulating the innate and adaptive immune responses. The current outlook on the role of IL-23-IL-17 pathway in protective immunity to intracellular pathogens is discussed here.
Collapse
Affiliation(s)
- Shabaana A Khader
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | |
Collapse
|
21
|
Markel G, Bar-Haim E, Zahavy E, Cohen H, Cohen O, Shafferman A, Velan B. The involvement of IL-17A in the murine response to sub-lethal inhalational infection with Francisella tularensis. PLoS One 2010; 5:e11176. [PMID: 20585449 PMCID: PMC2887844 DOI: 10.1371/journal.pone.0011176] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 05/19/2010] [Indexed: 01/16/2023] Open
Abstract
Background Francisella tularensis is an intercellular bacterium often causing fatal disease when inhaled. Previous reports have underlined the role of cell-mediated immunity and IFNγ in the host response to Francisella tularensis infection. Methodology/Principal Findings Here we provide evidence for the involvement of IL-17A in host defense to inhalational tularemia, using a mouse model of intranasal infection with the Live Vaccine Strain (LVS). We demonstrate the kinetics of IL-17A production in lavage fluids of infected lungs and identify the IL-17A-producing lymphocytes as pulmonary γδ and Th17 cells. The peak of IL-17A production appears early during sub-lethal infection, it precedes the peak of immune activation and the nadir of the disease, and then subsides subsequently. Exogenous airway administration of IL-17A or of IL-23 had a limited yet consistent effect of delaying the onset of death from a lethal dose of LVS, implying that IL-17A may be involved in restraining the infection. The protective role for IL-17A was directly demonstrated by in vivo neutralization of IL-17A. Administration of anti IL-17A antibodies concomitantly to a sub-lethal airway infection with 0.1×LD50 resulted in a fatal disease. Conclusion In summary, these data characterize the involvement and underline the protective key role of the IL-17A axis in the lungs from inhalational tularemia.
Collapse
Affiliation(s)
- Gal Markel
- Department of Biochemistry and Molecular Genetics, Israel Institute of Biological Research, Ness Ziona, Israel
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics, Israel Institute of Biological Research, Ness Ziona, Israel
| | - Eran Zahavy
- Department of Infectious Diseases, Israel Institute of Biological Research, Ness Ziona, Israel
| | - Hila Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute of Biological Research, Ness Ziona, Israel
| | - Ofer Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute of Biological Research, Ness Ziona, Israel
| | - Avigdor Shafferman
- Department of Biochemistry and Molecular Genetics, Israel Institute of Biological Research, Ness Ziona, Israel
| | - Baruch Velan
- Department of Biochemistry and Molecular Genetics, Israel Institute of Biological Research, Ness Ziona, Israel
- * E-mail:
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Compelling evidence suggests that the Th17 lineage and other IL-17-producing cells play critical roles in host defense against pathogens at mucosal sites. However, IL-17 can also contribute to inflammatory responses at mucosal sites. In this review, we will discuss the recent progress in our understanding of the role of Th17 and other IL-17-producing cells in defining the fine balance between immunity and inflammation at different mucosal sites. RECENT FNDINGS: Recent findings have highlighted that Th17 cytokines are important for the induction of innate and adaptive host responses and contribute to host defense against pathogens at mucosal sites. More recent developments have probed how the Th17 responses are generated in vivo in response to infections and their requirement in maintaining barrier function at mucosal sites. Most importantly, it is becoming apparent that there is a fine balance between protective and pathological manifestation of Th17 responses at mucosal sites that defines immunity or inflammation. SUMMARY In this review, we have summarized the recent advances in our understanding of Th17 cytokines and how they contribute to immunity versus inflammation at mucosal sites.
