51
|
The Enigmatic Role of Viruses in Multiple Sclerosis: Molecular Mimicry or Disturbed Immune Surveillance? Trends Immunol 2017; 38:498-512. [PMID: 28549714 PMCID: PMC7185415 DOI: 10.1016/j.it.2017.04.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 01/24/2023]
Abstract
Multiple sclerosis (MS) is a T cell driven autoimmune disease of the central nervous system (CNS). Despite its association with Epstein-Barr Virus (EBV), how viral infections promote MS remains unclear. However, there is increasing evidence that the CNS is continuously surveyed by virus-specific T cells, which protect against reactivating neurotropic viruses. Here, we discuss how viral infections could lead to the breakdown of self-tolerance in genetically predisposed individuals, and how the reactivations of viruses in the CNS could induce the recruitment of both autoaggressive and virus-specific T cell subsets, causing relapses and progressive disability. A disturbed immune surveillance in MS would explain several experimental findings, and has important implications for prognosis and therapy. A huge body of evidence suggests that viral infections promote MS; however, no single causal virus has been identified. Multiple viruses could promote MS via bystander effects. Molecular mimicry is an established pathogenic mechanism in selected autoimmune diseases. It is also well documented in MS, but its contribution to MS pathogenesis is still unclear. Bystander activation upon viral infection could be involved in the generation of the autoreactive and potentially encephalitogenic T helper (Th)-1/17 central memory (Th1/17CM) cells found in the circulation of patients with MS. Autoreactive Th1/17CM cells could expand at the cost of antiviral Th1CM cells in patients with MS, in particular in those undergoing natalizumab therapy, because these cells are expected to compete for the same homeostatic niche. Autoreactive Th1/17 cells and antiviral Th1 cells are recruited to the CSF of patients with MS following attacks, suggesting that viral reactivations in the CNS induce the recruitment of pathogenic Th1/17 cells. Autoreactive Th1/17 cells in the CNS might also induce de novo viral reactivations in a circuit of self-induced inflammation.
Collapse
|
52
|
Sharma J, Bhar S, Devi CS. A review on interleukins: The key manipulators in rheumatoid arthritis. Mod Rheumatol 2017; 27:723-746. [DOI: 10.1080/14397595.2016.1266071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jatin Sharma
- School of Biosciences and Technology, VIT University, Vellore, India
| | - Sutonuka Bhar
- School of Biosciences and Technology, VIT University, Vellore, India
| | - C. Subathra Devi
- School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|
53
|
Tahara M, Tsuboi H, Segawa S, Asashima H, Iizuka-Koga M, Hirota T, Takahashi H, Kondo Y, Matsui M, Matsumoto I, Sumida T. RORγt antagonist suppresses M3 muscarinic acetylcholine receptor-induced Sjögren's syndrome-like sialadenitis. Clin Exp Immunol 2016; 187:213-224. [PMID: 27643385 DOI: 10.1111/cei.12868] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2016] [Indexed: 01/09/2023] Open
Abstract
We showed recently that M3 muscarinic acetylcholine receptor (M3R)-reactive CD3+ T cells play a pathogenic role in the development of murine autoimmune sialadenitis (MIS), which mimics Sjögren's syndrome (SS). The aim of this study was to determine the effectiveness and mechanism of action of retinoic acid-related orphan receptor-gamma t (RORγt) antagonist (A213) in MIS. Splenocytes from M3R knockout (M3R-/- ) mice immunized with murine M3R peptide mixture were inoculated into recombination-activating gene 1 knockout (Rag-1-/- ) mice (M3R-/- →Rag-1-/- ) with MIS. Immunized M3R-/- mice (pretransfer treatment) and M3R-/- →Rag-1-/- mice (post-transfer treatment) were treated with A213 every 3 days. Salivary volume, severity of sialadenitis and cytokine production from M3R peptide-stimulated splenocytes and lymph node cells were examined. Effects of A213 on cytokine production were analysed by enzyme-linked immunosorbent assay (ELISA) and on T helper type 1 (Th1), Th17 and Th2 differentiation from CD4+ T cells by flow cytometry. Pretransfer A213 treatment maintained salivary volume, improved MIS and reduced interferon (IFN)-γ and interleukin (IL)-17 production significantly compared with phosphate-buffered saline (PBS) (P < 0·05). These suppressive effects involved CD4+ T cells rather than CD11c+ cells. Post-transfer treatment with A213 increased salivary volume (P < 0·05), suppressed MIS (P < 0·005) and reduced IFN-γ and IL-17 production (P < 0·05). In vitro, A213 suppressed IFN-γ and IL-17 production from M3R-stimulated splenocytes and CD4+ T cells of immunized M3R-/- mice (P < 0·05). In contrast with M3R specific responses, A213 suppressed only IL-17 production from Th17 differentiated CD4+ T cells without any effect on Th1 and Th2 differentiation in vitro. Our findings suggested that RORγt antagonism is potentially suitable treatment strategy for SS-like sialadenitis through suppression of IL-17 and IFN-γ production by M3R-specific T cells.
Collapse
Affiliation(s)
- M Tahara
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - H Tsuboi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - S Segawa
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - H Asashima
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - M Iizuka-Koga
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - T Hirota
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - H Takahashi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - Y Kondo
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - M Matsui
- Department of Internal Medicine, Fureai Higashitotsuka Hospital, Yokohama, Kanagawa, Japan
| | - I Matsumoto
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| | - T Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki
| |
Collapse
|
54
|
Kaiser Y, Lepzien R, Kullberg S, Eklund A, Smed-Sörensen A, Grunewald J. Expanded lung T-bet+RORγT+ CD4+ T-cells in sarcoidosis patients with a favourable disease phenotype. Eur Respir J 2016; 48:484-94. [DOI: 10.1183/13993003.00092-2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/29/2016] [Indexed: 11/05/2022]
Abstract
Disease phenotypes of pulmonary sarcoidosis are distinguished by clinical rather than immunological criteria. We aimed to characterise patterns of CD4+ T-cell lineage plasticity underlying the differences in clinical presentation and disease course between the acute form, Löfgren's syndrome, and the heterogeneous, potentially progressive “non-Löfgren” form.33 pulmonary sarcoidosis patients and nine controls underwent bronchoscopy with bronchoalveolar lavage. CD4+ T-cell transcription factor, chemokine receptor and T-cell receptor expression, proliferation and cytokine production were assessed in the lavage fluid and peripheral blood using flow cytometry and multicolour FluoroSpot.CD4+ T-cells simultaneously expressing the T-helper cell (Th)1 and Th17 transcriptional regulators T-bet and RORγT (T-bet+RORγT+) were identified in the lavage, but not blood, of all subjects, and to a significantly higher degree in Löfgren's patients. T-bet+RORγT+ cells proliferated actively, produced interferon (IFN)γ and interleukin (IL)-17A, co-expressed the chemokine receptors CXCR3 and CCR6, and correlated with nonchronic disease. T-cell receptor-restricted Vα2.3+Vβ22+ T-cells strongly co-expressed T-bet/RORγT and CXCR3/CCR6. Cytokine production was more heterogeneous in Löfgren's patients, with significantly higher IL-17A, IL-10, IL-22 and IL-2, but lower IFNγ.Here we demonstrate the presence of lung T-bet+RORγT+CXCR3+CCR6+ CD4+ T-cells and Th17-associated cytokines especially in sarcoidosis patients with a favourable prognosis, suggesting a Th1/Th17-permissive environment in the lung with implications for disease resolution.
Collapse
|
55
|
Smith C, Buhlmann JE, Wang X, Bartlett A, Lim B, Barrington RA. CD275-Independent IL-17-Producing T Follicular Helper-like Cells in Lymphopenic Autoimmune-Prone Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:4935-46. [PMID: 27183569 DOI: 10.4049/jimmunol.1402193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 04/15/2016] [Indexed: 12/27/2022]
Abstract
T cells undergo homeostatic expansion and acquire an activated phenotype in lymphopenic microenvironments. Restoration of normal lymphocyte numbers typically re-establishes normal homeostasis, and proinflammatory cytokine production returns to baseline. Mice deficient in guanine nucleotide exchange factor RasGRP1 exhibit dysregulated homeostatic expansion, which manifests as lymphoproliferative disease with autoantibody production. Our previous work revealed that autoreactive B cells lacking RasGRP1 break tolerance early during development, as well as during germinal center responses, suggesting that T cell-independent and T cell-dependent mechanisms are responsible. Examination of whether a particular T cell subset is involved in the breach of B cell tolerance revealed increased Th17 cells in Rasgrp1-deficient mice relative to control mice. Rasgrp1-deficient mice lacking IL-17R had fewer germinal centers, and germinal centers that formed contained fewer autoreactive B cells, suggesting that IL-17 signaling is required for a break in B cell tolerance in germinal centers. Interestingly, a fraction of Th17 cells from Rasgrp1-deficient mice were CXCR5(+) and upregulated levels of CD278 coordinate with their appearance in germinal centers, all attributes of T follicular helper cells (Tfh17). To determine whether CD278-CD275 interactions were required for the development of Tfh17 cells and for autoantibody, Rasgrp1-deficient mice were crossed with CD275-deficient mice. Surprisingly, mice deficient in RasGRP1 and CD275 formed Tfh17 cells and germinal centers and produced similar titers of autoantibodies as mice deficient in only RasGRP1. Therefore, these studies suggest that requirements for Tfh cell development change in lymphopenia-associated autoimmune settings.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| | - Janet E Buhlmann
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | - Xiaogan Wang
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| | - Amber Bartlett
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| | - Bing Lim
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | - Robert A Barrington
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| |
Collapse
|
56
|
Layman AAK, Oliver PM. Ubiquitin Ligases and Deubiquitinating Enzymes in CD4+ T Cell Effector Fate Choice and Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:3975-82. [PMID: 27183634 PMCID: PMC5738552 DOI: 10.4049/jimmunol.1502660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/10/2016] [Indexed: 12/22/2022]
Abstract
The human body is exposed to potentially pathogenic microorganisms at barrier sites such as the skin, lungs, and gastrointestinal tract. To mount an effective response against these pathogens, the immune system must recruit the right cells with effector responses that are appropriate for the task at hand. Several types of CD4(+) T cells can be recruited, including Th cells (Th1, Th2, and Th17), T follicular helper cells, and regulatory T cells. These cells help to maintain normal immune homeostasis in the face of constantly changing microbes in the environment. Because these cells differentiate from a common progenitor, the composition of their intracellular milieu of proteins changes to appropriately guide their effector function. One underappreciated process that impacts the levels and functions of effector fate-determining factors is ubiquitylation. This review details our current understanding of how ubiquitylation regulates CD4(+) T cell effector identity and function.
