51
|
Wlodarczyk A, Cédile O, Jensen KN, Jasson A, Mony JT, Khorooshi R, Owens T. Pathologic and Protective Roles for Microglial Subsets and Bone Marrow- and Blood-Derived Myeloid Cells in Central Nervous System Inflammation. Front Immunol 2015; 6:463. [PMID: 26441968 PMCID: PMC4562247 DOI: 10.3389/fimmu.2015.00463] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a series of processes designed for eventual clearance of pathogens and repair of damaged tissue. In the context of autoimmune recognition, inflammatory processes are usually considered to be pathological. This is also true for inflammatory responses in the central nervous system (CNS). However, as in other tissues, neuroinflammation can have beneficial as well as pathological outcomes. The complex role of encephalitogenic T cells in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE) may derive from heterogeneity of the myeloid cells with which these T cells interact within the CNS. Myeloid cells, including resident microglia and infiltrating bone marrow-derived cells, such as dendritic cells (DC) and monocytes/macrophages [bone marrow-derived macrophages (BMDM)], are highly heterogeneous populations that may be involved in neurotoxicity and also immunoregulation and regenerative processes. Better understanding and characterization of myeloid cell heterogeneity is essential for future development of treatments controlling inflammation and inducing neuroprotection and neuroregeneration in diseased CNS. Here, we describe and compare three populations of myeloid cells: CD11c+ microglia, CD11c− microglia, and CD11c+ blood-derived cells in terms of their pathological versus protective functions in the CNS of mice with EAE. Our data show that CNS-resident microglia include functionally distinct subsets that can be distinguished by their expression of CD11c. These subsets differ in their expression of Arg-1, YM1, iNOS, IL-10, and IGF-1. Moreover, in contrast to BMDM/DC, both subsets of microglia express protective interferon-beta (IFNβ), high levels of colony-stimulating factor-1 receptor, and do not express the Th1-associated transcription factor T-bet. Taken together, our data suggest that CD11c+ microglia, CD11c− microglia, and infiltrating BMDM/DC represent separate and distinct populations and illustrate the heterogeneity of the CNS inflammatory environment.
Collapse
Affiliation(s)
- Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Oriane Cédile
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Kirstine Nolling Jensen
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Agathe Jasson
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark ; Department of Biology, École Normale Supérieure de Lyon , Lyon , France
| | - Jyothi Thyagabhavan Mony
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| |
Collapse
|
52
|
Ji N, Kovalovsky A, Fingerle-Rowson G, Guentzel MN, Forsthuber TG. Macrophage migration inhibitory factor promotes resistance to glucocorticoid treatment in EAE. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e139. [PMID: 26280015 PMCID: PMC4529283 DOI: 10.1212/nxi.0000000000000139] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 06/12/2015] [Indexed: 02/04/2023]
Abstract
Objective: Glucocorticoids (GCs) are used as standard treatment for acute attacks of multiple sclerosis (MS). However, GCs eventually lose efficacy and do not prevent disease progression. Macrophage migration inhibitory factor (MIF) is the only known proinflammatory cytokine induced by GCs that inhibits their anti-inflammatory effects. Therefore, we investigated whether MIF plays a role in resistance to GC treatment in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Methods: EAE was induced in wild-type (Wt) and MIF knockout (MIF−/−) mice followed by treatment with dexamethasone (Dex) before or upon disease onset. Splenocytes and brain mononuclear cells were harvested for cytokine ELISPOT assay and flow cytometry analysis. Results: Treatment of EAE with Dex was substantially more efficacious in MIF−/− mice than Wt mice. Dex treatment decreased MOG35-55–induced cytokine production by Wt or MIF−/− CD4+ T cells only at the onset of EAE but inhibited upregulation of T-bet during acute and chronic phases of disease, particularly in MIF−/− mice. Furthermore, passive EAE induced by adoptive transfer of T cells showed that Dex was highly effective in ameliorating disease induced by MIF−/− CD4+ T cells but not by Wt CD4+ T cells. The expression of T-bet and VLA-4 was decreased in CD4+ T cells in MIF−/− mice compared with Wt mice. Conclusions: Our data establish MIF as a key molecule in resistance of pathogenic CD4+ T cells to GC treatment in EAE and as a potential target to enhance the effectiveness of steroid treatment in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Niannian Ji
- Department of Biology (N.J., M.N.G., T.G.F.), University of Texas at San Antonio; Department of Pathology (A.K.), Frederick Memorial Hospital, Frederick, MD; and Clinic I of Internal Medicine and Center of Integrative Oncology Cologne-Bonn (G.F.-R.), Cologne, Germany
| | - Andra Kovalovsky
- Department of Biology (N.J., M.N.G., T.G.F.), University of Texas at San Antonio; Department of Pathology (A.K.), Frederick Memorial Hospital, Frederick, MD; and Clinic I of Internal Medicine and Center of Integrative Oncology Cologne-Bonn (G.F.-R.), Cologne, Germany
| | - Günter Fingerle-Rowson
- Department of Biology (N.J., M.N.G., T.G.F.), University of Texas at San Antonio; Department of Pathology (A.K.), Frederick Memorial Hospital, Frederick, MD; and Clinic I of Internal Medicine and Center of Integrative Oncology Cologne-Bonn (G.F.-R.), Cologne, Germany
| | - M Neal Guentzel
- Department of Biology (N.J., M.N.G., T.G.F.), University of Texas at San Antonio; Department of Pathology (A.K.), Frederick Memorial Hospital, Frederick, MD; and Clinic I of Internal Medicine and Center of Integrative Oncology Cologne-Bonn (G.F.-R.), Cologne, Germany
| | - Thomas G Forsthuber
- Department of Biology (N.J., M.N.G., T.G.F.), University of Texas at San Antonio; Department of Pathology (A.K.), Frederick Memorial Hospital, Frederick, MD; and Clinic I of Internal Medicine and Center of Integrative Oncology Cologne-Bonn (G.F.-R.), Cologne, Germany
| |
Collapse
|
53
|
Jimeno R, Leceta J, Garín M, Ortiz AM, Mellado M, Rodríguez-Frade JM, Martínez C, Pérez-García S, Gomariz RP, Juarranz Y. Th17 polarization of memory Th cells in early arthritis: the vasoactive intestinal peptide effect. J Leukoc Biol 2015; 98:257-69. [PMID: 25957307 DOI: 10.1189/jlb.3a0714-327r] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 04/12/2015] [Indexed: 01/15/2023] Open
Abstract
Several studies in humans indicate the implication of Th17 cells in RA. Therapies targeting their pathogenicity, as well as their plasticity to the Th17/1 phenotype, could ameliorate the progression of the pathology. The neuroendocrine environment has a major impact on the differentiation of lymphoid cells. VIP is present in the microenvironment of the joint, and its known therapeutic effects are supported by several studies on RA. We examine the ability of VIP to modulate the differentiation of Th17 cells. Peripheral blood CD4(+)CD45RO(+) T cells from HD and eRA patients were expanded under Th17-polarizing conditions in the presence of TGF-β. After 7 days, the higher IL-17A, IL-21, and IL-9 levels and lower IL-22 levels indicate the nonpathogenic profile for Th17 cells in HD. In contrast, Th17 cells from eRA patients produced significantly more IL-22 and IFN-γ, and these cells show a more Th17/1 profile, indicating a pathogenic phenotype. Interestingly, when VIP was present in the Th17 conditioned medium, increased levels of IL-10 and IL-9 were detected in HD and eRA patients. VIP also reduced the levels of IL-22 in eRA patients. These data suggest that VIP reduces the pathogenic profile of the Th17-polarized cells. This effect was accompanied by an increased in the Treg/Th17 profile, as shown by the increase levels of Foxp3. In conclusion, this report addresses a novel and interesting question on the effect of VIP on human Th17 cells and adds clinical relevance by analyzing, in parallel, HD and eRA patients.
Collapse
Affiliation(s)
- Rebeca Jimeno
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Javier Leceta
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Marina Garín
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Ana M Ortiz
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Mario Mellado
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Jose Miguel Rodríguez-Frade
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carmen Martínez
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Selene Pérez-García
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Rosa P Gomariz
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Yasmina Juarranz
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
54
|
Brück J, Pascolo S, Fuchs K, Kellerer C, Glocova I, Geisel J, Dengler K, Yazdi AS, Röcken M, Ghoreschi K. Cholesterol Modification of p40-Specific Small Interfering RNA Enables Therapeutic Targeting of Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2216-23. [PMID: 26232431 DOI: 10.4049/jimmunol.1402989] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 07/01/2015] [Indexed: 11/19/2022]
Abstract
Small interfering RNA (siRNA)-based therapies allow targeted correction of molecular defects in distinct cell populations. Although efficient in multiple cell populations, dendritic cells (DCs) seem to resist siRNA delivery. Using fluorescence labeling and radiolabeling, we show that cholesterol modification enables siRNA uptake by DCs in vitro and in vivo. Delivery of cholesterol-modified p40 siRNA selectively abolished p40 transcription and suppressed TLR-triggered p40 production by DCs. During immunization with peptide in CFA, cholesterol-modified p40 siRNA generated p40-deficient, IL-10-producing DCs that prevented IL-17/Th17 and IFN-γ/Th1 responses. Only cholesterol-modified p40-siRNA established protective immunity against experimental autoimmune encephalomyelitis and suppressed IFN-γ and IL-17 expression by CNS-infiltrating mononuclear cells without inducing regulatory T cells. Because cholesterol-modified siRNA can thus modify selected DC functions in vivo, it is intriguing for targeted immune therapy of allergic, autoimmune, or neoplastic diseases.
Collapse
Affiliation(s)
- Jürgen Brück
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Steve Pascolo
- Department of Oncology, University Hospital Zurich, CH-8044 Zurich, Switzerland
| | - Kerstin Fuchs
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Christina Kellerer
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Ivana Glocova
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Julia Geisel
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Katja Dengler
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Amir S Yazdi
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Kamran Ghoreschi
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| |
Collapse
|
55
|
Benevides L, da Fonseca DM, Donate PB, Tiezzi DG, De Carvalho DD, de Andrade JM, Martins GA, Silva JS. IL17 Promotes Mammary Tumor Progression by Changing the Behavior of Tumor Cells and Eliciting Tumorigenic Neutrophils Recruitment. Cancer Res 2015. [PMID: 26208902 DOI: 10.1158/0008-5472.can-15-0054] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aggressiveness of invasive ductal carcinoma (IDC) of the breast is associated with increased IL17 levels. Studying the role of IL17 in invasive breast tumor pathogenesis, we found that metastatic primary tumor-infiltrating T lymphocytes produced elevated levels of IL17, whereas IL17 neutralization inhibited tumor growth and prevented the migration of neutrophils and tumor cells to secondary disease sites. Tumorigenic neutrophils promote disease progression, producing CXCL1, MMP9, VEGF, and TNFα, and their depletion suppressed tumor growth. IL17A also induced IL6 and CCL20 production in metastatic tumor cells, favoring the recruitment and differentiation of Th17. In addition, IL17A changed the gene-expression profile and the behavior of nonmetastatic tumor cells, causing tumor growth in vivo, confirming the protumor role of IL17. Furthermore, high IL17 expression was associated with lower disease-free survival and worse prognosis in IDC patients. Thus, IL17 blockade represents an attractive approach for the control of invasive breast tumors.