Collapse
Affiliation(s)
- Lokesh Guglani
- Division of Pulmonary Medicine, Allergy and Immunology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Shabaana A. Khader
- Division of Pulmonary Medicine, Allergy and Immunology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| |
Collapse
|
23
|
van de Wetering D, de Paus RA, van Dissel JT, van de Vosse E. Salmonella induced IL-23 and IL-1beta allow for IL-12 production by monocytes and Mphi1 through induction of IFN-gamma in CD56 NK/NK-like T cells. PLoS One 2009; 4:e8396. [PMID: 20027291 PMCID: PMC2791865 DOI: 10.1371/journal.pone.0008396] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2009] [Accepted: 11/23/2009] [Indexed: 11/18/2022] Open
Abstract
Background The type-1 cytokine pathway plays a pivotal role in immunity against intracellular bacterial pathogens such as Salmonellae and Mycobacteria. Bacterial stimulation of pattern recognition receptors on monocytes, macrophages and dendritic cells initiates this pathway, and results in the production of cytokines that activate lymphocytes to produce interferon (IFN)-γ. Interleukin (IL)-12 and IL-23 are thought to be the key cytokines required for initiating a type-1 cytokine immune response to Mycobacteria and Salmonellae. The relative contribution of IL-23 and IL-12 to this process is uncertain. Methodology/Principal Findings We show that various TLR agonists induce the production of IL-23 but not IL-12 in freshly isolated human monocytes and cultured human macrophages. In addition, type 1 pro-inflammatory macrophages (Mϕ1) differentiated in the presence of GM-CSF and infected with live Salmonella produce IL-23, IL-1β and IL-18, but not IL-12. Supernatants of Salmonella-infected Mϕ1 contained more IL-18 and IL-1β as compared with supernatants of Mϕ1 stimulated with isolated TLR agonists, and induced IFN-γ production in human CD56+ cells in an IL-23 and IL-1β-dependent but IL-12-independent manner. In addition, IL-23 together with IL-18 or IL-1β led to the production of GM-CSF in CD56+ cells. Both IFN-γ and GM-CSF enhanced IL-23 production by monocytes in response to TLR agonists, as well as induced IL-12 production. Conclusions/Significance The findings implicate a positive feedback loop in which IL-23 can enhance its release via induction of IFN-γ and GM-CSF. The IL-23 induced cytokines allow for the subsequent production of IL-12 and amplify the IFN-γ production in the type-1 cytokine pathway.
Collapse
Affiliation(s)
| | - Roelof A. de Paus
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap T. van Dissel
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther van de Vosse
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
24
|
Rajaram MVS, Butchar JP, Parsa KVL, Cremer TJ, Amer A, Schlesinger LS, Tridandapani S. Akt and SHIP modulate Francisella escape from the phagosome and induction of the Fas-mediated death pathway. PLoS One 2009; 4:e7919. [PMID: 19936232 PMCID: PMC2775408 DOI: 10.1371/journal.pone.0007919] [Citation(s) in RCA: 29] [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: 07/01/2009] [Accepted: 10/27/2009] [Indexed: 12/13/2022] Open
Abstract
Francisella tularensis infects macrophages and escapes phago-lysosomal fusion to replicate within the host cytosol, resulting in host cell apoptosis. Here we show that the Fas-mediated death pathway is activated in infected cells and correlates with escape of the bacterium from the phagosome and the bacterial burden. Our studies also demonstrate that constitutive activation of Akt, or deletion of SHIP, promotes phago-lysosomal fusion and limits bacterial burden in the host cytosol, and the subsequent induction of Fas expression and cell death. Finally, we show that phagosomal escape/intracellular bacterial burden regulate activation of the transcription factors sp1/sp3, leading to Fas expression and cell death. These data identify for the first time host cell signaling pathways that regulate the phagosomal escape of Francisella, leading to the induction of Fas and subsequent host cell death.