Collapse
Affiliation(s)
- Awo A K Layman
- Medical Scientist Training Program, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104; and
| | - Paula M Oliver
- Department of Pathology, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
57
|
IL-33 in T Cell Differentiation, Function, and Immune Homeostasis. Trends Immunol 2016; 37:321-333. [DOI: 10.1016/j.it.2016.03.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 01/01/2023]
|
58
|
Rathore JS, Wang Y. Protective role of Th17 cells in pulmonary infection. Vaccine 2016; 34:1504-1514. [PMID: 26878294 DOI: 10.1016/j.vaccine.2016.02.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 01/14/2023]
Abstract
Th17 cells are characterized as preferential producer of interleukins including IL-17A, IL-17F, IL-21 and IL-22. Corresponding receptors of these cytokines are expressed on number of cell types found in the mucosa, including epithelial cells and fibroblasts which constitute the prime targets of the Th17-associated cytokines. Binding of IL-17 family members to their corresponding receptors lead to modulation of antimicrobial functions of target cells including alveolar epithelial cells. Stimulated alveolar epithelial cells produce antimicrobial peptides and are involved in granulepoesis, neutrophil recruitment and tissue repair. Mucosal immunity mediated by Th17 cells is protective against numerous pulmonary pathogens including extracellular bacterial and fungal pathogens. This review focuses on the protective role of Th17 cells during pulmonary infection, highlighting subset differentiation, effector cytokines production, followed by study of the binding of these cytokines to their corresponding receptors, the subsequent signaling pathway they engender and their effector role in host defense.
Collapse
Affiliation(s)
- Jitendra Singh Rathore
- University of Pennsylvania, Perelman School of Medicine, Department of Microbiology, Philadelphia, PA, USA; Gautam Buddha University, School of Biotechnology, Greater Noida, Yamuna Expressway, Uttar Pradesh, India.
| | - Yan Wang
- University of Pennsylvania, Perelman School of Medicine, Department of Microbiology, Philadelphia, PA, USA
| |
Collapse
|
59
|
Abstract
Microscopic colitis (MC) is a common cause of chronic diarrhea. The 2 most frequent forms of MC are collagenous colitis and lymphocytic colitis. Over the past years, the incidence and prevalence of microscopic colitis are rising and this is largely attributed to a greater awareness, and concomitantly an increasing number of diagnoses. Patients with microscopic colitis report watery, nonbloody diarrhea of chronic course, abdominal pain, weight loss, and fatigue that may impair patient's health-related quality of life. The underlying mechanisms involved in the pathogenesis of microscopic colitis remain unspecified but is probably multifactorial. Collagenous colitis and lymphocytic colitis may represent specific mucosal responses to different luminal agents in predisposed individuals, resulting in an uncontrolled immune response. Genetic predisposition, altered modulation of cytokines and miRNAs, and aberrant response to drugs seem to be involved in the development of MC. Despite the progress of knowledge, still many questions remain unsolved regarding the etiology, pathophysiology, and optimal management of MC. This review gives an update on the immunological aspects of collagenous colitis and lymphocytic colitis.
Collapse
|
60
|
Xie C, Ciric B, Yu S, Zhang GX, Rostami A. IL-12Rβ2 has a protective role in relapsing-remitting experimental autoimmune encephalomyelitis. J Neuroimmunol 2015; 291:59-69. [PMID: 26857496 DOI: 10.1016/j.jneuroim.2015.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 12/10/2015] [Accepted: 12/17/2015] [Indexed: 12/11/2022]
Abstract
IL-12Rβ2 is a common receptor subunit of heterodimeric receptors for IL-12 and IL-35, two cytokines that are implicated in immunopathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. We evaluated the role of IL-12Rβ2 in relapsing-remitting EAE (RR-EAE). IL-12Rβ2-deficient SJL/J mice developed markedly more severe clinical EAE, and had greater mortality and more severe relapses compared with wild-type controls. IL-12Rβ2-deficient EAE mice also had more infiltrating mononuclear cells in the CNS, as well as higher T cell proliferative capacity and decreased IFN-γ production at the periphery. These findings demonstrate a protective role of IL-12Rβ2 in RR-EAE.
Collapse
Affiliation(s)
- Chong Xie
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shuo Yu
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
61
|
Kara EE, McKenzie DR, Bastow CR, Gregor CE, Fenix KA, Ogunniyi AD, Paton JC, Mack M, Pombal DR, Seillet C, Dubois B, Liston A, MacDonald KPA, Belz GT, Smyth MJ, Hill GR, Comerford I, McColl SR. CCR2 defines in vivo development and homing of IL-23-driven GM-CSF-producing Th17 cells. Nat Commun 2015; 6:8644. [PMID: 26511769 PMCID: PMC4639903 DOI: 10.1038/ncomms9644] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/15/2015] [Indexed: 12/22/2022] Open
Abstract
IL-17-producing helper T (Th17) cells are critical for host defense against extracellular pathogens but also drive numerous autoimmune diseases. Th17 cells that differ in their inflammatory potential have been described including IL-10-producing Th17 cells that are weak inducers of inflammation and highly inflammatory, IL-23-driven, GM-CSF/IFNγ-producing Th17 cells. However, their distinct developmental requirements, functions and trafficking mechanisms in vivo remain poorly understood. Here we identify a temporally regulated IL-23-dependent switch from CCR6 to CCR2 usage by developing Th17 cells that is critical for pathogenic Th17 cell-driven inflammation in experimental autoimmune encephalomyelitis (EAE). This switch defines a unique in vivo cell surface signature (CCR6−CCR2+) of GM-CSF/IFNγ-producing Th17 cells in EAE and experimental persistent extracellular bacterial infection, and in humans. Using this signature, we identify an IL-23/IL-1/IFNγ/TNFα/T-bet/Eomesodermin-driven circuit driving GM-CSF/IFNγ-producing Th17 cell formation in vivo. Thus, our data identify a unique cell surface signature, trafficking mechanism and T-cell intrinsic regulators of GM-CSF/IFNγ-producing Th17 cells. Little is known regarding migration of Th17 cells that produce distinct cytokines implicated in protection and pathology. Kara et al. show that a switch from CCR6 to CCR2 by Th17 cells defines a signature (CCR6−CCR2+) of GM-CSF+ Th17 cells and drives pathology in a mouse model of autoimmunity.
Collapse
Affiliation(s)
- Ervin E Kara
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Duncan R McKenzie
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Cameron R Bastow
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Carly E Gregor
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kevin A Fenix
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Abiodun D Ogunniyi
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia.,Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - James C Paton
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia.,Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg 93042, Germany
| | - Diana R Pombal
- Department of Microbiology and Immunology, VIB and University of Leuven, B-3000 Leuven, Belgium
| | - Cyrill Seillet
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Bénédicte Dubois
- Department of Neurosciences, KU-Leuven-University of Leuven, B-3000 Leuven, Belgium
| | - Adrian Liston
- Department of Microbiology and Immunology, VIB and University of Leuven, B-3000 Leuven, Belgium
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Gabrielle T Belz
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia.,School of Medicine, University of Queensland, Herston, Queensland 4006, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia.,The Royal Brisbane and Women's Hospital, Herston, Queensland 4029, Australia
| | - Iain Comerford
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Shaun R McColl
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia.,Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
62
|
Lurati A, Laria A, Gatti A, Brando B, Scarpellini M. Different T cells' distribution and activation degree of Th17 CD4+ cells in peripheral blood in patients with osteoarthritis, rheumatoid arthritis, and healthy donors: preliminary results of the MAGENTA CLICAO study. Open Access Rheumatol 2015; 7:63-68. [PMID: 27790046 PMCID: PMC5045119 DOI: 10.2147/oarrr.s81905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective To determine distribution of T cells and activation degree of Th CD4+ cells in peripheral blood of patients with osteoarthritis (OA), rheumatoid arthritis (RA), and healthy donors. Methods Patients with established diagnosis of RA according to American College of Rheumatology/European League Against Rheumatism 2010 criteria, knee or hip OA according to American College of Rheumatology criteria, and healthy blood donor volunteers were eligible. Multi-channel flow cytometry and monoclonal antibodies against CD3, CD4, CD8, CCR6, CD38, CXCR3, and HLA DR were used to distinguish and evaluate T cells’ subpopulation. Results We analyzed blood samples of 15 patients with well-defined RA, 56 with hip or knee OA, and 20 healthy age matched controls. Blood samples from RA patients showed significantly higher counts of CD4+ CD38+ DR+ (activated CD4 T cells) and Th17 (CCR6+ CXCR3−) cells as compared to OA patients and control group (P<0.01). Furthermore the samples from the OA patients showed a higher percentage of activated CD4 T cells and Th17 cells as compared to control group (P<0.05). Interestingly there was no difference between Th1 (CD4+ CXCR3+ CCR6−) and Th2 (CD4+ CXCR3− CCR6−) between the three groups (P>0.1). Conclusion According to the latest view of OA disease pathogenesis, our preliminary results support the hypothesis that OA may also be a disease with an immunological/inflammatory involvement like RA. It seems that there is a quantitative but non-qualitative difference in Th17 cells’ profile, including the expression of activation markers, between RA and OA.
Collapse
Affiliation(s)
| | | | - Arianna Gatti
- Transfusional Centre, Legnano Hospital, Legnano, Italy
| | - Bruno Brando
- Transfusional Centre, Legnano Hospital, Legnano, Italy
| | | |
Collapse
|
63
|
Arbelaez CA, Glatigny S, Duhen R, Eberl G, Oukka M, Bettelli E. IL-7/IL-7 Receptor Signaling Differentially Affects Effector CD4+ T Cell Subsets Involved in Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26223651 DOI: 10.4049/jimmunol.1403135] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-17-producing CD4(+) T (Th17) cells, along with IFN-γ-expressing Th1 cells, represent two major pathogenic T cell subsets in experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis (MS). The cytokines and transcription factors involved in the development and effector functions of Th1 and Th17 cells have been largely characterized. Among them, IL-23 is essential for the generation of stable and encephalitogenic Th17 cells and for the development of EAE. The IL-7/IL-7R signaling axis participates in cell survival, and perturbation of this pathway has been associated with enhanced susceptibility to MS. A link between IL-23-driven pathogenic T cells and IL-7/IL-7R signaling has previously been proposed, but has not been formally addressed. In the current study, we showed that Th17 cells from mice with EAE express high levels of IL-7Rα compared with Th1 cells. Using mice that constitutively express IL-7Rα on T cells, we determined that sustained IL-7R expression in IL-23R-deficient mice could not drive pathogenic T cells and the development of EAE. IL-7 inhibited the differentiation of Th17 cells, but promoted IFN-γ and GM-CSF secretion in vitro. In vivo IL-7/anti-IL-7 mAb complexes selectively expanded and enhanced the proliferation of CXCR3-expressing Th1 cells, but did not impact Th17 cells and EAE development in wild-type and IL-23R-deficient mice. Importantly, high IL-7 expression was detected in the CNS during EAE and could drive the plasticity of Th17 cells to IFN-γ-producing T cells. Together, these data address the contribution of IL-23/IL-23R and IL-7/IL-7R signaling in Th17 and Th1 cell dynamics during CNS autoimmunity.
