1
|
Brook B, Checkervarty AK, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The BNT162b2 mRNA vaccine demonstrates reduced age-associated T H1 support in vitro and in vivo. iScience 2024; 27:111055. [PMID: 39569372 PMCID: PMC11576392 DOI: 10.1016/j.isci.2024.111055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 07/05/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024] Open
Abstract
mRNA vaccines demonstrate impaired immunogenicity and durability in vulnerable older populations. We hypothesized that human in vitro modeling and proteomics could elucidate age-specific mRNA vaccine actions. BNT162b2-stimulation changed the plasma proteome of blood samples from young (18-50Y) and older adult (≥60Y) participants, assessed by mass spectrometry, proximity extension assay, and multiplex. Young adult up-regulation (e.g., PSMC6, CPN1) contrasted reduced induction in older adults (e.g., TPM4, APOF, APOC2, CPN1, PI16). 30-85% lower TH1-polarizing cytokines and chemokines were induced in elderly blood (e.g., IFNγ, CXCL10). Analytes lower in older adult samples included human in vivo mRNA immunogenicity biomarkers (e.g., IFNγ, CXCL10, CCL4, IL-1RA). BNT162b2 also demonstrated reduced CD4+ TH1 responses in aged vs. young adult mice. Our study demonstrates the utility of human in vitro platforms modeling age-specific mRNA vaccine immunogenicity, highlights impaired support of TH1 polarization in older adults, and provides a rationale for precision mRNA vaccine adjuvantation to induce greater immunogenicity.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC V6Z 2K5, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amy C Sherman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine- Chair of Pediatrics, University of Rome, 00133 Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dheeraj Soni
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hanno Steen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
2
|
Lee JY, Kim S, Sohn HJ, Kim CH, Kim TG, Lee HS. Local Myeloid-Derived Suppressor Cells Impair Progression of Experimental Autoimmune Uveitis by Alleviating Oxidative Stress and Inflammation. Invest Ophthalmol Vis Sci 2023; 64:39. [PMID: 37878302 PMCID: PMC10615146 DOI: 10.1167/iovs.64.13.39] [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] [Received: 05/03/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
Purpose To evaluate the immune regulatory effect of human cord blood myeloid-derived suppressor cells (MDSCs) in experimental autoimmune uveitis (EAU) models. Methods MDSCs (1 × 106) or PBS were injected into established C57BL/6 EAU mice via the subconjunctival route on days 0 and 7. The severity of intraocular inflammation was evaluated for up to 3 weeks. Tissue injury and inflammation were analyzed using immunolabelled staining, real-time PCR, and ELISA. In addition, immune cells in draining lymph nodes (LNs) were quantified using flow cytometry. Results After 21 days, the clinical scores and histopathological grades of EAU were lower in the MDSCs group compared with the PBS group. Local administration of MDSCs suppressed the oxidative stress and the expression of TNF-α and IL-1β in the retinal tissues. In addition, it inhibited the activation of pathogenic T helper 1 (Th1) and Th17 cells in draining LNs. MDSCs increased the frequency of CD25+ Foxp3+ regulatory T cells and the mRNA expression of IL-10, as an immune modulator. Conclusions MDSCs suppressed inflammation and oxidative stress in the retina and inhibited pathogenic T cells in the LNs in EAU. Therefore, ocular administration of MDSCs has therapeutic potential for uveitis.
Collapse
Affiliation(s)
- Jae-Young Lee
- Department of Ophthalmology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sueon Kim
- ViGenCell Inc., Seoul, Republic of Korea
| | | | | | - Tai-Gyu Kim
- ViGenCell Inc., Seoul, Republic of Korea
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Soo Lee
- Department of Ophthalmology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| |
Collapse
|
3
|
Jiang Z, Zhu H, Wang P, Que W, Zhong L, Li X, Du F. Different subpopulations of regulatory T cells in human autoimmune disease, transplantation, and tumor immunity. MedComm (Beijing) 2022; 3:e137. [PMID: 35474948 PMCID: PMC9023873 DOI: 10.1002/mco2.137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/11/2022] Open
Abstract
CD4+CD25+ regulatory T cells (Tregs), a subpopulation of naturally CD4+ T cells that characteristically express transcription factor Forkhead box P3 (FOXP3), play a pivotal role in the maintenance of immune homeostasis and the prevention of autoimmunity. With the development of biological technology, the understanding of plasticity and stability of Tregs has been further developed. Recent studies have suggested that human Tregs are functionally and phenotypically diverse. The functions and mechanisms of different phenotypes of Tregs in different disease settings, such as tumor microenvironment, autoimmune diseases, and transplantation, have gradually become hot spots of immunology research that arouse extensive attention. Among the complex functions, CD4+CD25+FOXP3+ Tregs possess a potent immunosuppressive capacity and can produce various cytokines, such as IL‐2, IL‐10, and TGF‐β, to regulate immune homeostasis. They can alleviate the progression of diseases by resisting inflammatory immune responses, whereas promoting the poor prognosis of diseases by helping cells evade immune surveillance or suppressing effector T cells activity. Therefore, methods for targeting Tregs to regulate their functions in the immune microenvironment, such as depleting them to strengthen tumor immunity or expanding them to treat immunological diseases, need to be developed. Here, we discuss that different subpopulations of Tregs are essential for the development of immunotherapeutic strategies involving Tregs in human diseases.
Collapse
Affiliation(s)
- Zhongyi Jiang
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Haitao Zhu
- Department of Hepatobiliary Surgery The Affiliated Hospital of Guizhou Medical University Guizhou P. R. China
| | - Pusen Wang
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Weitao Que
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Lin Zhong
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
| | - Xiao‐Kang Li
- Department of General Surgery Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai P. R. China
- Division of Transplantation Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Futian Du
- Department of Hepatobiliary Surgery Weifang People's Hospital Shandong P. R. China
| |
Collapse
|
4
|
Russell AE, Liao Z, Tkach M, Tarwater PM, Ostrowski M, Théry C, Witwer KW. Cigarette smoke-induced extracellular vesicles from dendritic cells alter T-cell activation and HIV replication. Toxicol Lett 2022; 360:33-43. [PMID: 35181468 PMCID: PMC9014967 DOI: 10.1016/j.toxlet.2022.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/26/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
Abstract
Despite decreased rates of tobacco smoking in many areas, cigarette smoking remains a major contributor to many health problems. Cigarette smoking can reduce immune system functioning while concurrently increasing inflammation. Dendritic cells in the lung exposed to cigarette smoke become stimulated and go on to activate T-cells. Extracellular vesicles (EVs) are nano-sized particles released by cells. They participate in intercellular communication by transferring functional proteins and nucleic acids to recipient cells and have been implicated in immune responses. For example, they can display MHC-peptide complexes to activate T-cells. In the current study, we sought to understand the role of cigarette smoke extract (CSE) on dendritic cell-derived EVs and their capacity to activate and differentiate T-cells. Primary human dendritic cells (iDCs) were exposed to CSE and EVs were separated and characterized. We exposed autologous primary CD4 + T-cells to iDC-EVs and observed T helper cell populations skewing towards Th1 and Th17 phenotypes. As HIV + individuals are disproportionately likely to be current smokers, we also examined the effects of iDC-EVs on acutely infected T-cells as well as on a cell model of HIV latency (ACH-2). We found that in most cases, iDC-CSE EVs tended to reduce p24 release from the acutely infected primary T-cells, albeit with great variability. We did not observe large effects of iDC-EVs or direct CSE exposure on p24 release from the ACH-2 cell line. Together, these data suggest that iDC-CSE EVs have the capacity to modulate the immune responses, in part by pushing T-cells towards Th1 and Th17 phenotypes.
Collapse
Affiliation(s)
- Ashley E Russell
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biology, School of Science, Penn State Erie, The Behrend College, Erie, PA, United States.
| | - Zhaohao Liao
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mercedes Tkach
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Patrick M Tarwater
- Department of Epidemiology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Matias Ostrowski
- Instituto INBIRS, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Clotilde Théry
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease.
| |
Collapse
|
5
|
Tan KT, Li S, Panny L, Lin CC, Lin SC. Galangin ameliorates experimental autoimmune encephalomyelitis in mice via modulation of cellular immunity. J Immunotoxicol 2021; 18:50-60. [PMID: 33770444 DOI: 10.1080/1547691x.2021.1890863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Multiple sclerosis (MS) causes neurologic disabilities that effect musculature, sensory systems, and vision. This is largely due to demyelination of nerve fibers caused by chronic inflammation. Corticosteroid treatments ameliorate symptoms of MS, but do not successfully cure the disease itself. In the current study, the application of galangin, a phytochemical flavonoid extracted from the ginger family of Alpinis officinarum, on experimental autoimmune encephalomyelitis (EAE; mouse model for MS) was explored. This study investigated prophylactic and therapeutic activity of the drug and mechanisms by which it acts. The results revealed that galangin at 40 and 80 mg/kg could lower the incidence rate of MS, and alleviate clinical/pathological manifestations. Mice administered galangin presented with less limb paralysis, lower levels of inflammatory cell infiltrates, and decreased demyelination compared to vehicle controls. Levels of CD4+IFNγ+ (TH1) and CD4+IL-17A+ (TH17) cells in the spinal cords of EAE mice administered galangin were reduced and both cell types were not capable of expansion. More surprisingly, galangin inhibited antigen presentation and cytokine production by dendritic cells (DC). Formation of cytokines like IL-6, IL-12, and IL-23 were significantly decreased due to galangin in co-culture models of DC and T-cells. Taken together, the data lead one to conclude that galangin could potentially be used as a potent immunoregulatory agent to alleviate clinical symptoms and reduce the prevalence of MS.
Collapse
Affiliation(s)
- Kok-Tong Tan
- Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shiming Li
- Hubei Key Laboratory for Processing and Application of Catalytic Materials, College of Chemistry & Chemical Engineering, Huangang Normal University, Hubei, China
| | - Lauren Panny
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Chi-Chien Lin
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Chao Lin
- Bachelor Degree Program in Marine Biotechnology, College of Life Sciences, National Taiwan Ocean University, Keelung, Taiwan
| |
Collapse
|
6
|
Karan S, Choudhury D, Dixit A. Immunogenic characterization and protective efficacy of recombinant CsgA, major subunit of curli fibers, against Vibrio parahaemolyticus. Appl Microbiol Biotechnol 2021; 105:599-616. [PMID: 33404830 DOI: 10.1007/s00253-020-11038-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022]
Abstract
Vibrio parahaemolyticus is one of the major pathogens responsible for vibriosis and zoonotic infections in teleosts, marine invertebrates, and also humans through consumption of contaminated or unprocessed seafood. Emergence of resistance against current accessible antibiotics and spread to the food chain and environment necessitate the development of safe and effective subunit vaccine against this bacterium. Many bacteria including V. parahaemolyticus produce extracellular curli fibrils, heteropolymeric filaments of major and minor subunit, which have been implicated in adhesion, biofilm formation, and virulence. Adhesins are the primary contact points with the host which help in establishing infection and colonization. CsgA, an adhesin, is the major structural component of the curli fiber that forms homopolymers of several hundred units. Due to their exposure on the cell surface, the curli fibers are recognized by the host's immune system, would generate high immune response, and therefore can serve as effective vaccine candidate. In the present study, we describe characterization of the csgA gene, and preparation of recombinant soluble CsgA of V. parahaemolyticus (rVpCsgA), and evaluation of its vaccine potential. Immunization of BALB/c mice with the rVpCsgA mounted a strong immune response. Cellular immune assays such as antibody isotyping, in vitro splenocyte proliferation analysis, and cytokine profiling revealed mixed T-helper cell immune response. The anti-rVpCsgA antiserum was agglutination positive and specifically cross-reacted with the curli CsgA present on the outer membrane of V. parahaemolyticus cells, thus demonstrating its neutralization potential. One hundred percent survival of the immunized mice upon challenge with the lethal dosage of the bacterium established that the rVpCsgA could serve as an effective vaccine against the bacterium. KEY POINTS: • Recombinant histidine-tagged CsgA of V. parahaemolyticus, rVpCsgA, was purified. • The rVpCsgA immunization produced mixed immune response and agglutinating antibodies. • Immunization with the rVpCsgA protected mice against V. parahaemolyticus challenge.