Collapse
Affiliation(s)
- Murugesan V. S. Rajaram
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Jonathan P. Butchar
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Kishore V. L. Parsa
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Thomas J. Cremer
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Amal Amer
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Larry S. Schlesinger
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Susheela Tridandapani
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
- Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
25
|
Santic M, Pavokovic G, Jones S, Asare R, Kwaik YA. Regulation of apoptosis and anti-apoptosis signalling by Francisella tularensis. Microbes Infect 2009; 12:126-34. [PMID: 19925880 DOI: 10.1016/j.micinf.2009.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 11/05/2009] [Accepted: 11/05/2009] [Indexed: 02/06/2023]
Abstract
Francisella tularensis induces apoptosis within macrophages but the temporal and spatial modulation through activation of caspase-1, caspase-3, and the anti-apoptosis nuclear transcription factor B (NF-kappaB) is not known. Whether escape of the bacteria into the cytosol is sufficient and/or essential for activation of NF-kappaB is not known. Our results show that F. tularensis subsp. novicida induces sustained nuclear translocation of NF-kappaB at early time points after infection of human monocytes derived macrophages (hMDMs). The sustained nuclear translocation of NF-kappaB is defective in the iglC mutant that fails to escape into the cytosol of macrophages. Nuclear translocation of NF-kappaB by the wild type strain is abolished upon treatment with the NF-kappaB inhibitor caffein acid phenyl ester. While the wild type strain triggers caspase-3 and caspase-1 activation by 6 h post-infection the iglC mutant is defective in triggering both caspases. In hMDMs treated with the apoptosis-inducing agent, staurosporin, there is an induction of cell death in the iglC mutant-infected macrophages despite reduced frequency of caspase-1 and caspase-3 activity. The wt-infected macrophages are resistant to cell death-induced agent. We conclude that although caspase-1 and capsase-3 are triggered within F. tularensis-infected hMDMs during early stages of infection, cell death is delayed, which is correlated with simultaneous activation of NF-kappaB.
Collapse
Affiliation(s)
- Marina Santic
- Department of Microbiology and Parasitology, University of Rijeka, Medical Faculty, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | | | | | | | | |
Collapse
|
26
|
Lin Y, Ritchea S, Logar A, Slight S, Messmer M, Rangel-Moreno J, Guglani L, Alcorn JF, Strawbridge H, Park SM, Onishi R, Nyugen N, Walter MJ, Pociask D, Randall TD, Gaffen SL, Iwakura Y, Kolls JK, Khader SA. Interleukin-17 is required for T helper 1 cell immunity and host resistance to the intracellular pathogen Francisella tularensis. Immunity 2009; 31:799-810. [PMID: 19853481 DOI: 10.1016/j.immuni.2009.08.025] [Citation(s) in RCA: 237] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 05/28/2009] [Accepted: 08/21/2009] [Indexed: 02/06/2023]
Abstract
The importance of T helper type 1 (Th1) cell immunity in host resistance to the intracellular bacterium Francisella tularensis is well established. However, the relative roles of interleukin (IL)-12-Th1 and IL-23-Th17 cell responses in immunity to F. tularensis have not been studied. The IL-23-Th17 cell pathway is critical for protective immunity against extracellular bacterial infections. In contrast, the IL-23-Th17 cell pathway is dispensable for protection against intracellular pathogens such as Mycobacteria. Here we show that the IL-23-Th17 pathway regulates the IL-12-Th1 cell pathway and was required for protective immunity against F.tularensis live vaccine strain. We show that IL-17A, but not IL-17F or IL-22, induced IL-12 production in dendritic cells and mediated Th1 responses. Furthermore, we show that IL-17A also induced IL-12 and interferon-gamma production in macrophages and mediated bacterial killing. Together, these findings illustrate a biological function for IL-17A in regulating IL-12-Th1 cell immunity and host responses to an intracellular pathogen.
Collapse
Affiliation(s)
- Yinyao Lin
- Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal Immunol 2009; 2:403-11. [PMID: 19587639 PMCID: PMC2811522 DOI: 10.1038/mi.2009.100] [Citation(s) in RCA: 354] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
T helper type 17 (Th17) cells are a distinct lineage of T cells that produce the effector molecules IL-17, IL-17F, IL-21, and IL-22. Although the role of Th17 cells in autoimmunity is well documented, there is growing evidence that the Th17 lineage and other interleukin (IL)-17-producing cells are critical for host defense against bacterial, fungal, and viral infections at mucosal surfaces. Here we summarize recent progress in our understanding of the function of IL-17-producing cells as a bridge between innate and adaptive immunity against infectious diseases at the mucosa.
