1
|
Chia JE, Rousseau RP, Ozturk M, Poswayo SKL, Lucas R, Brombacher F, Parihar SP. The divergent outcome of IL-4Rα signalling on Foxp3 T regulatory cells in listeriosis and tuberculosis. Front Immunol 2024; 15:1427055. [PMID: 39483462 PMCID: PMC11524857 DOI: 10.3389/fimmu.2024.1427055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/18/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Forkhead box P3 (Foxp3) T regulatory cells are critical for maintaining self-tolerance, immune homeostasis, and regulating the immune system. Methods We investigated interleukin-4 receptor alpha (IL-4Rα) signalling on T regulatory cells (Tregs) during Listeria monocytogenes (L. monocytogenes) infection using a mouse model on a BALB/c background, specifically with IL-4Rα knockdown in Tregs (Foxp3creIL-4Rα-/lox). Results We showed an impairment of Treg responses, along with a decreased bacterial burden and diminished tissue pathology in the liver and spleen, which translated into better survival. Mechanistically, we observed an enhancement of the Th1 signature, characterised by increased expression of the T-bet transcription factor and a greater number of effector T cells producing IFN-γ, IL-2 following ex-vivo stimulation with heat-killed L. monocytogenes in Foxp3creIL-4Rα-/lox mice. Furthermore, CD8 T cells from Foxp3creIL-4Rα-/lox mice displayed increased cytotoxicity (Granzyme-B) with higher proliferation capacity (Ki-67), better survival (Bcl-2) with concomitant reduced apoptosis (activated caspase 3). In contrast to L. monocytogenes, Foxp3creIL-4Rα-/lox mice displayed similar bacterial burdens, lung pathology and survival during Mycobacterium tuberculosis (M. tuberculosis) infection, despite increased T cell numbers and IFN-γ, TNF and IL-17 production. Conclusion Our results demonstrated that the diminished IL-4Rα signalling on Foxp3+ T regulatory cells resulted in a loss of their functionality, leading to survival benefits in listeriosis but not in tuberculosis.
Collapse
Affiliation(s)
- Julius E. Chia
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Robert P. Rousseau
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sibongiseni K. L. Poswayo
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rodney Lucas
- Research Animal Facility (RAF), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Suraj P. Parihar
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Medical Microbiology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
2
|
Cetinkaya PG, Abras IF, Evcili I, Yildirim T, Ceylan Y, Kara Eroglu F, Kayaoglu B, İpekoglu EM, Akarsu A, Yıldırım M, Kahraman T, Cengiz AB, Sahiner UM, Sekerel BE, Ozsurekci Y, Soyer O, Gursel I. Plasma Extracellular Vesicles Derived from Pediatric COVID-19 Patients Modulate Monocyte and T Cell Immune Responses Based on Disease Severity. Immunol Invest 2024; 53:1141-1175. [PMID: 39115924 DOI: 10.1080/08820139.2024.2385992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
BACKGROUND The COVID-19 pandemic has caused significant morbidity and mortality globally. The role of plasma-derived extracellular vesicles (EVs) in pediatric COVID-19 patients remains unclear. METHODS We isolated EVs from healthy controls (n = 13) and pediatric COVID-19 patients (n = 104) with varying severity during acute and convalescent phases using serial ultracentrifugation. EV effects on healthy PBMCs, naïve CD4+ T cells, and monocytes were assessed through in vitro assays, flow cytometry, and ELISA. RESULTS Our findings indicate that COVID-19 severity correlates with diverse immune responses. Severe acute cases exhibited increased cytokine levels, decreased IFNγ levels, and lower CD4+ T cell and monocyte counts, suggesting immunosuppression. EVs from severe acute patients stimulated healthy cells to express higher PDL1, increased Th2 and Treg cells, reduced IFNγ secretion, and altered Th1/Th17 ratios. Patient-derived EVs significantly reduced proinflammatory cytokine production by monocytes (p < .001 for mild, p = .0025 for severe cases) and decreased CD4+ T cell (p = .043) and monocyte (p = .033) populations in stimulated healthy PBMCs. CONCLUSION This study reveals the complex relationship between immunological responses and EV-mediated effects, emphasizing the impact of COVID-19 severity. We highlight the potential role of plasma-derived EVs in early-stage immunosuppression in severe COVID-19 patients.
Collapse
Affiliation(s)
- Pınar Gur Cetinkaya
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Irem Fatma Abras
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Irem Evcili
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Tugçe Yildirim
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Yasemin Ceylan
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Fehime Kara Eroglu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Başak Kayaoglu
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Emre Mert İpekoglu
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Aysegul Akarsu
- Division of Pediatric Allergy and Asthma Unit, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Muzaffer Yıldırım
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Tamer Kahraman
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Ali Bülent Cengiz
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Umit Murat Sahiner
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Bulent Enis Sekerel
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Yasemin Ozsurekci
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ozge Soyer
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ihsan Gursel
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| |
Collapse
|
3
|
Baris S, Benamar M, Chen Q, Catak MC, Martínez-Blanco M, Wang M, Fong J, Massaad MJ, Sefer AP, Kara A, Babayeva R, Eltan SB, Yucelten AD, Bozkurtlar E, Cinel L, Karakoc-Aydiner E, Zheng Y, Wu H, Ozen A, Schmitz-Abe K, Chatila TA. Severe allergic dysregulation due to a gain of function mutation in the transcription factor STAT6. J Allergy Clin Immunol 2023; 152:182-194.e7. [PMID: 36758835 PMCID: PMC10330134 DOI: 10.1016/j.jaci.2023.01.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Inborn errors of immunity have been implicated in causing immune dysregulation, including allergic diseases. STAT6 is a key regulator of allergic responses. OBJECTIVES This study sought to characterize a novel gain-of-function STAT6 mutation identified in a child with severe allergic manifestations. METHODS Whole-exome and targeted gene sequencing, lymphocyte characterization, and molecular and functional analyses of mutated STAT6 were performed. RESULTS This study reports a child with a missense mutation in the DNA binding domain of STAT6 (c.1114G>A, p.E372K) who presented with severe atopic dermatitis, eosinophilia, and elevated IgE. Naive lymphocytes from the affected patient displayed increased TH2- and suppressed TH1- and TH17-cell responses. The mutation augmented both basal and cytokine-induced STAT6 phosphorylation without affecting dephosphorylation kinetics. Treatment with the Janus kinase 1/2 inhibitor ruxolitinib reversed STAT6 hyperresponsiveness to IL-4, normalized TH1 and TH17 cells, suppressed the eosinophilia, and improved the patient's atopic dermatitis. CONCLUSIONS This study identified a novel inborn error of immunity due to a STAT6 gain-of-function mutation that gave rise to severe allergic dysregulation. Janus kinase inhibitor therapy could represent an effective targeted treatment for this disorder.
Collapse
Affiliation(s)
- Safa Baris
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Qian Chen
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Mehmet Cihangir Catak
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Mónica Martínez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Muyun Wang
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Jason Fong
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Michel J Massaad
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Asena Pinar Sefer
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Altan Kara
- TUBITAK Marmara Research Center, Gene Engineering and Biotechnology Institute, Gebze, Turkey
| | - Royala Babayeva
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Sevgi Bilgic Eltan
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ayse Deniz Yucelten
- Department of Dermatology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Emine Bozkurtlar
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Leyla Cinel
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Yumei Zheng
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Mass
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Mass
| | - Ahmet Ozen
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Klaus Schmitz-Abe
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Mass
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
4
|
Okamura T, Hamaguchi M, Tominaga H, Kitagawa N, Hashimoto Y, Majima S, Senmaru T, Okada H, Ushigome E, Nakanishi N, Shichino S, Fukui M. Characterization of Peripheral Blood TCR in Patients with Type 1 Diabetes Mellitus by BD RhapsodyTM VDJ CDR3 Assay. Cells 2022; 11:cells11101623. [PMID: 35626661 PMCID: PMC9139223 DOI: 10.3390/cells11101623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 02/05/2023] Open
Abstract
The sequence of complementarity-determining region 3 of the T-cell receptor (TCR) varies widely due to the insertion of random bases during V-(D)-J recombination. In this study, we used single-cell VDJ sequencing using the latest technology, BD Rhapsody, to identify the TCR sequences of autoreactive T-cells characteristic of Japanese type 1 diabetes mellitus (T1DM) and to clarify the pairing of TCR of peripheral blood mononuclear cells from four patients with T1DM at the single-cell level. The expression levels of the TCR alpha variable (TRAV) 17 and TRAV21 in T1DM patients were higher than those in healthy Japanese subjects. Furthermore, the Shannon index of CD8+ T cells and FOXP3+ cells in T1DM patients was lower than that of healthy subjects. The gene expression of PRF1, GZMH, ITGB2, NKG7, CTSW, and CST7 was increased, while the expression of CD4, CD7, CD5, HLA-A, CD27, and IL-32 was decreased in the CD8+ T cells of T1DM patients. The upregulated gene expression was IL4R and TNFRSF4 in FOXP3+ cells of T1DM patients. Overall, these findings demonstrate that TCR diversity and gene expression of CD8+ and FOXP3+ cells are different in patients with T1DM and healthy subjects.
Collapse
Affiliation(s)
- Takuro Okamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Hiroyuki Tominaga
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Noriyuki Kitagawa
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Yoshitaka Hashimoto
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Saori Majima
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Takafumi Senmaru
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Hiroshi Okada
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan;
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.O.); (M.H.); (H.T.); (N.K.); (Y.H.); (S.M.); (T.S.); (H.O.); (E.U.); (N.N.)
- Correspondence: ; Tel.: +81-75-251-5505
| |
Collapse
|
5
|
Raimondo TM, Mooney DJ. Anti-inflammatory nanoparticles significantly improve muscle function in a murine model of advanced muscular dystrophy. SCIENCE ADVANCES 2021; 7:7/26/eabh3693. [PMID: 34162554 PMCID: PMC8221619 DOI: 10.1126/sciadv.abh3693] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/10/2021] [Indexed: 05/24/2023]
Abstract
Chronic inflammation contributes to the pathogenesis of all muscular dystrophies. Inflammatory T cells damage muscle, while regulatory T cells (Tregs) promote regeneration. We hypothesized that providing anti-inflammatory cytokines in dystrophic muscle would promote proregenerative immune phenotypes and improve function. Primary T cells from dystrophic (mdx) mice responded appropriately to inflammatory or suppressive cytokines. Subsequently, interleukin-4 (IL-4)- or IL-10-conjugated gold nanoparticles (PA4, PA10) were injected into chronically injured, aged, mdx muscle. PA4 and PA10 increased T cell recruitment, with PA4 doubling CD4+/CD8- T cells versus controls. Further, 50% of CD4+/CD8- T cells were immunosuppressive Tregs following PA4, versus 20% in controls. Concomitant with Treg recruitment, muscles exhibited increased fiber area and fourfold increases in contraction force and velocity versus controls. The ability of PA4 to shift immune responses, and improve dystrophic muscle function, suggests that immunomodulatory treatment may benefit many genetically diverse muscular dystrophies, all of which share inflammatory pathology.