Collapse
Affiliation(s)
- Luciana Benevides
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Denise Morais da Fonseca
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Paula Barbim Donate
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Daniel Guimarães Tiezzi
- Department of Gynecology and Obstetrics, Breast Disease Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Daniel D De Carvalho
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, and Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Jurandyr M de Andrade
- Department of Gynecology and Obstetrics, Breast Disease Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gislaine A Martins
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute and Department of Medicine and Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, California
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
56
|
Khan D, Ansar Ahmed S. Regulation of IL-17 in autoimmune diseases by transcriptional factors and microRNAs. Front Genet 2015; 6:236. [PMID: 26236331 PMCID: PMC4500956 DOI: 10.3389/fgene.2015.00236] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/22/2015] [Indexed: 12/21/2022] Open
Abstract
In recent years, IL-17A (IL-17), a pro-inflammatory cytokine, has received intense attention of researchers and clinicians alike with documented effects in inflammation and autoimmune diseases. IL-17 mobilizes, recruits and activates different cells to increase inflammation. Although protective in infections, overproduction of IL-17 promotes inflammation in autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, psoriasis, among others. Regulating IL-17 levels or action by using IL-17-blocking antibodies or IL-17R antagonist has shown to attenuate experimental autoimmune diseases. It is now known that in addition to IL-17-specific transcription factor, RORγt, several other transcription factors and select microRNAs (miRNA) regulate IL-17. Given that miRNAs are dysregulated in autoimmune diseases, a better understanding of transcriptional factors and miRNA regulation of IL-17 expression and function will be essential for devising potential new therapies. In this review, we will overview IL-17 induction and function in relation to autoimmune diseases. In addition, current findings on transcriptional regulation of IL-17 induction and plausible interplay between IL-17 and miRNA in autoimmune diseases are highlighted.
Collapse
Affiliation(s)
- Deena Khan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University Blacksburg, VA, USA
| | - S Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University Blacksburg, VA, USA
| |
Collapse
|
57
|
Grishkan IV, Tosi DM, Bowman MD, Harary M, Calabresi PA, Gocke AR. Antigenic Stimulation of Kv1.3-Deficient Th Cells Gives Rise to a Population of Foxp3-Independent T Cells with Suppressive Properties. THE JOURNAL OF IMMUNOLOGY 2015; 195:1399-1407. [PMID: 26150529 DOI: 10.4049/jimmunol.1403024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 06/12/2015] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the CNS that has been linked with defects in regulatory T cell function. Therefore, strategies to selectively target pathogenic cells via enhanced regulatory T cell activity may provide therapeutic benefit. Kv1.3 is a voltage-gated potassium channel expressed on myelin-reactive T cells from MS patients. Kv1.3-knockout (KO) mice are protected from experimental autoimmune encephalomyelitis, an animal model of MS, and Kv1.3-KO Th cells display suppressive capacity associated with increased IL-10. In this article, we demonstrate that myelin oligodendrocyte glycoprotein-specific Kv1.3-KO Th cells exhibit a unique regulatory phenotype characterized by high CD25, CTLA4, pSTAT5, FoxO1, and GATA1 expression without a corresponding increase in Foxp3. These phenotypic changes result from increased signaling through IL-2R. Moreover, myelin oligodendrocyte glycoprotein-specific Kv1.3-KO Th cells can ameliorate experimental autoimmune encephalomyelitis following transfer to wild-type recipients in a manner that is partially dependent on IL-2R and STAT5 signaling. The present study identifies a population of Foxp3(-) T cells with suppressive properties that arises in the absence of Kv1.3 and enhances the understanding of the molecular mechanism by which these cells are generated. This increased understanding could contribute to the development of novel therapies for MS patients that promote heightened immune regulation.
Collapse
Affiliation(s)
- Inna V Grishkan
- Department of Neurology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, USA
| | - Dominique M Tosi
- Department of Neurology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, USA
| | - Melissa D Bowman
- Department of Neurology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, USA
| | - Maya Harary
- Department of Neurology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, USA
| | - Anne R Gocke
- Department of Neurology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, USA
| |
Collapse
|
58
|
Xiao J, Liu W, Chen Y, Deng W. Recombinant human PDCD5 (rhPDCD5) protein is protective in a mouse model of multiple sclerosis. J Neuroinflammation 2015; 12:117. [PMID: 26068104 PMCID: PMC4474568 DOI: 10.1186/s12974-015-0338-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 06/04/2015] [Indexed: 12/21/2022] Open
Abstract
Background In multiple sclerosis (MS) and its widely used animal model, experimental autoimmune encephalomyelitis (EAE), autoreactive T cells contribute importantly to central nervous system (CNS) tissue damage and disease progression. Promoting apoptosis of autoreactive T cells may help eliminate cells responsible for inflammation and may delay disease progression and decrease the frequency and severity of relapse. Programmed cell death 5 (PDCD5) is a protein known to accelerate apoptosis in response to various stimuli. However, the effects of recombinant human PDCD5 (rhPDCD5) on encephalitogenic T cell-mediated inflammation remain unknown. Methods We examined the effects of intraperitoneal injection of rhPDCD5 (10 mg/kg) on EAE both prophylactically (started on day 0 post-EAE induction) and therapeutically (started on the onset of EAE disease at day 8), with both of the treatment paradigms being given every other day until day 25. Repeated measures two-way analysis of variance was used for statistical analysis. Results We showed that the anti-inflammatory effects of rhPDCD5 were due to a decrease in Th1/Th17 cell frequency, accompanied by a reduction of proinflammatory cytokines, including IFN-γ and IL-17A, and were observed in both prophylactic and therapeutic regimens of rhPDCD5 treatment in EAE mice. Moreover, rhPDCD5-induced apoptosis of myelin-reactive CD4+ T cells, along with the upregulation of Bax and downregulation of Bcl-2, and with activated caspase 3. Conclusions Our data demonstrate that rhPDCD5 ameliorates the autoimmune CNS disease by inhibiting Th1/Th17 differentiation and inducing apoptosis of predominantly pathogenic T cells. This study provides a novel mechanism to explain the effects of rhPDCD5 on neural inflammation. The work represents a translational demonstration that rhPDCD5 has prophylactic and therapeutic properties in a model of multiple sclerosis.
Collapse
Affiliation(s)
- Juan Xiao
- Medical College, Hubei University of Arts and Science, Xiangyang, 441053, China. .,Department of Immunology, Peking University School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, 38 Xueyuan Road, Beijing, 100191, China. .,Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| | - Wenwei Liu
- Medical College, Hubei University of Arts and Science, Xiangyang, 441053, China.
| | - Yingyu Chen
- Department of Immunology, Peking University School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, 38 Xueyuan Road, Beijing, 100191, China.
| | - Wenbin Deng
- Medical College, Hubei University of Arts and Science, Xiangyang, 441053, China. .,Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| |
Collapse
|
59
|
Lee PW, Yang Y, Racke MK, Lovett-Racke AE. Analysis of TGF-β1 and TGF-β3 as regulators of encephalitogenic Th17 cells: Implications for multiple sclerosis. Brain Behav Immun 2015; 46:44-9. [PMID: 25499467 PMCID: PMC4414699 DOI: 10.1016/j.bbi.2014.12.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/04/2014] [Accepted: 12/05/2014] [Indexed: 12/11/2022] Open
Abstract
The phenotype of the CD4(+) T cells that mediate the CNS pathology in multiple sclerosis is still unclear, and yet a vital question for developing therapies. One of the conundrums is the role of TGF-β in the development of encephalitogenic Th17 cells. In the present study, TGF-β1 and TGF-β3 were directly compared in their capacity to promote the differentiation of myelin-specific Th17 cells that could induce experimental autoimmune encephalomyelitis (EAE). Myelin-specific CD4(+) T cell receptor transgenic cells differentiated with antigen in the presence of IL-6+TGF-β1 or IL-6+TGF-β3 generated T cells that produced robust amounts of IL-17, but were incapable of inducing EAE when transferred into mice. Further analysis of these non-encephalitogenic Th17 cells found that they expressed lower amounts of GM-CSF or IL-23R, both molecules necessary for encephalitogenicity. Thus, TGF-β, irrespective of isoform, negatively regulates the differentiation of encephalitogenic Th17 cells.
Collapse
Affiliation(s)
- Priscilla W. Lee
- Molecular Cellular and Developmental Biology Graduate Program, Department of Microbial Infection and Immunity, The Ohio State University, 460 W 12th Ave, Room 660, Columbus, OH, USA
| | - Yuhong Yang
- Department of Neurology, The Ohio State University Wexner Medical Center, 460 W 12th Ave, Room 660, Columbus, OH, USA.
| | - Michael K. Racke
- Department of Neurology, Department of Neuroscience, The Ohio State University Wexner Medical Center, 460 W 12th Ave, Room 686, Columbus, OH, USA
| | - Amy E. Lovett-Racke
- Corresponding Author: Amy E. Lovett-Racke, Department of Microbial Infection and Immunity, Department of Neuroscience, The Ohio State University Wexner Medical Center, Biomedical Research Tower, 460 W 12th Ave, Room 684, Columbus, OH 43210, USA. Phone: (614) 688-5647; Fax: (614) 292-7544,
| |
Collapse
|
60
|
Racke MK, Yang Y, Lovett-Racke AE. Is T-bet a potential therapeutic target in multiple sclerosis? J Interferon Cytokine Res 2015; 34:623-32. [PMID: 25084179 DOI: 10.1089/jir.2014.0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Treatments for multiple sclerosis (MS) have changed over the past years as our understanding of immunology and neuroscience has evolved. Experimental autoimmune encephalomyelitis (EAE) continues to remain the major model for MS and has been a major vehicle in the development of new therapeutic targets for MS, including new agents such as natalizumab, fingolimod, and dimethyl fumarate. As progress in the molecular understanding of immunology continues, many observations in EAE are pursued with the ultimate goal of defining the pathophysiology of MS and development of innovative treatments for the disease. Although many consider MS to be a T cell-mediated autoimmune disease directed against myelin antigens, the exact cause of the disease is still unknown. For many years, it was thought that myelin-specific T cells that secreted interferon-γ and were proinflammatory were the major T cell subset that mediated the disease, but recent studies on the cytokine phenotype of pathogenic T cells in EAE and MS have opened debate on this issue. Work over the past several years suggests that the transcription factor T-bet appears to be an important factor in T cell encephalitogenicity; however, recent data suggest that it is also dispensable in certain situations, particularly for Th17 cells. Understanding the molecular mechanisms responsible for T cell encephalitogenicity in MS and other autoimmune diseases will be essential in the development of specific therapies for these inflammatory diseases.