Collapse
Affiliation(s)
- S A Khader
- Department of Pediatrics (Pulmonology) and Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
28
|
Paidipally P, Periasamy S, Barnes PF, Dhiman R, Indramohan M, Griffith DE, Cosman D, Vankayalapati R. NKG2D-Dependent IL-17 Production by Human T Cells in Response to an Intracellular Pathogen. THE JOURNAL OF IMMUNOLOGY 2009; 183:1940-5. [DOI: 10.4049/jimmunol.0803578] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Abstract
SUMMARY Our understanding of the role of T cells in human disease is undergoing revision as a result of the discovery of T-helper 17 (Th17) cells, a unique CD4(+) T-cell subset characterized by production of interleukin-17 (IL-17). IL-17 is a highly inflammatory cytokine with robust effects on stromal cells in many tissues. Recent data in humans and mice suggest that Th17 cells play an important role in the pathogenesis of a diverse group of immune-mediated diseases, including psoriasis, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and asthma. Initial reports also propose a role for Th17 cells in tumorigenesis and transplant rejection. Important differences, as well as many similarities, are emerging when the biology of Th17 cells in the mouse is compared with corresponding phenomena in humans. As our understanding of human Th17 biology grows, the mechanisms underlying many diseases are becoming more apparent, resulting in a new appreciation for both previously known and more recently discovered cytokines, chemokines, and feedback mechanisms. Given the strong association between excessive Th17 activity and human disease, new therapeutic approaches targeting Th17 cells are highly promising, but the potential safety of such treatments may be limited by the role of these cells in normal host defenses against infection.
Collapse
Affiliation(s)
- Laura A Tesmer
- Department of Internal Medicine, Division of Rheumatology, Rheumatic Disease Core Center, University of Michigan, Ann Arbor, MI 48109-5358, USA
| | | | | | | |
Collapse
|
30
|
Twenhafel NA, Alves DA, Purcell BK. Pathology of inhalational Francisella tularensis spp. tularensis SCHU S4 infection in African green monkeys (Chlorocebus aethiops). Vet Pathol 2009; 46:698-706. [PMID: 19276059 DOI: 10.1354/vp.08-vp-0302-t-am] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tularemia, caused by Francisella tularensis, is a sporadic zoonotic disease with the potential to be an agent of biowarfare or bioterrorism. We describe here the gross, histologic, immunohistochemical, and ultrastructural findings in a group of 5 African green monkeys (AGMs) that received an average inhaled dose of 729 colony-forming units of F. tularensis and died or were euthanatized between days 7 and 11 post infection. Clinical changes were evident by 48 hours post infection, and key physiologic abnormalities included increases in body temperature, heart rate, peak cardiac pressure, and mean blood pressure. Prominent gross changes in all cases included numerous pinpoint to 1-cm, well-demarcated, necrotic foci present consistently in the lungs, mediastinal lymph nodes, and spleen but also seen in the heart, mediastinum, diaphragm, liver, urinary bladder, urethra, and mesentery. The lungs, mediastinal lymph nodes, and spleen were most severely affected, with as much as 50% of the tissue replaced by necrotic foci. Histologic changes in all tissues consisted of well-delineated foci of necrosis and neutrophilic and histiocytic inflammation, with varying amounts of hemorrhage, edema, fibrin, and vasculitis. Some lesions were immature pyogranulomas. Strong immunoreactivity was identified primarily within macrophages. Ultrastructurally, bacteria were present within cytoplasmic vacuoles of alveolar macrophages, many of which were degenerate. In summary, AGMs infected with F. tularensis by aerosol develop lethal multisystemic disease that particularly targets the lungs and lymphoid tissues. Thus, AGMs should serve as a suitable and reliable animal model for further studies of tularemia.