Collapse
Affiliation(s)
- Theresa M Raimondo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
6
|
Amiset L, Fend L, Gatard-Scheikl T, Rittner K, Duong V, Rooke R, Muller S, Bonnefoy JY, Préville X, Haegel H. TLR2 ligation protects effector T cells from regulatory T-cell mediated suppression and repolarizes T helper responses following MVA-based cancer immunotherapy. Oncoimmunology 2021; 1:1271-1280. [PMID: 23243590 PMCID: PMC3518499 DOI: 10.4161/onci.21479] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cancer immunotherapy is hampered by the immunosuppression maintained by regulatory T cells (Tregs) in tumor-bearing hosts. Stimulation of the Toll-like receptor 2 (TLR2) by Pam3Cys is known to affect Treg-mediated suppression. We found that Pam3Cys increases the proliferation of both CD4+ effector T cells (Teffs) and Tregs co-cultured in vitro, but did not induce the proliferation of Tregs alone upon CD3 and CD28 stimulation. In a mouse model of RMA-MUC1 tumors, Pam3Cys was administered either alone or in combination with a modified vaccinia ankara (MVA)-based mucin 1 (MUC1) therapeutic vaccine. The combination of Pam3Cys with MVA-MUC1 (1) diminished splenic Treg/CD4+ T-cell ratios to those found in tumor-free mice, (2) stimulated a specific anti-MUC1 interferon γ (IFNγ) response and (3) had a significant therapeutic effect on tumor growth and mouse survival. When CD4+ Teffs and Tregs were isolated from Pam3Cys-treated mice, Teffs had become resistant to Treg-mediated suppression while upregulating the expression of BclL-xL. Tregs from Pam3Cys-treated mice were fully suppressive for Teffs from naïve mice. Bcl-xL was induced by Pam3Cys with different kinetics in Tregs and Teffs. Teff from Pam3Cys-treated mice produced increased levels of Th1 and Th2-type cytokines and an interleukin (IL)-6-dependent secretion of IL-17 was observed in Teff:Treg co-cultures, suggesting that TLR2 stimulation had skewed the immune response toward a Th17 profile. Our results show for the first time that in a tumor-bearing host, TLR2 stimulation with Pam3Cys affects both Tregs and Teffs, protects Teff from Treg-mediated suppression and has strong therapeutic effects when combined with an MVA-based antitumor vaccine.
Collapse
Affiliation(s)
- Laurent Amiset
- Département d'Immunopharmacologie; Transgene S.A.; Parc d'Innovation; Illkirch-Graffenstaden, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Chu KH, Lin SY, Chiang BL. STAT6 Pathway Is Critical for the Induction and Function of Regulatory T Cells Induced by Mucosal B Cells. Front Immunol 2021; 11:615868. [PMID: 33584704 PMCID: PMC7878545 DOI: 10.3389/fimmu.2020.615868] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
B cells could convert naïve T cells into regulatory T cells (so-called Treg-of-B cells) which have the ability to treat animal models of inflammatory diseases, including allergic asthma, collagen-induced arthritis and colitis; however, the mechanisms of Treg-of-B cell generation remain unclear. In this study, we investigated the role of STAT6 in the generation of Treg-of-B (P) cells, which Treg cells were generated by Peyer’s patch B cells (P stands for Peyer’s patch). CD4+CD25- T cells from wild type, STAT6 knockout and IL-4 knockout mice were cocultured with wild type Peyer’s patch B cells for Treg-of-B (P) cell generation. A murine asthmatic model was used to analyze the in vivo regulatory function of Treg-of-B (P) cells. The data demonstrated that STAT6 played a critical role in the generation of Treg-of-B (P) cells, which confirmed with STAT6-deficient T cells and the STAT6 inhibitor AS1517499. When STAT6 was lacking, Treg-of-B (P) cells exerted impaired suppressive ability with decreased LAG3 expression. Furthermore, Peyer’s patch B cells played an essential role in regulatory T cell generation. In the absence of Peyer’s patch B cells, T cells expressed decreased phosphorylated STAT6, which was followed by decreased LAG3 expression and impaired suppressive ability, suggesting that Peyer’s patch B cells provided the critical signal to activate STAT6 phosphorylation in T cells. Moreover, STAT6 deficient Treg-of-B (P) cells could not alleviate inflammation in an animal model of asthma in vivo. IL-4 was downstream of phosphorylated STAT6 and maintained Treg-of-B (P) cell survival with increased expression of Bcl-2 and BclXL. We reported a novel finding that the STAT6-LAG3 signaling axis is important for the induction and function of Treg-of-B (P) cells.
Collapse
Affiliation(s)
- Kuan-Hua Chu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-Yu Lin
- Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan
| | - Bor-Luen Chiang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan.,Allergy Center, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
Khumalo J, Kirstein F, Hadebe S, Brombacher F. IL-4Rα signaling in CD4+CD25+FoxP3+ T regulatory cells restrains airway inflammation via limiting local tissue IL-33. JCI Insight 2020; 5:136206. [PMID: 32931477 PMCID: PMC7605533 DOI: 10.1172/jci.insight.136206] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 09/09/2020] [Indexed: 01/05/2023] Open
Abstract
Impaired tolerance to innocuous particles during allergic asthma has been linked to increased plasticity of FoxP3+ regulatory T cells (Tregs) reprogramming into pathogenic effector cells, thus exacerbating airway disease. However, failure of tolerance mechanisms is driven by Th2 inflammatory signals. Therefore, the in vivo role of canonical IL-4 receptor α (IL-4Rα) signaling, an essential driver of Th2-type airway responses to allergens, on the regulatory function of FoxP3+ Tregs in allergic asthma was explored. Here, we used transgenic Foxp3cre IL-4Rα-/lox and littermate control mice to investigate the role of IL-4 and IL-13 signaling via Tregs in house dust mite-induced (HDM-induced) allergic airway disease. We sensitized mice intratracheally on day 0, challenged them on days 6-10, and analyzed airway hyperresponsiveness (AHR), airway inflammation, mucus production, and cellular profile on day 14. In the absence of IL-4Rα responsiveness on FoxP3+ Tregs, exacerbated AHR and airway inflammation were shown in HDM-sensitized mice. Interestingly, reduced induction of FoxP3+ Tregs accompanied increased IL-33 alarmin production and type 2 innate lymphoid cell activation in the lung, exacerbating airway hyperreactivity and lung eosinophilia. Taken together, our findings indicate that IL-4Rα-unresponsive FoxP3+ Tregs result in exaggerated innate Th2-type, IL-33-dependent airway inflammation and a break in tolerance during allergic asthma.
Collapse
Affiliation(s)
- Jermaine Khumalo
- Division of Immunology, and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Department of Pathology.,International Centre for Genetic Engineering and Biotechnology (ICGEB), and
| | - Frank Kirstein
- Division of Immunology, and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Department of Pathology
| | - Sabelo Hadebe
- Division of Immunology, and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Department of Pathology
| | - Frank Brombacher
- Division of Immunology, and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Department of Pathology.,International Centre for Genetic Engineering and Biotechnology (ICGEB), and.,Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
9
|
Joshi RN, Fernandes SJ, Shang MM, Kiani NA, Gomez-Cabrero D, Tegnér J, Schmidt A. Phosphatase inhibitor PPP1R11 modulates resistance of human T cells toward Treg-mediated suppression of cytokine expression. J Leukoc Biol 2019; 106:413-430. [PMID: 30882958 PMCID: PMC6850362 DOI: 10.1002/jlb.2a0618-228r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 01/15/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Regulatory T cells (Tregs) act as indispensable unit for maintaining peripheral immune tolerance mainly by regulating effector T cells. T cells resistant to suppression by Tregs pose therapeutic challenges in the treatment of autoimmune diseases, while augmenting susceptibility to suppression may be desirable for cancer therapy. To understand the cell intrinsic signals in T cells during suppression by Tregs, we have previously performed a global phosphoproteomic characterization. We revealed altered phosphorylation of protein phosphatase 1 regulatory subunit 11 (PPP1R11; Inhibitor‐3) in conventional T cells upon suppression by Tregs. Here, we show that silencing of PPP1R11 renders T cells resistant toward Treg‐mediated suppression of TCR‐induced cytokine expression. Furthermore, whole‐transcriptome sequencing revealed that PPP1R11 differentially regulates not only the expression of specific T cell stimulation‐induced cytokines but also other molecules and pathways in T cells. We further confirmed the target of PPP1R11, PP1, to augment TCR‐induced cytokine expression. In conclusion, we present PPP1R11 as a novel negative regulator of T cell activation‐induced cytokine expression. Targeting PPP1R11 may have therapeutic potential to regulate the T cell activation status including modulating the susceptibility of T cells toward Treg‐mediated suppression, specifically altering the stimulation‐induced T cell cytokine milieu.
Collapse
Affiliation(s)
- Rubin N Joshi
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Sunjay Jude Fernandes
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Ming-Mei Shang
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Narsis A Kiani
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - David Gomez-Cabrero
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Mucosal and Salivary Biology Division, King's College London Dental Institute, London, United Kingdom.,Translational Bioinformatics Unit, NavarraBiomed, Departamento de Salud-Universidad Pública de Navarra, Pamplona, Navarra, Spain
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Biological and Environmental Sciences and Engineering Division, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Angelika Schmidt
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Abdel Aziz N, Nono JK, Mpotje T, Brombacher F. The Foxp3+ regulatory T-cell population requires IL-4Rα signaling to control inflammation during helminth infections. PLoS Biol 2018; 16:e2005850. [PMID: 30379806 PMCID: PMC6231676 DOI: 10.1371/journal.pbio.2005850] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 11/12/2018] [Accepted: 10/17/2018] [Indexed: 12/27/2022] Open
Abstract
Forkhead box P3 (Foxp3+) regulatory T (Treg)-cell function is controlled by environmental cues of which cytokine-mediated signaling is a dominant component. In vivo, interleukin-4 (IL-4)-mediated signaling via IL-4 receptor alpha (IL-4Rα) mediates Treg cell transdifferentiation into ex-Foxp3 T helper 2 (Th2) or T helper 17 (Th17) cells. However, IL-4-mediated signaling also reinforces the Foxp3 Treg compartment in vitro. We generated Foxp3-specific IL-4Rα-deficient mice and demonstrated differential efficiency of IL-4Rα deletion in male (approximately 90%) and female (approximately 40%) animals, because of cyclic recombinase (Cre)-mediated X-linked foxp3 inactivation. Irrespective of the degree of IL-4Rα deletion within the Foxp3+ Treg cell population, mice showed exacerbation of immune effector responses with aggravated tissue pathology in tissue-dwelling helminth infections (Schistosoma mansoni or Nippostrongylus brasiliensis). Mechanistically, IL-4Rα deletion in males and females led to a reduced expression of Foxp3 and subsequently an impaired accumulation of Foxp3+ Treg cells to inflamed tissues. In-depth cellular typing by flow cytometry revealed that the impairment of IL-4Rα-mediated signaling during helminth infections decreased the ability of central Treg cells to convert into effector Treg (eTreg) cells and caused a significant down-regulation of markers associated with Treg cell migration (C-X-C motif chemokine receptor 3 [CXCR3]) and accumulation in inflamed tissues (GATA binding protein 3 [GATA3]) as well as survival (B cell lymphoma 2 [Bcl-2]). These findings unprecedentedly, to our knowledge, uncover a role for IL-4Rα signaling in the positive regulation of Foxp3+ Treg cell function in vivo. Complementing our past knowledge on a widely reported role for IL-4Rα signaling in the negative regulation and transdifferentiation of Foxp3+ Treg cells in vivo, our present findings reveal the host requirement for an intact, but not reduced or potentiated, IL-4Rα-mediated signaling on Foxp3+ Treg cells to optimally control inflammation during helminth infections. Host soluble mediators such as cytokines play a key role in the regulation of the immune response. Forkhead box P3 (Foxp3+) regulatory T (Treg) cells, which are involved in maintaining self-tolerance and immune system homeostasis, are influenced by cytokines, including interleukin-4 (IL-4). However, opposing reports have emerged on the effect of this cytokine on Treg cells. Some evidence suggests IL-4 inhibits Treg cells, whereas other studies indicate a supportive role for this cytokine in Treg cell biology and function. To unambiguously address this question, we generated mice with IL-4 receptor specifically removed from the Treg cell population. Our newly generated mice did not show any sign of spontaneous inflammation during homeostasis, but when challenged with an experimental infection by parasitic worms, deletion of the IL-4 receptor from the Treg cell population led to increased inflammation and aggravated tissue pathology. Several defects such as poor activation, reduced promigratory marker expression, and reduced survival were apparent in Treg cells with impaired IL-4 responsiveness. Our evidence presents a strong case for a supportive role of IL-4 via IL-4 receptor in the biology and optimal regulatory function of Treg cells during worm infections.