Collapse
Affiliation(s)
- Michael K Racke
- 1 Department of Neurology, The Ohio State University Wexner Medical Center , Columbus, Ohio
| | | | | |
Collapse
|
61
|
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: 2.9] [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
|
62
|
Yang Y, Winger RC, Lee PW, Nuro-Gyina PK, Minc A, Larson M, Liu Y, Pei W, Rieser E, Racke MK, Lovett-Racke AE. Impact of suppressing retinoic acid-related orphan receptor gamma t (ROR)γt in ameliorating central nervous system autoimmunity. Clin Exp Immunol 2015; 179:108-18. [PMID: 25142403 DOI: 10.1111/cei.12441] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2014] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated chronic central nervous system (CNS) disease affecting more than 400 000 people in the United States. Myelin-reactive CD4 T cells play critical roles in the formation of acute inflammatory lesions and disease progression in MS and experimental autoimmune encephalomyelitis (EAE), a well-defined mouse model for MS. Current MS therapies are only partially effective, making it necessary to develop more effective therapies that specifically target pathogenic myelin-specific CD4 T cells for MS treatment. While suppressing T-bet, the key transcription factor in T helper type 1 (Th1) cells, has been demonstrated to be beneficial in prevention and treatment of EAE, the therapeutic potential of retinoic acid-related orphan receptor gamma t (ROR)γt, the key transcription factor for Th17 cells, has not been well-characterized. In this study, we characterized the correlation between RORγt expression and other factors affecting T cell encephalitogenicity and evaluated the therapeutic potential of targeting RORγt by siRNA inhibition of RORγt. Our data showed that RORγt expression correlates with interleukin (IL)-17 production, but not with the encephalitogenicity of myelin-specific CD4 T cells. IL-23, a cytokine that enhances encephalitogenicity, does not enhance RORγt expression significantly. Additionally, granulocyte-macrophage colony-stimulating factor (GM-CSF) levels, which correlate with the encephalitogenicity of different myelin-specific CD4 T cell populations, do not correlate with RORγt. More importantly, inhibiting RORγt expression in myelin-specific CD4 T cells with an siRNA does not reduce disease severity significantly in adoptively transferred EAE. Thus, RORγt is unlikely to be a more effective therapeutic target for ameliorating pathogenicity of encephalitogenic CD4 T cells.
Collapse
Affiliation(s)
- Y Yang
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Dubey D, Kieseier BC, Hartung HP, Hemmer B, Miller-Little WA, Stuve O. Clinical management of multiple sclerosis and neuromyelitis optica with therapeutic monoclonal antibodies: approved therapies and emerging candidates. Expert Rev Clin Immunol 2014; 11:93-108. [PMID: 25495182 DOI: 10.1586/1744666x.2015.992881] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Therapeutic monoclonal antibodies (mAbs) are a relatively novel class of drugs that has substantially advanced immunotherapy for patients with multiple sclerosis. The advantage of these agents is that they bind specifically and exclusively to predetermined proteins or cells. Natalizumab was the first mAb in neurology to obtain approval. It is also considered one of the most potent options for annualized relapse rate reduction among available therapeutic options. Alemtuzumab is currently also approved in several countries. Several mAbs have been tested in clinical studies in multiple sclerosis. Here, we review the history of drug development of therapeutic mAbs and their classification. Furthermore, we outline the putative mechanisms of action, clinical evidence and safety of approved mAbs and those in different stages of clinical development in multiple sclerosis and neuromyelitis optica.
Collapse
|
64
|
Hussain RZ, Hayardeny L, Cravens PC, Yarovinsky F, Eagar TN, Arellano B, Deason K, Castro-Rojas C, Stüve O. Immune surveillance of the central nervous system in multiple sclerosis--relevance for therapy and experimental models. J Neuroimmunol 2014; 276:9-17. [PMID: 25282087 PMCID: PMC4301841 DOI: 10.1016/j.jneuroim.2014.08.622] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/15/2014] [Accepted: 08/20/2014] [Indexed: 12/25/2022]
Abstract
Treatment of central nervous system (CNS) autoimmune disorders frequently involves the reduction, or depletion of immune-competent cells. Alternatively, immune cells are being sequestered away from the target organ by interfering with their movement from secondary lymphoid organs, or their migration into tissues. These therapeutic strategies have been successful in multiple sclerosis (MS), the most prevalent autoimmune inflammatory disorder of the CNS. However, many of the agents that are currently approved or in clinical development also have severe potential adverse effects that stem from the very mechanisms that mediate their beneficial effects by interfering with CNS immune surveillance. This review will outline the main cellular components of the innate and adaptive immune system that participate in host defense and maintain immune surveillance of the CNS. Their pathogenic role in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also discussed. Furthermore, an experimental model is introduced that may assist in evaluating the effect of therapeutic interventions on leukocyte homeostasis and function within the CNS. This model or similar models may become a useful tool in the repertoire of pre-clinical tests of pharmacological agents to better explore their potential for adverse events.
Collapse
Affiliation(s)
- Rehana Z Hussain
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | | | - Petra C Cravens
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Felix Yarovinsky
- Department of Immunology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Todd N Eagar
- Histocompatibility and Transplant Immunology, Department of Pathology and Genomic Medicine, The Methodist Hospital Physician Organization, Houston, TX, USA
| | - Benjamine Arellano
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Krystin Deason
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Cyd Castro-Rojas
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany.
| |
Collapse
|
65
|
Nicholas JA, Boster AL, Imitola J, O'Connell C, Racke MK. Design of oral agents for the management of multiple sclerosis: benefit and risk assessment for dimethyl fumarate. Drug Des Devel Ther 2014; 8:897-908. [PMID: 25045248 PMCID: PMC4094574 DOI: 10.2147/dddt.s50962] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Dimethyl fumarate (DMF) is the most recent oral disease-modifying therapy approved by the US Food and Drug Administration and is indicated for the treatment of relapsing forms of multiple sclerosis (MS). Prior to approval for use in MS, DMF and its active metabolite, monomethyl fumarate, had been used for decades as two of the fumaric acid esters in Fumaderm, a medication used in Europe for the treatment of psoriasis. The unique mechanism of action of DMF remains under evaluation; however, it has been shown to act through multiple pathways leading to shifts away from the Th1 proinflammatory response to the less inflammatory Th2 response. Preliminary data suggest that DMF may induce neuroprotective effects in central nervous system white matter, although further studies are needed to demonstrate these effects on inflammatory demyelination. The DMF Phase III clinical trials demonstrated its efficacy with regard to a reduction in the annualized relapse rate and reductions in new or enlarging T2 lesions and numbers of gadolinium-enhancing lesions on magnetic resonance imaging. DMF has a well-defined safety profile, given the experience with its use in the treatment of psoriasis, and more recently from the DMF clinical trials program and post-marketing era for treatment of MS. The safety profile and oral mode of administration of DMF place it as an attractive first-line therapy option for the treatment of relapsing forms of MS. Long-term observational studies will be needed to determine the effects of DMF on progression of disability in MS.
Collapse
Affiliation(s)
- Jacqueline Ann Nicholas
- Department of Neurology and Multiple Sclerosis Center, The Ohio State University, Columbus, OH, USA
| | - Aaron Lee Boster
- Department of Neurology and Multiple Sclerosis Center, The Ohio State University, Columbus, OH, USA
| | - Jaime Imitola
- Department of Neurology and Multiple Sclerosis Center, The Ohio State University, Columbus, OH, USA ; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Colleen O'Connell
- Department of Neurology and Multiple Sclerosis Center, The Ohio State University, Columbus, OH, USA
| | - Michael Karl Racke
- Department of Neurology and Multiple Sclerosis Center, The Ohio State University, Columbus, OH, USA ; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
66
|
Lin H, Tong ZH, Xu QQ, Wu XZ, Wang XJ, Jin XG, Ma WL, Cheng X, Zhou Q, Shi HZ. Interplay of Th1 and Th17 cells in murine models of malignant pleural effusion. Am J Respir Crit Care Med 2014; 189:697-706. [PMID: 24410406 DOI: 10.1164/rccm.201310-1776oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE IFN-γ-producing CD4(+) T (Th1) cells and IL-17-producing CD4(+) T (Th17) cells have been found to be involved in multiple malignancies; however, the reciprocal relationship between Th1 and Th17 cells in malignant pleural effusion (MPE) remains to be elucidated. OBJECTIVES To explore the differentiation and immune regulation of Th1 and Th17 cells in the development of MPE in murine models. METHODS The distribution and differentiation of Th1 and Th17 cells in MPE were investigated in IFN-γ(-/-), IL-17(-/-), and wild-type mice. The effects of Th1 and Th17 cells on the development of MPE and the survival of mice bearing MPE were also investigated. MEASUREMENTS AND MAIN RESULTS We have demonstrated that increased Th1 and Th17 cells could be found in MPE as compared with blood and spleen. Compared with wild-type mice, Th17 cells were markedly augmented in MPE from IFN-γ(-/-) mice, and improved survival could be seen in IFN-γ(-/-) mice. Th1 cell numbers were elevated in MPE from IL-17(-/-) mice, and decreased survival could be seen in IL-17(-/-) mice. The in vitro experiments showed that IFN-γ deficiency promoted Th17-cell differentiation by suppressing the STAT3 pathway and that IL-17 deficiency promoted Th1-cell differentiation by suppressing the STAT1 pathway. CONCLUSIONS In mouse models of MPE, IFN-γ inhibited Th17-cell differentiation, whereas IL-17 inhibited Th1-cell differentiation. IL-17 inhibited the formation of MPE and improved the survival of mice bearing MPE; in contrast, IFN-γ promoted MPE formation and mouse death.
Collapse
Affiliation(s)
- Hua Lin
- 1 Department of Respiratory and Critical Care Medicine and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Azizi G, Jadidi-Niaragh F, Mirshafiey A. Th17 Cells in Immunopathogenesis and treatment of rheumatoid arthritis. Int J Rheum Dis 2014; 16:243-53. [PMID: 23981743 DOI: 10.1111/1756-185x.12132] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by the sequestration of various leukocyte subpopulations within both the developing pannus and synovial space. The chronic nature of this disease results in inflammation of multiple joints, with subsequent destruction of the joint cartilage and erosion of bone. Identification of T helper (Th)17 cells led to breaking the dichotomy of the Th1/Th2 axis in immunopathogenesis of autoimmune diseases such as RA, and its experimental model, collagen-induced arthritis (CIA). Th17 cells produce cytokines, including interleukin (IL)-17, IL-6, IL-21, IL-22 and tumor necrosis factor (TNF)-α, with pro-inflammatory effects, which appear to have a role in immunopathogenesis of RA. Regarding the wide ranging production of pro-inflammatory cytokines and chemokines by Th17 cells, it is expected that Th17 cell could be a potent pathogenic factor in disease immunopathophysiology. Thus the identification of effector mechanisms used by Th17 cells in induction of disease lesions may open new prospects for designing a new therapeutic strategy for treatment of RA.