Collapse
Affiliation(s)
- Carlos A Arbelaez
- Department of Immunology, University of Washington, Seattle, WA 98101; Immunology Program, Benaroya Research Institute, Seattle, WA 98101
| | - Simon Glatigny
- Department of Immunology, University of Washington, Seattle, WA 98101; Immunology Program, Benaroya Research Institute, Seattle, WA 98101
| | - Rebekka Duhen
- Department of Immunology, University of Washington, Seattle, WA 98101; Immunology Program, Benaroya Research Institute, Seattle, WA 98101
| | - Gerard Eberl
- Lymphoid Tissue Development Unit, Pasteur Institute, Paris 75724, France; and
| | - Mohamed Oukka
- Department of Immunology, University of Washington, Seattle, WA 98101; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101
| | - Estelle Bettelli
- Department of Immunology, University of Washington, Seattle, WA 98101; Immunology Program, Benaroya Research Institute, Seattle, WA 98101;
| |
Collapse
|
64
|
Li P, Spolski R, Liao W, Leonard WJ. Complex interactions of transcription factors in mediating cytokine biology in T cells. Immunol Rev 2015; 261:141-56. [PMID: 25123282 DOI: 10.1111/imr.12199] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
T-helper (Th) cells play critical roles within the mammalian immune system, and the differentiation of naive CD4(+) T cells into distinct T-helper subsets is critical for normal immunoregulation and host defense. These carefully regulated differentiation processes are controlled by networks of cytokines, transcription factors, and epigenetic modifications, resulting in the generation of multiple CD4(+) T-cell subsets, including Th1, Th2, Th9, Th17, Treg, and Tfh cells. In this review, we discuss the roles of transcription factors in determining the specific type of differentiation and in particular the role of interleukin-2 (IL-2) in promoting or inhibiting Th differentiation. In addition to discussing master regulators and subset-specific transcription factors for distinct T-helper cell populations, we focus on signal transducer and activator of transcription (STAT) proteins and on the cooperative action of interferon regulatory factor 4 (IRF4) with activator protein 1 (AP-1) family proteins and STAT3 in the assembly of complexes that broadly influence T-cell differentiation.
Collapse
Affiliation(s)
- Peng Li
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
65
|
Schilbach K, Alkhaled M, Welker C, Eckert F, Blank G, Ziegler H, Sterk M, Müller F, Sonntag K, Wieder T, Braumüller H, Schmitt J, Eyrich M, Schleicher S, Seitz C, Erbacher A, Pichler BJ, Müller H, Tighe R, Lim A, Gillies SD, Strittmatter W, Röcken M, Handgretinger R. Cancer-targeted IL-12 controls human rhabdomyosarcoma by senescence induction and myogenic differentiation. Oncoimmunology 2015; 4:e1014760. [PMID: 26140238 PMCID: PMC4485786 DOI: 10.1080/2162402x.2015.1014760] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/27/2015] [Accepted: 01/29/2015] [Indexed: 02/07/2023] Open
Abstract
Stimulating the immune system to attack cancer is a promising approach, even for the control of advanced cancers. Several cytokines that promote interferon-γ-dominated immune responses show antitumor activity, with interleukin 12 (IL-12) being of major importance. Here, we used an antibody-IL-12 fusion protein (NHS-IL12) that binds histones of necrotic cells to treat human sarcoma in humanized mice. Following sarcoma engraftment, NHS-IL12 therapy was combined with either engineered IL-7 (FcIL-7) or IL-2 (IL-2MAB602) for continuous cytokine bioavailability. NHS-IL12 strongly induced innate and adaptive antitumor immunity when combined with IL-7 or IL-2. NHS-IL12 therapy significantly improved survival of sarcoma-bearing mice and caused long-term remissions when combined with IL-2. NHS-IL12 induced pronounced cancer cell senescence, as documented by strong expression of senescence-associated p16INK4a and nuclear translocation of p-HP1γ, and permanent arrest of cancer cell proliferation. In addition, this cancer immunotherapy initiated the induction of myogenic differentiation, further promoting the hypothesis that efficient antitumor immunity includes mechanisms different from cytotoxicity for efficient cancer control in vivo.
Collapse
Key Words
- CIP1, CDK-interacting protein 1
- DNAM-1, DNAX accessory molecule-1
- KIR, killer-cell immunoglobulin-like receptor
- M1/M2 macrophages
- MICA/B, MHC class I polypeptide-related sequence A/B
- NKG, natural killer group
- NSG, NOD SCID gamma chain knock out mouse
- PCNA, proliferating cell nuclear antigen
- PVR, poliovirus receptor
- RMS, rhabdomyosarcoma, (eRMS: embryonal, aRMS: alveolar)
- ROI, region of interest
- RORC, RAR-related orphan receptor C
- SCT, stem cell transplantation
- SPECT/CT, single-photon emission computed tomography
- TH1-induced senescence
- TH17 cells
- TRBV, T-cell receptor beta chain
- ULBP, UL16 binding protein
- WAF, wild-type activating fragment
- cancer-targeted IL-12
- differentiation
- humanized mice
- immunocytokine
- immunotherapy
- pHP1γ, phosphorylated heterochromatin protein 1 gamma
- rhabdomyosarcoma
- tumor-infiltrating lymphocytes
Collapse
Affiliation(s)
- Karin Schilbach
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Mohammed Alkhaled
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Christian Welker
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology; Eberhard Karls University ; Tübingen, Germany
| | - Gregor Blank
- Department of General, Visceral and Transplant Surgery; University Hospital ; Tübingen, Germany
| | - Hendrik Ziegler
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Marco Sterk
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Friederike Müller
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Katja Sonntag
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Thomas Wieder
- Department of Dermatology; Eberhard Karls University ; Tübingen, Germany
| | - Heidi Braumüller
- Department of Dermatology; Eberhard Karls University ; Tübingen, Germany
| | - Julia Schmitt
- Werner Siemens Imaging Center; Department for Preclinical Imaging and Radiopharmacy; Eberhard Karls University ; Tübingen, Germany
| | - Matthias Eyrich
- University of Würzburg; Department of Pediatrics; Interdisciplinary Stem Cell Laboratory ; Würzburg, Germany
| | - Sabine Schleicher
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Christian Seitz
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Annika Erbacher
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center; Department for Preclinical Imaging and Radiopharmacy; Eberhard Karls University ; Tübingen, Germany
| | - Hartmut Müller
- Department of General Pathology; Institute of Pathology; Eberhard Karls University ; Tübingen, Germany
| | - Robert Tighe
- EMD Serono Research Institute ; Billerica, MA USA
| | - Annick Lim
- Départment d'Immunologie; Institute Pasteur ; Paris, France
| | | | | | - Martin Röcken
- Department of Dermatology; Eberhard Karls University ; Tübingen, Germany
| | - Rupert Handgretinger
- Department of General Pediatrics; Oncology/Hematology; University Children's Hospital ; Tübingen, Germany
| |
Collapse
|
66
|
Mazzoni A, Santarlasci V, Maggi L, Capone M, Rossi MC, Querci V, De Palma R, Chang HD, Thiel A, Cimaz R, Liotta F, Cosmi L, Maggi E, Radbruch A, Romagnani S, Dong J, Annunziato F. Demethylation of the RORC2 and IL17A in Human CD4+ T Lymphocytes Defines Th17 Origin of Nonclassic Th1 Cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:3116-26. [DOI: 10.4049/jimmunol.1401303] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
67
|
Christie D, Zhu J. Transcriptional regulatory networks for CD4 T cell differentiation. Curr Top Microbiol Immunol 2015; 381:125-72. [PMID: 24839135 DOI: 10.1007/82_2014_372] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD4(+) T cells play a central role in controlling the adaptive immune response by secreting cytokines to activate target cells. Naïve CD4(+) T cells differentiate into at least four subsets, Th1Th1 , Th2Th2 , Th17Th17 , and inducible regulatory T cellsregulatory T cells , each with unique functions for pathogen elimination. The differentiation of these subsets is induced in response to cytokine stimulation, which is translated into Stat activation, followed by induction of master regulator transcription factorstranscription factors . In addition to these factors, multiple other transcription factors, both subset specific and shared, are also involved in promoting subset differentiation. This review will focus on the network of transcription factors that control CD4(+) T cell differentiation.
Collapse
Affiliation(s)
- Darah Christie
- Molecular and Cellular Immunoregulation Unit, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA,
| | | |
Collapse
|
68
|
McAllister F, Kolls JK. Th17 cytokines in non-melanoma skin cancer. Eur J Immunol 2015; 45:692-4. [PMID: 25655439 PMCID: PMC4461870 DOI: 10.1002/eji.201545456] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 01/28/2015] [Accepted: 02/04/2015] [Indexed: 11/10/2022]
Abstract
T-helper-type 17 cytokines have been implicated in epithelial cancer progression at mucosal sites. In this issue of the European Journal of Immunology, Nardinocchi et al. [Eur. J. Immunol. 2015. 45: 922-931] show that the Th17 cytokines IL-17 and IL-22 can both signal to nonmelanoma skin cancer cells, inducing both cellular proliferation and enhanced migration of human basal cell carcinoma and squamous cell carcinoma cell lines in vitro. These cytokines were also shown to exacerbate tumor growth in mice injected with the squamous cell carcinoma line, CAL27. Thus, IL-17 and IL-22 may be key factors in skin cancer progression and may provide novel prognostic markers in nonmelanoma skin cancer.
Collapse
Affiliation(s)
- Florencia McAllister
- Department of Clinical Cancer Prevention. The University of Texas MD Anderson Cancer Center. Houston, TX
| | - Jay K. Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
69
|
Dietz L, Frommer F, Vogel AL, Vaeth M, Serfling E, Waisman A, Buttmann M, Berberich-Siebelt F. NFAT1 deficit and NFAT2 deficit attenuate EAE via different mechanisms. Eur J Immunol 2015; 45:1377-89. [DOI: 10.1002/eji.201444638] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 09/30/2014] [Accepted: 01/27/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Lena Dietz
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Friederike Frommer
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
- Institute for Molecular Medicine; University Medical Center of the Johannes Gutenberg; University of Mainz; Mainz Germany
| | - Anna-Lena Vogel
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Martin Vaeth
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Edgar Serfling
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Ari Waisman
- Institute for Molecular Medicine; University Medical Center of the Johannes Gutenberg; University of Mainz; Mainz Germany
| | - Mathias Buttmann
- Department of Neurology; University of Wuerzburg; Wuerzburg Germany
| | - Friederike Berberich-Siebelt
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
- Comprehensive Cancer Center Mainfranken; University of Wuerzburg; Wuerzburg Germany
| |
Collapse
|
70
|
Hoppmann N, Graetz C, Paterka M, Poisa-Beiro L, Larochelle C, Hasan M, Lill CM, Zipp F, Siffrin V. New candidates for CD4 T cell pathogenicity in experimental neuroinflammation and multiple sclerosis. Brain 2015; 138:902-17. [DOI: 10.1093/brain/awu408] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
71
|
Abstract
BACKGROUND Although the present understanding of the immunopathogenesis of rheumatoid inflammation is still incomplete, there is substantial evidence that effector CD4+ T helper (Th) cells play a central role. RESULTS In recent years, in addition to the established Th cell subsets Th1 and Th2 cells, other subsets, such as Th9, Th17, Th22 and T follicular helper (Tfh) cells have been described. Defining the contribution of T cells in the initiation and maintenance of inflammation has been augmented by the identification of functionally distinct subsets of effector Th cells that can be classified based on their cytokine and transcription factor profiles. CONCLUSION Increasing knowledge of the role of these various T cell populations in chronic inflammation provides a better understanding and insights into the pathogenic mechanisms and chronification of rheumatic diseases.