Collapse
Affiliation(s)
- Sweta Karan
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru Universitys, New Delhi, 110067, India
| | - Devapriya Choudhury
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru Universitys, New Delhi, 110067, India.
| | - Aparna Dixit
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru Universitys, New Delhi, 110067, India.
| |
Collapse
|
7
|
Loos J, Schmaul S, Noll TM, Paterka M, Schillner M, Löffel JT, Zipp F, Bittner S. Functional characteristics of Th1, Th17, and ex-Th17 cells in EAE revealed by intravital two-photon microscopy. J Neuroinflammation 2020; 17:357. [PMID: 33243290 PMCID: PMC7694901 DOI: 10.1186/s12974-020-02021-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
Background T helper (Th) 17 cells are a highly plastic subset of T cells, which in the context of neuroinflammation, are able to acquire pathogenic features originally attributed to Th1 cells (resulting in so called ex-Th17 cells). Thus, a strict separation between the two T cell subsets in the context of experimental autoimmune encephalomyelitis (EAE) is difficult. High variability in culture and EAE induction protocols contributed to previous conflicting results concerning the differential contribution of Th1 and Th17 cells in EAE. Here, we systematically evaluate the role of different T cell differentiation and transfer protocols for EAE disease development and investigate the functional dynamics of encephalitogenic T cells directly within the inflamed central nervous system (CNS) tissue. Methods We compiled the currently used EAE induction protocols reported in literature and investigated the influence of the different Th1 and Th17 differentiation protocols as well as EAE induction protocols on the EAE disease course. Moreover, we assessed the cytokine profile and functional dynamics of both encephalitogenic Th1 and Th17 cells in the inflamed CNS using flow cytometry and intravital two-photon laser scanning microscopy. Lastly, we used astrocyte culture and adoptive transfer EAE to evaluate the impact of Th1 and Th17 cells on astrocyte adhesion molecule expression in vitro and in vivo. Results We show that EAE courses are highly dependent on in vitro differentiation and transfer protocols. Moreover, using genetically encoded reporter mice (B6.IL17A-EGFP.acRFP x 2d2/2d2.RFP), we show that the motility of interferon (IFN)γ-producing ex-Th17 cells more closely resembles Th1 cells than Th17 cells in transfer EAE. Mechanistically, IFNγ-producing Th1 cells selectively induce the expression of cellular adhesion molecules I-CAM1 while Th1 as well as ex-Th17 induce V-CAM1 on astrocytes. Conclusions The behavior of ex-Th17 cells in EAE lesions in vivo resembles Th1 rather than Th17 cells, underlining that their change in cytokine production is associated with functional phenotype alterations of these cells. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02021-x.
Collapse
Affiliation(s)
- Julia Loos
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Samantha Schmaul
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Theresa Marie Noll
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Magdalena Paterka
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Miriam Schillner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Julian T Löffel
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany.
| |
Collapse
|
8
|
Li X, Gao Q, Yang L, Han M, Zhou C, Mu H. Matairesinol ameliorates experimental autoimmune uveitis by suppression of IRBP-specific Th17 cells. J Neuroimmunol 2020; 345:577286. [PMID: 32559555 DOI: 10.1016/j.jneuroim.2020.577286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
We investigated the effects of matairesinol (MAT) in the experimental autoimmune uveitis (EAU), a classical animal model of uveitis. We found that treatment with MAT could alleviate intraocular inflammation of EAU. Notably, Th17 cells in eyes of EAU mice could be predominantly restrained by MAT. Furthermore, MAT could inhibit Th17 differentiation in vitro. In addition, MAT inhibited the signaling of MAPK and ROR-γt, a pivotal transcription factor for Th17 cell differentiation in vitro and in vivo. Taken together, these results suggested that MAT had immune-suppressive effects on autoimmune inflammation through Th17 cells.
Collapse
Affiliation(s)
- Xin Li
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Qiang Gao
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Lei Yang
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Meng Han
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Chunlei Zhou
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Hong Mu
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China.
| |
Collapse
|
9
|
Bittner-Eddy PD, Fischer LA, Costalonga M. Transient Expression of IL-17A in Foxp3 Fate-Tracked Cells in Porphyromonas gingivalis-Mediated Oral Dysbiosis. Front Immunol 2020; 11:677. [PMID: 32391008 PMCID: PMC7190800 DOI: 10.3389/fimmu.2020.00677] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/26/2020] [Indexed: 01/26/2023] Open
Abstract
In periodontitis Porphyromonas gingivalis contributes to the development of a dysbiotic oral microbiome. This altered ecosystem elicits a diverse innate and adaptive immune response that simultaneously involves Th1, Th17, and Treg cells. It has been shown that Th17 cells can alter their gene expression to produce interferon-gamma (IFN-γ). Forkhead box P3 (Foxp3) is considered the master regulator of Treg cells that produce inhibitory cytokines like IL-10. Differentiation pathways that lead to Th17 and Treg cells from naïve progenitors are considered antagonistic. However, it has been reported that Treg cells expressing IL-17A as well as IFN-γ producing Th17 cells have been observed in several inflammatory conditions. Each scenario appears plausible with T cell transdifferentiation resulting from persistent microbial challenge and consequent inflammation. We established that oral colonization with P. gingivalis drives an initial IL-17A dominated Th17 response in the oral mucosa that is dependent on intraepithelial Langerhans cells (LCs). We hypothesized that Treg cells contribute to this initial IL-17A response through transient expression of IL-17A and that persistent mucosal colonization with P. gingivalis drives Th17 cells toward an IFN-γ phenotype at later stages of infection. We utilized fate-tracking mice where IL-17A- or Foxp3-promoter activity drives the permanent expression of red fluorescent protein tdTomato to test our hypothesis. At day 28 of infection timeline, Th17 cells dominated in the oral mucosa, outnumbering Th1 cells by 3:1. By day 48 this dominance was inverted with Th1 cells outnumbering Th17 cells by nearly 2:1. Tracking tdTomato+ Th17 cells revealed only sporadic transdifferentiation to an IFN-γ-producing phenotype by day 48; the appearance of Th1 cells at day 48 was due to a late de novo Th1 response. tdTomato+ Foxp3+ T cells were 35% of the total live CD4+T cells in the oral mucosa and 3.9% of them developed a transient IL-17A-producing phenotype by day 28. Interestingly, by day 48 these IL-17A-producing Foxp3+ T cells had disappeared. Therefore, persistent oral P. gingivalis infection stimulates an initial IL-17A-biased response led by Th17 cells and a small but significant number of IL-17A-expressing Treg cells that changes into a late de novo Th1 response with only sporadic transdifferentiation of Th17 cells.
Collapse
|
10
|
Abdelaziz MH, Wang H, Cheng J, Xu H. Th2 cells as an intermediate for the differentiation of naïve T cells into Th9 cells, associated with the Smad3/Smad4 and IRF4 pathway. Exp Ther Med 2020; 19:1947-1954. [PMID: 32104253 DOI: 10.3892/etm.2020.8420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 06/26/2019] [Indexed: 01/12/2023] Open
Abstract
Type 9 T-helper (Th9) cells are associated with atopic and inflammatory diseases. Their increased levels and functions contribute to a number of inflammatory disorders, where they are accompanied by enhanced Th2-cell activity. However, there is currently no consensus regarding the association between Th9 and Th2 cells. Th9 cells may be induced from naïve T (Th0) cells under specific polarization conditions in vitro, a process driven by the addition of specific cytokines. In the present study, Th0 cells were cultured under Th9-polarizing conditions to promote differentiation into interleukin (IL)-4+ IL-9- or IL-4- IL-9+ T cells after 3 or 5 days in culture, respectively; the mRNA expression levels of IL-9 and IL-4 were consistent with the induced cell types. Simultaneously, the levels of interferon-regulatory factor 4 (IRF-4) and Smad3/Smad4 were significantly increased following Th9-cell polarization. It was therefore proposed that Th2 cells may be generated in the early stages of Th9-cell differentiation, and then ultimately differentiate into Th9 cells via the Smad3/Smad4 and IRF-4 activation pathway.
Collapse
Affiliation(s)
- Mohamed Hamed Abdelaziz
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Huixuan Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jianjun Cheng
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Laboratory Medicine, the Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Huaxi Xu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
11
|
Bittner-Eddy PD, Fischer LA, Costalonga M. Cre-loxP Reporter Mouse Reveals Stochastic Activity of the Foxp3 Promoter. Front Immunol 2019; 10:2228. [PMID: 31616418 PMCID: PMC6763954 DOI: 10.3389/fimmu.2019.02228] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
Mouse models that combine specific loxP-flanked gene sequences with Cre recombinase expressed from cell-regulated promoters have become important tools to investigate gene function. Critically however, expression of Cre recombinase may not always be restricted to the target cell or tissue of interest due to promiscuous activity of the driving promoter. Expression of Cre recombinase and, by extension, excision of the loxP-flanked gene may occur in non-target cells and may not be readily apparent. Here we report on the fidelity of Cre recombinase expressed from the il17a or Foxp3 promoters by combining them with a constitutively expressed floxed-stopped tdTomato reporter gene. Foxp3-driven Cre recombinase in F1 mice induced tdTomato red fluorescent protein in Treg cells but also in a range of other immune cells. Frequency of tdTomato expression was variable but positively correlated (p < 0.0001) amongst lymphoid (B cells and CD8 T cells) and blood-resident myeloid cells (dendritic cells, monocytes, neutrophils) suggesting stochastic activity of the Foxp3 promoter rather than developmental regulation in common ancestral progenitors. Interestingly, frequency of tdTomato+ dendritic cells, monocytes and neutrophils did not correlate with the tdTomato+ fraction in eosinophils, indicating that activity of the Foxp3 promoter in eosinophils occurred after the split from a common multipotent progenitor. When these F1 mice were crossed to achieve homozygosity of the promoter and reporter gene, a novel visually red phenotype was observed segregating amongst littermates. The red coloration was widespread and prevalent in non-immune tissues. Thymocytes examined from these red mice showed that all four subsets of immature thymocytes (CD4− CD8−) based on differential expression of CD25 and CD44 were expressing tdTomato. Finally, we show evidence of Foxp3 Cre recombinase independent tdTomato expression, suggesting germ line transmission of an activated tdTomato reporter gene. Our data highlights potential issues with conclusions drawn from using specifically the B6.129(Cg)-Foxp3tm4(YFP/Cre)Ayr/J mice.
Collapse
Affiliation(s)
- Peter D Bittner-Eddy
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Lori A Fischer
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Massimo Costalonga
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
12
|
Wang W, Chong WP, Li C, Chen Z, Wu S, Zhou H, Wan Y, Chen W, Gery I, Liu Y, Caspi RR, Chen J. Type I Interferon Therapy Limits CNS Autoimmunity by Inhibiting CXCR3-Mediated Trafficking of Pathogenic Effector T Cells. Cell Rep 2019; 28:486-497.e4. [PMID: 31291583 PMCID: PMC6748389 DOI: 10.1016/j.celrep.2019.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/13/2019] [Accepted: 06/05/2019] [Indexed: 12/22/2022] Open
Abstract
Type I interferons (IFNs) have therapeutic potential in CNS autoimmune diseases, such as uveitis, but the molecular mechanisms remain unclear. Using a T cell-transfer model of experimental autoimmune uveitis (EAU), we found that IFN-α/β treatment inhibited the migration of IFN-γ-producing pathogenic CD4+ T cells to effector sites. IFN-α/β upregulated the expression of the cognate ligands CXCL9, CXCL10, and CXCL11, causing ligand-mediated downregulation of CXCR3 expression and effector T cell retention in the spleen. Accordingly, type I IFN did not alter EAU progression in CXCR3-/- mice. In uveitis patients, disease exacerbations correlated with reduced serum IFN-α concentrations. IFN-α/β reduced CXCR3 expression and migration by human effector T cells, and these parameters were associated with the therapeutic efficacy of IFN-α in uveitis patients. Our findings provide insight into the molecular basis of type I IFN therapy for CNS autoimmune diseases and identify CXCR3 as a biomarker for effective type I IFN immunotherapy.