Collapse
Affiliation(s)
- N A Twenhafel
- United States Army Medical Research Institute of Infectious Diseases, Pathology Division, Fort Detrick, MD 21702, USA.
| | | | | |
Collapse
|
31
|
LPS and poly I:C induce chromatin modifications at a novel upstream region of the IL-23 p19 promoter. Inflammation 2008; 31:235-46. [PMID: 18587636 DOI: 10.1007/s10753-008-9070-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IL-23, a heterodimer of IL-12 p40 and IL-23 p19, is critical for an effective immune response to many infections and has been implicated in several autoimmune diseases, however, little is known about the regulation of IL-23 gene expression in monocytes. We found that poly I:C, LPS, flagellin, and zymogen activated significant IL-23 production in primary human monocytes. Using chromatin immunoprecipitation, we found that a distal upstream region of the IL-23 p19 promoter at -601 to -521 underwent extensive histone modifications in response to stimuli. This distal region of the promoter is not highly conserved between species and has not been previously implicated in the regulation of IL-23 expression. Knockdown of CBP markedly decreased IL-23 p19 responses to poly I:C but had a less dramatic effect on LPS responses, confirming different chromatin responses to these two stimuli. Our data suggest that one of the mechanisms regulating IL-23 expression is the regulation of histone modifications at this distal upstream region of the promoter.
Collapse
|
32
|
Microarray analysis of human monocytes infected with Francisella tularensis identifies new targets of host response subversion. PLoS One 2008; 3:e2924. [PMID: 18698339 PMCID: PMC2488368 DOI: 10.1371/journal.pone.0002924] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Accepted: 07/18/2008] [Indexed: 12/11/2022] Open
Abstract
Francisella tularensis is a gram-negative facultative bacterium that causes the disease tularemia, even upon exposure to low numbers of bacteria. One critical characteristic of Francisella is its ability to dampen or subvert the host immune response. In order to help understand the mechanisms by which this occurs, we performed Affymetrix microarray analysis on transcripts from blood monocytes infected with the virulent Type A Schu S4 strain. Results showed that expression of several host response genes were reduced such as those associated with interferon signaling, Toll-like receptor signaling, autophagy and phagocytosis. When compared to microarrays from monocytes infected with the less virulent F. tularensis subsp. novicida, we found qualitative differences and also a general pattern of quantitatively reduced pro-inflammatory signaling pathway genes in the Schu S4 strain. Notably, the PI3K / Akt1 pathway appeared specifically down-regulated following Schu S4 infection and a concomitantly lower cytokine response was observed. This study identifies several new factors potentially important in host cell subversion by the virulent Type A F. tularensis that may serve as novel targets for drug discovery.
Collapse
|
33
|
Parsa KVL, Butchar JP, Rajaram MVS, Cremer TJ, Gunn JS, Schlesinger LS, Tridandapani S. Francisella gains a survival advantage within mononuclear phagocytes by suppressing the host IFNgamma response. Mol Immunol 2008; 45:3428-37. [PMID: 18514317 PMCID: PMC2577832 DOI: 10.1016/j.molimm.2008.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 03/31/2008] [Accepted: 04/02/2008] [Indexed: 10/22/2022]
Abstract
Tularemia is a zoonotic disease caused by the Gram-negative intracellular pathogen Francisella tularensis. These bacteria evade phagolysosomal fusion, escape from the phagosome and replicate in the host cell cytoplasm. IFNgamma has been shown to suppress the intra-macrophage growth of Francisella through both nitric oxide-dependent and -independent pathways. Since Francisella is known to subvert host immune responses, we hypothesized that this pathogen could interfere with IFNgamma signaling. Here, we report that infection with Francisella suppresses IFNgamma-induced STAT1 expression and phosphorylation in both human and murine mononuclear phagocytes. This suppressive effect of Francisella is independent of phagosomal escape or replication and is mediated by a heat-stable and constitutively expressed bacterial factor. An analysis of the molecular mechanism of STAT1 inhibition indicated that expression of SOCS3, an established negative regulator of IFNgamma signaling, is highly up-regulated during infection and suppresses STAT1 phosphorylation. Functional analyses revealed that this interference with IFNgamma signaling is accompanied by the suppression of IP-10 production and iNOS induction resulting in increased intracellular bacterial survival. Importantly, the suppressive effect on IFNgamma-mediated host cell protection is most effective when IFNgamma is added post infection, suggesting that the bacteria establish a permissive environment within the host cell.