Collapse
Affiliation(s)
- Nada Abdel Aziz
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Biotechnology/Biomolecular Chemistry Program, Chemistry Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Justin Komguep Nono
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- The Medical Research Centre, Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
- * E-mail: (FB); (JKN)
| | - Thabo Mpotje
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- * E-mail: (FB); (JKN)
| |
Collapse
|
11
|
Yang WC, Hwang YS, Chen YY, Liu CL, Shen CN, Hong WH, Lo SM, Shen CR. Interleukin-4 Supports the Suppressive Immune Responses Elicited by Regulatory T Cells. Front Immunol 2017; 8:1508. [PMID: 29184551 PMCID: PMC5694475 DOI: 10.3389/fimmu.2017.01508] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/25/2017] [Indexed: 11/13/2022] Open
Abstract
Interleukin-4 (IL-4) has been considered as one of the tolerogenic cytokines in many autoimmune animal models and clinical settings. Despite its role in antagonizing pathogenic Th1 responses, little is known about whether IL-4 possesses functions that affect regulatory T cells (Tregs). Tregs are specialized cells responsible for the maintenance of peripheral tolerance through their immune modulatory capabilities. Interestingly, it has been suggested that IL-4 supplement at a high concentration protects responder T cells (Tresps) from Treg-mediated immune suppression. In addition, such supplement also impedes TGF-β-induced Treg differentiation in vitro. However, these phenomena may contradict the tolerogenic role of IL-4, and the effects of IL-4 on Tregs are therefore needed to be further elucidated. In this study, we utilized IL-4 knockout (KO) mice to validate the role of IL-4 on Treg-mediated immune suppression. Although IL-4 KO and control animals harbor similar frequencies of Tregs, Tregs from IL-4 KO mice weakly suppressed autologous Tresp activation. In addition, IL-4 deprivation impaired the ability of Tregs to modulate immune response, whereas IL-4 supplementation reinforced IL-4 KO Tregs in their function in suppressing Tresps. Finally, the presence of IL-4 was associated with increased cell survival and granzyme expression of Tregs. These results suggest the essential role of IL-4 in supporting Treg-mediated immune suppression, which may benefit the development of therapeutic strategies for autoimmune diseases.
Collapse
Affiliation(s)
- Wei-Cheng Yang
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Yih-Shiou Hwang
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Ophthalmology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Ying-Yu Chen
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chao-Lin Liu
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.,College of Engineering, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Hsin Hong
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Sheng-Min Lo
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Rui Shen
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Ophthalmology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| |
Collapse
|
12
|
Hurdayal R, Brombacher F. Interleukin-4 Receptor Alpha: From Innate to Adaptive Immunity in Murine Models of Cutaneous Leishmaniasis. Front Immunol 2017; 8:1354. [PMID: 29176972 PMCID: PMC5686050 DOI: 10.3389/fimmu.2017.01354] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022] Open
Abstract
The interleukin (IL)-4 receptor alpha (IL-4Rα), ubiquitously expressed on both innate and adaptive immune cells, controls the signaling of archetypal type 2 immune regulators; IL-4 and IL-13, which elicit their signaling action by the type 1 IL-4Rα/gamma common and/or the type 2 IL-4Rα/IL-13Rα complexes. Global gene-deficient mouse models targeting IL-4, IL-13, or the IL-4Rα chain, followed by the development of conditional mice and generation of important cell-type-specific IL-4Rα-deficient mouse models, were indeed critical to gaining in-depth understanding of detrimental T helper (Th) 2 mechanisms in type 1-controlled diseases. A primary example being cutaneous leishmaniasis, which is caused by the protozoan parasite Leishmania major, among others. The disease is characterized by localized self-healing cutaneous lesions and necrosis for which, currently, not a single vaccine has made it to a stage that can be considered effective. The spectrum of human leishmaniasis belongs to the top 10 infectious diseases according to the World Health Organization. As such, 350 million humans are at risk of infection and disease, with an incidence of 1.5–2 million new cases being reported annually. A major aim of our research is to identify correlates of host protection and evasion, which may aid in vaccine design and therapeutic interventions. In this review, we focus on the immune-regulatory role of the IL-4Rα chain from innate immune responses to the development of beneficial type 1 and detrimental type 2 adaptive immune responses during cutaneous Leishmania infection. We discuss the cell-specific requirements of the IL-4Rα chain on crucial innate immune cells during L. major infection, including, IL-4Rα-responsive skin keratinocytes, macrophages, and neutrophils, as well as dendritic cells (DCs). The latter, contributing to one of the paradigm shifts with respect to the role of IL-4 instructing DCs in vivo, to promote Th1 responses against L. major. Finally, we extend these innate responses and mechanisms to control of adaptive immunity and the effect of IL-4Rα-responsiveness on T and B lymphocytes orchestrating the development of CD4+ Th1/Th2 and B effector 1/B effector 2 B cells in response to L. major infection in the murine host.
Collapse
Affiliation(s)
- Ramona Hurdayal
- Faculty of Health Sciences, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Department of Molecular and Cell Biology Faculty of Science, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Faculty of Health Sciences, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| |
Collapse
|
13
|
Kälin S, Becker M, Ott VB, Serr I, Hosp F, Mollah MMH, Keipert S, Lamp D, Rohner-Jeanrenaud F, Flynn VK, Scherm MG, Nascimento LFR, Gerlach K, Popp V, Dietzen S, Bopp T, Krishnamurthy P, Kaplan MH, Serrano M, Woods SC, Tripal P, Palmisano R, Jastroch M, Blüher M, Wolfrum C, Weigmann B, Ziegler AG, Mann M, Tschöp MH, Daniel C. A Stat6/Pten Axis Links Regulatory T Cells with Adipose Tissue Function. Cell Metab 2017; 26:475-492.e7. [PMID: 28877454 PMCID: PMC5627977 DOI: 10.1016/j.cmet.2017.08.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 07/21/2017] [Accepted: 08/08/2017] [Indexed: 12/22/2022]
Abstract
Obesity and type 2 diabetes are associated with metabolic defects and adipose tissue inflammation. Foxp3+ regulatory T cells (Tregs) control tissue homeostasis by counteracting local inflammation. However, if and how T cells interlink environmental influences with adipocyte function remains unknown. Here, we report that enhancing sympathetic tone by cold exposure, beta3-adrenergic receptor (ADRB3) stimulation or a short-term high-calorie diet enhances Treg induction in vitro and in vivo. CD4+ T cell proteomes revealed higher expression of Foxp3 regulatory networks in response to cold or ADRB3 stimulation in vivo reflecting Treg induction. Specifically, Ragulator-interacting protein C17orf59, which limits mTORC1 activity, was upregulated in CD4+ T cells by either ADRB3 stimulation or cold exposure, suggesting contribution to Treg induction. By loss- and gain-of-function studies, including Treg depletion and transfers in vivo, we demonstrated that a T cell-specific Stat6/Pten axis links cold exposure or ADRB3 stimulation with Foxp3+ Treg induction and adipose tissue function. Our findings offer a new mechanistic model in which tissue-specific Tregs maintain adipose tissue function.
Collapse
Affiliation(s)
- Stefanie Kälin
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Maike Becker
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Verena B Ott
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Advanced Study, Technische Universität München, 85748 Garching, Germany
| | - Isabelle Serr
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Fabian Hosp
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Mohammad M H Mollah
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Susanne Keipert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Daniel Lamp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Francoise Rohner-Jeanrenaud
- Laboratory of Metabolism, Division of Endocrinology, Diabetology, Hypertension, and Nutrition, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Victoria K Flynn
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Martin G Scherm
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Lucas F R Nascimento
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Katharina Gerlach
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052 Erlangen, Germany
| | - Vanessa Popp
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052 Erlangen, Germany
| | - Sarah Dietzen
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Tobias Bopp
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Purna Krishnamurthy
- Department of Pediatrics and HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark H Kaplan
- Department of Pediatrics and HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Manuel Serrano
- Tumour Suppression Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Philipp Tripal
- OICE (Optical Imaging Centre Erlangen), University Erlangen, 91052 Erlangen, Germany
| | - Ralf Palmisano
- OICE (Optical Imaging Centre Erlangen), University Erlangen, 91052 Erlangen, Germany
| | - Martin Jastroch
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Matthias Blüher
- Department of Medicine, Research Group Molecular Endocrinology, University of Leipzig, 04103 Leipzig, Germany
| | - Christian Wolfrum
- Swiss Federal Institute of Technology, Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| | - Benno Weigmann
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052 Erlangen, Germany
| | - Anette-Gabriele Ziegler
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Klinikum rechts der Isar, Technische Universität München, 80333 Munich, Germany
| | - Matthias Mann
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany.
| | - Carolin Daniel
- Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany.
| |
Collapse
|
14
|
The transcription factor musculin promotes the unidirectional development of peripheral T reg cells by suppressing the T H2 transcriptional program. Nat Immunol 2017; 18:344-353. [PMID: 28114290 DOI: 10.1038/ni.3667] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022]
Abstract
Although master transcription factors (TFs) are key to the development of specific T cell subsets, whether additional transcriptional regulators are induced by the same stimuli that dominantly repress the development of other, non-specific T cell lineages has not been fully elucidated. Through the use of regulatory T cells (Treg cells) induced by transforming growth factor-β (TGF-β), we identified the TF musculin (MSC) as being critical for the development of induced Treg cells (iTreg cells) by repression of the T helper type 2 (TH2) transcriptional program. Loss of MSC reduced expression of the Treg cell master TF Foxp3 and induced TH2 differentiation even under iTreg-cell-differentiation conditions. MSC interrupted binding of the TF GATA-3 to the locus encoding TH2-cell-related cytokines and diminished intrachromosomal interactions within that locus. MSC-deficient (Msc-/-) iTreg cells were unable to suppress TH2 responses, and Msc-/- mice spontaneously developed gut and lung inflammation with age. MSC therefore enforced Foxp3 expression and promoted the unidirectional induction of iTreg cells by repressing the TH2 developmental program.
Collapse
|
15
|
Crucial Contributions by T Lymphocytes (Effector, Regulatory, and Checkpoint Inhibitor) and Cytokines (TH1, TH2, and TH17) to a Pathological Complete Response Induced by Neoadjuvant Chemotherapy in Women with Breast Cancer. J Immunol Res 2016; 2016:4757405. [PMID: 27777963 PMCID: PMC5061970 DOI: 10.1155/2016/4757405] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 08/19/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023] Open
Abstract
The tumour microenvironment consists of malignant cells, stroma, and immune cells. Prominent tumour-infiltrating lymphocytes (TILs) in breast cancer are associated with a good prognosis and are predictors of a pathological complete response (pCR) with neoadjuvant chemotherapy (NAC). The contribution of different T effector/regulatory cells and cytokines to tumour cell death with NAC requires further characterisation and was investigated in this study. Breast tumours from 33 women with large and locally advanced breast cancers undergoing NAC were immunohistochemically (intratumoural, stromal) assessed for T cell subsets and cytokine expression using labelled antibodies, employing established semiquantitative methods. Prominent levels of TILs and CD4+, CD8+, and CTLA-4+ (stromal) T cells and CD8+ : FOXP3+ ratios were associated with a significant pCR; no association was seen with FOXP3+, CTLA-4+ (intratumoural), and PD-1+ T cells. NAC significantly reduced CD4+, FOXP3+, CTLA-4+ (stromal) (concurrently blood FOXP3+, CTLA-4+ Tregs), and PD-1+ T cells; no reduction was seen with CD8+ and CTLA-4+ (intratumoural) T cells. High post-NAC tumour levels of FOXP3+ T cells, IL-10, and IL-17 were associated with a failed pCR. Our study has characterised further the contribution of T effector/regulatory cells and cytokines to tumour cell death with NAC.