Collapse
Affiliation(s)
- J Leipe
- Sektion für Rheumatologie und Klinische Immunologie, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Pettenkoferstr. 8a, 80336, München, Deutschland,
| | | |
Collapse
|
72
|
Perez S, Fishman S, Bordowitz A, Margalit A, Wong FS, Gross G. Selective immunotargeting of diabetogenic CD4 T cells by genetically redirected T cells. Immunology 2015; 143:609-17. [PMID: 24943731 DOI: 10.1111/imm.12340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 01/10/2023] Open
Abstract
The key role played by islet-reactive CD8 and CD4 T cells in type 1 diabetes calls for new immunotherapies that target pathogenic T cells in a selective manner. We previously demonstrated that genetically linking the signalling portion of CD3-ζ onto the C-terminus of β2 -microglobulin and an autoantigenic peptide to its N-terminus converts MHC-I complexes into functional T-cell receptor-specific receptors. CD8 T cells expressing such receptors specifically killed diabetogenic CD8 T cells, blocked T-cell-induced diabetes in immunodeficient NOD.SCID mice and suppressed disease in wild-type NOD mice. Here we describe the immunotargeting of CD4 T cells by chimeric MHC-II receptors. To this end we chose the diabetogenic NOD CD4 T-cell clone BDC2.5, which recognizes the I-A(g7) -bound 1040-31 mimotope. We assembled several constructs encoding I-A(g7) α- and β-chains, the latter carrying mim or hen egg lysozyme peptide as control, each supplemented with CD3-ζ intracellular portion, either with or without its transmembrane domain. Following mRNA co-transfection of reporter B3Z T cells and mouse CD8 and CD4 T cells, these constructs triggered robust activation upon I-A(g7) cross-linking. A BDC2.5 T-cell hybridoma activated B3Z transfectants expressing the mimotope, but not the control peptide, in both configurations. Potent two-way activation was also evident with transgenic BDC2.5 CD4 T cells, but peptide-specific activation required the CD3-ζ transmembrane domain. Chimeric MHC-II/CD3-ζ complexes therefore allow the selective immunotargeting of islet-reactive CD4 T cells, which take part in the pathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Shira Perez
- Laboratory of Immunology, MIGAL Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | | | | | | | | | | |
Collapse
|
73
|
Nelson MH, Kundimi S, Bowers JS, Rogers CE, Huff LW, Schwartz KM, Thyagarajan K, Little EC, Mehrotra S, Cole DJ, Rubinstein MP, Paulos CM. The inducible costimulator augments Tc17 cell responses to self and tumor tissue. THE JOURNAL OF IMMUNOLOGY 2015; 194:1737-47. [PMID: 25576595 DOI: 10.4049/jimmunol.1401082] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The inducible costimulator (ICOS) plays a key role in the development of Th17 cells, but its role in the development and antitumor activity of IL-17-producing CD8(+) T cells (Tc17) remains unknown. We found that ICOS costimulation was important for the functional maintenance, but not differentiation, of Tc17 cells in vitro. Blocking the ICOS pathway using an antagonist mAb or by using recipient mice genetically deficient in the ICOS ligand reduced the antitumor activity of adoptively transferred Tc17 cells. Conversely, activating Tc17 cells with an ICOS agonist in vitro enhanced their capacity to eradicate melanoma and induce autoimmune vitiligo when infused into mice. However, ICOS stimulation did not augment the antitumor activity of IL-2 expanded T cells. Additional investigation revealed that ICOS stimulation not only increased IL-2Rα, CXCR3, and IL-23R expression on Tc17 cells, but also dampened their expression of suppressive molecule CD39. Although Tc17 cells activated with an ICOS agonist cosecreted heightened IL-17A, IL-9, and IFN-γ, their therapeutic effectiveness was critically dependent on IFN-γ production. Depletion of IL-17A and IL-9 had little impact on antitumor Tc17 cells activated with an ICOS agonist. Collectively, our work reveals that the ICOS pathway potentiates the antitumor activity of adoptively transferred Tc17 cells. This work has major implications for the design of vaccine, Ab and cell-based therapies for autoimmunity, infectious disease, and cancer.
Collapse
Affiliation(s)
- Michelle H Nelson
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and
| | - Sreenath Kundimi
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and
| | - Jacob S Bowers
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and
| | - Carolyn E Rogers
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and
| | - Logan W Huff
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and
| | - Kristina M Schwartz
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and
| | - Krishnamurthy Thyagarajan
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
| | - Elizabeth C Little
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
| | - Shikhar Mehrotra
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
| | - David J Cole
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
| | - Mark P Rubinstein
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
74
|
Geginat J, Paroni M, Maglie S, Alfen JS, Kastirr I, Gruarin P, De Simone M, Pagani M, Abrignani S. Plasticity of human CD4 T cell subsets. Front Immunol 2014; 5:630. [PMID: 25566245 PMCID: PMC4267263 DOI: 10.3389/fimmu.2014.00630] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 11/25/2014] [Indexed: 12/23/2022] Open
Abstract
Human beings are exposed to a variety of different pathogens, which induce tailored immune responses and consequently generate highly diverse populations of pathogen-specific T cells. CD4(+) T cells have a central role in adaptive immunity, since they provide essential help for both cytotoxic T cell- and antibody-mediated responses. In addition, CD4(+) regulatory T cells are required to maintain self-tolerance and to inhibit immune responses that could damage the host. Initially, two subsets of CD4(+) helper T cells were identified that secrete characteristic effector cytokines and mediate responses against different types of pathogens, i.e., IFN-γ secreting Th1 cells that fight intracellular pathogens, and IL-4 producing Th2 cells that target extracellular parasites. It is now well established that this dichotomy is insufficient to describe the complexity of CD4(+) T cell differentiation, and in particular the human CD4 compartment contains a myriad of T cell subsets with characteristic capacities to produce cytokines and to home to involved tissues. Moreover, it has become increasingly clear that these T cell subsets are not all terminally differentiated cells, but that the majority is plastic and that in particular central memory T cells can acquire different properties and functions in secondary immune responses. In addition, there is compelling evidence that helper T cells can acquire regulatory functions upon chronic stimulation in inflamed tissues. The plasticity of antigen-experienced human T cell subsets is highly relevant for translational medicine, since it opens new perspectives for immune-modulatory therapies for chronic infections, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Jens Geginat
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Moira Paroni
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Stefano Maglie
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Johanna Sophie Alfen
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Ilko Kastirr
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Paola Gruarin
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Marco De Simone
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Massimiliano Pagani
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| | - Sergio Abrignani
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" INGM , Milan , Italy
| |
Collapse
|
75
|
Acerbi E, Zelante T, Narang V, Stella F. Gene network inference using continuous time Bayesian networks: a comparative study and application to Th17 cell differentiation. BMC Bioinformatics 2014; 15:387. [PMID: 25495206 PMCID: PMC4267461 DOI: 10.1186/s12859-014-0387-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 11/17/2014] [Indexed: 12/17/2022] Open
Abstract
Background Dynamic aspects of gene regulatory networks are typically investigated by measuring system variables at multiple time points. Current state-of-the-art computational approaches for reconstructing gene networks directly build on such data, making a strong assumption that the system evolves in a synchronous fashion at fixed points in time. However, nowadays omics data are being generated with increasing time course granularity. Thus, modellers now have the possibility to represent the system as evolving in continuous time and to improve the models’ expressiveness. Results Continuous time Bayesian networks are proposed as a new approach for gene network reconstruction from time course expression data. Their performance was compared to two state-of-the-art methods: dynamic Bayesian networks and Granger causality analysis. On simulated data, the methods comparison was carried out for networks of increasing size, for measurements taken at different time granularity densities and for measurements unevenly spaced over time. Continuous time Bayesian networks outperformed the other methods in terms of the accuracy of regulatory interactions learnt from data for all network sizes. Furthermore, their performance degraded smoothly as the size of the network increased. Continuous time Bayesian networks were significantly better than dynamic Bayesian networks for all time granularities tested and better than Granger causality for dense time series. Both continuous time Bayesian networks and Granger causality performed robustly for unevenly spaced time series, with no significant loss of performance compared to the evenly spaced case, while the same did not hold true for dynamic Bayesian networks. The comparison included the IRMA experimental datasets which confirmed the effectiveness of the proposed method. Continuous time Bayesian networks were then applied to elucidate the regulatory mechanisms controlling murine T helper 17 (Th17) cell differentiation and were found to be effective in discovering well-known regulatory mechanisms, as well as new plausible biological insights. Conclusions Continuous time Bayesian networks were effective on networks of both small and large size and were particularly feasible when the measurements were not evenly distributed over time. Reconstruction of the murine Th17 cell differentiation network using continuous time Bayesian networks revealed several autocrine loops, suggesting that Th17 cells may be auto regulating their own differentiation process.
Collapse
Affiliation(s)
- Enzo Acerbi
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Immunos Building, Level 4 138648, Singapore.
| | | | | | | |
Collapse
|
76
|
Reinert-Hartwall L, Honkanen J, Salo HM, Nieminen JK, Luopajärvi K, Härkönen T, Veijola R, Simell O, Ilonen J, Peet A, Tillmann V, Knip M, Vaarala O. Th1/Th17 plasticity is a marker of advanced β cell autoimmunity and impaired glucose tolerance in humans. THE JOURNAL OF IMMUNOLOGY 2014; 194:68-75. [PMID: 25480564 DOI: 10.4049/jimmunol.1401653] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Upregulation of IL-17 immunity and detrimental effects of IL-17 on human islets have been implicated in human type 1 diabetes. In animal models, the plasticity of Th1/Th17 cells contributes to the development of autoimmune diabetes. In this study, we demonstrate that the upregulation of the IL-17 pathway and Th1/Th17 plasticity in peripheral blood are markers of advanced β cell autoimmunity and impaired β cell function in human type 1 diabetes. Activated Th17 immunity was observed in the late stage of preclinical diabetes in children with β cell autoimmunity and impaired glucose tolerance, but not in children with early β cell autoimmunity. We found an increased ratio of IFN-γ/IL-17 expression in Th17 cells in children with advanced β cell autoimmunity, which correlated with HbA1c and plasma glucose concentrations in an oral glucose tolerance test, and thus impaired β cell function. Low expression of Helios was seen in Th17 cells, suggesting that Th1/Th17 cells are not converted thymus-derived regulatory T cells. Our results suggest that the development of Th1/Th17 plasticity may serve as a biomarker of disease progression from β cell autoantibody positivity to type 1 diabetes. These data in human type 1 diabetes emphasize the role of Th1/Th17 plasticity as a potential contributor to tissue destruction in autoimmune conditions.