Collapse
Affiliation(s)
- Kishore V L Parsa
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
SUMMARY Our understanding of the role of T cells in human disease is undergoing revision as a result of the discovery of T-helper 17 (Th17) cells, a unique CD4(+) T-cell subset characterized by production of interleukin-17 (IL-17). IL-17 is a highly inflammatory cytokine with robust effects on stromal cells in many tissues. Recent data in humans and mice suggest that Th17 cells play an important role in the pathogenesis of a diverse group of immune-mediated diseases, including psoriasis, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and asthma. Initial reports also propose a role for Th17 cells in tumorigenesis and transplant rejection. Important differences, as well as many similarities, are emerging when the biology of Th17 cells in the mouse is compared with corresponding phenomena in humans. As our understanding of human Th17 biology grows, the mechanisms underlying many diseases are becoming more apparent, resulting in a new appreciation for both previously known and more recently discovered cytokines, chemokines, and feedback mechanisms. Given the strong association between excessive Th17 activity and human disease, new therapeutic approaches targeting Th17 cells are highly promising, but the potential safety of such treatments may be limited by the role of these cells in normal host defenses against infection.
Collapse
Affiliation(s)
- Laura A. Tesmer
- Division of Rheumatology, Department of Internal Medicine and Rheumatic Disease Core Center, University of Michigan, Ann Arbor, MI, USA
| | - Steven K. Lundy
- Division of Rheumatology, Department of Internal Medicine and Rheumatic Disease Core Center, University of Michigan, Ann Arbor, MI, USA
| | - Sujata Sarkar
- Division of Rheumatology and the Arizona Arthritis Center Department of Internal Medicine, University of Arizona, Tucson, AZ, USA
| | - David A. Fox
- Division of Rheumatology, Department of Internal Medicine and Rheumatic Disease Core Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
35
|
Tournier JN, Mohamadzadeh M. Microenvironmental impact on lung cell homeostasis and immunity during infection. Expert Rev Vaccines 2008; 7:457-66. [PMID: 18444892 DOI: 10.1586/14760584.7.4.457] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The lung is a vital organ devoted mainly to gas exchange with an external environment that may be contaminated with various life-threatening pathogens and inert particles. Lung immunity must be permanently balanced between costimulatory and coinhibitory signals, thus controlling potential pathogens while avoiding detrimental inflammation. The lung harbors macrophages and dendritic cells (myeloid and plasmacytoid), which orchestrate the primary defense against microbial invaders. During an infection involving host-microbial synapses, microbes either escape by using host cell physiology or are eliminated by a robust immune response. We thus focus on the dynamics of such cellular interactions within the lung and stress the critical role played by airway epithelial cells in modulating immunity.
Collapse
Affiliation(s)
- Jean-Nicolas Tournier
- Centre de Recherches du Service de Santédes Armées, Unité Interactions Hôte-Pathogéne, La Tronche, France.
| | | |
Collapse
|
36
|
Dobreva ZG, Stanilova SA, Miteva LD. Differences in the inducible gene expression and protein production of IL-12p40, IL-12p70 and IL-23: involvement of p38 and JNK kinase pathways. Cytokine 2008; 43:76-82. [PMID: 18499470 DOI: 10.1016/j.cyto.2008.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 03/14/2008] [Accepted: 04/05/2008] [Indexed: 11/18/2022]
Abstract
The proper balance between IL-12p40-related cytokines controls the appearance of normal and pathological Th1 immune responses. In this study, we examined the inducible IL-12p40, IL-12p35 and IL-23p19 mRNA expression and protein production in human peripheral blood mononuclear cells (PBMC) and purified monocytes, isolated from healthy donors. We investigated how JNK and p38 MAPKs inhibitors influenced IL-12p40, IL-12p70 and IL-23 production. The cytokines' quantity determination was performed by ELISA. qRT-PCR was performed for mRNA transcripts detection. All stimuli tested induced higher level of IL-12p40 and IL-12p19 mRNAs. LPS was the strongest inducer of IL-12p40 mRNA, whereas C3bgp stimulated the highest expression of IL-23p19 mRNA in human monocytes. IL-12p40 and IL-23 protein production observed was increased in the highest level after C3bgp stimulation. The inhibition of both JNK and p38 augmented IL-12p40 production. The inhibition of p38 MAPK downregulated IL-23 production and upregulated IL-12p40 production in stimulated monocytes and PBMC. These results provide evidence that in human monocytes and PBMC p38 MAP kinase activation has an opposite effect on the IL-12p40 and IL-23 expression.