Collapse
Affiliation(s)
- Gholamreza Azizi
- Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | | | | |
Collapse
|
68
|
Effector molecules released by Th1 but not Th17 cells drive an M1 response in microglia. Brain Behav Immun 2014; 37:248-59. [PMID: 24412213 DOI: 10.1016/j.bbi.2014.01.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 12/15/2022] Open
Abstract
Microglia act as sensors of inflammation in the central nervous system (CNS) and respond to many stimuli. Other key players in neuroinflammatory diseases are CD4+ T helper cell (Th) subsets that characteristically secrete IFN-γ (Th1) or IL-17 (Th17). However, the potential of a distinct cytokine milieu generated by these effector T cell subsets to modulate microglial phenotype and function is poorly understood. We therefore investigated the ability of factors secreted by Th1 and Th17 cells to induce microglial activation. In vitro experiments wherein microglia were cultured in the presence of supernatants derived from polarized Th1 or Th17 cultures, revealed that Th1-associated factors could directly activate and trigger a proinflammatory M1-type gene expression profile in microglia that was cell-cell contact independent, whereas Th17 cells or its associated factors did not have any direct influence on microglia. To assess the effects of the key Th17 effector cytokine IL-17A in vivo we used transgenic mice in which IL-17A is specifically expressed in astrocytes. Flow cytometric and histological analysis revealed only subtle changes in the phenotype of microglia suggesting only minimal effects of constitutively produced IL-17A on microglia in vivo. Neither IL-23 signaling nor addition of GM-CSF, a recently described effector molecule of Th17 cells, changed the incapacity of Th17 cells to activate microglia. These findings demonstrate a potent effect of Th1 cells on microglia, however, the mechanism of how Th17 cells achieve their effect in CNS inflammation remains unclear.
Collapse
|
69
|
Tatano Y, Shimizu T, Tomioka H. Unique macrophages different from M1/M2 macrophages inhibit T cell mitogenesis while upregulating Th17 polarization. Sci Rep 2014; 4:4146. [PMID: 24553452 PMCID: PMC3930092 DOI: 10.1038/srep04146] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/05/2014] [Indexed: 02/06/2023] Open
Abstract
Mycobacterial infection induces suppressor macrophages (MΦs), causing disease exacerbation. There are two major MΦ subsets (M1 and M2 MΦs) that are phenotypically and functionally different. Here, we examined which of the MΦ subsets the mycobacterial infection-induced suppressor MΦs (MIS-MΦs) belong to. MIS-MΦs down-regulated T cell production of Th1 and Th2 cytokines but markedly increased production of interleukin (IL)-17A and IL-22 through up-regulation of Th17 cell expansion. In this phenomenon, a novel MΦ population, which is functionally distinguishable from M1 and M2 MΦ subsets and possesses unique phenotypes (IL-12(+), IL-1β(high), IL-6(+), tumor necrosis factor (TNF)-α(+), nitric oxide synthase (NOS) 2(+), CCR7(high), IL-10(high), arginase (Arg)-1(-), mannose receptor (MR)(low), Ym1(high), Fizz(low), and CD163(high)), played central roles through the action of IL-6 and transforming growth factor (TGF)-β but not IL-21 and IL-23. This new type of MΦ population was induced in infected mice and actively supported the in vivo expansion of Th17 cells.
Collapse
Affiliation(s)
- Yutaka Tatano
- Department of Microbiology and Immunology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Toshiaki Shimizu
- Department of Microbiology and Immunology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| | - Haruaki Tomioka
- Department of Microbiology and Immunology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan
| |
Collapse
|
70
|
Shi G, Field DJ, Ko KA, Ture S, Srivastava K, Levy S, Kowalska MA, Poncz M, Fowell DJ, Morrell CN. Platelet factor 4 limits Th17 differentiation and cardiac allograft rejection. J Clin Invest 2014; 124:543-52. [PMID: 24463452 DOI: 10.1172/jci71858] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 11/07/2013] [Indexed: 01/12/2023] Open
Abstract
Th cells are the major effector cells in transplant rejection and can be divided into Th1, Th2, Th17, and Treg subsets. Th differentiation is controlled by transcription factor expression, which is driven by positive and negative cytokine and chemokine stimuli at the time of T cell activation. Here we discovered that chemokine platelet factor 4 (PF4) is a negative regulator of Th17 differentiation. PF4-deficient and platelet-deficient mice had exaggerated immune responses to cardiac transplantation, including increased numbers of infiltrating Th17 cells and increased plasma IL-17. Although PF4 has been described as a platelet-specific molecule, we found that activated T cells also express PF4. Furthermore, bone marrow transplantation experiments revealed that T cell-derived PF4 contributes to a restriction in Th17 differentiation. Taken together, the results of this study demonstrate that PF4 is a key regulator of Th cell development that is necessary to limit Th17 differentiation. These data likely will impact our understanding of platelet-dependent regulation of T cell development, which is important in many diseases, in addition to transplantation.
Collapse
|
71
|
Xue Y, Yang Y, Su Z, Barnie PA, Zheng D, Zhang Y, Xu Y, Wang S, Xu H. Local delivery of T-bet shRNA reduces inflammation in collagen II-induced arthritis via downregulation of IFN-γ and IL-17. Mol Med Rep 2014; 9:899-903. [PMID: 24425064 DOI: 10.3892/mmr.2014.1893] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 01/06/2014] [Indexed: 11/06/2022] Open
Abstract
Th1 and Th17 cells are involved in the pathogenesis of rheumatoid arthritis (RA). T-bet, a Th1-specific transcription factor, appears to drive the maturation of Th1 and IFN-γ secretion. In the present study, we established the T-bet shRNA recombinant plasmid (p-T-shRNA) and explored its possible anti-inflammatory effect in a collagen-induced arthritis (CIA) model by local injection of plasmid vectors. For the initiation of CIA, DBA/1J mice were immunized with type II collagen (CII) in Freund's adjuvant and the CII-immunized mice were treated with p-T-shRNA. Levels of T-bet, IFN-γ, IL-17 and RORγt mRNA in splenocytes and synovial joints were measured by quantitative real-time PCR and T-bet expression in joint tissue was detected by immunohistochemistry staining. The intracellular IFN-γ and IL-17 were analyzed by flow cytometry (FCM). The results demonstrated that therapeutic administration on the local joints with p-T-shRNA significantly suppressed IFN-γ and IL-17 gene expression and improved the pathogenesis of arthritis in CIA mice, while administration of a plasmid expressing T-bet (pIRES-T-bet) accelerated the disease onset. Our study suggests that T-bet may be developed as a potential target for arthritis therapy.
Collapse
Affiliation(s)
- Yuan Xue
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yong Yang
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Zhaoliang Su
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Prince Amoatt Barnie
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Dong Zheng
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yun Zhang
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yan Xu
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Shengjun Wang
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Huaxi Xu
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
72
|
Hegab D, Saudi W, Ammo DA, El Bedewy M, Elhabian N, Gamei M. Role of interleukin-23 in the immunopathogenesis of systemic lupus erythematosus. EGYPTIAN JOURNAL OF DERMATOLOGY AND VENEROLOGY 2014. [DOI: 10.4103/1110-6530.150269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
73
|
Advances in siRNA delivery to T-cells: potential clinical applications for inflammatory disease, cancer and infection. Biochem J 2013; 455:133-47. [DOI: 10.1042/bj20130950] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The specificity of RNAi and its ability to silence ‘undruggable’ targets has made inhibition of gene expression in T-cells with siRNAs an attractive potential therapeutic strategy for the treatment of inflammatory disease, cancer and infection. However, delivery of siRNAs into primary T-cells represents a major hurdle to their use as potential therapeutic agents. Recent advances in siRNA delivery through the use of electroporation/nucleofection, viral vectors, peptides/proteins, nanoparticles, aptamers and other agents have now enabled efficient gene silencing in primary T-cells both in vitro and in vivo. Overcoming such barriers in siRNA delivery offers exciting new prospects for directly targeting T-cells systemically with siRNAs, or adoptively transferring T-cells back into patients following ex vivo manipulation with siRNAs. In the present review, we outline the challenges in delivering siRNAs into primary T-cells and discuss the mechanism and therapeutic opportunities of each delivery method. We emphasize studies that have exploited RNAi-mediated gene silencing in T-cells for the treatment of inflammatory disease, cancer and infection using mouse models. We also discuss the potential therapeutic benefits of manipulating T-cells using siRNAs for the treatment of human diseases.
Collapse
|
74
|
Nuclear factor kappa B (NF-κB) in multiple sclerosis pathology. Trends Mol Med 2013; 19:604-13. [PMID: 24007818 DOI: 10.1016/j.molmed.2013.08.001] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/12/2013] [Accepted: 08/02/2013] [Indexed: 12/15/2022]
Abstract
The nuclear factor kappa B (NF-κB) signaling cascade plays a critical role in the regulation of immune and inflammatory responses and has been implicated in the pathogenesis of autoimmune demyelinating diseases such as multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), the main animal model of MS. NF-κB is essential for peripheral immune cell activation and the induction of pathology, but also plays crucial roles in resident cells of the central nervous system (CNS) during disease development. Here we review recent evidence clarifying the role of NF-κB in the different cell compartments contributing to MS pathology and its implications for the development of therapeutic strategies for the treatment of MS and other demyelinating pathologies of the CNS.
Collapse
|
75
|
Grifka-Walk HM, Lalor SJ, Segal BM. Highly polarized Th17 cells induce EAE via a T-bet independent mechanism. Eur J Immunol 2013; 43:2824-31. [PMID: 23878008 DOI: 10.1002/eji.201343723] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/02/2013] [Accepted: 07/17/2013] [Indexed: 12/12/2022]
Abstract
In the MOG35-55 induced EAE model, autoreactive Th17 cells that accumulate in the central nervous system acquire Th1 characteristics via a T-bet dependent mechanism. It remains to be determined whether Th17 plasticity and encephalitogenicity are causally related to each other. Here, we show that IL-23 polarized T-bet(-/-) Th17 cells are unimpaired in either activation or proliferation, and induce higher quantities of the chemokines RANTES and CXCL2 than WT Th17 cells. Unlike their WT counterparts, T-bet(-/-) Th17 cells retain an IL-17(hi) IFN-γ(neg-lo) cytokine profile following adoptive transfer into syngeneic hosts. This population of highly polarized Th17 effectors is capable of mediating EAE, albeit with a milder clinical course. It has previously been reported that the signature Th1 and Th17 effector cytokines, IFN-γ and IL-17, are dispensable for the development of autoimmune demyelinating disease. The current study demonstrates that the "master regulator" transcription factor, T-bet, is also not universally required for encephalitogenicity. Our results contribute to a growing body of data showing heterogeneity of myelin-reactive T cells and the independent mechanisms they employ to inflict damage to central nervous system tissues, complicating the search for therapeutic targets relevant across the spectrum of individuals with multiple sclerosis.