Collapse
Affiliation(s)
- Weiwei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China
| | - Wai Po Chong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China; Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Chunmei Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China
| | - Zilin Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China
| | - Sihan Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China
| | - Hongyan Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China
| | - Ying Wan
- Biomedical Analysis Center, Third Military Medical University, Chongqing 40038, China
| | - Wanjun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Igal Gery
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA.
| | - Jun Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University (SYSU), Guangzhou 510060, China; Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
13
|
Bing SJ, Shemesh I, Chong WP, Horai R, Jittayasothorn Y, Silver PB, Sredni B, Caspi RR. AS101 ameliorates experimental autoimmune uveitis by regulating Th1 and Th17 responses and inducing Treg cells. J Autoimmun 2019; 100:52-61. [PMID: 30853312 DOI: 10.1016/j.jaut.2019.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 10/27/2022]
Abstract
AS101 is an organotellurium compound with multifaceted immunoregulatory properties that is remarkable for its lack of toxicity. We tested the therapeutic effect of AS101 in experimental autoimmune uveitis (EAU), a model for human autoimmune uveitis. Unexpectedly, treatment with AS101 elicited Treg generation in vivo in otherwise unmanipulated mice. Mice immunized for EAU with the retinal antigen IRBP and treated with AS101 developed attenuated disease, as did AS101-treated recipients of retina-specific T cells activated in vitro. In both settings, eye-infiltrating effector T cells were decreased, whereas regulatory T (Treg) cells in the spleen were increased. Mechanistic studies in vitro revealed that AS101 restricted polarization of retina-specific T cells towards Th1 or Th17 lineage by repressing activation of their respective lineage-specific transcription factors and downstream signals. Retina-specific T cells polarized in vitro towards Th1 or Th17 in the presence of AS101 had impaired ability to induce EAU in naïve recipients. Finally, AS101 promoted differentiation of retina-specific T cells to Tregs in vitro independently of TGF-β. We conclude that AS101 modulates autoimmune T cells by inhibiting acquisition and expression of effector function and by promoting Treg generation, and suggest that AS101 could be useful as a therapeutic approach for autoimmune uveitis.
Collapse
Affiliation(s)
- So Jin Bing
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Itay Shemesh
- C.A.I.R. Institute, Safdié AIDS and Immunology Research Center, Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Wai Po Chong
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Reiko Horai
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yingyos Jittayasothorn
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Phyllis B Silver
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Benjamin Sredni
- C.A.I.R. Institute, Safdié AIDS and Immunology Research Center, Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel.
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
14
|
Lyu C, Bing SJ, Wandu WS, Xu B, Shi G, Hinshaw SJ, Lobera M, Caspi RR, Lu L, Yang J, Gery I. TMP778, a selective inhibitor of RORγt, suppresses experimental autoimmune uveitis development, but affects both Th17 and Th1 cell populations. Eur J Immunol 2018; 48:1810-1816. [PMID: 30218573 DOI: 10.1002/eji.201747029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 08/03/2018] [Accepted: 09/12/2018] [Indexed: 12/24/2022]
Abstract
Experimental autoimmune uveitis (EAU), an animal model for severe intraocular inflammatory eye diseases, is mediated by both Th1 and Th17 cells. Here, we examined the capacity of TMP778, a selective inhibitor of RORγt, to inhibit the development of EAU, as well as the related immune responses. EAU was induced in B10.A mice by immunization with interphotoreceptor retinoid-binding protein (IRBP). Treatment with TMP778 significantly inhibited the development of EAU, determined by histological examination. In addition, the treatment suppressed the cellular immune response to IRBP, determined by reduced production of IL-17 and IFN-γ, as well as lower percentages of lymphocytes expressing these cytokines, as compared to vehicle-treated controls. The inhibition of IFN-γ expression by TMP778 is unexpected in view of this compound being a selective inhibitor of RORγt. The observation was further confirmed by the finding of reduced expression of the T-bet (Tbx21) gene, the transcription factor for IFN-γ, by cells of TMP778-treated mice. Thus, these data demonstrate the capacity of TMP778 to inhibit pathogenic autoimmunity in the eye and shed new light on its mode of action in vivo.
Collapse
Affiliation(s)
- Cancan Lyu
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - So Jin Bing
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wambui S Wandu
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Biying Xu
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Guangpu Shi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Samuel J Hinshaw
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | | | - Igal Gery
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Puniya BL, Todd RG, Mohammed A, Brown DM, Barberis M, Helikar T. A Mechanistic Computational Model Reveals That Plasticity of CD4 + T Cell Differentiation Is a Function of Cytokine Composition and Dosage. Front Physiol 2018; 9:878. [PMID: 30116195 PMCID: PMC6083813 DOI: 10.3389/fphys.2018.00878] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
CD4+ T cells provide cell-mediated immunity in response to various antigens. During an immune response, naïve CD4+ T cells differentiate into specialized effector T helper (Th1, Th2, and Th17) cells and induced regulatory (iTreg) cells based on a cytokine milieu. In recent studies, complex phenotypes resembling more than one classical T cell lineage have been experimentally observed. Herein, we sought to characterize the capacity of T cell differentiation in response to the complex extracellular environment. We constructed a comprehensive mechanistic (logical) computational model of the signal transduction that regulates T cell differentiation. The model's dynamics were characterized and analyzed under 511 different environmental conditions. Under these conditions, the model predicted the classical as well as the novel complex (mixed) T cell phenotypes that can co-express transcription factors (TFs) related to multiple differentiated T cell lineages. Analyses of the model suggest that the lineage decision is regulated by both compositions and dosage of signals that constitute the extracellular environment. In this regard, we first characterized the specific patterns of extracellular environments that result in novel T cell phenotypes. Next, we predicted the inputs that can regulate the transition between the canonical and complex T cell phenotypes in a dose-dependent manner. Finally, we predicted the optimal levels of inputs that can simultaneously maximize the activity of multiple lineage-specifying TFs and that can drive a phenotype toward one of the co-expressed TFs. In conclusion, our study provides new insights into the plasticity of CD4+ T cell differentiation, and also acts as a tool to design testable hypotheses for the generation of complex T cell phenotypes by various input combinations and dosages.
Collapse
Affiliation(s)
- Bhanwar Lal Puniya
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Robert G Todd
- Department of Natural and Applied Sciences, Mount Mercy University, Cedar Rapids, IA, United States
| | - Akram Mohammed
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States.,Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands.,Molecular Cell Physiology, VU University Amsterdam, Amsterdam, Netherlands
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
16
|
Regulatory Role of CD4 + T Cells in Myocarditis. J Immunol Res 2018; 2018:4396351. [PMID: 30035131 PMCID: PMC6032977 DOI: 10.1155/2018/4396351] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 12/13/2022] Open
Abstract
Myocarditis is an important cause of heart failure in young patients. Autoreactive, most often, infection-triggered CD4+ T cells were confirmed to be critical for myocarditis induction. Due to a defect in clonal deletion of heart-reactive CD4+ T cells in the thymus of mice and humans, significant numbers of heart-specific autoreactive CD4+ T cells circulate in the blood. Normally, regulatory T cells maintain peripheral tolerance and prevent spontaneous myocarditis development. In the presence of tissue damage and innate immune activation, however, activated self-antigen-loaded dendritic cells promote CD4+ effector T cell expansion and myocarditis. So far, a direct pathogenic role has been described for both activated Th17 and Th1 effector CD4+ T cell subsets, though Th1 effector T cell-derived interferon-gamma was shown to limit myocarditis severity and prevent transition to inflammatory dilated cardiomyopathy. Interestingly, recent observations point out that various CD4+ T cell subsets demonstrate high plasticity in maintaining immune homeostasis and modulating disease phenotypes in myocarditis. These subsets include Th1 and Th17 effector cells and regulatory T cells, despite the fact that there are still sparse and controversial data on the specific role of FOXP3-expressing Treg in myocarditis. Understanding the specific roles of these T cell populations at different stages of the disease progression might provide a key for the development of successful therapeutic strategies.
Collapse
|
17
|
The Immunoregulation of Th17 in Host against Intracellular Bacterial Infection. Mediators Inflamm 2018; 2018:6587296. [PMID: 29743811 PMCID: PMC5884031 DOI: 10.1155/2018/6587296] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/04/2018] [Indexed: 12/14/2022] Open
Abstract
T helper 17 cells (Th17) constitute a distinct subset of helper T cells with a unique transcriptional profile (STAT3, RORγ, and RORα), cytokine production pattern (IL17 family), and requirement of specific cytokines for their differentiation (TGF-β, IL6, IL21, and IL23). Recent studies involving experimental animals and humans have shown that Th17/IL17 plays a crucial role in host defense against a variety of pathogens, including bacteria and viruses. The underlying mechanisms by which Th17 performs include dendritic cell (DC) regulation, neutrophil recruitment, Th1 modulation, and T regulatory cell (Treg) balance. In recent years, researchers have generated an accumulating wealth of evidence on the role of Th17/IL17 in protective immunity to intracellular bacterial pathogens, such as Mycobacterium tuberculosis and Chlamydia trachomatis, which are one of the most important pathogens that inflict significant socioeconomic burden across the globe. In this article, we reviewed the current literature on the functions and mechanisms by which Th17/IL17 responds to intracellular bacterial infections. A better understanding of Th17/IL17 immunity to pathogens would be crucial for developing effective prophylactics and therapeutics.
Collapse
|
18
|
Rogier R, Evans-Marin H, Manasson J, van der Kraan PM, Walgreen B, Helsen MM, van den Bersselaar LA, van de Loo FA, van Lent PL, Abramson SB, van den Berg WB, Koenders MI, Scher JU, Abdollahi-Roodsaz S. Alteration of the intestinal microbiome characterizes preclinical inflammatory arthritis in mice and its modulation attenuates established arthritis. Sci Rep 2017; 7:15613. [PMID: 29142301 PMCID: PMC5688157 DOI: 10.1038/s41598-017-15802-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
Perturbations of the intestinal microbiome have been observed in patients with new-onset and chronic autoimmune inflammatory arthritis. However, it is currently unknown whether these alterations precede the development of arthritis or are rather a consequence of disease. Modulation of intestinal microbiota by oral antibiotics or germ-free condition can prevent arthritis in mice. Yet, the therapeutic potential of modulation of the microbiota after the onset of arthritis is not well characterized. We here show that the intestinal microbial community undergoes marked changes in the preclinical phase of collagen induced arthritis (CIA). The abundance of the phylum Bacteroidetes, specifically families S24-7 and Bacteroidaceae was reduced, whereas Firmicutes and Proteobacteria, such as Ruminococcaceae, Lachnospiraceae and Desulfovibrinocaceae, were expanded during the immune-priming phase of arthritis. In addition, we found that the abundance of lamina propria Th17, but not Th1, cells is highly correlated with the severity of arthritis. Elimination of the intestinal microbiota during established arthritis specifically reduced intestinal Th17 cells and attenuated arthritis. These effects were associated with reduced serum amyloid A expression in ileum and synovial tissue. Our observations suggest that intestinal microbiota perturbations precede arthritis, and that modulation of the intestinal microbiota after the onset of arthritis may offer therapeutic opportunities.