Collapse
Affiliation(s)
- Zlatka Georgieva Dobreva
- Department of Molecular Biology, Immunology & Genetics, Faculty of Medicine, Trakia University, Armeiska 11, 6000 Stara Zagora, Bulgaria.
| | | | | |
Collapse
|
37
|
Parsa KVL, Butchar JP, Rajaram MVS, Cremer TJ, Tridandapani S. The tyrosine kinase Syk promotes phagocytosis of Francisella through the activation of Erk. Mol Immunol 2008; 45:3012-21. [PMID: 18295889 PMCID: PMC2408723 DOI: 10.1016/j.molimm.2008.01.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Accepted: 01/15/2008] [Indexed: 11/29/2022]
Abstract
Francisella tularensis is a highly infectious, Gram-negative intra-cellular pathogen that can cause the zoonotic disease tularemia. Although the receptors critical for internalization of Francisella by macrophages are beginning to be defined, the identity of the downstream signaling pathways essential for the engulfment are not yet identified. In this study we have tested the role of Syk in the phagocytosis of Francisella. We report that Syk is activated during Francisella infection and is critical for the uptake of the organisms. Pharmacologic inhibition of Syk almost completely abrogated uptake, whereas the overexpression of Syk significantly enhanced uptake. However, Syk appears to be dispensable during initial host-pathogen contact. Further analyses of the molecular mechanism of Syk influence on Francisella uptake revealed that the MAPK Erk but not the phosphatidylinositol 3 kinase (PI3K)/Akt pathway is the downstream effector of Syk. Thus, the inhibition of Erk in Syk-overexpressing cells or the inhibition of Syk in Erk-overexpressing cells led to a significant attenuation of uptake. Collectively, these data identify Syk and Erk as key players in the phagocytosis of Francisella.
Collapse
Affiliation(s)
- Kishore V L Parsa
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
38
|
Butchar JP, Parsa KVL, Marsh CB, Tridandapani S. IFNgamma enhances IL-23 production during Francisella infection of human monocytes. FEBS Lett 2008; 582:1044-8. [PMID: 18319062 PMCID: PMC2376054 DOI: 10.1016/j.febslet.2008.02.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 02/21/2008] [Accepted: 02/22/2008] [Indexed: 11/16/2022]
Abstract
We previously demonstrated that monocytes produce IL-23 during Francisella infection, and that IL-23 induces IFNgamma from NK cells. Here, we demonstrate that IFNgamma-priming of monocytes enhances IL-23 production during Francisella infection. This effect was seen on the IL12/23 p40 subunit. Induction of IL-12/23 p40 is reported to be enhanced by IRF-1 and IRF-8. Consistently, microarray analysis of IFNgamma-treated monocytes revealed a significant induction of the IRFs. Interestingly, IFNgamma-primed monocytes produced IL-12 p70, a more potent inducer of IFNgamma than IL-23. We propose that there exists an amplification loop between monocyte IL-23 and NK/T cell IFNgamma that leads to IL-12 p70 production.
Collapse
Affiliation(s)
- Jonathan P. Butchar
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | | | - Clay B. Marsh
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Susheela Tridandapani
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
39
|
Mise-Omata S, Kuroda E, Niikura J, Yamashita U, Obata Y, Doi TS. A Proximal κB Site in the IL-23 p19 Promoter Is Responsible for RelA- and c-Rel-Dependent Transcription. THE JOURNAL OF IMMUNOLOGY 2007; 179:6596-603. [DOI: 10.4049/jimmunol.179.10.6596] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|