Collapse
Affiliation(s)
- Heather M Grifka-Walk
- Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | | |
Collapse
|
76
|
O'Connor RA, Cambrook H, Huettner K, Anderton SM. T-bet is essential for Th1-mediated, but not Th17-mediated, CNS autoimmune disease. Eur J Immunol 2013; 43:2818-23. [PMID: 23878019 PMCID: PMC4068221 DOI: 10.1002/eji.201343689] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/14/2013] [Accepted: 07/17/2013] [Indexed: 11/08/2022]
Abstract
T cells that produce both IL-17 and IFN-γ, and co-express ROR-γt and T-bet, are often found at sites of autoimmune inflammation. However, it is unknown whether this co-expression of T-bet with ROR-γt is a prerequisite for immunopathology. We show here that T-bet is not required for the development of Th17-driven experimental autoimmune encephalomyelitis (EAE). The disease was not impaired in T-bet(-/-) mice and was associated with low IFN-γ production and elevated IL-17 production among central nervous system (CNS) infiltrating CD4(+) T cells. T-bet(-/-) Th17 cells generated in the presence of IL-6/TGF-β/IL-1 and IL-23 produced GM-CSF and high levels of IL-17 and induced disease upon transfer to naïve mice. Unlike their WT counterparts, these T-bet(-/-) Th17 cells did not exhibit an IL-17→IFN-γ switch upon reencounter with antigen in the CNS, indicating that this functional change is not critical to disease development. In contrast, T-bet was absolutely required for the pathogenicity of myelin-responsive Th1 cells. T-bet-deficient Th1 cells failed to accumulate in the CNS upon transfer, despite being able to produce GM-CSF. Therefore, T-bet is essential for establishing Th1-mediated inflammation but is not required to drive IL-23-induced GM-CSF production, or Th17-mediated autoimmune inflammation.
Collapse
Affiliation(s)
- Richard A O'Connor
- Medical Research Council Centre for Inflammation Research, Centre for Multiple Sclerosis Research and Centre for Immunity Infection and Evolution, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | | | | |
Collapse
|
77
|
Podojil JR, Miller SD. Targeting the B7 family of co-stimulatory molecules: successes and challenges. BioDrugs 2013; 27:1-13. [PMID: 23329394 DOI: 10.1007/s40259-012-0001-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As more patient data is cross-referenced with animal models of disease, the primary focus on T(h)1 autoreactive effector cell function in autoimmune diseases, such as rheumatoid arthritis and multiple sclerosis, has shifted towards the role of T(h)17 autoreactive effector cells and the ability of regulatory T cells (T(reg)) to modulate the pro-inflammatory autoimmune response. Therefore, the currently favored hypothesis is that a delicate balance between T(h)1/17 effector cells and T(reg) cell function is critical in the regulation of inflammatory autoimmune disease. An intensive area of research with regard to the T(h)1/17:T(reg) cell balance is the utilization of blockade and/or ligation of various co-stimulatory or co-inhibitory molecules, respectively, during ongoing disease to skew the immune response toward a more tolerogenic/regulatory state. Currently, FDA-approved therapies for multiple sclerosis patients are all aimed at the suppression of immune cell function. The other favored method of treatment is a modulation or deletion of autoreactive immune cells via short-term blockade of activating co-stimulatory receptors via treatment with fusion proteins such as CTLA4-Ig and CTLA4-FasL. Based on the initial success of CTLA4-Ig, there are additional fusion proteins that are currently under development. Examples of the more recently identified B7/CD28 family members are PD-L1, PD-L2, inducible co-stimulatory molecule-ligand (ICOS-L), B7-H3, and B7-H4, all of which may emerge as potential fusion protein therapeutics, each with unique, yet often overlapping functions. The expression of both stimulatory and inhibitory B7 molecules seems to play an essential role in modulating immune cell function through a variety of mechanisms, which is supported by findings that suggest each B7 molecule has developed its own indispensable niche in the immune system. As more data are generated, the diagnostic and therapeutic potential of the above B7 family-member-derived fusion proteins becomes ever more apparent. Besides defining the biology of these B7/CD28 family members in vivo, additional difficulty in the development of these therapies lies in maintaining the normal immune functions of recognition and reaction to non-self-antigens following viral or bacterial infection in the patient. Further complicating the clinical translation of these therapies, the mechanism of action identified for a particular reagent may depend upon the method of immune-cell activation and the subset of immune cells targeted in the study.
Collapse
Affiliation(s)
- Joseph R Podojil
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Tarry 6-718, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | | |
Collapse
|
78
|
Paintlia AS, Paintlia MK, Singh AK, Singh I. Modulation of Rho-Rock signaling pathway protects oligodendrocytes against cytokine toxicity via PPAR-α-dependent mechanism. Glia 2013; 61:1500-1517. [PMID: 23839981 DOI: 10.1002/glia.22537] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/06/2013] [Accepted: 05/07/2013] [Indexed: 12/13/2022]
Abstract
We earlier documented that lovastatin (LOV)-mediated inhibition of small Rho GTPases activity protects vulnerable oligodendrocytes (OLs) in mixed glial cell cultures stimulated with Th1 cytokines and in a murine model of multiple sclerosis (MS). However, the precise mechanism of OL protection remains unclear. We here employed genetic and biochemical approaches to elucidate the underlying mechanism that protects LOV treated OLs from Th1 (tumor necrosis factor-α) and Th17 (interleukin-17) cytokines toxicity in in vitro. Cytokines enhanced the reactive oxygen species (ROS) generation and mitochondrial membrane depolarization with corresponding lowering of glutathione (reduced) level in OLs and that were reverted by LOV. In addition, the expression of ROS detoxifying enzymes (catalase and superoxide-dismutase 2) and the transactivation of peroxisome proliferators-activated receptor (PPAR)-α/-β/-γ including PPAR-γ coactivator-1α were enhanced by LOV in similarly treated OLs. Interestingly, LOV-mediated inhibition of small Rho GTPases, i.e., RhoA and cdc42, and Rho-associated kinase (ROCK) activity enhanced the levels of PPAR ligands in OLs via extracellular signal regulated kinase (1/2)/p38 mitogen-activated protein kinase/cytoplasmic phospholipase 2/cyclooxygenase-2 signaling cascade activation. Small hairpin RNA transfection-based studies established that LOV mainly enhances PPAR-α and less so of PPAR-β and PPAR-γ transactivation that enhances ROS detoxifying defense in OLs. In support of this, the observed LOV-mediated protection was lacking in PPAR-α-deficient OLs exposed to cytokines. Collectively, these data provide unprecedented evidence that LOV-mediated inhibition of the Rho-ROCK signaling pathway boosts ROS detoxifying defense in OLs via PPAR-α-dependent mechanism that has implication in neurodegenerative disorders including MS.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina
| | - Manjeet K Paintlia
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Ralph H. Johnson VA Medical Center, Charleston, South Carolina
| | - Inderjit Singh
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina
| |
Collapse
|
79
|
Rouse M, Nagarkatti M, Nagarkatti PS. The role of IL-2 in the activation and expansion of regulatory T-cells and the development of experimental autoimmune encephalomyelitis. Immunobiology 2013; 218:674-82. [PMID: 22954711 PMCID: PMC3582788 DOI: 10.1016/j.imbio.2012.08.269] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 08/02/2012] [Indexed: 10/28/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease that affects ≈ 400,000 people in the US. It is a chronic, disabling disease with no cure, and the current treatment includes use of immunosuppressive drugs that often exhibit toxic side effects. Thus, there is a pressing need for alternate and more effective treatment strategies that target the components of inflammatory cells. In recent years, regulatory T-cells (Tregs) have been found to play an important role in preventing the development of autoimmunity. Thus, expansion of Tregs in vivo has the therapeutic potential against autoimmune diseases. Because Tregs constitutively express IL-2 receptors (IL-2Rs), we tested the effect of administration of IL-2 on the development of experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). We used IL-2 both before (pre-treatment) or after (post-treatment) immunization with myelin oligodendrocyte glycoprotein (MOG35-55) peptide to induce EAE. The data demonstrated that pre-treatment with a moderate dose of IL-2 caused significant amelioration of EAE. Tissue histopathology of the central nervous system also confirmed the effectiveness of IL-2 pre-treatment by decreasing cellular infiltration in the spinal cord and preserving tissue integrity. IL-2 pretreatment expanded Treg cells while preventing the induction of Th17 during EAE development. In contrast, post-treatment with IL-2 failed to suppress EAE despite induction of Tregs. Together, these studies demonstrate that while expansion of Tregs using IL-2, prior to immunization or the onset of disease, can suppress the immune response, their role is limited after the antigen-specific response is triggered. Because IL-2 is used to treat certain types of cancers, and Tregs have applications in preventing the rejection of transplants, our studies also provide useful information on the use and limitations of Tregs in such clinical manifestations.
Collapse
Affiliation(s)
- Michael Rouse
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Prakash S. Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| |
Collapse
|
80
|
Dutta A, Miaw SC, Yu JS, Chen TC, Lin CY, Lin YC, Chang CS, He YC, Chuang SH, Yen MI, Huang CT. Altered T-bet dominance in IFN-γ-decoupled CD4+ T cells with attenuated cytokine storm and preserved memory in influenza. THE JOURNAL OF IMMUNOLOGY 2013; 190:4205-14. [PMID: 23509355 DOI: 10.4049/jimmunol.1202434] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cytokine storm has been postulated as one of the major causes of mortality in patients with severe respiratory viral infections such as influenza. With the help of an influenza Ag- specific mouse experimental system, we report that CD4(+) T cells contribute effector cytokines leading to lung inflammation in acute influenza. Although virus can no longer be detected from tissues 14 d postinfection, virus-derived Ag continues to drive a CD4(+) T cell response after viral clearance. Ag-specific CD4(+) T cells proliferate and evolve into memory CD4(+) T cells efficiently, but the production of effector cytokines is seriously hampered during this phase. This decoupling of proliferation and effector cytokine production doesn't appear in conjunction with increased suppression by regulatory T cells or decreased induction of transcription factors. Rather, GATA-3 and ROR-γt levels are elevated when compared with cells that have effector cytokine production. T-bet dominance over GATA-3 and ROR-γt decreases with the disarmament of effector cytokine production. Importantly, upon reinfection, these decoupled cells produce elevated levels of IFN-γ and were effective in virus eradication. These results provide a mechanism through altered T-bet dominance to dampen the cytokine storm without impeding the generation of memory T cells in influenza virus infection.