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW The aims of this review are to discuss the impact of chronic high-dose allergen exposure on allergen-specific CD4(+) T-cell subset during allergen-specific immunotherapy (AIT) and discuss recent advances supporting novel mechanisms for desensitization and tolerance induction during AIT. (Figure is included in full-text article.) RECENT FINDINGS New technologies for direct molecular and cellular analysis have now provided an unprecedented opportunity to compare the functions and phenotypes of allergen-specific T cells at a single cell level, both in the context of disease and clinical intervention. Recent studies have demonstrated that AIT may restore tolerance by transiently inducing interleukin (IL)-10 producing T cells followed by selective deletion of allergen-specific TH2 cell subset. SUMMARY With antigen-specific TH2 cells at the core of the allergic process in atopic individuals, the duration and dose of antigen exposure can be the driving force behind current AIT protocol. Mechanisms modulating allergen-specific CD4(+) T-cell responses may include autocrine IL-10 production to limit excessive TH2 cell effector responses, T-cell exhaustion, and preferential proallergic TH2 cell deletion allowing concurrent downregulating T-cell responses to emerge. Administration of AIT in the context of immune modulating strategies able to induce counter-regulatory immune response may lead to improved AIT with durable clinical benefit.
Collapse
|
17
|
Harakal J, Rival C, Qiao H, Tung KS. Regulatory T Cells Control Th2-Dominant Murine Autoimmune Gastritis. THE JOURNAL OF IMMUNOLOGY 2016; 197:27-41. [PMID: 27259856 DOI: 10.4049/jimmunol.1502344] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/18/2016] [Indexed: 12/12/2022]
Abstract
Pernicious anemia and gastric carcinoma are serious sequelae of autoimmune gastritis (AIG). Our study indicates that in adult C57BL/6-DEREG mice expressing a transgenic diphtheria toxin receptor under the Foxp3 promoter, transient regulatory T cell (Treg) depletion results in long-lasting AIG associated with both H(+)K(+)ATPase and intrinsic factor autoantibody responses. Although functional Tregs emerge over time during AIG occurrence, the effector T cells rapidly become less susceptible to Treg-mediated suppression. Whereas previous studies have implicated dysregulated Th1 cell responses in AIG pathogenesis, eosinophils have been detected in gastric biopsy specimens from patients with AIG. Indeed, AIG in DEREG mice is associated with strong Th2 cell responses, including dominant IgG1 autoantibodies, elevated serum IgE, increased Th2 cytokine production, and eosinophil infiltration in the stomach-draining lymph nodes. In addition, the stomachs exhibit severe mucosal and muscular hypertrophy, parietal cell loss, mucinous epithelial cell metaplasia, and massive eosinophilic inflammation. Notably, the Th2 responses and gastritis severity are significantly ameliorated in IL-4- or eosinophil-deficient mice. Furthermore, expansion of both Th2-promoting IFN regulatory factor 4(+) programmed death ligand 2(+) dendritic cells and ILT3(+) rebounded Tregs was detected after transient Treg depletion. Collectively, these data suggest that Tregs maintain physiological tolerance to clinically relevant gastric autoantigens, and Th2 responses can be a pathogenic mechanism in AIG.
Collapse
Affiliation(s)
- Jessica Harakal
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and
| | - Claudia Rival
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Hui Qiao
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Kenneth S Tung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
18
|
Mercadante ER, Lorenz UM. Breaking Free of Control: How Conventional T Cells Overcome Regulatory T Cell Suppression. Front Immunol 2016; 7:193. [PMID: 27242798 PMCID: PMC4870238 DOI: 10.3389/fimmu.2016.00193] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/02/2016] [Indexed: 01/10/2023] Open
Abstract
Conventional T (Tcon) cells are crucial in shaping the immune response, whether it is protection against a pathogen, a cytotoxic attack on tumor cells, or an unwanted response to self-antigens in the context of autoimmunity. In each of these immune settings, regulatory T cells (Tregs) can potentially exert control over the Tcon cell response, resulting in either suppression or activation of the Tcon cells. Under physiological conditions, Tcon cells are able to transiently overcome Treg-imposed restraints to mount a protective response against an infectious threat, achieving clonal expansion, differentiation, and effector function. However, evidence has accumulated in recent years to suggest that Tcon cell resistance to Treg-mediated suppression centrally contributes to the pathogenesis of autoimmune disease. Tipping the balance too far in the other direction, cancerous tumors utilize Tregs to establish an overly suppressive microenvironment, preventing antitumor Tcon cell responses. Given the wide-ranging clinical importance of the Tcon/Treg interaction, this review aims to provide a better understanding of what determines whether a Tcon cell is susceptible to Treg-mediated suppression and how perturbations to this finely tuned balance play a role in pathological conditions. Here, we focus in detail on the complex array of factors that confer Tcon cells with resistance to Treg suppression, which we have divided into two categories: (1) extracellular factor-mediated signaling and (2) intracellular signaling molecules. Further, we explore the therapeutic implications of manipulating the phosphatidylinositol-3 kinase (PI3K)/Akt signaling pathway, which is proposed to be the convergence point of signaling pathways that mediate Tcon resistance to suppression. Finally, we address important unresolved questions on the timing and location of acquisition of resistance, and the stability of the “Treg-resistant” phenotype.
Collapse
Affiliation(s)
- Emily R Mercadante
- Department of Microbiology Immunology and Cancer Biology, Beirne Carter Center for Immunology Research, University of Virginia , Charlottesville, VA , USA
| | - Ulrike M Lorenz
- Department of Microbiology Immunology and Cancer Biology, Beirne Carter Center for Immunology Research, University of Virginia , Charlottesville, VA , USA
| |
Collapse
|
19
|
Lambracht-Washington D, Rosenberg RN. Co-stimulation with TNF receptor superfamily 4/25 antibodies enhances in-vivo expansion of CD4+CD25+Foxp3+ T cells (Tregs) in a mouse study for active DNA Aβ42 immunotherapy. J Neuroimmunol 2014; 278:90-9. [PMID: 25595257 DOI: 10.1016/j.jneuroim.2014.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 01/09/2023]
Abstract
The study was designed to test DNA Aβ42 immunization in mice as alternative approach for possible active immunotherapy in Alzheimer patients. As results, we found polarized Th2 immune responses, efficient Aβ42 antibody levels, and disappearance of antigen specific T cells. In-vivo TNFRSF4/25 antibody co-stimulation enhanced Aβ42 specific T cell responses with initial Th2 expansion and subsequent development of Aβ42 specific CD4+CD25+Foxp3+ cells. It showed that Th2 biased responses due to gene gun immunizations propagate the development of regulatory T cells. In conclusion, full-length DNA Aβ42 immunization into skin results in a regulatory response with minimal risk of inflammation and autoimmunity.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
20
|
Chapman TJ, Georas SN. Regulatory tone and mucosal immunity in asthma. Int Immunopharmacol 2014; 23:330-6. [PMID: 24975833 DOI: 10.1016/j.intimp.2014.05.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 12/21/2022]
Abstract
The lung is constantly exposed to a variety of inhaled foreign antigens, many of which are harmless to the body. Therefore, the mucosal immune system must not only have the capacity to distinguish self from non-self, but also harmless versus dangerous non-self. To address this, mucosal immune cells establish an anti-inflammatory steady state in the lung that must be overcome by inflammatory signals in order to mount an effector immune response. In the case of inhaled allergens, the false detection of dangerous non-self results in inappropriate immune activation and eventual allergic asthma. Both basic and clinical studies suggest that the balance between tolerogenic and inflammatory immune responses is a key feature in the outcome of health or disease. This review is focused on what we term 'regulatory tone': the immunosuppressive environment in the lung that must be overcome to induce inflammatory responses. We will summarize the current literature on this topic, with a particular focus on the role of regulatory T cells in preventing allergic disease of the lung. We propose that inter-individual differences in regulatory tone have the potential to not only establish the threshold for immune activation in the lung, but also shape the quality of resulting effector responses following tolerance breakdown.
Collapse
Affiliation(s)
- Timothy J Chapman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14610, United States
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14610, United States.
| |
Collapse
|
21
|
Chatterjee P, Chiasson VL, Bounds KR, Mitchell BM. Regulation of the Anti-Inflammatory Cytokines Interleukin-4 and Interleukin-10 during Pregnancy. Front Immunol 2014; 5:253. [PMID: 24904596 PMCID: PMC4034149 DOI: 10.3389/fimmu.2014.00253] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/14/2014] [Indexed: 01/05/2023] Open
Abstract
Inflammation mediated by both innate and adaptive immune cells is necessary for several important processes during pregnancy. Pro-inflammatory immune cell activation plays a critical role in embryo implantation, placentation, and parturition; however dysregulation of these cells can lead to detrimental pregnancy outcomes including spontaneous abortion, fetal growth restriction, maternal pathology including hypertensive disorders, or fetal and maternal death. The resolution of inflammation plays an important role throughout pregnancy and is largely mediated by immune cells that produce interleukin (IL)-4 and IL-10. The temporal and spatial aspects of reducing inflammation during pregnancy represent a complex process that if not functioning optimally can lead to persistent inflammation and pregnancy complications. In this review, we examine how immune cells that produce IL-4 and IL-10 are regulated throughout pregnancy as well as the effects that reduced IL-4 and IL-10 signaling has on fetal and maternal physiology.
Collapse
Affiliation(s)
- Piyali Chatterjee
- Department of Internal Medicine, Texas A&M Health Science Center , Temple, TX , USA ; Baylor Scott and White Health , Temple, TX , USA
| | - Valorie L Chiasson
- Department of Internal Medicine, Texas A&M Health Science Center , Temple, TX , USA
| | - Kelsey R Bounds
- Department of Internal Medicine, Texas A&M Health Science Center , Temple, TX , USA
| | - Brett M Mitchell
- Department of Internal Medicine, Texas A&M Health Science Center , Temple, TX , USA ; Baylor Scott and White Health , Temple, TX , USA
| |
Collapse
|
22
|
Zhang H, Kong H, Zeng X, Guo L, Sun X, He S. Subsets of regulatory T cells and their roles in allergy. J Transl Med 2014; 12:125. [PMID: 24886492 PMCID: PMC4023533 DOI: 10.1186/1479-5876-12-125] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/28/2014] [Indexed: 12/17/2022] Open
Abstract
In recent years, it is recognized that acquired immunity is controlled by regulatory T cell (Treg). Since fundamental pathophysiological changes of allergy are mainly caused by hyperresponsiveness of immune system to allergens that acquires after birth, Tregs likely play key roles in the pathogenesis of allergy, particularly during the sensitization phase. However, accumulated information indicate that there are several distinctive subtypes of Tregs in man, and each of them seems to play different role in controlling immune system, which complicates the involvement of Tregs in allergy. The aim of the present study is to attempt to classify subtypes of Tregs and summarize their roles in allergy. Tregs should include natural Tregs (nTreg) including inducible costimulator (ICOS)(+) Tregs, inducible/adaptive Tregs (iTreg), interleukin (IL)-10-producing type 1 Tregs (Tr1 cells), CD8(+) Tregs and IL-17-producing Tregs. These cells share some common features including expression of Foxp3 (except for Tr1 cells), and secretion of inhibitory cytokine IL-10 and/or TGF-β. Furthermore, it is noticeable that Tregs likely contribute to allergic disorders such as dermatitis and airway inflammation, and play a crucial role in the treatment of allergy through their actions on suppression of effector T cells and inhibition of activation of mast cells and basophils. Modulation of functions of Tregs may provide a novel strategy to prevent and treat allergic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Shaoheng He
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No, 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning 121001, People's Republic of China.
| |
Collapse
|
23
|
Li W, Holsinger RMD, Kruse CA, Flügel A, Graeber MB. The potential for genetically altered microglia to influence glioma treatment. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2014; 12:750-62. [PMID: 24047526 DOI: 10.2174/18715273113126660171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/06/2012] [Accepted: 12/06/2012] [Indexed: 01/06/2023]
Abstract
Diffuse and unstoppable infiltration of brain and spinal cord tissue by neoplastic glial cells is the single most important therapeutic problem posed by the common glioma group of tumors: astrocytoma, oligoastrocytoma, oligodendroglioma, their malignant variants and glioblastoma. These neoplasms account for more than two thirds of all malignant central nervous system tumors. However, most glioma research focuses on an examination of the tumor cells rather than on host-specific, tumor micro-environmental cells and factors. This can explain why existing diffuse glioma therapies fail and why these tumors have remained incurable. Thus, there is a great need for innovation. We describe a novel strategy for the development of a more effective treatment of diffuse glioma. Our approach centers on gaining control over the behavior of the microglia, the defense cells of the CNS, which are manipulated by malignant glioma and support its growth. Armoring microglia against the influences from glioma is one of our research goals. We further discuss how microglia precursors may be genetically enhanced to track down infiltrating glioma cells.