Collapse
Affiliation(s)
- Rebecca Rogier
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Heather Evans-Marin
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Julia Manasson
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Peter M van der Kraan
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Birgitte Walgreen
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Monique M Helsen
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Fons A van de Loo
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter L van Lent
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Steven B Abramson
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Wim B van den Berg
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije I Koenders
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Shahla Abdollahi-Roodsaz
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States.
| |
Collapse
|
19
|
Tan C, Wandu WS, St Leger A, Kielczewski J, Wawrousek EF, Chan CC, Gery I. Unlike Th1/Th17 cells, Th2/Th9 cells selectively migrate to the limbus/conjunctiva and initiate an eosinophilic infiltration process. Exp Eye Res 2017; 166:116-119. [PMID: 29074386 DOI: 10.1016/j.exer.2017.10.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 07/27/2017] [Accepted: 10/18/2017] [Indexed: 12/24/2022]
Abstract
In this study we compared polarized mouse T-helper (Th) lymphocytes of four populations, sensitized against an ocular antigen, for their patterns of migration and induction of inflammatory processes in recipient mouse eyes expressing the target antigen. Th1, Th2, Th9 and Th17 cells transgenically expressing T-cell receptor (TCR) specific against hen egg lysozyme (HEL) were adoptively transferred to recipient mice expressing HEL in their eyes. Recipient eyes collected 4 or 7 days post injection were analyzed for histopathological changes. Th1 and Th17 cells induced moderate to severe intraocular inflammation in the recipient mouse eyes, but essentially did not migrate into the conjunctiva. In contrast, Th2 and Th9 cells invaded minimally the intraocular space of recipient eyes, but accumulated in the limbus and migrated into the conjunctiva of the recipient mice and initiated allergy-like inflammatory responses, as indicated by remarkable eosinophil involvement. These data thus shed new light on the differences between the migration patterns and ocular pathogenic processes mediated by Th1/Th17 and by Th2/Th9 populations.
Collapse
Affiliation(s)
- Cuiyan Tan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wambui S Wandu
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony St Leger
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Kielczewski
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eric F Wawrousek
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chi-Chao Chan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Igal Gery
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
20
|
The Th17 Lineage: From Barrier Surfaces Homeostasis to Autoimmunity, Cancer, and HIV-1 Pathogenesis. Viruses 2017; 9:v9100303. [PMID: 29048384 PMCID: PMC5691654 DOI: 10.3390/v9100303] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022] Open
Abstract
The T helper 17 (Th17) cells represent a subset of CD4+ T-cells with unique effector functions, developmental plasticity, and stem-cell features. Th17 cells bridge innate and adaptive immunity against fungal and bacterial infections at skin and mucosal barrier surfaces. Although Th17 cells have been extensively studied in the context of autoimmunity, their role in various other pathologies is underexplored and remains an area of open investigation. This review summarizes the history of Th17 cell discovery and the current knowledge relative to the beneficial role of Th17 cells in maintaining mucosal immunity homeostasis. We further discuss the concept of Th17 pathogenicity in the context of autoimmunity, cancer, and HIV infection, and we review the most recent discoveries on molecular mechanisms regulating HIV replication/persistence in pathogenic Th17 cells. Finally, we stress the need for novel fundamental research discovery-based Th17-specific therapeutic interventions to treat pathogenic conditions associated with Th17 abnormalities, including HIV infection.
Collapse
|
21
|
Prajeeth CK, Kronisch J, Khorooshi R, Knier B, Toft-Hansen H, Gudi V, Floess S, Huehn J, Owens T, Korn T, Stangel M. Effectors of Th1 and Th17 cells act on astrocytes and augment their neuroinflammatory properties. J Neuroinflammation 2017; 14:204. [PMID: 29037246 PMCID: PMC5644084 DOI: 10.1186/s12974-017-0978-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 10/06/2017] [Indexed: 12/30/2022] Open
Abstract
Background Autoreactive Th1 and Th17 cells are believed to mediate the pathology of multiple sclerosis in the central nervous system (CNS). Their interaction with microglia and astrocytes in the CNS is crucial for the regulation of the neuroinflammation. Previously, we have shown that only Th1 but not Th17 effectors activate microglia. However, it is not clear which cells are targets of Th17 effectors in the CNS. Methods To understand the effects driven by Th17 cells in the CNS, we induced experimental autoimmune encephalomyelitis in wild-type mice and CD4+ T cell-specific integrin α4-deficient mice where trafficking of Th1 cells into the CNS was affected. We compared microglial and astrocyte response in the brain and spinal cord of these mice. We further treated astrocytes with supernatants from highly pure Th1 and Th17 cultures and assessed the messenger RNA expression of neurotrophic factors, cytokines and chemokines, using real-time PCR. Data obtained was analyzed using the Kruskal-Wallis test. Results We observed in α4-deficient mice weak microglial activation but comparable astrogliosis to that of wild-type mice in the regions of the brain populated with Th17 infiltrates, suggesting that Th17 cells target astrocytes and not microglia. In vitro, in response to supernatants from Th1 and Th17 cultures, astrocytes showed altered expression of neurotrophic factors, pro-inflammatory cytokines and chemokines. Furthermore, increased expression of chemokines in Th1- and Th17-treated astrocytes enhanced recruitment of microglia and transendothelial migration of Th17 cells in vitro. Conclusion Our results demonstrate the delicate interaction between T cell subsets and glial cells and how they communicate to mediate their effects. Effectors of Th1 act on both microglia and astrocytes whereas Th17 effectors preferentially target astrocytes to promote neuroinflammation. Electronic supplementary material The online version of this article (10.1186/s12974-017-0978-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chittappen K Prajeeth
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Julius Kronisch
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Benjamin Knier
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Henrik Toft-Hansen
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark.,Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Stefan Floess
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Trevor Owens
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Thomas Korn
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany. .,Center of Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
22
|
Tahmasebinia F, Pourgholaminejad A. The role of Th17 cells in auto-inflammatory neurological disorders. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:408-416. [PMID: 28760387 DOI: 10.1016/j.pnpbp.2017.07.023] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 07/26/2017] [Accepted: 07/27/2017] [Indexed: 01/13/2023]
Abstract
The role of T helper 17 (Th17) cells in auto-inflammatory neurological disorders such as Multiple Sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and schizophrenia has not been clarified completely. Th17-derived pro-inflammatory cytokines including IL-17, IL-21, IL-22, IL-23, GM-CSF, and IFN-γ have a critical role in the pathogenesis of these disorders. In this review, we demonstrate the role of Th17 cells and their related cytokines in the immunopathology of above-mentioned disorders to get a better understanding of neuroinflammatory mechanisms mediated by Th17 cells associated with events leading to neurodegeneration.
Collapse
Affiliation(s)
- Foozhan Tahmasebinia
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Arash Pourgholaminejad
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
23
|
Downs-Canner S, Berkey S, Delgoffe GM, Edwards RP, Curiel T, Odunsi K, Bartlett DL, Obermajer N. Suppressive IL-17A +Foxp3 + and ex-Th17 IL-17A negFoxp3 + T reg cells are a source of tumour-associated T reg cells. Nat Commun 2017; 8:14649. [PMID: 28290453 PMCID: PMC5355894 DOI: 10.1038/ncomms14649] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/20/2017] [Indexed: 01/05/2023] Open
Abstract
Th17 and regulatory T (Treg) cells are integral in maintaining immune homeostasis and Th17–Treg imbalance is associated with inflammatory immunosuppression in cancer. Here we show that Th17 cells are a source of tumour-induced Foxp3+ cells. In addition to natural (n)Treg and induced (i)Treg cells that develop from naive precursors, suppressive IL-17A+Foxp3+ and ex-Th17 Foxp3+ cells are converted from IL-17A+Foxp3neg cells in tumour-bearing mice. Metabolic phenotyping of Foxp3-expressing IL-17A+, ex-Th17 and iTreg cells demonstrates the dissociation between the metabolic fitness and the suppressive function of Foxp3-expressing Treg cell subsets. Although all Foxp3-expressing subsets are immunosuppressive, glycolysis is a prominent metabolic pathway exerted only by IL-17A+Foxp3+ cells. Transcriptome analysis and flow cytometry of IL-17A+Foxp3+ cells indicate that Folr4, GARP, Itgb8, Pglyrp1, Il1rl1, Itgae, TIGIT and ICOS are Th17-to-Treg cell transdifferentiation-associated markers. Tumour-associated Th17-to-Treg cell conversion identified here provides insights for targeting the dynamism of Th17–Treg cells in cancer immunotherapy. Th17 cells can transdifferentiate into regulatory T (Treg) cells. Here the authors characterize tumour-driven Th17-to-Treg cell transdifferentiation and identify potential cancer therapy targets.
Collapse
Affiliation(s)
- Stephanie Downs-Canner
- Department of Surgical Oncology, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA
| | - Sara Berkey
- Department of Surgical Oncology, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA
| | - Greg M Delgoffe
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA.,Tumour Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, 5115 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA.,Magee-Womens Research Institute Ovarian Cancer Center of Excellence, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA
| | - Robert P Edwards
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA.,Magee-Womens Research Institute Ovarian Cancer Center of Excellence, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA.,Peritoneal/Ovarian Cancer Specialty Care Center, Pittsburgh, Pennsylvania 15213, USA
| | - Tyler Curiel
- UT Health Science Center at San Antonio, 8403 Floyd Curl Drive, San Antonio, Texas 78229, USA
| | - Kunle Odunsi
- Departments of Gynecologic Oncology and Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | - David L Bartlett
- Department of Surgical Oncology, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA
| | - Nataša Obermajer
- Department of Surgical Oncology, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
24
|
Mondal S, Rangasamy SB, Ghosh S, Watson RL, Pahan K. Nebulization of RNS60, a Physically-Modified Saline, Attenuates the Adoptive Transfer of Experimental Allergic Encephalomyelitis in Mice: Implications for Multiple Sclerosis Therapy. Neurochem Res 2017; 42:1555-1570. [PMID: 28271325 DOI: 10.1007/s11064-017-2214-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/20/2017] [Accepted: 02/20/2017] [Indexed: 01/12/2023]
Abstract
Developing a new and effective therapeutic approach against multiple sclerosis (MS) is always an important area of research. RNS60 is a bioactive aqueous solution generated by subjecting normal saline to Taylor-Couette-Poiseuille flow under elevated oxygen pressure. Recently we have demonstrated that RNS60, administered through intraperitoneal injection, ameliorated clinical symptoms and disease progression of experimental allergic encephalomyelitis (EAE), an animal model of MS. Since the intravenous route is not preferred for treating a chronic condition, we tested if nebulization of RNS60 could attenuate the disease process of adoptively-transferred EAE in mice. Although we could not directly image RNS60 after nebulization, nebulized Alexa680 reached spleen, spinal cord and different parts of the brain. Nebulization of RNS60 starting from the acute phase attenuated clinical symptoms of relapsing-remitting EAE in female SJL/J mice. RNS60 nebulization also inhibited perivascular cuffing, maintained the integrity of blood-brain and blood-spinal cord barriers, suppressed inflammation, normalized the expression of myelin genes, and blocked demyelination in the CNS of EAE mice. On the immunomodulatory front, nebulization of RNS60 to EAE mice led to the enrichment of anti-autoimmune regulatory T cells (Tregs) and suppression of autoimmune Th17 cells. Together, these results suggest that nebulization of RNS60 may be used to control aberrant immune responses in MS and other autoimmune disorders.