Collapse
Affiliation(s)
- Avijit Dutta
- Division of Infectious Diseases, Department of Medicine, Chang Gung University and Chang Gung Memorial Hospital, Kweishan, Taoyuan 33333, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Abstract
T cells surviving the clonal selection process emigrate from the thymus to the periphery as immature naive T cells. In the periphery, upon activation under specific cytokine milieus, naive T cells adopt specific effector phenotypes, e.g. T-helper 1 (Th1), Th2, or Th17, and acquire diverse functions to control a myriad of pathogens, tissue injuries, and other immunological insults. Interleukin-23 (IL-23) is one of the key cytokines that shapes the development and function of Th17 cells with characteristic expression of retinoic acid receptor-related orphan receptor γ-t (RORγt), IL-17, IL-22, and granulocyte macrophage colony-stimulating factor (GM-CSF). More recently, emerging data suggest that IL-23 also promotes development of 'natural Th17' (nTh17) cells that arise from the thymus, analogous to natural regulatory T cells (nTreg). We are just beginning to understand the unique thymic developmental path of nTh17 cells, which are distinct from antigen-experienced memory Th17 cells. In this review, we explore the differentiation and function of inducible, natural, and memory Th17 subsets, which encompass a broad range of immune functions while maintaining tissue hemostasis, and highlight the participation of IL-23 during the life cycle of Th17 cells.
Collapse
Affiliation(s)
- Luis A Zúñiga
- Pathway Biology, Merck Research Laboratories, Palo Alto, CA 94304-1104, USA
| | | | | | | |
Collapse
|
82
|
Abstract
During the past decade, it has been firmly established that IL-23 is essential for disease development in several models of autoimmune disease, including psoriatic skin inflammation, inflammatory bowel disease (IBD), and experimental autoimmune encephalomyelitis (EAE). The mechanism by which IL-23 exerts its pathogenic role has been mostly scrutinized in the context of Th17 cells, which were thought to mediate autoimmunity by secretion of IL-17 family cytokines. However, the picture emerging now is one of multiple IL-23-responsive cell types, pro-inflammatory cytokine induction, and pathogenic "licensing" following an IL-23-dominated interaction between the T cell and the antigen-presenting cell (APC). This review will focus on our changing view of IL-23-dependent autoimmune pathologies with a particular emphasis on the responder cells and their IL-23-induced factors that ultimately mediate tissue destruction.
Collapse
Affiliation(s)
- Andrew L Croxford
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
83
|
Wu W, Shi S, Ljunggren HG, Cava AL, Van Kaer L, Shi FD, Liu R. NK cells inhibit T-bet-deficient, autoreactive Th17 cells. Scand J Immunol 2013; 76:559-66. [PMID: 22928727 DOI: 10.1111/j.1365-3083.2012.02773.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The differentiation and maintenance of Th17 cells require a unique cytokine milieu and activation of lineage-specific transcription factors. This process appears to be antagonized by the transcription factor T-bet, which controls the differentiation of Th1 cells. Considering that T-bet-deficient (T-bet(-/-) ) mice are largely devoid of natural killer (NK) cells due to a defect in the terminal maturation of these cells, and because NK cells can influence the differentiation of T helper cells, we investigated whether the absence of NK cells in T-bet-deficient mice contributes to the augmentation of autoreactive Th17 cell responses. We show that the loss of T-bet renders the transcription factors Rorc and STAT3 highly responsive to activation by stimuli provided by NK cells. Furthermore, reconstitution of T-bet(-/-) mice with wild-type NK cells inhibited the development of autoreactive Th17 cells through NK cell-derived production of IFN-γ. These results identify NK cells as critical regulators in the development of autoreactive Th17 cells and Th17-mediated pathology.
Collapse
Affiliation(s)
- W Wu
- Department of Neurology and Tianjin Neurologic Institute, Tianjin Medical University General Hospital, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
84
|
Qi ZX, Wang LY, Fan YC, Zhang JJ, Li T, Wang K. Increased peripheral RORα and RORγt mRNA expression is associated with acute-on-chronic hepatitis B liver failure. J Viral Hepat 2012; 19:811-822. [PMID: 23043388 DOI: 10.1111/j.1365-2893.2012.01603.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
T helper cells17 (Th17) have accurate but inconclusive roles in the pathogenesis of acute-on-chronic hepatitis B liver failure (ACHBLF). Retinoic acid-related orphan receptor γ t(RORγt) and RORα are two lineage-specific nuclear receptors directly mediating Th17 differentiation. This study was aimed to evaluate the gene expression of RORα and RORγt and their potential role in ACHBLF. Forty patients with liver failure, 30 with chronic hepatitis B (CHB) and 20 healthy controls were studied. The mRNA levels of RORα and RORγt in peripheral mononuclear cells were determined by quantitative real-time polymerase chain reaction. The frequency of peripheral Th17 cells was determined using flow cytometry. The serum levels of interleukin-6(IL-6), transforming growth factor -β (TGF-β), interleukin-17(IL-17), interleukin-23(IL-23) and interferon-γ (IFN-γ) were measured by enzyme-linked immunosorbent assay. The frequency of peripheral Th17 cells in patients with liver failure was significantly increased compared to patients with CHB and controls. The peripheral mRNA levels of RORα and RORγt in hepatitis B-associated acute-on-chronic liver failure were significantly higher than in patients with CHB and controls as were the serum levels of IL-6 and TGF-β. The serum level of IFN-γ in patients with acute-on-chronic liver failure from HBV was significantly higher than patients with CHB but lower than controls. In patients with acute-on-chronic liver failure associated with HBV, RORγt, IL-6 and IL-23 were positively correlated with the frequency of Th17 cells, while RORα, TGF-β and IFN-γ had no correlation with the latter. The mRNA level of RORγt was positively correlated with model of end-stage liver disease (MELD) score, but there was no correlation of RORα and MELD score. RORγt plays an important role in the pathogenesis of acute-on-chronic HBV-associated liver failure and might be considered to be a candidate factor consistent with the severity of disease.
Collapse
MESH Headings
- Adult
- Female
- Hepatitis B, Chronic/genetics
- Hepatitis B, Chronic/immunology
- Hepatitis B, Chronic/metabolism
- Humans
- Interferon-gamma/blood
- Interleukin-17/blood
- Interleukin-23/blood
- Interleukin-6/blood
- Liver Failure, Acute/genetics
- Liver Failure, Acute/immunology
- Liver Failure, Acute/metabolism
- Lymphocyte Count
- Male
- Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis
- Nuclear Receptor Subfamily 1, Group F, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Severity of Illness Index
- Th17 Cells/immunology
- Transforming Growth Factor beta/blood
Collapse
Affiliation(s)
- Z-X Qi
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | | | | | | | | | | |
Collapse
|
85
|
Lee Y, Awasthi A, Yosef N, Quintana FJ, Xiao S, Peters A, Wu C, Kleinewietfeld M, Kunder S, Hafler DA, Sobel RA, Regev A, Kuchroo VK. Induction and molecular signature of pathogenic TH17 cells. Nat Immunol 2012; 13:991-9. [PMID: 22961052 PMCID: PMC3459594 DOI: 10.1038/ni.2416] [Citation(s) in RCA: 914] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 07/12/2012] [Indexed: 12/12/2022]
Abstract
Interleukin 17 (IL-17)-producing helper T cells (T(H)17 cells) are often present at the sites of tissue inflammation in autoimmune diseases, which has led to the conclusion that T(H)17 cells are main drivers of autoimmune tissue injury. However, not all T(H)17 cells are pathogenic; in fact, T(H)17 cells generated with transforming growth factor-β1 (TGF-β1) and IL-6 produce IL-17 but do not readily induce autoimmune disease without further exposure to IL-23. Here we found that the production of TGF-β3 by developing T(H)17 cells was dependent on IL-23, which together with IL-6 induced very pathogenic T(H)17 cells. Moreover, TGF-β3-induced T(H)17 cells were functionally and molecularly distinct from TGF-β1-induced T(H)17 cells and had a molecular signature that defined pathogenic effector T(H)17 cells in autoimmune disease.
Collapse
Affiliation(s)
- Youjin Lee
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Solt LA, Kamenecka TM, Burris TP. LXR-mediated inhibition of CD4+ T helper cells. PLoS One 2012; 7:e46615. [PMID: 23029557 PMCID: PMC3460920 DOI: 10.1371/journal.pone.0046615] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/05/2012] [Indexed: 11/21/2022] Open
Abstract
TH17 cells, which require the expression of both retinoic acid receptor-related orphan receptors α and γt (RORαand RORγt) for full differentiation and function, have been implicated as major effectors in the pathogenesis of inflammatory and autoimmune diseases. We recently demonstrated that the Liver X Receptor (LXR) agonist, T0901317 (T09), also displays high-affinity RORα and RORγ inverse activity, potentially explaining its effectiveness in various TH17-mediated autoimmune disease models. However, recent studies suggest that in conjunction with the RORs, LXR mediates a negative regulatory effect on TH17 cell differentiation. Since T09 acts on both LXRs and RORs, it presents as a valuable tool to understand how compounds with mixed pharmacology affect potential pathological cell types. Therefore, using T09, we investigated the mechanism by which the LXRs and RORs affect TH17 cell differentiation and function. Here we demonstrate that T09 activity at RORα and γ, not LXR, is facilitating the inhibition of TH17 cell differentiation and function. We also demonstrate that LXR activity inhibits the differentiation and function of TH1, TH2 and iTreg cells. Finally, T09 inhibited T cell proliferation and induced cell death. These data help explain much of the efficacy of T09 in inflammatory models and suggest that the generation of synthetic ligands with graded, combined LXR and ROR activity may hold utility in the treatment of inflammatory and autoimmune diseases where targeting both TH17 and TH1 cells is required.