Collapse
Affiliation(s)
- W Li
- Brain and Mind Research Institute, The University of Sydney, Camperdown, NSW, Australia.
| | | | | | | | | |
Collapse
|
24
|
Steinke JW, Lawrence MG. T-cell biology in immunotherapy. Ann Allergy Asthma Immunol 2014; 112:195-9. [PMID: 24565594 DOI: 10.1016/j.anai.2013.12.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/18/2013] [Accepted: 12/23/2013] [Indexed: 11/29/2022]
Abstract
OBJECTIVE This review discusses the current state of immunotherapy and how the CD4 T-cell response is pivotal in altering the allergic response. DATA SOURCES PubMed literature review. STUDY SELECTIONS Articles pertaining to subcutaneous, sublingual, and oral immunotherapies, with specific emphasis on those describing the T-cell response. RESULTS Although many drugs are available that help ameliorate allergic symptoms, the only intervention that has proved to provide long-term benefit and modulation of disease is immunotherapy. Many routes of immunotherapy are being pursued, including subcutaneous, sublingual, and oral immunotherapies; however, subcutaneous immunotherapy has the historical record of leading to immune changes that alter the immune response at subsequent allergen exposure. These changes are mediated by the induction of peripherally derived T-regulatory cells and appear to occur only after high-dose therapy for 3 to 5 years. Newer methods of sublingual and oral immunotherapies are currently being investigated, but their efficacy is not yet on par with subcutaneous immunotherapy. CONCLUSION The primary cells ultimately responsible for successful immunomodulation are CD4 T cells, specifically peripherally derived T-regulatory cells.
Collapse
Affiliation(s)
- John W Steinke
- Asthma and Allergic Disease Center, Carter Immunology Center, University of Virginia Health Systems, Charlottesville, Virginia.
| | - Monica G Lawrence
- Asthma and Allergic Disease Center, Carter Immunology Center, University of Virginia Health Systems, Charlottesville, Virginia
| |
Collapse
|
25
|
Plain KM, Verma ND, Tran GT, Nomura M, Boyd R, Robinson CM, Hodgkinson SJ, Hall BM. Cytokines affecting CD4(+) T regulatory cells in transplant tolerance. Interleukin-4 does not maintain alloantigen specific CD4(+)CD25(+) Treg. Transpl Immunol 2013; 29:51-9. [PMID: 24139939 DOI: 10.1016/j.trim.2013.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/03/2013] [Accepted: 10/03/2013] [Indexed: 12/31/2022]
Abstract
IL-4 is thought to promote induction of transplantation tolerance and alloantigen-specific CD4(+)CD25(+) T regulatory cells (Treg). This study examined the effect of IL-4 on the induction and maintenance of the CD4(+) T regulatory cells (Treg) that mediate transplantation tolerance. Tolerance was induced in DA rats with PVG heterotopic cardiac allografts by a short course of cyclosporine. Naïve and tolerant lymphocytes, including the CD4(+) and CD4(+)CD25(+) T cell subsets, were assayed in mixed lymphocyte cultures with or without recombinant (r)IL-4 or other cytokines. The proliferation, cell surface and cytokine phenotype of these cells was examined, as was their capacity to adoptively transfer tolerance. rIL-4 enhanced the proliferation of naïve and tolerant lymphoid cells, including CD4(+) and CD4(+)CD25(+) T cells, but this was not alloantigen specific. Naïve or tolerant CD4(+) T cells cultured with rIL-4 and donor PVG antigen effected rapid graft rejection, even though before culture tolerant CD4(+) T cells transferred antigen-specific tolerance. These rIL-4 cultured CD4(+) T cells had a phenotype consistent with activated CD4(+)CD25(+)FoxP3(-) Th2 cells. While naïve natural CD4(+)CD25(+) T cells (nTreg) cultured with alloantigen and rIL-4 had enhanced proliferation and capacity to suppress rejection in vivo, the culture of tolerant CD4(+)CD25(+) T cells with alloantigen and rIL-4 could not sustain their proliferation against specific donor, nor their capacity to transfer tolerance to specific donor allograft. Thus, IL-4 promotes both regulatory and effector T cells early in the immune response, but once alloimmune tolerance is established, IL-4 promoted the activation of effector cells to mediate rejection and did not support alloantigen-specific Treg that could transfer specific tolerance.
Collapse
Affiliation(s)
- Karren M Plain
- Immune Tolerance Laboratory Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Hall BM, Tran GT, Verma ND, Plain KM, Robinson CM, Nomura M, Hodgkinson SJ. Do Natural T Regulatory Cells become Activated to Antigen Specific T Regulatory Cells in Transplantation and in Autoimmunity? Front Immunol 2013; 4:208. [PMID: 23935597 PMCID: PMC3731939 DOI: 10.3389/fimmu.2013.00208] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/08/2013] [Indexed: 12/20/2022] Open
Abstract
Antigen specific T regulatory cells (Treg) are often CD4+CD25+FoxP3+ T cells, with a phenotype similar to natural Treg (nTreg). It is assumed that nTreg cannot develop into an antigen specific Treg as repeated culture with IL-2 and a specific antigen does not increase the capacity or potency of nTreg to promote immune tolerance or suppress in vitro. This has led to an assumption that antigen specific Treg mainly develop from CD4+CD25−FoxP3− T cells, by activation with antigen and TGF-β in the absence of inflammatory cytokines such as IL-6 and IL-1β. Our studies on antigen specific CD4+CD25+ T cells from animals with tolerance to an allograft, identified that the antigen specific and Treg are dividing, and need continuous stimulation with specific antigen T cell derived cytokines. We identified that a variety of cytokines, especially IL-5 and IFN-γ but not IL-2 or IL-4 promoted survival of antigen specific CD4+CD25+FoxP3+ Treg. To examine if nTreg could be activated to antigen specific Treg, we activated nTreg in culture with either IL-2 or IL-4. Within 3 days, antigen specific Treg are activated and there is induction of new cytokine receptors on these cells. Specifically nTreg activated by IL-2 and antigen express the interferon-γ receptor (IFNGR) and IL-12p70 (IL-12Rβ2) receptor but not the IL-5 receptor (IL-5Rα). These cells were responsive to IFN-γ or IL-12p70. nTreg activated by IL-4 and alloantigen express IL-5Rα not IFNGR or IL-12p70Rβ2 and become responsive to IL-5. These early activated antigen specific Treg, were respectively named Ts1 and Ts2 cells, as they depend on Th1 or Th2 responses. Further culture of Ts1 cells with IL-12p70 induced Th1-like Treg, expressing IFN-γ, and T-bet as well as FoxP3. Our studies suggest that activation of nTreg with Th1 or Th2 responses induced separate lineages of antigen specific Treg, that are dependent on late Th1 and Th2 cytokines, not the early cytokines IL-2 and IL-4.
Collapse
Affiliation(s)
- Bruce M Hall
- Immune Tolerance Laboratory, Medicine, University of New South Wales , Sydney, NSW , Australia
| | | | | | | | | | | | | |
Collapse
|
27
|
Dorsey NJ, Chapoval SP, Smith EP, Skupsky J, Scott DW, Keegan AD. STAT6 controls the number of regulatory T cells in vivo, thereby regulating allergic lung inflammation. THE JOURNAL OF IMMUNOLOGY 2013; 191:1517-28. [PMID: 23825312 DOI: 10.4049/jimmunol.1300486] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
STAT6 plays a central role in IL-4-mediated allergic responses. Several studies indicate that regulatory T cells (Tregs) can be modulated by IL-4 in vitro. We previously showed that STAT6(-/-) mice are highly resistant to allergic lung inflammation even when wild-type Th2 effectors were provided and that they have increased numbers of Tregs. However, the role of STAT6 in modulating Tregs in vivo during allergic lung inflammation has not been thoroughly investigated. To examine Treg and STAT6 interaction during allergic inflammation, STAT6(-/-), STAT6xRAG2(-/-), and RAG2(-/-) mice were subjected to OVA sensitization and challenge following adoptive transfer of OVA-specific, wild-type Th2 effectors with or without prior Treg depletion/inactivation, using anti-CD25 (PC61). As expected, STAT6(-/-) mice were highly resistant to airway inflammation and remodeling. In contrast, allergic lung inflammation was partially restored in STAT6(-/-) mice treated with PC61 to levels observed in STAT6xRAG2(-/-) mice. In some cases, STAT6xRAG2(-/-) mice were also given natural Tregs along with Th2 effectors. Adoptive transfer of natural Tregs caused a substantial reduction in bronchoalveolar lavage eosinophil composition and suppressed airway remodeling and T cell migration into the lung in STAT6xRAG2(-/-) mice to levels comparable to those in STAT6(-/-) mice. These results demonstrate the STAT6-dependent suppression of Tregs in vivo to promote allergic airway inflammation.
Collapse
Affiliation(s)
- Nicolas J Dorsey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
28
|
Reyes NJ, Chen PW, Niederkorn JY. Allergic conjunctivitis renders CD4(+) T cells resistant to t regulatory cells and exacerbates corneal allograft rejection. Am J Transplant 2013; 13:1181-92. [PMID: 23489547 PMCID: PMC3640580 DOI: 10.1111/ajt.12198] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 12/24/2012] [Accepted: 01/10/2013] [Indexed: 01/25/2023]
Abstract
Allergic diseases rob corneal allografts of immune privilege and increase immune rejection. Corneal allograft rejection in BALB/c allergic hosts was analyzed using a short ragweed (SWR) pollen model of allergic conjunctivitis. Allergic conjunctivitis did not induce exaggerated T-cell responses to donor C57BL/6 (B6) alloantigens or stimulate cytotoxic T lymphocyte (CTL) responses. Allergic conjunctivitis did affect T regulatory cells (Tregs) that support graft survival. Exogenous IL-4, but not IL-5 or IL-13, prevented Treg suppression of CD4(+) effector T cells isolated from naïve mice. However, mice with allergic conjunctivitis developed Tregs that suppressed CD4(+) effector T-cell proliferation. In addition, IL-4 did not inhibit Treg suppression of IL-4Rα(-/-) CD4(+) T-cell responses, suggesting that IL-4 rendered effector T cells resistant to Tregs. SRW-sensitized IL-4Rα(-/-) mice displayed the same 50% graft survival as nonallergic WT mice, that was significantly less than the 100% rejection that occurred in allergic WT hosts, supporting the role of IL-4 in the abrogation of immune privilege. Moreover, exacerbation of corneal allograft rejection in allergic mice was reversed by administering anti-IL-4 antibody. Thus, allergy-induced exacerbation of corneal graft rejection is due to the production of IL-4, which renders effector T cells resistant to Treg suppression of alloimmune responses.