Collapse
Affiliation(s)
- Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Suresh B Rangasamy
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Supurna Ghosh
- Revalesio Corporation, 1200 East D Street, Tacoma, WA, 98421, USA
| | - Richard L Watson
- Revalesio Corporation, 1200 East D Street, Tacoma, WA, 98421, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA.
| |
Collapse
|
25
|
Johnson MO, Siska PJ, Contreras DC, Rathmell JC. Nutrients and the microenvironment to feed a T cell army. Semin Immunol 2016; 28:505-513. [PMID: 27712958 PMCID: PMC5154770 DOI: 10.1016/j.smim.2016.09.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/28/2016] [Accepted: 09/30/2016] [Indexed: 02/04/2023]
Abstract
T cells have dramatic functional and proliferative shifts in the course of maintaining immune protection from pathogens and cancer. To support these changes, T cells undergo metabolic reprogramming upon stimulation and again after antigen clearance. Depending on the extrinsic cell signals, T cells can differentiate into functionally distinct subsets that utilize and require diverse metabolic programs. Effector T cells (Teff) enhance glucose and glutamine uptake, whereas regulatory T cells (Treg) do not rely on significant rates of glycolysis. The dependence of these subsets on specific metabolic programs makes T cells reliant on these signaling pathways and nutrients. Metabolic pathways, such as those regulated by mTOR and Myc, augment T cell glycolysis and glutaminolysis programs to promote T cell activity. These pathways respond to signals and control metabolism through both transcriptional or post-transcriptional mechanisms. Epigenetic modifications also play an important role by stabilizing the transcription factors that define subset specific reprogramming. In addition, circadian rhythm cycling may also influence energy use, immune surveillance, and function of T cells. In this review, we focus on the metabolic and nutrient requirements of T cells, and how canonical pathways of growth and metabolism regulate nutrients that are essential for T cell function.
Collapse
Affiliation(s)
- Marc O Johnson
- Department of Pathology, Microbiology, and Immunology, and Cancer Biology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Peter J Siska
- Department of Pathology, Microbiology, and Immunology, and Cancer Biology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Diana C Contreras
- Department of Pathology, Microbiology, and Immunology, and Cancer Biology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, and Cancer Biology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States.
| |
Collapse
|
26
|
HLA-DPB1 mismatch alleles represent powerful leukemia rejection antigens in CD4 T-cell immunotherapy after allogeneic stem-cell transplantation. Leukemia 2016; 31:434-445. [DOI: 10.1038/leu.2016.210] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 06/03/2016] [Accepted: 07/04/2016] [Indexed: 12/24/2022]
|
27
|
Wang X, Chen X, Tang H, Zhu J, Zhou S, Xu Z, Liu F, Su C. Increased Frequency of Th17 Cells in Children With Mycoplasma pneumoniae Pneumonia. J Clin Lab Anal 2016; 30:1214-1219. [PMID: 27240139 DOI: 10.1002/jcla.22005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/02/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Mycoplasma pneumoniae (M. pneumoniae, MP) is recognized globally as a significant cause of primary atypical pneumonia in humans, particularly in children. Overzealous host immune responses are viewed as key mediators of the pathogenesis of M. pneumoniae infection. Although Th17 cells have been identified as key modulators in the clearance of pathogens and induction of autoimmunity caused by excessive immune responses, little is known about the role of Th17 cells in patients with M. pneumoniae infection. METHODS The percentages of T cells, CD4+ T cells and Th17 cells in children with M. pneumoniae infection were measured by flow cytometry. RESULTS We documented an increased frequency of Th17 cells in children with M. pneumoniae infection. Furthermore, we found a significantly higher percentage of Th17 cells in M. pneumoniae-infected children with extrapulmonary manifestations, compared with children without extrapulmonary manifestations. In addition, patients who experienced a short course of Mycoplasma pneumoniae pneumonia (MPP) showed an increase in the percentage of Th17 cells. CONCLUSION Our findings suggest that Th17 cells may be involved in the clearance of M. pneumoniae during an acute infection. Excessive Th17 cell responses may also contribute to the immuno-pathological damage observed during persistent infection.
Collapse
Affiliation(s)
- Xiaowei Wang
- Department of Blood Transfusion, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xiaojun Chen
- Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Heng Tang
- Department of Respiratory, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jifeng Zhu
- Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Sha Zhou
- Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhipeng Xu
- Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Feng Liu
- Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chuan Su
- Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China. ,
| |
Collapse
|
28
|
Bossowski A, Moniuszko M, Idźkowska E, Grubczak K, Singh P, Bossowska A, Diana T, Kahaly GJ. Decreased proportions of CD4 + IL17+/CD4 + CD25 + CD127- and CD4 + IL17+/CD4 + CD25 + CD127 - FoxP3+ T cells in children with autoimmune thyroid diseases (.). Autoimmunity 2016; 49:320-8. [PMID: 27206624 DOI: 10.1080/08916934.2016.1183654] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Until now, altered balance of Th1 and Th2 immune cells has been postulated to play an important role in the pathogenesis of autoimmune thyroid diseases (AITD). However, recent studies on thyroid diseases have suggested a new role for Th17 cells that have been classified as a new lineage, distinct from Th1, Th2 and Treg cells. Despite wide interest, the role of Th17 cells in the pathogenesis of inflammatory and autoimmune diseases is still debated. The aim of the study was to estimate the proportions of Th17/Treg T cells in peripheral blood from patients with Graves' disease (GD; n = 29, mean age 15.4 ± 5.1 years), Hashimoto's thyroiditis (HT; n = 39, mean age 15.2 ± 4.1 years) and in healthy controls (n = 49, mean age 14.8 ± 3 years). Polychromatic flow cytometry and several fluorochrome-conjugated monoclonal antibodies were applied to delineate Th17 and Treg cells. The analysis of Th17/Treg T cell proportions in peripheral blood from patients with Graves' disease revealed significantly lower ratios of CD4 + IL17+/CD4 + CD25 + CD127 - (p < 0.0021) and CD4 + IL17+/CD4 + CD25 + CD127 - FoxP3 + (p < 0.0031) than in the control group. In addition, in the case of HT, we observed a significant decrease in the ratios of CD4 + IL17+/CD4 + CD25 + CD127 - (p < 0.0001) and CD4 + IL17+/CD4 + CD25 + CD127 - FoxP3 + (p < 0.0001) T cells in comparison to healthy children. In patients with untreated GD, a statistically significant positive correlation was found between the proportions of CD4 + IL17+/CD4 + CD25 + CD127-, CD4 + IL17+/CD4 + CD25 + CD127 - FoxP3+ T cells and the TRAbs (R = 0.71, p < 0.029; R = 0.72, p < 0.026, respectively) and a positive correlation was noted between the percentage of CD4 + CD - IL - 17 + T cells and the level of TSAbs (R = 0.66, p < 0.037). We conclude that the changes in the proportion of Th17/Treg T cells in peripheral blood and their significant relationship with the level of anti-thyroid antibodies indicate an involvement of these cells in the pathogenesis of AITD.
Collapse
Affiliation(s)
- Artur Bossowski
- a Department of Pediatric Endocrinology , Diabetology with Cardiology Division, Medical University of Białystok , Białystok , Poland
| | - Marcin Moniuszko
- b Department of Regenerative Medicine and Immune Regulation , Medical University of Bialystok , Białystok , Poland
| | - Ewelina Idźkowska
- a Department of Pediatric Endocrinology , Diabetology with Cardiology Division, Medical University of Białystok , Białystok , Poland
| | - Kamil Grubczak
- b Department of Regenerative Medicine and Immune Regulation , Medical University of Bialystok , Białystok , Poland
| | - Paulina Singh
- b Department of Regenerative Medicine and Immune Regulation , Medical University of Bialystok , Białystok , Poland
| | - Anna Bossowska
- c Division of Cardiology , Internal Affairs Ministry Hospital in Białystok , Białystok , Poland , and
| | - Tanja Diana
- d Molecular Thyroid Research Laboratory, Johannes Gutenberg University (JGU) Medical Center , Mainz , Germany
| | - George J Kahaly
- d Molecular Thyroid Research Laboratory, Johannes Gutenberg University (JGU) Medical Center , Mainz , Germany
| |
Collapse
|
29
|
Hinshaw SJH, Ogbeifun O, Wandu WS, Lyu C, Shi G, Li Y, Qian H, Gery I. Digoxin Inhibits Induction of Experimental Autoimmune Uveitis in Mice, but Causes Severe Retinal Degeneration. Invest Ophthalmol Vis Sci 2016; 57:1441-7. [PMID: 27028065 PMCID: PMC4821074 DOI: 10.1167/iovs.15-19040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/24/2016] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Digoxin, a major medication for heart disease, was recently reported to have immunosuppressive capacity. Here, we determined the immunosuppressive capacity of digoxin on the development of experimental autoimmune uveitis (EAU) and on related immune responses. METHODS The B10.A mice were immunized with interphotoreceptor retinoid-binding protein (IRBP) and were treated daily with digoxin or vehicle control. On postimmunization day 14, the mouse eyes were examined histologically, while spleen cells were tested for cytokine production in response to IRBP and purified protein derivative. The immunosuppressive activity of digoxin was also tested in vitro, by its capacity to inhibit development of Th1 or Th17 cells. To investigate the degenerative effect of digoxin on the retina, naïve (FVB/N × B10.BR)F1 mice were similarly treated with digoxin and tested histologically and by ERG. RESULTS Treatment with digoxin inhibited the development of EAU, as well as the cellular response to IRBP. Unexpectedly, treatment with digoxin suppressed the production of interferon-γ to a larger extent than the production of interleukin 17. Importantly, digoxin treatment induced severe retinal degeneration, determined by histologic analysis with thinning across all layers of the retina. Digoxin treatment also induced dose-dependent vision loss monitored by ERG on naïve mice without induction of EAU. CONCLUSIONS Treatment of mice with digoxin inhibited the development of EAU and cellular immune response to IRBP. However, the treatment induced severe damage to the retina. Thus, the use of digoxin in humans should be avoided due to its toxicity to the retina.
Collapse
Affiliation(s)
- Samuel J. H. Hinshaw
- Laboratory of Immunology National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Osato Ogbeifun
- Laboratory of Immunology National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Wambui S. Wandu
- Laboratory of Immunology National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Cancan Lyu
- Laboratory of Immunology National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Guangpu Shi
- Laboratory of Immunology National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Yichao Li
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Igal Gery
- Laboratory of Immunology National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
30
|
Ryan ES, Micci L, Fromentin R, Paganini S, McGary CS, Easley K, Chomont N, Paiardini M. Loss of Function of Intestinal IL-17 and IL-22 Producing Cells Contributes to Inflammation and Viral Persistence in SIV-Infected Rhesus Macaques. PLoS Pathog 2016; 12:e1005412. [PMID: 26829644 PMCID: PMC4735119 DOI: 10.1371/journal.ppat.1005412] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/04/2016] [Indexed: 01/19/2023] Open
Abstract
In HIV/SIV-infected humans and rhesus macaques (RMs), a severe depletion of intestinal CD4(+) T-cells producing interleukin IL-17 and IL-22 associates with loss of mucosal integrity and chronic immune activation. However, little is known about the function of IL-17 and IL-22 producing cells during lentiviral infections. Here, we longitudinally determined the levels and functions of IL-17, IL-22 and IL-17/IL-22 producing CD4(+) T-cells in blood, lymph node and colorectum of SIV-infected RMs, as well as how they recover during effective ART and are affected by ART interruption. Intestinal IL-17 and IL-22 producing CD4(+) T-cells are polyfunctional in SIV-uninfected RMs, with the large majority of cells producing four or five cytokines. SIV infection induced a severe dysfunction of colorectal IL-17, IL-22 and IL-17/IL-22 producing CD4(+) T-cells, the extent of which associated with the levels of immune activation (HLA-DR(+)CD38(+)), proliferation (Ki-67+) and CD4(+) T-cell counts before and during ART. Additionally, Th17 cell function during ART negatively correlated with residual plasma viremia and levels of sCD163, a soluble marker of inflammation and disease progression. Furthermore, IL-17 and IL-22 producing cell frequency and function at various pre, on, and off-ART experimental points associated with and predicted total SIV-DNA content in the colorectum and blood. While ART restored Th22 cell function to levels similar to pre-infection, it did not fully restore Th17 cell function, and all cell types were rapidly and severely affected--both quantitatively and qualitatively--after ART interruption. In conclusion, intestinal IL-17 producing cell function is severely impaired by SIV infection, not fully normalized despite effective ART, and strongly associates with inflammation as well as SIV persistence off and on ART. As such, strategies able to preserve and/or regenerate the functions of these CD4(+) T-cells central for mucosal immunity are critically needed in future HIV cure research.