Collapse
Affiliation(s)
- Linnea Reinert-Hartwall
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Jarno Honkanen
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Harri M Salo
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Janne K Nieminen
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Kristiina Luopajärvi
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland
| | - Taina Härkönen
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland
| | - Riitta Veijola
- Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, 90014 Oulu, Finland
| | - Olli Simell
- Department of Pediatrics, University of Turku, 20520 Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, 20520 Turku, Finland; Department of Clinical Microbiology, University of Eastern Finland, 70211 Kuopio, Finland
| | - Aleksandr Peet
- Department of Pediatrics, University of Tartu and Children's Clinic of Tartu University Hospital, Tartu 51014, Estonia
| | - Vallo Tillmann
- Department of Pediatrics, University of Tartu and Children's Clinic of Tartu University Hospital, Tartu 51014, Estonia
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland; Folkhälsan Research Center, 00290 Helsinki, Finland; Diabetes and Obesity Research Program, University of Helsinki, 00290 Helsinki, Finland; Department of Pediatrics, Tampere University Hospital, 33521 Tampere, Finland; and
| | - Outi Vaarala
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland; Respiratory, Inflammatory and Autoimmune Diseases, Innovative Medicine, AstraZeneca, 43183 Mölndal, Sweden
| | | | | |
Collapse
|
77
|
Jimeno R, Gomariz RP, Garín M, Gutiérrez-Cañas I, González-Álvaro I, Carrión M, Galindo M, Leceta J, Juarranz Y. The pathogenic Th profile of human activated memory Th cells in early rheumatoid arthritis can be modulated by VIP. J Mol Med (Berl) 2014; 93:457-67. [PMID: 25430993 PMCID: PMC4366555 DOI: 10.1007/s00109-014-1232-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/15/2014] [Accepted: 11/12/2014] [Indexed: 12/30/2022]
Abstract
UNLABELLED Our aim is to study the behavior of memory Th cells (Th17, Th17/1, and Th1 profiles) from early rheumatoid arthritis (eRA) patients after their in vitro activation/expansion to provide information about its contribution to RA chronicity. Moreover, we analyzed the potential involvement of vasoactive intestinal peptide (VIP) as an endogenous healing mediator. CD4(+)CD45RO(+) T cells from PBMCs of HD and eRA were activated/expanded in vitro in the presence/absence of VIP. FACS, ELISA, RT-PCR, and immunocytochemistry analyses were performed. An increase in CCR6(+)/RORC(+) cells and in RORC-proliferating cells and a decrease in T-bet-proliferating cells and T-bet(+)/RORC(+) cells were shown in eRA. mRNA expression of IL-17, IL-2, RORC, RORA, STAT3, and Tbx21 and protein secretion of IL-17, IFNγ, and GM-CSF were higher in eRA. VIP decreased the mRNA expression of IL-22, IL-2, STAT3, Tbx21, IL-12Rβ2, IL-23R, and IL-21R in HD and it decreased IL-21, IL-2, and STAT3 in eRA. VIP decreased IL-22 and GM-CSF secretion and increased IL-9 secretion in HD and it decreased IL-21 secretion in eRA. VPAC2/VPAC1 ratio expression was increased in eRA. All in all, memory Th cells from eRA patients show a greater proportion of Th17 cells with a pathogenic Th17 and Th17/1 profile compared to HD. VIP is able to modulate the pathogenic profile, mostly in HD. Our results are promising for therapy in the early stages of RA because they suggest that targeting molecules involved in the pathogenic Th17, Th17/1, and Th1 phenotypes and targeting VIP receptors could have a therapeutic effect modulating these subsets. KEY MESSAGES Th17 cells are more important than Th1 in the contribution to pathogenesis in eRA patients. Pathogenic Th17 and Th17/1 profile are abundant in activated/expanded memory Th cells from eRA patients. VIP decreases the pathogenic Th17, Th1, and Th17/1 profiles, mainly in healthy donors. The expression of VIP receptors is reduced in eRA patients respect to healthy donors, whereas the ratio of VPAC2/VPAC1 expression is higher.
Collapse
Affiliation(s)
- Rebeca Jimeno
- Departamento de Biología Celular, Facultad de Biología, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
105. Cytokine 2014. [DOI: 10.1016/j.cyto.2014.07.112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
79
|
Brucklacher-Waldert V, Carr EJ, Linterman MA, Veldhoen M. Cellular Plasticity of CD4+ T Cells in the Intestine. Front Immunol 2014; 5:488. [PMID: 25339956 PMCID: PMC4188036 DOI: 10.3389/fimmu.2014.00488] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/22/2014] [Indexed: 01/07/2023] Open
Abstract
Barrier sites such as the gastrointestinal tract are in constant contact with the environment, which contains both beneficial and harmful components. The immune system at the epithelia must make the distinction between these components to balance tolerance, protection, and immunopathology. This is achieved via multifaceted immune recognition, highly organized lymphoid structures, and the interaction of many types of immune cells. The adaptive immune response in the gut is orchestrated by CD4+ helper T (Th) cells, which are integral to gut immunity. In recent years, it has become apparent that the functional identity of these Th cells is not as fixed as initially thought. Plasticity in differentiated T cell subsets has now been firmly established, in both health and disease. The gut, in particular, utilizes CD4+ T cell plasticity to mold CD4+ T cell phenotypes to maintain its finely poised balance of tolerance and inflammation and to encourage biodiversity within the enteric microbiome. In this review, we will discuss intestinal helper T cell plasticity and our current understanding of its mechanisms, including our growing knowledge of an evolutionarily ancient symbiosis between microbiota and malleable CD4+ T cell effectors.
Collapse
Affiliation(s)
| | - Edward J Carr
- Laboratory for Lymphocyte Signalling and Development, The Babraham Institute , Cambridge , UK
| | - Michelle A Linterman
- Laboratory for Lymphocyte Signalling and Development, The Babraham Institute , Cambridge , UK
| | - Marc Veldhoen
- Laboratory for Lymphocyte Signalling and Development, The Babraham Institute , Cambridge , UK
| |
Collapse
|
80
|
Annunziato F, Cosmi L, Liotta F, Maggi E, Romagnani S. Human Th1 dichotomy: origin, phenotype and biologic activities. Immunology 2014; 144:343-351. [PMID: 25284714 PMCID: PMC4557671 DOI: 10.1111/imm.12399] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 12/14/2022] Open
Abstract
The great variety of pathogens present in the environment has obliged the immune system to evolve different mechanisms for tailored and maximally protective responses. Initially, two major types of CD4+ T helper (Th) effector cells were identified, and named as type 1 (Th1) and type (Th2) cells because of the different cytokines they produce. More recently, a third type of CD4+ Th effectors has been identified and named as Th17 cells. Th17 cells, however, have been found to exhibit high plasticity because they rapidly shift into the Th1 phenotype in the inflammatory sites. Therefore, in these sites usually there is a dichotomic mixture of classic and non classic (Th17-derived) Th1 cells. In humans, non classic Th1 cells express CD161, as well as the retinoic acid orphan receptor C, IL-17 receptor E, IL-1RI, CCR6, and IL-4-induced gene 1 and Tob-1, which are all virtually absent from classic Th1 cells. The possibility to distinguish these two cell subsets may allow the opportunity to better establish their respective pathogenic role in different chronic inflammatory disorders. In this review, we discuss the different origin, the distinctive phenotypic features and the major biologic activities of classic and non classic Th1 cells. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Centre, University of FlorenceFlorence, Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit, Azienda Ospedaliera CareggiFlorence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine and DENOTHE Centre, University of FlorenceFlorence, Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit, Azienda Ospedaliera CareggiFlorence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine and DENOTHE Centre, University of FlorenceFlorence, Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit, Azienda Ospedaliera CareggiFlorence, Italy
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Centre, University of FlorenceFlorence, Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit, Azienda Ospedaliera CareggiFlorence, Italy
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine and DENOTHE Centre, University of FlorenceFlorence, Italy
| |
Collapse
|
81
|
Chao K, Zhang S, Yao J, He Y, Chen B, Zeng Z, Zhong B, Chen M. Imbalances of CD4(+) T-cell subgroups in Crohn's disease and their relationship with disease activity and prognosis. J Gastroenterol Hepatol 2014; 29:1808-14. [PMID: 24720272 DOI: 10.1111/jgh.12592] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIM The CD4(+) T-cell subgroups play central pathophysiological roles in Crohn's disease (CD); however, their clinical relevance requires additional clarification and remains controversial. We investigated their balance in Chinese CD patients and explored their clinical significance. METHODS Peripheral blood mononuclear cells and serum were collected from 46 Chinese CD patients and 23 healthy donors. Circulating Treg, Th1, Th2, and Th17 cells were flow cytometrically analyzed. Subgroup-restricted transcription factor expression was determined by real-time polymerase chain reaction. Serum concentrations of the main cytokines produced by each subgroup were measured by cytometric bead arrays or enzyme-linked immunosorbent assay. RESULTS Lower Treg proportion (6.0 ± 1.2% vs 7.8 ± 1.5%, P = 0.030), FOXP3 mRNA expression (0.58-fold, P = 0.030), and circulating soluble TGFβ-1 (19.1 ± 9.9 vs 32.7 ± 16.8 ng/mL, P = 0.038) were observed in CD patients versus controls. The Th1 and Th17 proportions were higher in CD patients (17.8 ± 6.6% vs 7.8 ± 1.5%, P < 0.001; and 3.7 ± 1.8% vs 1.8 ± 0.7%, P = 0.022, respectively), as were transcription factors T-bet (4.6-fold, P = 0.043) and RORγt (14-fold, P < 0.001) and related cytokines (P < 0.05). Th2 proportion, GATA3 mRNA expression, and serum interleukin-4 concentration in CD patients were similar to controls (P > 0.05). Treg/Th1 and Treg/Th17 ratios were higher in inactive versus active CD patients (0.6 ± 0.4 vs 0.3 ± 0.1, P = 0.022; and 3.7 ± 2.0 vs 1.7 ± 1.4, P = 0.013, respectively). During follow-up, patients with lower Treg/Th1 and Treg/Th17 ratios were at higher recurrence risk. CONCLUSIONS Imbalances among Treg, Th1, and Th17 subgroups were found in Chinese CD patients. Treg/Th1 and Treg/Th17 ratios are associated with disease activity and are potential prognostic indicators for predicting CD recurrence.