Collapse
Affiliation(s)
- N J Reyes
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | |
Collapse
|
29
|
Rosen MJ, Chaturvedi R, Washington MK, Kuhnhein LA, Moore PD, Coggeshall SS, McDonough EM, Weitkamp JH, Singh AB, Coburn LA, Williams CS, Yan F, Van Kaer L, Peebles RS, Wilson KT. STAT6 deficiency ameliorates severity of oxazolone colitis by decreasing expression of claudin-2 and Th2-inducing cytokines. THE JOURNAL OF IMMUNOLOGY 2013; 190:1849-58. [PMID: 23303670 DOI: 10.4049/jimmunol.1201373] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Patients suffering from ulcerative colitis (UC) exhibit chronic colonic inflammation caused by a dysregulated mucosal immune response and epithelial barrier disruption. Th2 cytokines, including IL-13, have been implicated in the pathogenesis of UC. IL-13 induces phosphorylation of STAT6, and we previously demonstrated increased epithelial p-STAT6 in children with UC. In this study, we investigated the role of STAT6 in oxazolone colitis, a murine model of UC, by inducing colitis in STAT6-deficient (STAT6(-/-)) and wild type (WT) mice. We observed increased epithelial cell, T cell, macrophage, and NKT cell STAT6 phosphorylation, as well as increased p-STAT6(+) IL-13-producing NKT cells, in colitic WT mice. Colitis was attenuated in STAT6(-/-) mice, with improvements in weight, colon length, and histopathology. There was decreased induction of the pore-forming tight junction protein claudin-2 in STAT6(-/-) mice. Similarly, short hairpin RNA STAT6 knockdown reduced claudin-2 induction and transepithelial resistance decrease in IL-13-treated human T84 cells. Tissue expression of IL-13, IFN-γ, IL-17, and IL-10 mRNA was similarly induced in WT and STAT6(-/-) colitic mice; however, we observed increased mRNA expression for the Th2-inducing cytokines IL-33 and thymic stromal lymphopoietin in WT mice with colitis, which was abrogated in STAT6(-/-) mice. Mesenteric lymph node cells from STAT6(-/-) mice with colitis exhibited reduced secretion of IL-4, IL-5, IL-13, and IFN-γ. IL-33 augmented mesenteric lymph node cell secretion of IL-5, IL-13, IL-6, and IFN-γ. These data implicate STAT6 in the pathogenesis of colitis in vivo with important roles in altering epithelial barrier function and regulating Th2-inducing cytokine production.
Collapse
Affiliation(s)
- Michael J Rosen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sawant DV, Sehra S, Nguyen ET, Jadhav R, Englert K, Shinnakasu R, Hangoc G, Broxmeyer HE, Nakayama T, Perumal NB, Kaplan MH, Dent AL. Bcl6 controls the Th2 inflammatory activity of regulatory T cells by repressing Gata3 function. THE JOURNAL OF IMMUNOLOGY 2012; 189:4759-69. [PMID: 23053511 DOI: 10.4049/jimmunol.1201794] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The transcriptional repressor Bcl6 is a critical arbiter of Th cell fate, promoting the follicular Th lineage while repressing other Th cell lineages. Bcl6-deficient (Bcl6(-/-)) mice develop a spontaneous and severe Th2-type inflammatory disease, thus warranting assessment of Bcl6 in regulatory T cell (Treg) function. Bcl6(-/-) Tregs were competent at suppressing T cell proliferation in vitro and Th1-type colitogenic T cell responses in vivo. In contrast, Bcl6(-/-) Tregs strongly exacerbated lung inflammation in a model of allergic airway disease and promoted higher Th2 responses, including systemic upregulation of microRNA-21. Further, Bcl6(-/-) Tregs were selectively impaired at controlling Th2 responses, but not Th1 and Th17 responses, in mixed chimeras of Bcl6(-/-) bone marrow with Foxp3(-/-) bone marrow. Bcl6(-/-) Tregs displayed increased levels of the Th2 transcription factor Gata3 and other Th2 and Treg genes. Bcl6 potently repressed Gata3 transcriptional transactivation, providing a mechanism for the increased expression of Th2 genes by Bcl6(-/-) Tregs. Gata3 has a critical role in regulating Foxp3 expression and functional fitness of Tregs; however, the signal that regulates Gata3 and restricts its transactivation of Th2 cytokines in Tregs has remained unexplored. Our results identify Bcl6 as an essential transcription factor regulating Gata3 activity in Tregs. Thus, Bcl6 represents a crucial regulatory layer in the Treg functional program that is required for specific suppression of Gata3 and Th2 effector responses by Tregs.
Collapse
Affiliation(s)
- Deepali V Sawant
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wambre E, James EA, Kwok WW. Characterization of CD4+ T cell subsets in allergy. Curr Opin Immunol 2012; 24:700-6. [PMID: 22889592 DOI: 10.1016/j.coi.2012.07.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 07/25/2012] [Indexed: 12/16/2022]
Abstract
Allergen specific T(H)2 cells are a key component of allergic disease, but their characterization has been hindered by technical limitations and lack of epitope data. Knowledge about the factors that drive the differentiation of naïve T cells into allergy-promoting T(H)2 cells and the influence of allergen specific immunotherapy on the phenotype and function of allergen-specific T cells have also been limited. Recent advances indicate that innate and adaptive immune factors drive the development of diverse subsets of allergen-specific T cells. While allergen-specific T cells are present even in non-allergic subjects, highly differentiated T(H)2 cells are present only in allergic subjects and their disappearance correlates with successful immunotherapy. Therefore, elimination of pathogenic T(H)2 cells is an essential step in tolerance induction.
Collapse
Affiliation(s)
- Erik Wambre
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101-2795, USA
| | | | | |
Collapse
|
32
|
Sule G, Suzuki M, Guse K, Cela R, Rodgers JR, Lee B. Cytokine-conditioned dendritic cells induce humoral tolerance to protein therapy in mice. Hum Gene Ther 2012; 23:769-80. [PMID: 22468961 DOI: 10.1089/hum.2011.225] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A major obstacle in the genetic therapy of inherited metabolic disease is host immune responses to the therapeutic protein. This is best exemplified by inhibitor formation in the protein therapy for hemophilia A. An approach to overcoming this is induction of immunological tolerance to the therapeutic protein. Tolerogenic dendritic cells (DCtols) have been reported to induce tolerance. In addition, cytokines such as interleukin (IL)-10 and transforming growth factor (TGF)-β(1) are known to induce tolerance. To model protein therapy, we used ovalbumin (OVA) as antigen in BALB/c mice and their transgenic derivative, DO11.10 mice. In this study we show that adoptive transfer of antigen-pulsed dendritic cells (DCs) treated with a combination of IL-10 and TGF-β(1) can suppress the antibody response in mice. Adoptive transfer of cytokine-conditioned DCs in preimmunized mice results in reduction of antibody response in the mice. Furthermore, the effect is antigen specific, as the recipient mice were able to mount a potent antibody response to the control antigen. Last, we show that TGF-β(1) and IL-10-conditioned DCs are able to inhibit anti-FVIII antibody responses in FVIII knockout (KO) mice. Analysis of the contribution of IL-10 and TGF-β(1) to the DCtol phenotype shows that IL-10 treatment of DCs is sufficient for inducing OVA-specific tolerance in BALB/c mice, but we observed a requirement for treatment with both human TGF-β(1) and human IL-10 to significantly inhibit anti-FVIII antibody responses in FVIII KO mice. This paper demonstrates that autologous cell therapy for antigen-targeted immune suppression may be developed to facilitate long-term therapy.
Collapse
Affiliation(s)
- Gautam Sule
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
33
|
Wambre E, DeLong JH, James EA, LaFond RE, Robinson D, Kwok WW. Differentiation stage determines pathologic and protective allergen-specific CD4+ T-cell outcomes during specific immunotherapy. J Allergy Clin Immunol 2011; 129:544-51, 551.e1-7. [PMID: 21975176 DOI: 10.1016/j.jaci.2011.08.034] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 07/23/2011] [Accepted: 08/29/2011] [Indexed: 10/16/2022]
Abstract
BACKGROUND The main obstacle to elucidating the role of CD4(+) T cells in allergen-specific immunotherapy (SIT) has been the absence of an adequately sensitive approach to directly characterize rare allergen-specific T cells without introducing substantial phenotypic modifications by means of in vitro amplification. OBJECTIVE We sought to monitor, in physiological conditions, the allergen-specific CD4(+) T cells generated during natural pollen exposure and during allergy vaccination. METHODS Alder pollen allergy was used as a model for studying seasonal allergies. Allergen-specific CD4(+) T cells were tracked and characterized in 12 subjects with alder pollen allergy, 6 nonallergic subjects, and 9 allergy vaccine-treated subjects by using peptide-MHC class II tetramers. RESULTS Allergen-specific CD4(+) T cells were detected in all of the subjects with alder pollen allergy and nonallergic subjects tested. Pathogenic responses--chemoattractant receptor homologous molecule expressed on T(H)2 lymphocytes (CRTH2) expression and T(H)2 cytokine production--are specifically associated with terminally differentiated (CD27(-)) allergen-specific CD4(+) T cells, which dominate in allergic subjects but are absent in nonallergic subjects. In contrast, CD27(+) allergen-specific CD4(+) T cells are present at low frequencies in both allergic and nonallergic subjects and reflect classical features of the protective immune response with high expression of IL-10 and IFN-γ. Restoration of a protective response during SIT appears to be due to the preferential deletion of pathogenic (CD27(-)) allergen-specific CD4(+) T cells accompanied by IL-10 induction in surviving CD27(+) allergen-specific CD4(+) T cells. CONCLUSIONS Differentiation stage divides allergen-specific CD4(+) T cells into 2 distinct subpopulations with unique functional properties and different fates during SIT.
Collapse
Affiliation(s)
- Erik Wambre
- Benaroya Research Institute at Virginia Mason, Seattle, Wash 98101, USA
| | | | | | | | | | | |
Collapse
|
34
|
Cunnusamy K, Chen PW, Niederkorn JY. IL-17A-dependent CD4+CD25+ regulatory T cells promote immune privilege of corneal allografts. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:6737-45. [PMID: 21551366 PMCID: PMC3110606 DOI: 10.4049/jimmunol.1100101] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
IL-17A is a proinflammatory cytokine that has received attention for its role in the pathogenesis of several autoimmune diseases. IL-17A has also been implicated in cardiac and renal allograft rejection. Accordingly, we hypothesized that depletion of IL-17A would enhance corneal allograft survival. Instead, our results demonstrate that blocking IL-17A in a mouse model of keratoplasty accelerated the tempo and increased the incidence of allograft rejection from 50 to 90%. We describe a novel mechanism by which CD4(+)CD25(+) regulatory T cells (Tregs) respond to IL-17A and enhance corneal allograft survival. Our findings suggest the following: 1) IL-17A is necessary for ocular immune privilege; 2) IL-17A is not required for the induction of anterior chamber-associated immune deviation; 3) Tregs require IL-17A to mediate a contact-dependent suppression; 4) corneal allograft Tregs suppress the efferent arm of the immune response and are Ag specific; 5) Tregs are not required for corneal allograft survival beyond day 30; and 6) corneal allograft-induced Treg-mediated suppression is transient. Our findings identify IL-17A as a cytokine essential for the maintenance of corneal immune privilege and establish a new paradigm whereby interplay between IL-17A and CD4(+)CD25(+) Tregs is necessary for survival of corneal allografts.