Collapse
Affiliation(s)
- Emily S. Ryan
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Luca Micci
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Rémi Fromentin
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Faculty of Medicine, and Centre de Recherche du CHUM, Montreal, Québec, Canada
| | - Sara Paganini
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Colleen S. McGary
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kirk Easley
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Nicolas Chomont
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Faculty of Medicine, and Centre de Recherche du CHUM, Montreal, Québec, Canada
| | - Mirko Paiardini
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
31
|
Richard AC, Tan C, Hawley ET, Gomez-Rodriguez J, Goswami R, Yang XP, Cruz AC, Penumetcha P, Hayes ET, Pelletier M, Gabay O, Walsh M, Ferdinand JR, Keane-Myers A, Choi Y, O'Shea JJ, Al-Shamkhani A, Kaplan MH, Gery I, Siegel RM, Meylan F. The TNF-family ligand TL1A and its receptor DR3 promote T cell-mediated allergic immunopathology by enhancing differentiation and pathogenicity of IL-9-producing T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:3567-82. [PMID: 25786692 PMCID: PMC5112176 DOI: 10.4049/jimmunol.1401220] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 02/09/2015] [Indexed: 11/19/2022]
Abstract
The TNF family cytokine TL1A (Tnfsf15) costimulates T cells and type 2 innate lymphocytes (ILC2) through its receptor DR3 (Tnfrsf25). DR3-deficient mice have reduced T cell accumulation at the site of inflammation and reduced ILC2-dependent immune responses in a number of models of autoimmune and allergic diseases. In allergic lung disease models, immunopathology and local Th2 and ILC2 accumulation is reduced in DR3-deficient mice despite normal systemic priming of Th2 responses and generation of T cells secreting IL-13 and IL-4, prompting the question of whether TL1A promotes the development of other T cell subsets that secrete cytokines to drive allergic disease. In this study, we find that TL1A potently promotes generation of murine T cells producing IL-9 (Th9) by signaling through DR3 in a cell-intrinsic manner. TL1A enhances Th9 differentiation through an IL-2 and STAT5-dependent mechanism, unlike the TNF-family member OX40, which promotes Th9 through IL-4 and STAT6. Th9 differentiated in the presence of TL1A are more pathogenic, and endogenous TL1A signaling through DR3 on T cells is required for maximal pathology and IL-9 production in allergic lung inflammation. Taken together, these data identify TL1A-DR3 interactions as a novel pathway that promotes Th9 differentiation and pathogenicity. TL1A may be a potential therapeutic target in diseases dependent on IL-9.
Collapse
Affiliation(s)
- Arianne C Richard
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Cuiyan Tan
- Experimental Immunology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Eric T Hawley
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Julio Gomez-Rodriguez
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ritobrata Goswami
- Department of Pediatrics and Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Xiang-Ping Yang
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Anthony C Cruz
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Pallavi Penumetcha
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Erika T Hayes
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Martin Pelletier
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Odile Gabay
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Matthew Walsh
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19102
| | - John R Ferdinand
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892; Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom; and
| | - Andrea Keane-Myers
- Biological Defense Research Directorate, Naval Medical Research Center-Frederick, Fort Detrick, MD 21702
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19102
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Aymen Al-Shamkhani
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom; and
| | - Mark H Kaplan
- Department of Pediatrics and Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Igal Gery
- Experimental Immunology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Richard M Siegel
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892;
| | - Françoise Meylan
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
32
|
Glucocorticoid-resistant Th17 cells are selectively attenuated by cyclosporine A. Proc Natl Acad Sci U S A 2015; 112:4080-5. [PMID: 25775512 DOI: 10.1073/pnas.1418316112] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Glucocorticoids remain the cornerstone of treatment for inflammatory conditions, but their utility is limited by a plethora of side effects. One of the key goals of immunotherapy across medical disciplines is to minimize patients' glucocorticoid use. Increasing evidence suggests that variations in the adaptive immune response play a critical role in defining the dose of glucocorticoids required to control an individual's disease, and Th17 cells are strong candidate drivers for nonresponsiveness [also called steroid resistance (SR)]. Here we use gene-expression profiling to further characterize the SR phenotype in T cells and show that Th17 cells generated from both SR and steroid-sensitive individuals exhibit restricted genome-wide responses to glucocorticoids in vitro, and that this is independent of glucocorticoid receptor translocation or isoform expression. In addition, we demonstrate, both in transgenic murine T cells in vitro and in an in vivo murine model of autoimmunity, that Th17 cells are reciprocally sensitive to suppression with the calcineurin inhibitor, cyclosporine A. This result was replicated in human Th17 cells in vitro, which were found to have a conversely large genome-wide shift in response to cyclosporine A. These observations suggest that the clinical efficacy of cyclosporine A in the treatment of SR diseases may be because of its selective attenuation of Th17 cells, and also that novel therapeutics, which target either Th17 cells themselves or the effector memory T-helper cell population from which they are derived, would be strong candidates for drug development in the context of SR inflammation.
Collapse
|
33
|
Christie D, Zhu J. Transcriptional regulatory networks for CD4 T cell differentiation. Curr Top Microbiol Immunol 2015; 381:125-72. [PMID: 24839135 DOI: 10.1007/82_2014_372] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD4(+) T cells play a central role in controlling the adaptive immune response by secreting cytokines to activate target cells. Naïve CD4(+) T cells differentiate into at least four subsets, Th1Th1 , Th2Th2 , Th17Th17 , and inducible regulatory T cellsregulatory T cells , each with unique functions for pathogen elimination. The differentiation of these subsets is induced in response to cytokine stimulation, which is translated into Stat activation, followed by induction of master regulator transcription factorstranscription factors . In addition to these factors, multiple other transcription factors, both subset specific and shared, are also involved in promoting subset differentiation. This review will focus on the network of transcription factors that control CD4(+) T cell differentiation.
Collapse
Affiliation(s)
- Darah Christie
- Molecular and Cellular Immunoregulation Unit, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA,
| | | |
Collapse
|
34
|
Cinnamon ameliorates experimental allergic encephalomyelitis in mice via regulatory T cells: implications for multiple sclerosis therapy. PLoS One 2015; 10:e0116566. [PMID: 25569428 PMCID: PMC4287621 DOI: 10.1371/journal.pone.0116566] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 12/11/2014] [Indexed: 12/11/2022] Open
Abstract
Upregulation and/or maintenance of regulatory T cells (Tregs) during an autoimmune insult may have therapeutic efficacy in autoimmune diseases. Although several immunomodulatory drugs and molecules are available, most present significant side effects over long-term use. Cinnamon is a commonly used natural spice and flavoring material used for centuries throughout the world. Here, we have explored a novel use of cinnamon powder in protecting Tregs and treating the disease process of experimental allergic encephalomyelitis (EAE), an animal model of MS. Oral feeding of cinnamon (Cinnamonum verum) powder suppresses clinical symptoms of relapsing-remitting EAE in female PLP-TCR transgenic mice and adoptive transfer mouse model. Cinnamon also inhibited clinical symptoms of chronic EAE in male C57/BL6 mice. Dose-dependent study shows that cinnamon powder at a dose of 50 mg/kg body wt/d or higher significantly suppresses clinical symptoms of EAE in mice. Accordingly, oral administration of cinnamon also inhibited perivascular cuffing, maintained the integrity of blood-brain barrier and blood-spinal cord barrier, suppressed inflammation, normalized the expression of myelin genes, and blocked demyelination in the central nervous system of EAE mice. Interestingly, cinnamon treatment upregulated Tregs via reduction of nitric oxide production. Furthermore, we demonstrate that blocking of Tregs by neutralizing antibodies against CD25 abrogates cinnamon-mediated protection of EAE. Taken together, our results suggest that oral administration of cinnamon powder may be beneficial in MS patients and that no other existing anti-MS therapies could be so economical and trouble-free as this approach.
Collapse
|
35
|
A systems model of phosphorylation for inflammatory signaling events. PLoS One 2014; 9:e110913. [PMID: 25333362 PMCID: PMC4205014 DOI: 10.1371/journal.pone.0110913] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/19/2014] [Indexed: 12/24/2022] Open
Abstract
Phosphorylation is a fundamental biochemical reaction that modulates protein activity in cells. While a single phosphorylation event is relatively easy to understand, multisite phosphorylation requires systems approaches for deeper elucidation of the underlying molecular mechanisms. In this paper we develop a mechanistic model for single- and multi-site phosphorylation. The proposed model is compared with previously reported studies. We compare the predictions of our model with experiments published in the literature in the context of inflammatory signaling events in order to provide a mechanistic description of the multisite phosphorylation-mediated regulation of Signal Transducer and Activator of Transcription 3 (STAT3) and Interferon Regulatory Factor 5 (IRF-5) proteins. The presented model makes crucial predictions for transcription factor phosphorylation events in the immune system. The model proposes potential mechanisms for T cell phenotype switching and production of cytokines. This study also provides a generic framework for the better understanding of a large number of multisite phosphorylation-regulated biochemical circuits.
Collapse
|
36
|
Aktas Cetin E, Cosan F, Cefle A, Deniz G. IL-22-secreting Th22 and IFN-γ-secreting Th17 cells in Behçet's disease. Mod Rheumatol 2014; 24:802-7. [PMID: 24498963 DOI: 10.3109/14397595.2013.879414] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Behçet's disease (BD) is a systemic inflammatory disease with unknown etiology. Studies have shown that some T helper (Th) 1-associated cytokines have role in the inflammation of BD. The CD4(+) Th cells can be differentiated into Th1, Th2, Th17 and Th22 secrete different cytokines to regulate immune system. In this study, cytokine secretion of Th subsets in BD was investigated. METHODS The study group consisted of 26 BD patients with mucocutaneous involvement and 12 healthy subjects. Lymphocyte subpopulations, IL-5, IL-10, IL-17, IL-22 and IFN-γ secretion of CD4(+) T and Foxp3(+) Treg cells were determined by flow cytometry. RESULTS Compared with healthy subjects, Th1 (IL-17A(-)IL-22(-)IFN-γ(+)), Th22 (IL-17A(-)IL-22(+)IFN-γ) and IL-17A(+)IFN-γ(+)-secreting cells were significantly increased, and the percentage of Treg cells were dramatically reduced in BD patients. The frequency of recurrent oral ulcers was associated with increased Th22 cells. CONCLUSIONS Our study describes an association between Th22 cell subset and IL-17A(+) IFNγ(+)-secreting cells with mucocutaneous BD. These findings revealed that reduced levels of Tregs and increased levels of Th1 and Th22 cells as well as Th17/Th1 cells might be associated with the pathogenesis of BD.
Collapse
Affiliation(s)
- Esin Aktas Cetin
- Department of Immunology, Istanbul University, Institute of Experimental Medicine (DETAE) , Istanbul , Turkey
| | | | | | | |
Collapse
|
37
|
Rebhahn JA, Deng N, Sharma G, Livingstone AM, Huang S, Mosmann TR. An animated landscape representation of CD4+ T-cell differentiation, variability, and plasticity: insights into the behavior of populations versus cells. Eur J Immunol 2014; 44:2216-29. [PMID: 24945794 PMCID: PMC4209377 DOI: 10.1002/eji.201444645] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 12/12/2022]
Abstract
Recent advances in understanding CD4(+) T-cell differentiation suggest that previous models of a few distinct, stable effector phenotypes were too simplistic. Although several well-characterized phenotypes are still recognized, some states display plasticity, and intermediate phenotypes exist. As a framework for reexamining these concepts, we use Waddington's landscape paradigm, augmented with explicit consideration of stochastic variations. Our animation program "LAVA" visualizes T-cell differentiation as cells moving across a landscape of hills and valleys, leading to attractor basins representing stable or semistable differentiation states. The model illustrates several principles, including: (i) cell populations may behave more predictably than individual cells; (ii) analogous to reticulate evolution, differentiation may proceed through a network of interconnected states, rather than a single well-defined pathway; (iii) relatively minor changes in the barriers between attractor basins can change the stability or plasticity of a population; (iv) intrapopulation variability of gene expression may be an important regulator of differentiation, rather than inconsequential noise; (v) the behavior of some populations may be defined mainly by the behavior of outlier cells. While not a quantitative representation of actual differentiation, our model is intended to provoke discussion of T-cell differentiation pathways, particularly highlighting a probabilistic view of transitions between states.