Collapse
Affiliation(s)
- Laura A. Solt
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Theodore M. Kamenecka
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Thomas P. Burris
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida, United States of America
- * E-mail:
| |
Collapse
|
87
|
Niimi N, Kohyama K, Matsumoto Y. Therapeutic gene silencing with siRNA for IL-23 but not for IL-17 suppresses the development of experimental autoimmune encephalomyelitis in rats. J Neuroimmunol 2012; 254:39-45. [PMID: 22989513 DOI: 10.1016/j.jneuroim.2012.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 08/02/2012] [Accepted: 08/29/2012] [Indexed: 12/17/2022]
Abstract
Gene silencing with siRNAs is important as a therapeutic tool in autoimmune diseases. In this study, we administered siRNAs specific for cytokines that may be involved in pathogenesis of experimental autoimmune encephalomyelitis (EAE). siRNA specific for IL-23p19 (siRNA-IL-23) suppressed EAE almost completely, whereas siRNA-IL-17A did not modulate the clinical course. Flow cytometric analysis revealed that siRNA-IL-23 significantly reduced the proportion of both IFN-γ(+)IL-17(-) Th1 and IFN-γ(-)IL-17(+) Th17 cells in the spinal cord. Consistent with this finding, siRNA-IL-23 treatment downregulated IL-12, IL-17 and IL-23 mRNAs. These findings indicate that IL-23, but not IL-17, play an important role in the development of EAE.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Drug Delivery Systems
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Flow Cytometry
- Freund's Adjuvant/adverse effects
- Interleukin-12/genetics
- Interleukin-12/metabolism
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Myelin Basic Protein/immunology
- Myelin Basic Protein/metabolism
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
- Rats
- Rats, Inbred Lew
- Severity of Illness Index
- Spinal Cord/drug effects
- Spinal Cord/metabolism
- Statistics, Nonparametric
- Th1 Cells/drug effects
- Th1 Cells/metabolism
- Th17 Cells/drug effects
- Th17 Cells/metabolism
- Time Factors
Collapse
Affiliation(s)
- Naoko Niimi
- Department of Molecular Neuropathology, Tokyo Metropolitan Institute for Neuroscience, Tokyo, Japan
| | | | | |
Collapse
|
88
|
Hermann-Kleiter N, Meisel M, Fresser F, Thuille N, Müller M, Roth L, Katopodis A, Baier G. Nuclear orphan receptor NR2F6 directly antagonizes NFAT and RORγt binding to the Il17a promoter. J Autoimmun 2012; 39:428-40. [PMID: 22921335 PMCID: PMC3516707 DOI: 10.1016/j.jaut.2012.07.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 07/19/2012] [Accepted: 07/19/2012] [Indexed: 12/20/2022]
Abstract
Interleukin-17A (IL-17A) is the signature cytokine produced by Th17 CD4(+) T cells and has been tightly linked to autoimmune pathogenesis. In particular, the transcription factors NFAT and RORγt are known to activate Il17a transcription, although the detailed mechanism of action remains incompletely understood. Here, we show that the nuclear orphan receptor NR2F6 can attenuate the capacity of NFAT to bind to critical regions of the Il17a gene promoter. In addition, because NR2F6 binds to defined hormone response elements (HREs) within the Il17a locus, it interferes with the ability of RORγt to access the DNA. Consistently, NFAT and RORγt binding within the Il17a locus were enhanced in Nr2f6-deficient CD4(+) Th17 cells but decreased in Nr2f6-overexpressing transgenic CD4(+) Th17 cells. Taken together, our findings uncover an example of antagonistic regulation of Il17a transcription through the direct reciprocal actions of NR2F6 versus NFAT and RORγt.
Collapse
Affiliation(s)
- Natascha Hermann-Kleiter
- Department for Pharmacology and Genetics, Medical University Innsbruck, Peter Mayr Str. 1a, A-6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
89
|
The immunomodulatory and neuroprotective effects of mesenchymal stem cells (MSCs) in experimental autoimmune encephalomyelitis (EAE): a model of multiple sclerosis (MS). Int J Mol Sci 2012; 13:9298-9331. [PMID: 22942767 PMCID: PMC3430298 DOI: 10.3390/ijms13079298] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/11/2012] [Accepted: 07/11/2012] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that differentiate into the mesenchymal lineages of adipocytes, osteocytes and chondrocytes. MSCs can also transdifferentiate and thereby cross lineage barriers, differentiating for example into neurons under certain experimental conditions. MSCs have anti-proliferative, anti-inflammatory and anti-apoptotic effects on neurons. Therefore, MSCs were tested in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), for their effectiveness in modulating the pathogenic process in EAE to develop effective therapies for MS. The data in the literature have shown that MSCs can inhibit the functions of autoreactive T cells in EAE and that this immunomodulation can be neuroprotective. In addition, MSCs can rescue neural cells via a mechanism that is mediated by soluble factors, which provide a suitable environment for neuron regeneration, remyelination and cerebral blood flow improvement. In this review, we discuss the effectiveness of MSCs in modulating the immunopathogenic process and in providing neuroprotection in EAE.
Collapse
|
90
|
Dendritic cell are able to differentially recognize Sporothrix schenckii antigens and promote Th1/Th17 response in vitro. Immunobiology 2012; 217:788-94. [PMID: 22656886 DOI: 10.1016/j.imbio.2012.04.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 04/26/2012] [Accepted: 04/27/2012] [Indexed: 12/24/2022]
Abstract
Sporotrichosis is a disease caused by the dimorphic fungus Sporothrix schenckii. The main clinical manifestations occur in the skin, however the number of systemic and visceral cases has increased, especially in immunocompromised patients. Dendritic cells (DCs) are highly capable to recognize the fungus associated data and translate it into differential T cells responses both in vivo and in vitro. Although, the mechanisms involved in the interaction between DCs and S. schenckii are not fully elucidated. The present study investigated the phenotypic and functional changes in bone marrow dendritic cells (BMDCs) stimulated in vitro with the yeast form of S. schenckii or exoantigen (ExoAg) and its ability to trigger a cellular immune response in vitro. Our results demonstrated that the live yeast of S. schenckii and its exoantigen, at a higher dose, were able to activate BMDCs and made them capable of triggering T cell responses in vitro. Whereas the yeast group promoted more pronounced IFN-γ production rather than IL-17, the Exo100 group generated similar production of both cytokines. The exoantigen stimulus suggests a capability to deviate the immune response from an effector Th1 to an inflammatory Th17 response. Interestingly, only the Exo100 group promoted the production of IL-6 and a significant increase of TGF-β, in addition to IL-23 production. Interestingly, only Exo100 group was capable to promote the production of IL-6 and a significant increase on TGF-β, in addition with IL-23 detection. Our results demonstrated the plasticity of DCs in translating the data associated with the fungus S. schenckii and ExoAg into differential T cell responses in vitro. The possibility of using ex vivo-generated DCs as vaccinal and therapeutic tools for sporotrichosis is a challenge for the future.
Collapse
|
91
|
Stowe AM, Wacker BK, Cravens PD, Perfater JL, Li MK, Hu R, Freie AB, Stüve O, Gidday JM. CCL2 upregulation triggers hypoxic preconditioning-induced protection from stroke. J Neuroinflammation 2012; 9:33. [PMID: 22340958 PMCID: PMC3298779 DOI: 10.1186/1742-2094-9-33] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 02/16/2012] [Indexed: 02/04/2023] Open
Abstract
Background A brief exposure to systemic hypoxia (i.e., hypoxic preconditioning; HPC) prior to transient middle cerebral artery occlusion (tMCAo) reduces infarct volume, blood-brain barrier disruption, and leukocyte migration. CCL2 (MCP-1), typically regarded as a leukocyte-derived pro-inflammatory chemokine, can also be directly upregulated by hypoxia-induced transcription. We hypothesized that such a hypoxia-induced upregulation of CCL2 is required for HPC-induced ischemic tolerance. Methods Adult male SW/ND4, CCL2-null, and wild-type mice were used in these studies. Cortical CCL2/CCR2 message, protein, and cell-type specific immunoreactivity were determined following HPC (4 h, 8% O2) or room air control (21% O2) from 6 h through 2 weeks following HPC. Circulating leukocyte subsets were determined by multi-parameter flow cytometry in naïve mice and 12 h after HPC. CCL2-null and wild-type mice were exposed to HPC 2 days prior to tMCAo, with immunoneutralization of CCL2 during HPC achieved by a monoclonal CCL2 antibody. Results Cortical CCL2 mRNA and protein expression peaked at 12 h after HPC (both p < 0.01), predominantly in cortical neurons, and returned to baseline by 2 days. A delayed cerebral endothelial CCL2 message expression (p < 0.05) occurred 2 days after HPC. The levels of circulating monocytes (p < 0.0001), T lymphocytes (p < 0.0001), and granulocytes were decreased 12 h after HPC, and those of B lymphocytes were increased (p < 0.0001), but the magnitude of these respective changes did not differ between wild-type and CCL2-null mice. HPC did decrease the number of circulating CCR2+ monocytes (p < 0.0001) in a CCL2-dependent manner, but immunohistochemical analyses at this 12 h timepoint indicated that this leukocyte subpopulation did not move into the CNS. While HPC reduced infarct volumes by 27% (p < 0.01) in wild-type mice, CCL2-null mice subjected to tMCAo were not protected by HPC. Moreover, administration of a CCL2 immunoneutralizing antibody prior to HPC completely blocked (p < 0.0001 vs. HPC-treated mice) the development of ischemic tolerance. Conclusions The early expression of CCL2 in neurons, the delayed expression of CCL2 in cerebral endothelial cells, and CCL2-mediated actions on circulating CCR2+ monocytes, appear to be required to establish ischemic tolerance to focal stroke in response to HPC, and thus represent a novel role for this chemokine in endogenous neurovascular protection.
Collapse
Affiliation(s)
- Ann M Stowe
- Department of Neurological Surgery, Washington University School of Medicine, 660 S, Euclid Ave,, Box 8057, St, Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Immunopathological roles of cytokines, chemokines, signaling molecules, and pattern-recognition receptors in systemic lupus erythematosus. Clin Dev Immunol 2012; 2012:715190. [PMID: 22312407 PMCID: PMC3270521 DOI: 10.1155/2012/715190] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 10/11/2011] [Indexed: 11/26/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with unknown etiology affecting more than one million individuals each year. It is characterized by B- and T-cell hyperactivity and by defects in the clearance of apoptotic cells and immune complexes. Understanding the complex process involved and the interaction between various cytokines, chemokines, signaling molecules, and pattern-recognition receptors (PRRs) in the immune pathways will provide valuable information on the development of novel therapeutic targets for treating SLE. In this paper, we review the immunopathological roles of novel cytokines, chemokines, signaling molecules, PRRs, and their interactions in immunoregulatory networks and suggest how their disturbances may implicate pathological conditions in SLE.
Collapse
|
93
|
Lynch MA, Mills KHG. Immunology meets neuroscience--opportunities for immune intervention in neurodegenerative diseases. Brain Behav Immun 2012; 26:1-10. [PMID: 21664452 DOI: 10.1016/j.bbi.2011.05.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 05/05/2011] [Accepted: 05/24/2011] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammatory changes are characteristic of many, if not all, neurodegenerative diseases but the extent to which the immune system is involved in the pathogenesis of these diseases is unclear. The findings of several studies during the past decade has established that there is a well-developed communication between the central nervous system (CNS) and the peripheral immune system, but also has revealed that the immune system in the CNS is much more sophisticated that previously acknowledged. In this mini-review, we discuss two major neurodegenerative disorders, Alzheimer's disease (AD) and multiple sclerosis (MS), and consider whether the therapies most likely to succeed are those that are identified by studying the marriage of neuroscience and immunology.