Collapse
Affiliation(s)
- Khrishen Cunnusamy
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | |
Collapse
|
35
|
Chapoval SP, Dasgupta P, Smith EP, DeTolla LJ, Lipsky MM, Kelly-Welch AE, Keegan AD. STAT6 expression in multiple cell types mediates the cooperative development of allergic airway disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:2571-83. [PMID: 21242523 PMCID: PMC3139332 DOI: 10.4049/jimmunol.1002567] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Th2 cells induce asthma through the secretion of cytokines. Two such cytokines, IL-4 and IL-13, are critical mediators of many features of this disease. They both share a common receptor subunit, IL-4Rα, and signal through the STAT6 pathway. STAT6(-/-) mice have impaired Th2 differentiation and reduced airway response to allergen. Transferred Th2 cells were not able to elicit eosinophilia in response to OVA in STAT6(-/-) mice. To clarify the role of STAT6 in allergic airway inflammation, we generated mouse bone marrow (BM) chimeras. We observed little to no eosinophilia in OVA-treated STAT6(-/-) mice even when STAT6(+/+) BM or Th2 cells were provided. However, when Th2 cells were transferred to STAT6×Rag2(-/-) mice, we observed an eosinophilic response to OVA. Nevertheless, the expression of STAT6 on either BM-derived cells or lung resident cells enhanced the severity of OVA-induced eosinophilia. Moreover, when both the BM donor and recipient lacked lymphocytes, transferred Th2 cells were sufficient to induce the level of eosinophilia comparable with that of wild-type (WT) mice. The expression of STAT6 in BM-derived cells was more critical for the enhanced eosinophilic response. Furthermore, we found a significantly higher number of CD4(+)CD25(+)Foxp3(+) T cells (regulatory T cells [Tregs]) in PBS- and OVA-treated STAT6(-/-) mouse lungs compared with that in WT animals suggesting that STAT6 limits both naturally occurring and Ag-induced Tregs. Tregs obtained from either WT or STAT6(-/-) mice were equally efficient in suppressing CD4(+) T cell proliferation in vitro. Taken together, our studies demonstrate multiple STAT6-dependent and -independent features of allergic inflammation, which may impact treatments targeting STAT6.
Collapse
Affiliation(s)
- Svetlana P. Chapoval
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
- Program in Oncology, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Preeta Dasgupta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Elizabeth P. Smith
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Louis J. DeTolla
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Michael M. Lipsky
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Ann E. Kelly-Welch
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Achsah D. Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
- Program in Oncology, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
36
|
Abstract
Forkhead box P3 (FOXP3)(+) regulatory T (T(Reg)) cells prevent autoimmune disease, maintain immune homeostasis and modulate immune responses during infection. To accomplish these tasks, T(Reg) cell activity is precisely controlled, and this requires T(Reg) cells to alter their migratory, functional and homeostatic properties in response to specific cues in the immune environment. We review progress in understanding the diversity of T(Reg) cells, T(Reg) cell function in different anatomical and inflammatory settings, and the influence of the immune environment on T(Reg) cell activity. We also consider how these factors affect immune-mediated disease in the contexts of infection, autoimmunity, cancer and transplantation.
Collapse
Affiliation(s)
- Daniel J Campbell
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98103, USA.
| | | |
Collapse
|
37
|
Velavan TP, Bechlars S, Tomiuk J, Kremsner PG, Kun JFJ. Molecular characterization of regulatory polymorphisms in the promoter region of the STAT6 gene in a Gabonese population. Mem Inst Oswaldo Cruz 2011; 106:65-9. [DOI: 10.1590/s0074-02762011000100011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 11/12/2010] [Indexed: 01/23/2023] Open
Affiliation(s)
| | | | | | - Peter G Kremsner
- Institute for Tropical Medicine; Albert Schweitzer Hospital, Gabon
| | | |
Collapse
|
38
|
Follicular lymphoma cell niche: identification of a preeminent IL-4-dependent T(FH)-B cell axis. Leukemia 2010; 24:2080-9. [PMID: 20944673 DOI: 10.1038/leu.2010.223] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Follicular lymphoma (FL) B cells contract tight connections with their microenvironment, which governs the pathogenesis and progression of the disease. Indeed, specific immune response gene signatures, obtained from whole biopsy samples, have been associated with patient survival. In this study, we performed gene expression profiling of purified B cell and non-B cell compartments obtained from FL and reactive lymph nodes. We identified 677 non-redundant genes defining the FL interface and involving 26 FL-specific functional networks. This approach highlighted an interleukin-4 (IL-4)-centered pathway associated with an activation of signal transducer and activator of transcription 6 (STAT6), which favors overexpression of IL-4-target genes. In addition, FL microenvironment was characterized by a strong enrichment in follicular helper T cells (T(FH)), as demonstrated through transcriptomic and flow cytometry analyses. The majority of phospho-STAT6(pos) B cells were located at the vicinity of cells expressing the programmed death 1 (PD-1) T(FH) marker. Moreover, purified FL-derived T(FH), expressed IL4 at very high levels compared with purified tonsil-derived T(FH) or non-T(FH) microenvironment. Altogether, our study demonstrated that tumor-infiltrating T(FH) specifically express functional IL-4 in FL, creating an IL-4-dependent T(FH)-B cell axis. This cross talk could sustain FL pathogenesis and represent a new potential therapeutic target.
Collapse
|
39
|
Cunnusamy K, Chen PW, Niederkorn JY. IL-17 promotes immune privilege of corneal allografts. THE JOURNAL OF IMMUNOLOGY 2010; 185:4651-8. [PMID: 20844197 DOI: 10.4049/jimmunol.1001576] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Corneal allograft rejection has been described as a Th1-mediated process involving IFN-γ production. However, it has been reported that corneal allograft rejection soars in IFN-γ(-/-) mice or mice treated with anti-IFN-γ mAb. Th17 is a recently described IL-17A-producing Th cell population that has been linked to renal and cardiac graft rejection, which was originally thought to be Th1-mediated. We tested the hypothesis that Th17 cells mediate corneal allograft rejection in an IL-17A-dependent fashion and unexpectedly found that depletion of IL-17A increased the incidence of rejection to 90%. We demonstrate that the exacerbated rejection following depletion of IL-17A did not result from a loss of cross-regulation of Th1 cells or exaggerated delayed-type hypersensitivity responses. Instead, inhibition of the Th1 or Th17 cell lineages promoted the emergence of a Th2 cell subset that independently mediated allograft rejection. These findings demonstrate that IL-17A is not required for corneal allograft rejection and may instead contribute to the immune privilege of corneal allografts.
Collapse
Affiliation(s)
- Khrishen Cunnusamy
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | |
Collapse
|
40
|
Niederkorn JY, Chen PW, Mellon J, Stevens C, Mayhew E. Allergic conjunctivitis exacerbates corneal allograft rejection by activating Th1 and th2 alloimmune responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:6076-83. [PMID: 20410484 PMCID: PMC2910911 DOI: 10.4049/jimmunol.0902300] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Allergic conjunctivitis (AC) and airway hyperreactivity exacerbate corneal allograft rejection. Because AC and airway hyperreactivity are allergic diseases of mucosal tissues, we determined whether an allergic disease of a nonmucosal tissue would affect corneal allograft rejection and whether Th2 cells alone accounted for accelerated graft rejection in allergic mice. Hosts sensitized cutaneously with short ragweed pollen developed cutaneous immediate hypersensitivity but rejected corneal allografts at the same tempo and incidence as naive mice. Th2 immune deviation induced with keyhole limpet hemocyanin and IFA did not affect corneal allograft rejection. Thus, Th2 immune deviation alone does not account for the exacerbation of corneal allograft rejection that occurs in mice with AC. CD4(+) T cells from AC mice elaborated Th1 (IFN-gamma) and Th2 (IL-13) cytokines when challenged with donor alloantigens. Adoptive transfer of Th1 or Th2 cells to nude mice, from AC mice that had rejected corneal allografts, produced graft rejection in 70% and 20% of the hosts, respectively. In contrast, adoptive transfer of a combination of Th1 and Th2 cells produced 100% rejection. Administration of exogenous IFN-gamma could substitute for Th1 cells and produced 100% corneal allograft rejection in recipients of Th2 cells alone. By contrast, IFN-gamma did not significantly enhance corneal allograft rejection mediated by Th1 cells. Thus, exacerbation of corneal allograft rejection in mice with AC is associated with a mixed Th1 and Th2 alloimmune response, and the contribution of Th1 cells is through their production of IFN-gamma.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9057, USA.
| | | | | | | | | |
Collapse
|
41
|
Jyonouchi H. Autism spectrum disorders and allergy: observation from a pediatric allergy/immunology clinic. Expert Rev Clin Immunol 2010; 6:397-411. [PMID: 20441426 DOI: 10.1586/eci.10.18] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
IgE-mediated allergic diseases (e.g., allergic rhinoconjunctivitis, atopic asthma and food allergy) are prevalent (up to 30%) in the general population and are increasing in developed countries. In infants and young children, non-IgE-mediated food allergy is also prevalent. In addition to easily recognized organ-specific symptoms, allergic diseases can cause neuropsychiatric symptoms, such as irritability and hyperactivity, in otherwise healthy individuals. This is also likely to occur in children with autism spectrum disorder (ASD). Moreover, the discomfort and pain associated with allergic diseases could aggravate behavioral symptoms in ASD children. Allergic conditions are easily treatable; however, ASD children may be underdiagnosed and/or undertreated for allergic and other common childhood diseases, in part due to their impaired communication skills. Practicing physicians should be aware of the potential impact of allergic diseases on behavioral symptoms and cognitive activity in ASD children. However, they also need to be aware that certain symptoms often attributed to 'allergy' by caregivers may not be immune mediated and should understand that behavioral symptoms can also be affected by many non-IgE-mediated causes.
Collapse
Affiliation(s)
- Harumi Jyonouchi
- Pediatrics, University of Medicine and Dentistry of New Jersey (UMDNJ)-New Jersey Medical School (NJMS), 185 South Orange Ave, Newark, NJ 07101, USA.
| |
Collapse
|
42
|
Abstract
That regulatory T cells (Tregs) have a crucial role in controlling allergic diseases such as asthma is now undisputed. The cytokines most commonly implicated in Treg-mediated suppression of allergic asthma are transforming growth factor-beta (TGF-beta) and interleukin (IL)-10). In addition to naturally occurring Tregs, adaptive Tregs, induced in response to foreign antigens, have been shown in recent studies. The concept of inducible/adaptive Tregs (iTregs) has considerable significance in preventing asthma if generated early enough in life. This is because cytokines such as IL-4 and IL-6 inhibit Foxp3 induction in naive CD4+ T cells and therefore de novo generation of Tregs can be expected to be less efficient when it is concomitant with effector cell development in response to an allergen. However, if iTregs can be induced, the process of infectious tolerance would facilitate expansion of the iTreg pool as suggested in the recent literature. It is tempting to speculate that there is a window of opportunity in early life in the context of a relatively immature immune system that is permissive for the generation of iTregs specific to a spectrum of allergens that would regulate asthma for lifelong. The focus of this review is the relevance of nTregs and iTregs in controlling asthma from early life into adulthood, the mechanisms underlying Treg function, and the prospects for using our current concepts to harness the full potential of Tregs to limit disease development and progression.