Collapse
Affiliation(s)
- Kang Chao
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Roeleveld DM, van Nieuwenhuijze AEM, van den Berg WB, Koenders MI. The Th17 pathway as a therapeutic target in rheumatoid arthritis and other autoimmune and inflammatory disorders. BioDrugs 2014; 27:439-52. [PMID: 23620106 DOI: 10.1007/s40259-013-0035-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Production of the pro-inflammatory cytokine interleukin (IL)-17 by Th17 cells and other cells of the immune system protects the host against bacterial and fungal infections, but also promotes the development of rheumatoid arthritis (RA) and other autoimmune and inflammatory disorders. Several biologicals targeting IL-17, the IL-17 receptor, or IL-17-related pathways are being tested in clinical trials, and might ultimately lead to better treatment for patients suffering from various IL-17-mediated disorders. In this review, we provide a clear overview of current knowledge on Th17 cell regulation and the main Th17 effector cytokines in relation to IL-17-mediated conditions, as well as on recent IL-17-related drug developments. We demonstrate that targeting the Th17 pathway is a promising treatment for rheumatoid arthritis and various other autoimmune and inflammatory diseases. However, improvements in technical developments assisting in the identification of patients suffering from IL-17-driven disease are needed to enable the application of tailor-made, personalized medicine.
Collapse
|
83
|
Duhen T, Campbell DJ. IL-1β promotes the differentiation of polyfunctional human CCR6+CXCR3+ Th1/17 cells that are specific for pathogenic and commensal microbes. THE JOURNAL OF IMMUNOLOGY 2014; 193:120-9. [PMID: 24890729 DOI: 10.4049/jimmunol.1302734] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In humans, Th1/17 cells, identified by coexpression of the chemokine receptors CCR6 and CXCR3, are proposed to be highly pathogenic in several autoimmune disorders due in part to their expression of the proinflammatory cytokines IL-17, IFN-γ, and GM-CSF. However, their developmental requirements, relationship with "classic" Th17 and Th1 cells and physiological role in normal immune responses are not well understood. In this study, we examined CCR6+ CXCR3+ Th1/17 cells from healthy individuals and found that ex vivo these cells produced the effector cytokines IL-17, IL-22, and IFN-γ in all possible combinations and were highly responsive to both IL-12 and IL-23. Moreover, although the Ag specificity of CCR6+ CXCR3+ Th1/17 cells showed substantial overlap with that of Th1 and Th17 cells, this population was enriched in cells recognizing certain extracellular bacteria and expressing the intestinal homing receptor integrin β7. Finally, we identified IL-1β as a key cytokine that renders Th17 cells sensitive to IL-12, and both cytokines together potently induced the differentiation of cells that produce IL-17, IFN-γ, and GM-CSF. Therefore, interfering with IL-1β and IL-12 signaling in Th17 cells during inflammation may be a promising therapeutic approach to reduce their differentiation into "pathogenic" CCR6+ CXCR3+ Th1/17 cells in patients with autoimmune diseases.
Collapse
Affiliation(s)
- Thomas Duhen
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; andDepartment of Immunology, University of Washington School of Medicine, Seattle, WA 98195
| | - Daniel J Campbell
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; andDepartment of Immunology, University of Washington School of Medicine, Seattle, WA 98195
| |
Collapse
|
84
|
Vasoactive intestinal peptide maintains the nonpathogenic profile of human th17-polarized cells. J Mol Neurosci 2014; 54:512-25. [PMID: 24805298 DOI: 10.1007/s12031-014-0318-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/28/2014] [Indexed: 01/01/2023]
Abstract
The cytokine microenvironment modulates CD4 T cell differentiation causing the shift of naïve CD4 T cells into different cell subsets. This process is also regulated by modulators such as vasoactive intestinal peptide (VIP), a neuropeptide with known immunomodulatory properties on CD4 T cells that exert this action through specific receptors, vasoactive intestinal peptide receptor (VPAC)1 and VPAC2. Our results show that the pattern of VIP receptors expression ratio is modified during Th17 differentiation. In this report, we evaluate the capacity of VIP to modulate naïve human cells into Th17 cells in vitro by analyzing their functional phenotype. The presence of VIP maintains the nonpathogenic profile of Th17-polarized cells, increases the proliferation rate, and decreases their Th1 potential. VIP induces the upregulation of the STAT3 gene interaction with the VPAC1 receptor during the onset of Th17 differentiation. Moreover, RAR-related orphan receptor C (RORC), RAR-related orphan receptor A (RORA), and interleukin (IL)-17A genes are upregulated in the presence of VIP through interaction with VPAC1 and VPAC2 receptors. Interestingly, VIP induces the expression of the IL-23R gene through interaction with the VPAC2 receptor during the expansion phase. This is the first report that describes the differentiation of naïve human T cells to Th17-polarized cells in the presence of VIP and demonstrates how this differentiation regulates the expression of the VIP receptors.
Collapse
|
85
|
Bronchoalveolar lavage fluid IFN-γ+ Th17 cells and regulatory T cells in pulmonary sarcoidosis. Mediators Inflamm 2014; 2014:438070. [PMID: 24882950 PMCID: PMC4027000 DOI: 10.1155/2014/438070] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 04/09/2014] [Indexed: 12/19/2022] Open
Abstract
In sarcoidosis, increased Th17 cell fractions have been reported in bronchoalveolar lavage fluid, and elevated numbers of Th17 cells producing IFN-γ have been observed in peripheral blood. The balance between Th1, Th17, and FoxP3+ CD4+ T cell subsets in sarcoidosis remains unclear. Bronchoalveolar lavage fluid cells, from 30 patients with sarcoidosis, 18 patients with other diffuse parenchymal lung diseases, and 15 healthy controls, were investigated with flow cytometry for intracellular expression of FoxP3. In a subset of the patients, expression of the cytokines IL17A and IFN-γ was investigated. The fractions of FoxP3+ CD4+ T cells and Th17 cells were both lower in sarcoidosis compared to controls (P = 0.017 and P = 0.011, resp.). The proportion of Th17 cells positive for IFN-γ was greater in sarcoidosis than controls (median 72.4% versus 31%, P = 0.0005) and increased with radiologic stage (N = 23, rho = 0.45, and P = 0.03). IFN-γ+ Th17 cells were highly correlated with Th1 cells (N = 23, rho = 0.64, and P = 0.001), and the ratio of IFN-γ+ Th17/FoxP3+ CD4+ T cells was prominently increased in sarcoidosis. IFN-γ+ Th17 cells may represent a pathogenic subset of Th17 cells, yet their expression of IFN-γ could be a consequence of a Th1-polarized cytokine milieu. Our results indicate a possible immune cell imbalance in sarcoidosis.
Collapse
|
86
|
Santarlasci V, Maggi L, Mazzoni A, Capone M, Querci V, Rossi MC, Beltrame L, Cavalieri D, De Palma R, Liotta F, Cosmi L, Maggi E, Romagnani S, Annunziato F. IL-4-induced gene 1 maintains high Tob1 expression that contributes to TCR unresponsiveness in human T helper 17 cells. Eur J Immunol 2013; 44:654-61. [DOI: 10.1002/eji.201344047] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/08/2013] [Accepted: 11/28/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Veronica Santarlasci
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
| | - Valentina Querci
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
| | - Maria Caterina Rossi
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
| | - Luca Beltrame
- Department of Oncology Mario Negri Institute; Milano Italy
| | - Duccio Cavalieri
- Department of Computational Biology; Comparative Genomics Unit; Fondazione Edmund Mach (FEM); San Michele all'Adige Italy
| | - Raffaele De Palma
- Dept. of Clinical & Experimental Medicine; Second University of Naples and Centro di Competenza Regionale GEAR (Genomics for Applied Research); Naples Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit of Azienda Ospedaliera Careggi; Florence Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit of Azienda Ospedaliera Careggi; Florence Italy
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit of Azienda Ospedaliera Careggi; Florence Italy
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Center; University of Florence; Florence Italy
- Regenerative Medicine Unit and Immunology and Cellular Therapy Unit of Azienda Ospedaliera Careggi; Florence Italy
| |
Collapse
|
87
|
Geginat J, Paroni M, Facciotti F, Gruarin P, Kastirr I, Caprioli F, Pagani M, Abrignani. S. The CD4-centered universe of human T cell subsets. Semin Immunol 2013; 25:252-62. [DOI: 10.1016/j.smim.2013.10.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
88
|
Th17-cell plasticity in Helicobacter hepaticus-induced intestinal inflammation. Mucosal Immunol 2013; 6:1143-56. [PMID: 23462910 DOI: 10.1038/mi.2013.11] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 01/30/2013] [Indexed: 02/04/2023]
Abstract
Bacterial-induced intestinal inflammation is crucially dependent on interleukin (IL)-23 and is associated with CD4(+) T helper type 1 (Th1) and Th17 responses. However, the relative contributions of these subsets during the induction and resolution of colitis in T-cell-sufficient hosts remain unknown. We report that Helicobacter hepaticus-induced typhlocolitis in specific pathogen-free IL-10(-/-) mice is associated with elevated frequencies and numbers of large intestinal interferon (IFN)-γ(+) and IFN-γ(+)IL-17A(+) CD4(+) T cells. By assessing histone modifications and transcript levels in IFN-γ(+), IFN-γ(+)IL-17A(+), and IL-17A(+) CD4(+) T cells isolated from the inflamed intestine, we show that Th17 cells are predisposed to upregulate the Th1 program and that they express IL-23R but not IL-12R. Using IL-17A fate-reporter mice, we further demonstrate that H. hepaticus infection gives rise to Th17 cells that extinguish IL-17A secretion and turn on IFN-γ within 10 days post bacterial inoculation. Together, our results suggest that bacterial-induced Th17 cells arising in disease-susceptible hosts contribute to intestinal pathology by switching phenotype, transitioning via an IFN-γ(+)IL-17A(+) stage, to become IFN-γ(+) ex-Th17 cells.