Collapse
Affiliation(s)
- Jonathan A Rebhahn
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical SchoolRochester, NY, USA
| | - Nan Deng
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical SchoolRochester, NY, USA
| | - Gaurav Sharma
- Department of Electrical and Computer Engineering, University of RochesterRochester, NY, USA
| | - Alexandra M Livingstone
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical SchoolRochester, NY, USA
| | - Sui Huang
- Institute for Systems BiologySeattle, WA, USA
| | - Tim R Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical SchoolRochester, NY, USA
| |
Collapse
|
38
|
Mony JT, Khorooshi R, Owens T. Chemokine receptor expression by inflammatory T cells in EAE. Front Cell Neurosci 2014; 8:187. [PMID: 25071447 PMCID: PMC4081975 DOI: 10.3389/fncel.2014.00187] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/19/2014] [Indexed: 11/13/2022] Open
Abstract
Chemokines direct cellular infiltration to tissues, and their receptors and signaling pathways represent targets for therapy in diseases such as multiple sclerosis (MS). The chemokine CCL20 is expressed in choroid plexus, a site of entry of T cells to the central nervous system (CNS). The CCL20 receptor CCR6 has been reported to be selectively expressed by CD4(+) T cells that produce the cytokine IL-17 (Th17 cells). Th17 cells and interferon-gamma (IFNγ)-producing Th1 cells are implicated in induction of MS and its animal model experimental autoimmune encephalomyelitis (EAE). We have assessed whether CCR6 identifies specific inflammatory T cell subsets in EAE. Our approach was to induce EAE, and then examine chemokine receptor expression by cytokine-producing T cells sorted from CNS at peak disease. About 7% of CNS-infiltrating CD4(+) T cells produced IFNγ in flow cytometric cytokine assays, whereas less than 1% produced IL-17. About 1% of CD4(+) T cells produced both cytokines. CCR6 was expressed by Th1, Th1+17 and by Th17 cells, but not by CD8(+) T cells. CD8(+) T cells expressed CXCR3, which was also expressed by CD4(+) T cells, with no correlation to cytokine profile. Messenger RNA for IFNγ, IL-17A, and the Th1 and Th17-associated transcription factors T-bet and RORγt was detected in both CCR6(+) and CXCR3(+) CD4(+) T cells. IFNγ, but not IL-17A mRNA expression was detected in CD8(+) T cells in CNS. CCR6 and CD4 were co-localized in spinal cord infiltrates by double immunofluorescence. Consistent with flow cytometry data some but not all CD4(+) T cells expressed CCR6 within infiltrates. CD4-negative CCR6(+) cells included macrophage/microglial cells. Thus we have for the first time directly studied CD4(+) and CD8(+) T cells in the CNS of mice with peak EAE, and determined IFNγ and IL17 expression by cells expressing CCR6 and CXCR3. We show that neither CCR6 or CXCR3 align with CD4 T cell subsets, and Th1 or mixed Th1+17 predominate in EAE.
Collapse
Affiliation(s)
- Jyothi Thyagabhavan Mony
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Reza Khorooshi
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Trevor Owens
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| |
Collapse
|
39
|
Zhao C, Shi G, Vistica BP, Hinshaw SJH, Wandu WS, Tan C, Zhang M, Gery I. Induced regulatory T-cells (iTregs) generated by activation with anti-CD3/CD28 antibodies differ from those generated by the physiological-like activation with antigen/APC. Cell Immunol 2014; 290:179-84. [PMID: 25038545 DOI: 10.1016/j.cellimm.2014.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 03/31/2014] [Accepted: 06/10/2014] [Indexed: 11/29/2022]
Abstract
Regulatory T-cells (Tregs) are responsible for homeostasis of the immune system, as well as for inhibition of pathogenic autoimmune processes. Induced-(i)-Tregs, can be generated in vitro by activation of CD4 cells in the presence of TGF-β. A commonly used activation mechanism is by antibodies against CD3 and CD28. The physiological-like activation of T-cells, however, is with the specific target antigen presented by antigen-presenting cells (APC). The two modes of activation have been considered to yield the same populations of iTregs. Here, we compared between iTreg populations generated by either one of the two methods and found differences between their capacities to inhibit T-lymphocyte proliferative response, their expression of cell surface antigens and particularly, in their transcript expression profiles of certain chemokines and chemokine receptors. Our data thus indicate that iTregs generated by activation with anti-CD3/CD28 antibodies cannot be considered identical to iTregs generated by antigen/APC.
Collapse
Affiliation(s)
- Chan Zhao
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States; Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guangpu Shi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Barbara P Vistica
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Samuel J H Hinshaw
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Wambui S Wandu
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cuiyan Tan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Meifen Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Igal Gery
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
40
|
Fu J, Heinrichs J, Yu XZ. Helper T-cell differentiation in graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Arch Immunol Ther Exp (Warsz) 2014; 62:277-301. [PMID: 24699629 DOI: 10.1007/s00005-014-0284-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 01/27/2014] [Indexed: 02/07/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an effective therapeutic option for many malignant diseases. However, the efficacy of allo-HSCT is limited by the occurrence of destructive graft-versus-host disease (GVHD). Since allogeneic T cells are the driving force in the development of GVHD, their activation, proliferation, and differentiation are key factors to understanding GVHD pathogenesis. This review focuses on one critical aspect: the differentiation and function of helper T (Th) cells in acute GVHD. We first summarize well-established subsets including Th1, Th2, Th17, and T-regulatory cells; their flexibility, plasticity, and epigenetic modification; and newly identified subsets including Th9, Th22, and T follicular helper cells. Next, we extensively discuss preclinical findings of Th-cell lineages in GVHD: the networks of transcription factors involved in differentiation, the cytokine and signaling requirements for development, the reciprocal differentiation features, and the regulation of microRNAs on T-cell differentiation. Finally, we briefly summarize the recent findings on the roles of T-cell subsets in clinical GVHD and ongoing strategies to modify T-cell differentiation for controlling GVHD in patients. We believe further exploration and understanding of the immunobiology of T-cell differentiation in GVHD will expand therapeutic options for the continuing success of allo-HSCT.
Collapse
Affiliation(s)
- Jianing Fu
- Cancer Biology PhD Program, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL, 33612, USA
| | | | | |
Collapse
|
41
|
Peters A, Yosef N. Understanding Th17 cells through systematic genomic analyses. Curr Opin Immunol 2014; 28:42-8. [PMID: 24594517 DOI: 10.1016/j.coi.2014.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 12/12/2022]
Abstract
Th17 cells are a subset of CD4+ T helper cells that play an important role in host defense and have been strongly associated with the pathogenesis of autoimmunity. The immense research effort on Th17 cells has benefited in recent years from major breakthroughs in genomic profiling technologies. The picture emerging from these studies has led us away from thinking about T cell differentiation in terms of rigid, separate pathways that give rise to canonical 'types'. Instead, it has encouraged us to understand T cell differentiation and function through a complex network of transcriptional regulators that can lead to different and potentially plastic cell states. This review summarizes some of the lessons we have learned from these studies about the identity, interplay, and function of the factors that are involved in Th17 cell differentiation, effector functions and plasticity. It also highlights some applications, challenges, and limitations of large-scale systematic analyses of T cell function.
Collapse
Affiliation(s)
- Anneli Peters
- Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Nir Yosef
- Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States; Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, United States.
| |
Collapse
|
42
|
Tan C, Wei L, Vistica BP, Shi G, Wawrousek EF, Gery I. Phenotypes of Th lineages generated by the commonly used activation with anti-CD3/CD28 antibodies differ from those generated by the physiological activation with the specific antigen. Cell Mol Immunol 2014; 11:305-13. [PMID: 24583715 DOI: 10.1038/cmi.2014.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 01/02/2014] [Accepted: 01/20/2014] [Indexed: 12/29/2022] Open
Abstract
T-helper (Th) lineages have been generated in vitro by activating CD4 cells with anti-CD3/CD28 antibodies during polarization. Physiologically, however, the generation of Th lineages is by activation with the specific antigen presented by antigen-presenting cells (APC). Here, we used T-cell receptor (TCR)-transgenic mice to compare the phenotypes of Th1, Th9 and Th17 lineages when generated by either one of the two activation modes. Lineage Th cells specific against hen egg lysozyme (HEL), were adoptively transferred into recipient mice transgenically expressing HEL in their lens. Remarkable differences were found between lineages of Th1, Th9 or Th17, generated by either one of the two modes in their capacities to migrate to and proliferate in the recipient spleen and, importantly, to induce inflammation in the recipient mouse eyes. Substantial differences were also observed between the lineage pairs in their transcript expression profiles of certain chemokines and chemokine receptors. Surprisingly, however, close similarities were observed between the transcript expression profiles of lineages of the three phenotypes, activated by the same mode. Furthermore, Th cell lineages generated by the two activation modes differed considerably in their pattern of gene expression, as monitored by microarray analysis, but exhibited commonality with lineages of other phenotypes generated by the same activation mode. This study thus shows that (i) Th lineages generated by activation with anti-CD3/CD28 antibodies differ from lineages generated by antigen/APC; and (ii) the mode of activation determines to a large extent the expression profile of major transcripts.
Collapse
Affiliation(s)
- Cuiyan Tan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lai Wei
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Barbara P Vistica
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Guangpu Shi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eric F Wawrousek
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Igal Gery
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Moraes-Vieira PMM, Larocca RA, Bassi EJ, Peron JPS, Andrade-Oliveira V, Wasinski F, Araujo R, Thornley T, Quintana FJ, Basso AS, Strom TB, Câmara NOS. Leptin deficiency impairs maturation of dendritic cells and enhances induction of regulatory T and Th17 cells. Eur J Immunol 2014; 44:794-806. [PMID: 24271843 DOI: 10.1002/eji.201343592] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 11/08/2013] [Accepted: 11/20/2013] [Indexed: 01/09/2023]
Abstract
Leptin is an adipose-secreted hormone that plays an important role in both metabolism and immunity. Leptin has been shown to induce Th1-cell polarization and inhibit Th2-cell responses. Additionally, leptin induces Th17-cell responses, inhibits regulatory T (Treg) cells and modulates autoimmune diseases. Here, we investigated whether leptin mediates its activity on T cells by influencing dendritic cells (DCs) to promote Th17 and Treg-cell immune responses in mice. We observed that leptin deficiency (i) reduced the expression of DC maturation markers, (ii) decreased DC production of IL-12, TNF-α, and IL-6, (iii) increased DC production of TGF-β, and (iv) limited the capacity of DCs to induce syngeneic CD4(+) T-cell proliferation. As a consequence of this unique phenotype, DCs generated under leptin-free conditions induced Treg or TH 17 cells more efficiently than DCs generated in the presence of leptin. These data indicate important roles for leptin in DC homeostasis and the initiation and maintenance of inflammatory and regulatory immune responses by DCs.