Collapse
Affiliation(s)
- A Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | |
Collapse
|
43
|
Macey MR, Sturgill JL, Morales JK, Falanga YT, Morales J, Norton SK, Yerram N, Shim H, Fernando J, Gifillan AM, Gomez G, Schwartz L, Oskeritzian C, Spiegel S, Conrad D, Ryan JJ. IL-4 and TGF-beta 1 counterbalance one another while regulating mast cell homeostasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:4688-95. [PMID: 20304823 PMCID: PMC3339193 DOI: 10.4049/jimmunol.0903477] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mast cell responses can be altered by cytokines, including those secreted by Th2 and regulatory T cells (Treg). Given the important role of mast cells in Th2-mediated inflammation and recent demonstrations of Treg-mast cell interactions, we examined the ability of IL-4 and TGF-beta1 to regulate mast cell homeostasis. Using in vitro and in vivo studies of mouse and human mast cells, we demonstrate that IL-4 suppresses TGF-beta1 receptor expression and signaling, and vice versa. In vitro studies demonstrated that IL-4 and TGF-beta1 had balancing effects on mast cell survival, migration, and FcepsilonRI expression, with each cytokine cancelling the effects of the other. However, in vivo analysis of peritoneal inflammation during Nippostrongylus brasiliensis infection in mice revealed a dominant suppressive function for TGF-beta1. These data support the existence of a cytokine network involving the Th2 cytokine IL-4 and the Treg cytokine TGF-beta1 that can regulate mast cell homeostasis. Dysregulation of this balance may impact allergic disease and be amenable to targeted therapy.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Homeostasis/immunology
- Humans
- Interleukin-4/physiology
- Mast Cells/immunology
- Mast Cells/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/biosynthesis
- Protein Serine-Threonine Kinases/physiology
- Receptor, Transforming Growth Factor-beta Type I
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/physiology
- Receptors, Interleukin-4/antagonists & inhibitors
- Receptors, Interleukin-4/biosynthesis
- Receptors, Interleukin-4/physiology
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Receptors, Transforming Growth Factor beta/biosynthesis
- Receptors, Transforming Growth Factor beta/physiology
- Tissue Culture Techniques
- Transforming Growth Factor beta1/antagonists & inhibitors
- Transforming Growth Factor beta1/biosynthesis
- Transforming Growth Factor beta1/physiology
Collapse
Affiliation(s)
- Matthew R. Macey
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
| | - Jamie L. Sturgill
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Johanna K. Morales
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
| | - Yves T. Falanga
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
| | - Joshua Morales
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Sarah K. Norton
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Nitin Yerram
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
| | - Hoon Shim
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
| | - Josephine Fernando
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
| | - Alasdair M. Gifillan
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Gregorio Gomez
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Lawrence Schwartz
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Carole Oskeritzian
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Sarah Spiegel
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Daniel Conrad
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - John J. Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
- Virginia Commonwealth University Allergy and Allergic Disease Cooperative Research Center, Virginia Commonwealth University, Richmond, VA 23284
| |
Collapse
|
44
|
Chapoval S, Dasgupta P, Dorsey NJ, Keegan AD. Regulation of the T helper cell type 2 (Th2)/T regulatory cell (Treg) balance by IL-4 and STAT6. J Leukoc Biol 2010; 87:1011-8. [PMID: 20335310 DOI: 10.1189/jlb.1209772] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
During the development of immune responses to pathogens, self-antigens, or environmental allergens, naive CD4(+) T cells differentiate into subsets of effector cells including Th1, Th2, and Th17 cells. The differentiation into these subsets is controlled by specific transcription factors. The activity of these effector cells is limited by nTregs and iTregs, whose differentiation and maintenance are dependent on the transcription factor Foxp3. The regulation of autoimmune diseases mediated by Th1 and Th17 cells by Tregs has been studied and reviewed extensively. However, much less has been presented about the interplay between Tregs and Th2 cells and their contribution to allergic disease. In this perspective, we discuss the regulation of Th2 cells by Tregs and vice versa, focusing on the interplay between the IL-4-activated STAT6/GATA3 pathway and Foxp3.
Collapse
Affiliation(s)
- Svetlana Chapoval
- Department of Microbiology and Immunology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
45
|
Moraes-Vieira PMM, Silva HM, Takenaka MCS, Monteiro SM, Lemos F, Saitovitch D, Kalil J, Coelho V. Differential monocyte STAT6 activation and CD4(+)CD25(+)Foxp3(+) T cells in kidney operational tolerance transplanted individuals. Hum Immunol 2010; 71:442-50. [PMID: 20122976 DOI: 10.1016/j.humimm.2010.01.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 01/12/2010] [Accepted: 01/15/2010] [Indexed: 12/28/2022]
Abstract
In organ transplantation, the immunosuppression withdrawal leads, in most cases, to rejection. Nonetheless, a special group of patients maintain stable graft function after complete withdrawal of immunosuppression, achieving a state called "operational tolerance." The study of such patients may be important to understand the mechanisms involved in human transplantation tolerance. We compared the profile of CD4(+)CD25(+)Foxp3(+) T cells and the signaling pathways IL-6/STAT3 (signal transducers and activators of transcription) and IL-4/STAT6 in peripheral blood mononuclear cells of four kidney transplant groups: (i) operational tolerance (OT), (ii) chronic allograft nephropathy (CR), (iii) stable graft function under standard immunosuppression (Sta), (iv) stable graft function under low immunosuppression, and (v) healthy individuals. Both CR and Sta displayed lower numbers and percentages of CD4(+)CD25(+)Foxp3(+) T cells compared with all other groups (p < 0.05). The OT patients displayed a reduced activation of the IL-4/STAT6 pathway in monocytes, compared with all other groups (p < 0.05). The lower numbers of CD4(+)CD25(+)Foxp3(+) T cells observed in CR individuals may be a feature of chronic allograft nephropathy. The differential OT signaling profile, with reduced phosphorylation of STAT6, in monocytes' region, suggests that some altered function of STAT6 signaling may be important for the operational tolerance state.
Collapse
|
46
|
Rynda A, Maddaloni M, Ochoa-Repáraz J, Callis G, Pascual DW. IL-28 supplants requirement for T(reg) cells in protein sigma1-mediated protection against murine experimental autoimmune encephalomyelitis (EAE). PLoS One 2010; 5:e8720. [PMID: 20090936 PMCID: PMC2806841 DOI: 10.1371/journal.pone.0008720] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 12/09/2009] [Indexed: 01/10/2023] Open
Abstract
Conventional methods to induce tolerance in humans have met with limited success. Hence, efforts to redirect tolerogen uptake using reovirus adhesin, protein sigma 1 (pσ1), may circumvent these shortcomings based upon the recent finding that when reovirus pσ1 is engineered to deliver chicken ovalbumin (OVA) mucosally, tolerance is obtained, even with a single dose. To test whether single-dose tolerance can be induced to treat EAE, proteolipid protein (PLP130–151) was genetically fused to OVA to pσ1 (PLP:OVA-pσ1) and shown to significantly ameliorate EAE, suppressing proinflammatory cytokines by IL-10+ forkhead box P3 (FoxP3)+ CD25+CD4+ Treg and IL-4+CD25−CD4+ Th2 cells. IL-10R or IL-4 neutralization reversed protection to EAE conferred by PLP:OVA-pσ1, and adoptive transfer of Ag-specific Treg or Th2 cells restored protection against EAE in recipients. Upon assessment of each relative participant, functional inactivation of CD25 impaired PLP:OVA-pσ1's protective capacity, triggering TGF-β-mediated inflammation; however, concomitant inactivation of TGF-β and CD25 reestablished PLP:OVA-pσ1-mediated protection by IL-28-producing FoxP3+CD25−CD4+ T cells. Thus, pσ1-based therapy can resolve EAE independently of or dependently upon CD25 and assigns IL-28 as an alternative therapy for autoimmunity.
Collapse
Affiliation(s)
- Agnieszka Rynda
- Veterinary Molecular Biology, Montana State University, Bozeman, Montana, United States of America
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Massimo Maddaloni
- Veterinary Molecular Biology, Montana State University, Bozeman, Montana, United States of America
| | - Javier Ochoa-Repáraz
- Veterinary Molecular Biology, Montana State University, Bozeman, Montana, United States of America
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Gayle Callis
- Veterinary Molecular Biology, Montana State University, Bozeman, Montana, United States of America
| | - David W. Pascual
- Veterinary Molecular Biology, Montana State University, Bozeman, Montana, United States of America
- * E-mail:
| |
Collapse
|
47
|
Pierau M, Engelmann S, Reinhold D, Lapp T, Schraven B, Bommhardt UH. Protein kinase B/Akt signals impair Th17 differentiation and support natural regulatory T cell function and induced regulatory T cell formation. THE JOURNAL OF IMMUNOLOGY 2009; 183:6124-34. [PMID: 19841181 DOI: 10.4049/jimmunol.0900246] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Protein kinase B (PKB)/Akt signals control T cell proliferation and differentiation but their effect on the generation and function of regulatory T cells (Treg) and Th17 cells is not well understood. In this study, we show that elevated PKB signals antagonize the immunosuppressive effect of TGF-beta1 on cell size, CD25 and CD98 expression, and proliferation of CD3-stimulated naive CD4(+) T cells from wild-type and CD28-deficient mice. Conventional CD4(+) T cells expressing active PKB are less susceptible to suppression by natural regulatory T cells. Although PKB signals do not affect the development of natural regulatory T cells, they enhance their suppressor capacity. Upon TCR triggering and TGF-beta1 costimulation, wild-type and CD28-deficient CD4(+) T cells transgenic for PKB readily express Foxp3, thereby acquiring suppressor capacity. These effects of elevated PKB signals on T cell function involve a marked and sustained activation of STAT5 and Foxp3 and reduction in nuclear NFATc1 levels. In contrast, PKB signals impair TGF-beta1/IL-6-mediated differentiation of naive CD4(+) T cells into the Th17 lineage. This correlates with an increased signaling of ERK, STAT5, and STAT6. Finally, elevated PKB signals reduced the severity of experimental autoimmune encephalomyelitis in wild-type mice but induced experimental autoimmune encephalomyelitis in mice deficient for CD28. Altogether, these data indicate an important role of PKB signals on control of TGF-beta1-mediated T cell responses and, thereby, on tolerizing and inflammatory immune processes.
Collapse
Affiliation(s)
- Mandy Pierau
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Melgert BN, Oriss TB, Qi Z, Dixon-McCarthy B, Geerlings M, Hylkema MN, Ray A. Macrophages: regulators of sex differences in asthma? Am J Respir Cell Mol Biol 2009; 42:595-603. [PMID: 19574533 DOI: 10.1165/rcmb.2009-0016oc] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Females are more susceptible to development of asthma than are males. In a mouse model of ovalbumin-induced airway inflammation, with aggravated disease in females compared with males, we studied interactions between immune and resident lung cells during asthma development to elucidate which processes are affected by sex. We studied numbers of regulatory T cells (Tregs), effector T cells, myeloid dendritic cells (mDCs), and alternatively activated macrophages (AAMPhi), and their functional capabilities. Male and female mice had comparable Treg numbers in lung tissue and comparable Treg function, but effector T cells had expanded to a greater extent in lungs of females after ovalbumin exposure. This difference in T cell expansion was therefore not the result of lack of Treg control, but appeared to be driven by a greater number of inflammatory mDCs migrating from the lungs to lymph nodes in females. Resident lung cells can influence mDC migration, and AAMPhi in lung tissue were found to be involved. Artificially elevating the number of AAMPhi in lung tissue increased the migration of mDCs and airway inflammation. We found greater numbers of AAMPhi in female lungs than in males; we therefore postulate that AAMPhi are involved in increased airway inflammation found in female mice.
Collapse
Affiliation(s)
- Barbro N Melgert
- University of Pittsburgh Medical Center, Division of Pulmonary, Allergy, and Critical Care Medicine, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | |
Collapse
|