Collapse
|
89
|
Kumawat AK, Strid H, Tysk C, Bohr J, Hörnquist EH. Microscopic colitis patients demonstrate a mixed Th17/Tc17 and Th1/Tc1 mucosal cytokine profile. Mol Immunol 2013; 55:355-64. [PMID: 23566938 DOI: 10.1016/j.molimm.2013.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 03/13/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Microscopic colitis (MC) is a chronic inflammatory bowel disorder of unknown aetiology comprising collagenous colitis (CC) and lymphocytic colitis (LC). Data on the local cytokine profile in MC is limited. This study investigated the T helper (Th) cell and cytotoxic T lymphocyte (CTL) mucosal cytokine profile at messenger and protein levels in MC patients. METHODS Mucosal biopsies from CC (n=10), LC (n=5), and CC or LC patients in histopathological remission (CC-HR, n=4), (LC-HR, n=6), ulcerative colitis (UC, n=3) and controls (n=10) were analysed by real-time PCR and Luminex for expression/production of IL-1β, -4, -5, -6, -10, -12, -17, -21, -22, -23, IFN-γ, TNF-α, T-bet and RORC2. RESULTS Mucosal mRNA but not protein levels of IFN-γ and IL-12 were significantly up regulated in CC, LC as well as UC patients compared to controls. Transcription of the Th1 transcription factor T-bet was significantly enhanced in CC but not LC patients. mRNA levels for IL-17A, IL-21, IL-22 and IL-6 were significantly up regulated in CC and LC patients compared to controls, albeit less than in UC patients. Significantly enhanced IL-21 protein levels were noted in both CC and LC patients. IL-6 protein and IL-1β mRNA levels were increased in CC and UC but not LC patients. Increased mucosal mRNA levels of IFN-γ, IL-21 and IL-22 were correlated with higher clinical activity, recorded as the number of bowel movements per day, in MC patients. Although at lower magnitude, IL-23A mRNA was upregulated in CC and LC, whereas TNF-α protein was increased in CC, LC as well as in UC patients. Neither mRNA nor protein levels of IL-4, IL-5 or IL-10 were significantly changed in any of the colitis groups. LC-HR and especially CC-HR patients had normalized mRNA and protein levels of the above cytokines compared to LC and CC patients. No significant differences were found between LC and CC in cytokine expression/production. CONCLUSION LC and CC patients demonstrate a mixed Th17/Tc17 and Th1/Tc1 mucosal cytokine profile.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Case-Control Studies
- Colitis, Collagenous/genetics
- Colitis, Collagenous/immunology
- Colitis, Collagenous/pathology
- Colitis, Lymphocytic/genetics
- Colitis, Lymphocytic/immunology
- Colitis, Lymphocytic/pathology
- Colitis, Microscopic/genetics
- Colitis, Microscopic/immunology
- Colitis, Microscopic/pathology
- Cytokines/biosynthesis
- Cytokines/genetics
- Female
- Humans
- Immunity, Mucosal/genetics
- Male
- Middle Aged
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- Th1 Cells/immunology
- Th1 Cells/pathology
- Th17 Cells/immunology
- Th17 Cells/pathology
- Young Adult
Collapse
|
90
|
Kuriya G, Uchida T, Akazawa S, Kobayashi M, Nakamura K, Satoh T, Horie I, Kawasaki E, Yamasaki H, Yu L, Iwakura Y, Sasaki H, Nagayama Y, Kawakami A, Abiru N. Double deficiency in IL-17 and IFN-γ signalling significantly suppresses the development of diabetes in the NOD mouse. Diabetologia 2013; 56:1773-80. [PMID: 23699989 DOI: 10.1007/s00125-013-2935-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 04/22/2013] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS T helper type (Th) 17 cells have been shown to play important roles in mouse models of several autoimmune diseases that have been classified as Th1 diseases. In the NOD mouse, the relevance of Th1 and Th17 is controversial, because single-cytokine-deficient NOD mice develop diabetes similarly to wild-type NOD mice. METHODS We studied the impact of IL-17/IFN-γ receptor double deficiency in NOD mice on the development of insulitis/diabetes compared with IL-17 single-deficient mice and wild-type mice by monitoring diabetes-related phenotypes. The lymphocyte phenotypes were determined by flow cytometric analysis. RESULTS IL-17 single-deficient NOD mice showed delayed onset of diabetes and reduced severity of insulitis, but the cumulative incidence of longstanding diabetes in the IL-17-deficient mice was similar to that in wild-type mice. The IL-17/IFN-γ receptor double-deficient NOD mice showed an apparent decline in longstanding diabetes onset, but not in insulitis compared with that in the IL-17 single-deficient mice. We also found that double-deficient NOD mice had a severe lymphopenic phenotype and preferential increase in regulatory T cells among CD4(+) T cells compared with the IL-17 single-deficient mice and wild-type NOD mice. An adoptive transfer study with CD4(+)CD25(-) T cells from young non-diabetic IL-17 single-deficient NOD mice, but not those from older mice, showed significantly delayed disease onset in immune-deficient hosts compared with the corresponding wild-type mice. CONCLUSIONS/INTERPRETATION These results indicate that IL-17/Th17 participates in the development of insulitis and that both IL-17 and IFN-γ signalling may synergistically contribute to the development of diabetes in NOD mice.
Collapse
Affiliation(s)
- G Kuriya
- Department of Endocrinology and Metabolism, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Henriques A, Gomes V, Duarte C, Pedreiro S, Carvalheiro T, Areias M, Caseiro A, Gabriel AJ, Laranjeira P, Pais ML, da Silva JAP, Paiva A. Distribution and functional plasticity of peripheral blood Th(c)17 and Th(c)1 in rheumatoid arthritis. Rheumatol Int 2013; 33:2093-9. [PMID: 23412693 DOI: 10.1007/s00296-013-2703-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 02/05/2013] [Indexed: 10/27/2022]
Abstract
UNLABELLED With the discovery of Th17 cells, it became unclear whether rheumatoid arthritis (RA) is a Th1-mediated and/or a Th17-mediated disease. OBJECTIVE The aim of this study was to identify and characterize the pro-inflammatory function of IL-17-producing T cell subsets (Th(c)17) in RA. Flow cytometry analysis was performed on peripheral blood from RA patients with inactive or low disease activity (LDA, n = 19) and moderate to high disease activity (HDA, n = 13) to analyze the number and functional activity of Th(c)17 and Th(c)1 cell subsets according to the frequency of IL-2-, TNF-α- and IFN-γ-producers cells, as well as, their cytokine amount. Additionally, 13 age-matched healthy volunteers were added to the study. Our data point to a slight increase in Tc17 frequency in RA patients, more evident in HDA, and a higher ability of Th17 to produce IL-17, whereas a lower production of TNF-α was noted either in Th17 or Tc17 cells, particularly from HDA. A similar decrease was observed in Th(c)1 for almost all studied pro-inflammatory cytokines, with the exception of IL-2, which was increased in Tc1 from LDA patients. Analysing the proportion of pro-inflammatory cytokines-producing cells, a polarization to a Tc1 phenotype seemed to occur in CD8 T cells, while CD4 T cells appear to be decreased in their frequency of IFN-γ-producing cells. Taken together, the functional plasticity features of Th17 and Tc17 cells suggest a particular contribution to the local cytokine production, pointing an underestimated role, namely of Tc1 and Tc17 cells, in the RA pathophysiology.
Collapse
Affiliation(s)
- Ana Henriques
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Edifício São Jerónimo, 4º Piso Praceta Mota Pinto, 3001-301 Coimbra, Portugal
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Johnston A, Xing X, Swindell WR, Kochkodan J, Riblett M, Nair RP, Stuart PE, Ding J, Voorhees JJ, Elder JT, Gudjonsson JE. Susceptibility-associated genetic variation at IL12B enhances Th1 polarization in psoriasis. Hum Mol Genet 2013; 22:1807-15. [PMID: 23376980 DOI: 10.1093/hmg/ddt034] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The IL12B gene encodes the common p40 subunit of IL-12 and IL-23, cytokines with key roles in Th1 and Th17 biology, respectively, and genetic variation in this region significantly influences risk of psoriasis. Here, we demonstrate that a psoriasis-associated risk haplotype at the IL12B locus leads to increased expression of IL12B by monocytes and correlated with increased serum levels of IL-12, IFN-γ and the IFN-γ induced chemokine, CXCL10. In contrast, serum IL-23 levels were decreased in risk carriers when compared with non-carriers. We further demonstrate that IL-12 is increased in psoriatic skin and that risk carriers manifest a skewing of the inflammatory network toward stronger IFN-γ responses. Taken together, our data demonstrate that the risk variant in IL12B associates with its increased expression and predisposes to stronger Th1 polarization through deviation of the local inflammatory environment toward increased IL-12/IFN-γ at the expense of IL-23/IL-17 responses.
Collapse
Affiliation(s)
- Andrew Johnston
- Department of Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Savino MT, Ulivieri C, Emmi G, Prisco D, De Falco G, Ortensi B, Beccastrini E, Emmi L, Pelicci G, D'Elios MM, Baldari CT. The Shc family protein adaptor, Rai, acts as a negative regulator of Th17 and Th1 cell development. J Leukoc Biol 2013; 93:549-59. [PMID: 23345394 DOI: 10.1189/jlb.0712331] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rai, a Shc adapter family member, acts as a negative regulator of antigen receptor signaling in T and B cells. Rai(-/-) mice develop lupus-like autoimmunity associated to the spontaneous activation of self-reactive lymphocytes. Here, we have addressed the potential role of Rai in the development of the proinflammatory Th1 and Th17 subsets, which are centrally implicated in the pathogenesis of a number of autoimmune diseases, including lupus. We show that Rai(-/-) mice display a spontaneous Th1/Th17 bias. In vitro polarization experiments on naive and effector/memory CD4(+) T cells demonstrate that Rai(-/-) favors the development and expansion of Th17 but not Th1 cells, indicating that Rai modulates TCR signaling to antagonize the pathways driving naive CD4(+) T cell differentiation to the Th17 lineage, while indirectly limiting Th1 cell development in vivo. Th1 and Th17 cell infiltrates were found in the kidneys of Rai(-/-) mice, providing evidence that Rai(-/-) contributes to the development of lupus nephritis, not only by enhancing lymphocyte activation but also by promoting the development and expansion of proinflammatory effector T cells. Interestingly, T cells from SLE patients were found to have a defect in Rai expression, suggesting a role for Rai in disease pathogenesis.
Collapse
|
94
|
Szkaradkiewicz A, Karpiński T, Goślińska-Pawłowska O, Szkaradkiewicz A, Giedrys-Kalemba S. Cytokine Response in Autovaccine-Treated Patients with Chronic Staphylococcus Aureus Infections. EUR J INFLAMM 2013; 11:103-110. [DOI: 10.1177/1721727x1301100110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
This study aims to describe the levels of circulating cytokines produced by Th lymphocytes (IFN-γ, IL-4, IL-10, IL-17A), as well as the levels of cytokines produced by monocytes/macrophages (TNF-α, IL-1β, IL-12), in patients with chronic Staphylococcus aureus infections before treatment and following completion of autovaccine treatment. The study was carried out on adult individuals, including 25 healthy subjects (group 1, control, not treated), 50 patients with chronic suppurative dermatitis (group 2) and 40 patients with chronic infections of the upper respiratory tract (group 3). Blood serum cytokine levels were measured by enzyme–linked immunosorbent assay (ELISA). S. aureus was detected in cultures of suppurative dermal exudates or of pharyngeal smears. For every individual patient an autovaccine was prepared, containing a suspension of inactivated S. aureus bacteria (1.5 × 108 bacteria/ml) isolated from the patient. The autovaccine was administered subcutaneously for a period exceeding 3 months, for a total of 18 injections. The average level of IFN-γ and IL-17 was 2–2.5 times higher in the infected patients. This was not accompanied by an increase in TNF-α or IL-12 levels. A treatment with autovaccine eradicated S. aureus infection in 42 (84%) patients of group 2 and in 14 (35%) patients of group 3. A significant increase (two-fold) in IL-17A was observed in treated patients. Also, following the treatment with autovaccine, all patients demonstrated a significant increase in the levels of IFN-γ, TNF-α and IL-12. These studies showed for the first time that efficiency of the autovaccine treatment in patients with chronic S. aureus infection depends on an adequate secretory response of TH17 cells.