Collapse
Affiliation(s)
- Marina A Lynch
- Trinity Institute for Neuroscience, Trinity College, Dublin, Ireland.
| | | |
Collapse
|
94
|
Reppert S, Boross I, Koslowski M, Türeci Ö, Koch S, Lehr HA, Finotto S. A role for T-bet-mediated tumour immune surveillance in anti-IL-17A treatment of lung cancer. Nat Commun 2011; 2:600. [PMID: 22186896 DOI: 10.1038/ncomms1609] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 11/23/2011] [Indexed: 12/29/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. The cytokine interleukin-17A supports tumour vascularization and growth, however, its role in lung cancer is unknown. Here we show, in the lungs of patients with lung adenocarcinoma, an increase in interleukin-17A that is inversely correlated with the expression of T-bet and correlated with the T regulatory cell transcription factor Foxp3. Local targeting of interleukin-17A in experimental lung adenocarcinoma results in a reduction in tumour load, local expansion of interferon-γ-producing CD4(+) T cells and a reduction in lung CD4(+)CD25(+)Foxp3(+) regulatory T cells. T-bet((-/-)) mice have a significantly higher tumour load compared with wild-type mice. This is associated with the local upregulation of interleukin-23 and induction of interleukin-17A/interleukin-17R-expressing T cells infiltrating the tumour. Local anti-interleukin-17A antibody treatment partially improves the survival of T-bet((-/-)) mice. These results suggest that local anti-interleukin-17A antibody therapy could be considered for the treatment of lung tumours.
Collapse
Affiliation(s)
- S Reppert
- Department of Anesthesia, Laboratories of Cellular and Molecular Lung Immunology, Institute of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, Erlangen 91052, Germany
| | | | | | | | | | | | | |
Collapse
|
95
|
Alzabin S, Williams RO. Effector T cells in rheumatoid arthritis: lessons from animal models. FEBS Lett 2011; 585:3649-59. [PMID: 21515267 DOI: 10.1016/j.febslet.2011.04.034] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 04/13/2011] [Accepted: 04/14/2011] [Indexed: 01/19/2023]
Abstract
The development of an immune response to self antigens drives naive T cells to differentiate into subsets of CD8(+) and CD4(+) effector cells including T(H)1, T(H)2, cells and the more recently described T(H)17, and regulatory T cells (T(reg)). Rheumatoid arthritis is an autoimmune disease that engages an uncontrolled influx of inflammatory cells to the joints, eventually leading to joint damage. The role that effector T cells play in the local or systemic maintenance of, or protection against, inflammation and subsequent joint damage is now becoming better understoodthrough the use of animal models. In this review, we will explore the different animal models of RA, and their contribution to elucidating the role that effector T cells play in the regulation, induction, and maintenance of inflammatory joint disease. This understanding will aid in the design of more effective therapeutic strategies for rheumatoid arthritis and other autoimmune disorders.
Collapse
Affiliation(s)
- Saba Alzabin
- Imperial College London, Kennedy Institute of Rheumatology, London, United Kingdom.
| | | |
Collapse
|
96
|
Summers SA, Phoon RKS, Odobasic D, Dewage L, Kitching AR, Holdsworth SR. Signal transducer and activation of transcription 6 (STAT6) regulates T helper type 1 (Th1) and Th17 nephritogenic immunity in experimental crescentic glomerulonephritis. Clin Exp Immunol 2011; 166:227-34. [PMID: 21985369 DOI: 10.1111/j.1365-2249.2011.04437.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Experimental crescentic glomerulonephritis is driven by systemic cellular immune responses. A pathogenic role for T helper type 1 (Th1) and Th17 cells is well established. T-bet, a key transcription factor required for Th1 lineage commitment, and retinoic acid-related orphan receptor-γt (Rorγt), a key Th17 transcription factor, are required for full expression of disease. Similarly, several Th1- and Th17-associated cytokines have been implicated in disease augmentation. The role of Th2 cells in the disease is less clear, although Th2-associated cytokines, interleukin (IL)-4 and IL-10, are protective. We sought to determine the role of signal transducer and activation of transcription 6 (STAT6), a key regulator of Th2 responses, in experimental crescentic glomerulonephritis. Compared to wild-type mice, histological and functional renal injury was enhanced significantly in STAT6(-/-) mice 21 days after administration of sheep anti-mouse glomerular basement membrane globulin. Consistent with the enhanced renal injury, both Th1 and Th17 nephritogenic immune responses were increased in STAT6(-/-) mice. Conversely, production of IL-5, a key Th2-associated cytokine, was decreased significantly in STAT6(-/-) mice. Early in the disease process systemic mRNA expression of T-bet and Rorγ was increased in STAT6(-/-) mice. We conclude that STAT6 is required for attenuation of Th1 and Th17 nephritogenic immune responses and protection from crescentic glomerulonephritis.
Collapse
Affiliation(s)
- S A Summers
- Centre for Inflammatory Diseases, Monash University Department of Medicine, VIC, Australia
| | | | | | | | | | | |
Collapse
|
97
|
Hu L, Gocke AR, Knapp E, Rosenzweig JM, Grishkan IV, Baxi EG, Zhang H, Margolick JB, Whartenby KA, Calabresi PA. Functional blockade of the voltage-gated potassium channel Kv1.3 mediates reversion of T effector to central memory lymphocytes through SMAD3/p21cip1 signaling. J Biol Chem 2011; 287:1261-8. [PMID: 22110135 DOI: 10.1074/jbc.m111.296798] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The maintenance of T cell memory is critical for the development of rapid recall responses to pathogens, but may also have the undesired side effect of clonal expansion of T effector memory (T(EM)) cells in chronic autoimmune diseases. The mechanisms by which lineage differentiation of T cells is controlled have been investigated, but are not completely understood. Our previous work demonstrated a role of the voltage-gated potassium channel Kv1.3 in effector T cell function in autoimmune disease. In the present study, we have identified a mechanism by which Kv1.3 regulates the conversion of T central memory cells (T(CM)) into T(EM). Using a lentiviral-dominant negative approach, we show that loss of function of Kv1.3 mediates reversion of T(EM) into T(CM), via a delay in cell cycle progression at the G2/M stage. The inhibition of Kv1.3 signaling caused an up-regulation of SMAD3 phosphorylation and induction of nuclear p21(cip1) with resulting suppression of Cdk1 and cyclin B1. These data highlight a novel role for Kv1.3 in T cell differentiation and memory responses, and provide further support for the therapeutic potential of Kv1.3 specific channel blockers in T(EM)-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Lina Hu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Kim SJ, Lee H, Joung HY, Lee G, Lee HJ, Shin MK, Kim SH, Shim I, Bae H. T-bet deficient mice exhibit resistance to stress-induced development of depression-like behaviors. J Neuroimmunol 2011; 240-241:45-51. [PMID: 22018704 DOI: 10.1016/j.jneuroim.2011.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/25/2011] [Accepted: 09/17/2011] [Indexed: 11/25/2022]
Abstract
T-bet, a Th1-specific T-box transcription factor, regulates Th1 development by inducing endogenous Th1 cytokines and IFN-γ. This study was conducted to determine if T-bet knockout mice exhibit resistance to stress-induced development of depression-like behaviors. The T-bet knockout mice significantly reduced depressive-like behaviors provoked by repeated restraint stress in an elevated plus-maze test (EPM), tail suspension test (TST), and forced swim test (FST). Moreover, stress-induced elevations of the pro-inflammatory cytokines were attenuated in T-bet deficient group. These results suggest that T-bet directly mediated stress-induced depression. Therefore, understanding T-bet function during stress represents an additional treatment strategy for depression.
Collapse
Affiliation(s)
- Soo-Jeong Kim
- Department of Physiology, College of Oriental Medicine, Kyung-Hee University, #1 Hoeki-Dong, Dongdaemoon-gu, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Bai F, Villagra AV, Zou J, Painter JS, Connolly K, Blaskovich MA, Sokol L, Sebti S, Djeu JY, Loughran TP, Wei S, Sotomayor E, Epling-Burnette P. Tipifarnib-mediated suppression of T-bet-dependent signaling pathways. Cancer Immunol Immunother 2011; 61:523-33. [PMID: 21983879 DOI: 10.1007/s00262-011-1109-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 08/30/2011] [Indexed: 12/21/2022]
Abstract
Large granular lymphocyte (LGL) leukemia is a chronic lymphoproliferative disease in which T-bet [T-box transcription factor 21 gene (tbx21)] overexpression may play a pathogenic role. T-bet orchestrates the differentiation of mature peripheral T-cells into interferon-γ (IFN-γ) and tumor necrosis factor-α producing CD4+ T-helper type I (Th1) and CD8+ T cytotoxic cells that are necessary for antiviral responses. When IL-12 is produced by antigen-presenting cells, T-bet expression is induced, causing direct stimulation of ifng gene transcription while simultaneously acting as a transcriptional repressor of the IL4 gene, which then leads to Th1 dominance and T-helper type 2 differentiation blockade. Additionally, T-bet has been shown to regulate histone acetylation of the ifng promoter and enhancer to loosen condensed DNA, creating greater accessibility for other transcription factor binding, which further amplifies IFNγ production. We found that treatment with a farnesyltransferase inhibitor tipifarnib reduced Th1 cytokines in LGL leukemia patient T-cells and blocked T-bet protein expression and IL-12 responsiveness in T-cells from healthy donors. The mechanism of suppression was based on modulation of histone acetylation of the ifng gene, which culminated in Th1 blockade.
Collapse
Affiliation(s)
- Fanqi Bai
- Immunology Program, H. Lee Moffitt Cancer Center, SRB3, 12902 Magnolia Dr, Tampa, FL 33612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Yeh WI, McWilliams IL, Harrington LE. Autoreactive Tbet-positive CD4 T cells develop independent of classic Th1 cytokine signaling during experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2011; 187:4998-5006. [PMID: 21984703 DOI: 10.4049/jimmunol.1100031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Many autoimmune chronic inflammatory diseases, including multiple sclerosis, are associated with the presence of Th1 and Th17 effector CD4 T cells. Paradoxically, the principal Th1 cytokine IFN-γ does not appear necessary for disease, but the key Th1-associated transcription factor Tbet has been reported to be essential for disease development. This conundrum propelled us to investigate the regulation of this transcription factor during autoimmunity. Following the onset of experimental autoimmune encephalomyelitis, we observed a preferential upregulation of Tbet by CD4 T cells within the CNS, but not the secondary lymphoid organs. These Tbet-positive CD4 T cells were capable of producing the cytokine IFN-γ, and a proportion of these cells produced both IFN-γ and IL-17A. Interestingly, these Tbet-positive cells were present in high frequencies during disease in IFN-γ-deficient mice. Moreover, we found that CD4 T cells from IFN-γ-deficient/IFN-γ reporter mice upregulated the Thy1.1 reporter, indicating the presence of Th1 or Th1-like, Tbet-positive CD4 T cells even in the absence of the cardinal Th1 cytokine IFN-γ. These IFN-γ-deficient Th1-like cells not only maintain multiple Th1 properties but also exhibit increased expression of genes associated with the Th17 phenotype. We further examined the requirement of other Th1-associated molecules in controlling Tbet expression during experimental autoimmune encephalomyelitis and noted that STAT1, IL-12, and IFN-γ were dispensable for the induction of Tbet in vivo. Hence, this study highlights the complex regulation of Tbet and the potential unrecognized role for Th1 cells during autoimmunity.
Collapse
Affiliation(s)
- Wen-I Yeh
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|