Collapse
Affiliation(s)
- A. Szkaradkiewicz
- Department of Medical Microbiology, University of Medical Sciences in Poznan, Poland
| | - T.M. Karpiński
- Department of Medical Microbiology, University of Medical Sciences in Poznan, Poland
| | | | - A.K. Szkaradkiewicz
- Department of Conservative Dentistry and Periodontology, University of Medical Sciences in Poznan, Poland
| | - S. Giedrys-Kalemba
- Department of Microbiology and Immunology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
95
|
Anti-cancer versus cancer-promoting effects of the interleukin-17-producing T helper cells. Immunol Lett 2012; 149:123-33. [PMID: 23159638 DOI: 10.1016/j.imlet.2012.11.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 10/15/2012] [Accepted: 11/05/2012] [Indexed: 12/18/2022]
Abstract
Research on T helper 17 (Th17) cells with regard to immunoediting has revealed elusive results. Whereas enhanced Th17 response and related molecules such as interleukin (IL)-17, IL-21, IL-22, IL-23 and STAT3 accompanied tumor induction and progression, finding that tumor growth/stage was negatively correlated with increased infiltration of Th17 cells in the tumor mass has prompted elucidation of various antitumor mechanisms elicited by Th17 and their related molecules. The pro-tumor efficacy of Th17 response included promotion of neutrophilia and induction of angiogenic (e.g. VEGF, MMP2 and MMP9) and anti-apoptotic factors (e.g. Bcl-XL), as well as expansion and activation of myeloid-derived suppressor cells, which facilitate generation of tumor-specific regulatory T cells. Other tumor immunogenic settings revealed anti-tumor pathways including induction of cytotoxic activity, expression of MHC antigens, the ability Th17 cells to reside within the tumor, and to convert into IFN-γ producers. Notably, Th17 cell related molecules exert indirect pro- or anti-tumor effects via inducing viral persistence or mediating protective mechanisms against bacterial and viral infection. Herein, the recent literature revealing such immunoediting events mediated by Th17 cells and their associated molecules as delivered by various experimental regimens and observed in cancer patient are revised, with a focus on some proposed anti-cancer therapies.
Collapse
|
96
|
Aijö T, Edelman SM, Lönnberg T, Larjo A, Kallionpää H, Tuomela S, Engström E, Lahesmaa R, Lähdesmäki H. An integrative computational systems biology approach identifies differentially regulated dynamic transcriptome signatures which drive the initiation of human T helper cell differentiation. BMC Genomics 2012; 13:572. [PMID: 23110343 PMCID: PMC3526425 DOI: 10.1186/1471-2164-13-572] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 10/02/2012] [Indexed: 01/19/2023] Open
Abstract
Background A proper balance between different T helper (Th) cell subsets is necessary for normal functioning of the adaptive immune system. Revealing key genes and pathways driving the differentiation to distinct Th cell lineages provides important insight into underlying molecular mechanisms and new opportunities for modulating the immune response. Previous computational methods to quantify and visualize kinetic differential expression data of three or more lineages to identify reciprocally regulated genes have relied on clustering approaches and regression methods which have time as a factor, but have lacked methods which explicitly model temporal behavior. Results We studied transcriptional dynamics of human umbilical cord blood T helper cells cultured in absence and presence of cytokines promoting Th1 or Th2 differentiation. To identify genes that exhibit distinct lineage commitment dynamics and are specific for initiating differentiation to different Th cell subsets, we developed a novel computational methodology (LIGAP) allowing integrative analysis and visualization of multiple lineages over whole time-course profiles. Applying LIGAP to time-course data from multiple Th cell lineages, we identified and experimentally validated several differentially regulated Th cell subset specific genes as well as reciprocally regulated genes. Combining differentially regulated transcriptional profiles with transcription factor binding site and pathway information, we identified previously known and new putative transcriptional mechanisms involved in Th cell subset differentiation. All differentially regulated genes among the lineages together with an implementation of LIGAP are provided as an open-source resource. Conclusions The LIGAP method is widely applicable to quantify differential time-course dynamics of many types of datasets and generalizes to any number of conditions. It summarizes all the time-course measurements together with the associated uncertainty for visualization and manual assessment purposes. Here we identified novel human Th subset specific transcripts as well as regulatory mechanisms important for the initiation of the Th cell subset differentiation.
Collapse
Affiliation(s)
- Tarmo Aijö
- Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Bachmann M, Scheiermann P, Härdle L, Pfeilschifter J, Mühl H. IL-36γ/IL-1F9, an innate T-bet target in myeloid cells. J Biol Chem 2012; 287:41684-96. [PMID: 23095752 DOI: 10.1074/jbc.m112.385443] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
By concerted action in dendritic (DC) and T cells, T-box expressed in T cells (T-bet, Tbx21) is pivotal for initiation and perpetuation of Th1 immunity. Identification of novel T-bet-regulated genes is crucial for further understanding the biology of this transcription factor. By combining siRNA technology with genome-wide mRNA expression analysis, we sought to identify new T-bet-regulated genes in predendritic KG1 cells activated by IL-18. One gene robustly dependent on T-bet was IL-36γ, a recently described novel IL-1 family member. Promoter analysis revealed a T-bet binding site that, along with a κB site, enables efficient IL-36γ induction. Using knock-out animals, IL-36γ reliance on T-bet was extended to murine DC. IL-36γ expression by human myeloid cells was confirmed using monocyte-derived DC and M1 macrophages. The latter model was employed to substantiate dependence of IL-36γ on endogenous T-bet in human primary cells. Ectopic expression of T-bet likewise mediated IL-36γ production in HaCaT keratinocytes that otherwise lack this transcription factor. Additional experiments furthermore revealed that mature IL-36γ has the capability to establish an inflammatory gene expression profile in human primary keratinocytes that displays enhanced mRNA levels for TNFα, CCL20, S100A7, inducible NOS, and IL-36γ itself. Data presented herein shed further light on involvement of T-bet in innate immunity and suggest that IL-36γ, besides IFNγ, may contribute to functions of this transcription factor in immunopathology.
Collapse
Affiliation(s)
- Malte Bachmann
- Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
98
|
Anti-leukemic properties of IL-12, IL-23 and IL-27: Differences and similarities in the control of pediatric B acute lymphoblastic leukemia. Crit Rev Oncol Hematol 2012; 83:310-8. [DOI: 10.1016/j.critrevonc.2011.11.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 11/11/2011] [Accepted: 11/15/2011] [Indexed: 12/19/2022] Open
|
99
|
Poon AH, Eidelman DH, Martin JG, Laprise C, Hamid Q. Pathogenesis of severe asthma. Clin Exp Allergy 2012; 42:625-37. [PMID: 22515387 DOI: 10.1111/j.1365-2222.2012.03983.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patients with severe asthma have asthma symptoms which are difficult to control, require high dosages of medication, and continue to experience persistent symptoms, asthma exacerbations or airflow obstruction. Epidemiological and clinical evidences point to the fact that severe asthma is not a single phenotype. Cluster analyses have identified subclasses of severe asthma using parameters such as patient characteristics, and cytokine profiles have also been useful in classifying moderate and severe asthma. The IL-4/IL-13 signalling pathway accounts for the symptoms experienced by a subset of severe asthmatics with allergen-associated symptoms and high serum immunoglobulin E (IgE) levels, and these patients are generally responsive to anti-IgE treatment. The IL-5/IL-33 signalling pathway is likely to play a key role in the disease pathogenesis of those who are resistant to high doses of inhaled corticosteroid but responsive to systemic corticosteroids and anti-IL5 therapy. The IL-17 signalling pathway is thought to contribute to 'neutrophilic asthma'. Although traditionally viewed as players in the defence mechanism against viral and intracellular bacterial infection, mounting evidence supports a role for Th1 cytokines such as IL-18 and IFN-γ in severe asthma pathogenesis. Furthermore, these cytokine signalling pathways interact to contribute to the spectrum of clinical pathological outcomes in severe asthma. To date, glucocorticoids are the most effective anti-asthma drugs available, yet severe asthma patients are typically resistant to the effects of glucocorticoids. Glucocorticoid receptor dysfunction and histone deacetylase activity reduction are likely to contribute to glucocorticoid resistance in severe asthma patients. This review discusses recent development in different cytokine signalling pathways, their interactions and steroid resistance, in the context of severe asthma pathogenesis.
Collapse
Affiliation(s)
- A H Poon
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
100
|
Wan X, Guloglu FB, VanMorlan AM, Rowland LM, Jain R, Haymaker CL, Cascio JA, Dhakal M, Hoeman CM, Tartar DM, Zaghouani H. Mechanisms underlying antigen-specific tolerance of stable and convertible Th17 cells during suppression of autoimmune diabetes. Diabetes 2012; 61:2054-65. [PMID: 22751698 PMCID: PMC3402331 DOI: 10.2337/db11-1723] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes involves both T helper (Th)1 and Th17 cells. While the mechanisms underlying the control of Th1 cells are relatively well defined, those operating modulation of Th17 cells remain unknown. Moreover, given that Th17 cells are plastic and can drive disease as stable or convertible T cells, effective approaches to counter type 1 diabetes would have to alter Th17 function under both circumstances. Herein, we genetically incorporated the BDC2.5-reactive p79 mimotope into an Ig molecule, and the resulting Ig-p79 was used to investigate Th17 tolerance. Accordingly, diabetogenic BDC2.5 Th17 cells were transferred into NOD mice under convertible or stable conditions and their fate was evaluated upon induction of tolerance and disease suppression by Ig-p79. The findings show that convertible (Th17 to Th1) cells display downregulation of the chemokine (C-X-C motif) receptor 3 that was associated with diminished T-box transcription factor T-bet expression, retention in the spleen, and inhibition of trafficking to the pancreas. In contrast, stable Th17 cells downregulated orphan nuclear receptor ROR-γt but increased Fas ligand expression and died by apoptosis. Thus, the final signature transcription factor shapes the mechanism of tolerance in plastic Th17 cells. These findings suggest that effective strategies against type 1 diabetes will require regimens that could drive both mechanisms of tolerance to overcome the disease.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - F. Betul Guloglu
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
- Scientific and Technological Research Council of Turkey, Marmara Research Center, Genetic Engineering and Biotechnology Institute, Gebze, Kocaeli, Turkey
| | - Amie M. VanMorlan
- Department of Child Health, University of Missouri School of Medicine, Columbia, Missouri
| | - Linda M. Rowland
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Renu Jain
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
- Merck Research Laboratories, Palo Alto, California
| | - Cara L. Haymaker
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
- Department of Melanoma Medical Oncology, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Jason A. Cascio
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Mermagya Dhakal
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Christine M. Hoeman
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Danielle M. Tartar
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Habib Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
- Department of Child Health, University of Missouri School of Medicine, Columbia, Missouri
- Corresponding author: Habib Zaghouani,
| |
Collapse
|