Collapse
Affiliation(s)
- Pedro M M Moraes-Vieira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Department of Medicine, Harvard Medical School, Beth Israel Medical Deaconess Center, Transplant Institute, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gökmen MR, Dong R, Kanhere A, Powell N, Perucha E, Jackson I, Howard JK, Hernandez-Fuentes M, Jenner RG, Lord GM. Genome-wide regulatory analysis reveals that T-bet controls Th17 lineage differentiation through direct suppression of IRF4. THE JOURNAL OF IMMUNOLOGY 2013; 191:5925-32. [PMID: 24249732 DOI: 10.4049/jimmunol.1202254] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The complex relationship between Th1 and Th17 cells is incompletely understood. The transcription factor T-bet is best known as the master regulator of Th1 lineage commitment. However, attention is now focused on the repression of alternate T cell subsets mediated by T-bet, particularly the Th17 lineage. It has recently been suggested that pathogenic Th17 cells express T-bet and are dependent on IL-23. However, T-bet has previously been shown to be a negative regulator of Th17 cells. We have taken an unbiased approach to determine the functional impact of T-bet on Th17 lineage commitment. Genome-wide analysis of functional T-bet binding sites provides an improved understanding of the transcriptional regulation mediated by T-bet, and suggests novel mechanisms by which T-bet regulates Th cell differentiation. Specifically, we show that T-bet negatively regulates Th17 lineage commitment via direct repression of the transcription factor IFN regulatory factor-4 (IRF4). An in vivo analysis of the pathogenicity of T-bet-deficient T cells demonstrated that mucosal Th17 responses were augmented in the absence of T-bet, and we have demonstrated that the roles of T-bet in enforcing Th1 responses and suppressing Th17 responses are separable. The interplay of the two key transcription factors T-bet and IRF4 during the determination of T cell fate choice significantly advances our understanding of the mechanisms underlying the development of pathogenic T cells.
Collapse
Affiliation(s)
- M Refik Gökmen
- Department of Experimental Immunobiology, Division of Transplantation Immunology and Mucosal Biology and Medical Research Council Centre for Transplantation, Guy's Campus, King's College London, London SE1 9RT, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bharadwaj AS, Schewitz-Bowers LP, Wei L, Lee RWJ, Smith JR. Intercellular adhesion molecule 1 mediates migration of Th1 and Th17 cells across human retinal vascular endothelium. Invest Ophthalmol Vis Sci 2013; 54:6917-25. [PMID: 24022011 DOI: 10.1167/iovs.13-12058] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
PURPOSE Autoimmune inflammation of the retina causes vision loss in the majority of affected individuals. Th1 or Th17 cells initiate the disease on trafficking from the circulation into the eye across the retinal vascular endothelium. We investigated the ability of human Th1- and Th17-polarized cells to cross a simulated human retinal endothelium, and examined the role of IgG superfamily members in this process. METHODS Th1- and Th17-polarized cell populations were generated from human peripheral blood CD4(+) T cells, using two Th1- and Th17-polarizing protocols. Transendothelial migration assays were performed over 18 hours in Boyden chambers, after seeding the transwell membrane with human retinal endothelial cells. In some assays intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), or activated leukocyte cell adhesion molecule (ALCAM) blocking antibody, or isotype- and concentration-matched control antibody, was added to the upper chambers. RESULTS Th1- and Th17-polarized cells migrated equally efficiently across the human retinal endothelial monolayer. The percentage of IL-17(+) IFN-γ(-) Th17-polarized cells was reduced following migration. Blocking ICAM-1, but not VCAM-1 or ALCAM, significantly reduced migration of Th1- and Th17-polarized cells for a majority of human donors. CONCLUSIONS Taken in the context of other literature on transendothelial migration, our results illustrate the importance of investigating the specific tissue and vascular endothelium when considering helper T cell migration in autoimmune inflammation. Our findings further indicate that while generalizations about involvement of specific adhesion molecules in uveitis and other autoimmune disease may be possible, these may not apply to individual patients universally. The observations are relevant to the use of adhesion blockade for therapeutic purposes.
Collapse
Affiliation(s)
- Arpita S Bharadwaj
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | | | | | | | | |
Collapse
|
46
|
Shi G, Vistica BP, Nugent LF, Tan C, Wawrousek EF, Klinman DM, Gery I. Differential involvement of Th1 and Th17 in pathogenic autoimmune processes triggered by different TLR ligands. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:415-23. [PMID: 23720812 PMCID: PMC3749791 DOI: 10.4049/jimmunol.1201732] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The interaction between TLRs and their cognate ligands triggers both the innate and adaptive immune systems, and thus can play a pivotal role in the defense against pathogen invasion. This work investigates the differentiation of naive CD4 cells into Th1 or Th17 phenotypes in mice treated with different TLR ligands. We use a model system in which naive transgenic cells specific to hen egg lysozyme are adoptively transferred into recipients that express hen egg lysozyme in the lens of the eye. The transferred naive T cells induce ocular inflammation only in recipients treated with TLR ligands. Treatment with LPS preferentially stimulated IL-17 production, whereas CpG oligodeoxynucleotide and polyinosinic:polycytidylic acid primarily stimulated Th1 cells. Peptidoglycan stimulated the two Th subpopulations equally. The preferential induction of Th1 or Th17 by the four ligands was detected in the spleen (where a major portion of the adoptively transferred cells homed) and in the eyes, where activated Th cells initiate inflammation. Analysis of the cytokines present in recipient mice suggests that Th1 induction is elicited by IL-12 and/or IFN-α, whereas Th17 generation is preferentially mediated by IL-6. Importantly, we show in this article that treatment with LPS selectively promoted in the recipient mice the generation of IL-6-producing activated B cells. An inverse correlation was found between the level of regulatory T cells and severity of inflammation induced by the donor cells. Taken together, our data show that specific TLR ligands differentially activate the immune system as evidenced by the generation of distinct Th phenotypes from naive CD4 cells.
Collapse
Affiliation(s)
- Guangpu Shi
- Laboratory of Immunology, NIH, Bethesda, MD 20892
| | | | | | - Cuiyan Tan
- Laboratory of Immunology, NIH, Bethesda, MD 20892
| | - Eric F. Wawrousek
- Laboratory of Molecular and Developmental Biology of the National Eye Institute, NIH, Bethesda, MD 20892
| | - Dennis M. Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21702
| | - Igal Gery
- Laboratory of Immunology, NIH, Bethesda, MD 20892
| |
Collapse
|
47
|
Differential effects of activated human renal epithelial cells on T-cell migration. PLoS One 2013; 8:e64916. [PMID: 23717673 PMCID: PMC3661561 DOI: 10.1371/journal.pone.0064916] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 04/22/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Renal tubular epithelial cells (TECs) are one of the main targets of inflammatory insults during interstitial nephritis and kidney transplant rejection. While Th1 cells are know to be essential in the pathogenesis of rejection, the role of Th17 is still under debate. We hypothesize that TECs modulate the outcome of rejection process by production of distinct chemokines and cytokines that determine the attraction of different T-cell subsets. Therefore, we studied differential effects of activated human renal epithelial cells on T-cell migration. METHODS Human primary TECs were stimulated by IFN-γ and TNF-α in vitro. Chemokines and cytokines produced by activated TECs were measured using Luminex or ELISA. Chemotaxis assay was performed using activated peripheral blood mononuclear cells composed of CD4+CXCR3+ and CD4+CCR6+ T cells migrating towards stimulated and unstimulated TECs. RESULTS While activated TECs secreted abundant amounts of the pro-inflammatory cytokines IL-6 and IL-8, the T helper cell differentiation cytokines IL-1β, IL-12p70, IL-23 or TGF-β1 were not produced. The production of Th1 chemokines CXCL9, CXCL10 and CCL5 were significantly upregulated after TEC stimulation. In contrast, Th17 chemokine CCL20 could not be detected. Finally, activated TECs attracted significantly higher numbers of CD4+CXCR3+ T cells as compared to unstimulated TECs. No migration of CD4+CCR6+ T cells could be observed. CONCLUSION Activated primary renal tubular epithelial cells do not attract Th17 cells nor produce cytokines promoting Th17 cell differentiation in our experimental system mimicking the proinflammatory microenvironment of rejection.
Collapse
|
48
|
Hirahara K, Poholek A, Vahedi G, Laurence A, Kanno Y, Milner JD, O’Shea JJ. Mechanisms underlying helper T-cell plasticity: implications for immune-mediated disease. J Allergy Clin Immunol 2013; 131:1276-87. [PMID: 23622118 PMCID: PMC3677748 DOI: 10.1016/j.jaci.2013.03.015] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 03/11/2013] [Accepted: 03/18/2013] [Indexed: 12/13/2022]
Abstract
CD4 helper T cells are critical for proper immune cell homeostasis and host defense but are also major contributors to immune and inflammatory disease. Arising from a simple biphasic model of differentiation (ie, TH1 and TH2 cells). A bewildering number of fates seem possible for helper T cells. To what extent different helper cell subsets maintain their characteristic gene expression profiles or exhibit functional plasticity is a hotly debated topic. In this review we will discuss how the expression of "signature cytokines" and "master regulator" transcription factors do not neatly conform to a simple helper T-cell paradigm. Although this might seem confusing, the good news is that the newly recognized complexity fits better with our understanding of immunopathogenesis. Finally, we will discuss factors, including epigenetic regulation and metabolic alterations, that contribute to helper cell specificity and plasticity.
Collapse
Affiliation(s)
- Kiyoshi Hirahara
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases
| | - Amanda Poholek
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases
| | - Golnaz Vahedi
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases
| | - Arian Laurence
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases
| | - Yuka Kanno
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases
| | - Joshua D. Milner
- Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John J. O’Shea
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases
| |
Collapse
|
49
|
Vahedi G, Poholek A, Hand TW, Laurence A, Kann Y, O’Shea JJ, Hirahara K. Helper T-cell identity and evolution of differential transcriptomes and epigenomes. Immunol Rev 2013; 252:24-40. [PMID: 23405893 PMCID: PMC3577092 DOI: 10.1111/imr.12037] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD4(+) T cells are critical for the elimination of an immense array of microbial pathogens. Among the ways they accomplish this task is to generate progeny with specialized, characteristic patterns of gene expression. From this perspective, helper cells can be viewed as pluripotent precursors that adopt distinct cell fates. Although there are aspects of helper cell differentiation that can be modeled as a classic cell fate commitment, CD4(+) T cells also maintain considerable flexibility in their transcriptional program. This makes sense in terms of host defense, but raises the question of how these remarkable cells balance both these requirements, a high degree of specific gene expression and the capacity for plasticity. In this review, we discuss recent advances in our understanding of CD4(+) T-cell specification, focusing on how genomic perspectives have influenced our views of these processes. The relative contributions of sensors of the cytokine milieu, especially the signal transducer and activator of transcription family transcription factors, 'master regulators', and other transcription factors are considered as they relate to the helper cell transcriptome and epigenome.
Collapse
Affiliation(s)
- Golnaz Vahedi
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - Amanda Poholek
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - Timothy W. Hand
- Laboratory of parasitic diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arian Laurence
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - Yuka Kann
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - John J. O’Shea
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - Kiyoshi Hirahara
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| |
Collapse
|
50
|
Bharadwaj AS, Appukuttan B, Wilmarth PA, Pan Y, Stempel AJ, Chipps TJ, Benedetti EE, Zamora DO, Choi D, David LL, Smith JR. Role of the retinal vascular endothelial cell in ocular disease. Prog Retin Eye Res 2013; 32:102-80. [PMID: 22982179 PMCID: PMC3679193 DOI: 10.1016/j.preteyeres.2012.08.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 08/23/2012] [Accepted: 08/24/2012] [Indexed: 12/14/2022]
Abstract
Retinal endothelial cells line the arborizing microvasculature that supplies and drains the neural retina. The anatomical and physiological characteristics of these endothelial cells are consistent with nutritional requirements and protection of a tissue critical to vision. On the one hand, the endothelium must ensure the supply of oxygen and other nutrients to the metabolically active retina, and allow access to circulating cells that maintain the vasculature or survey the retina for the presence of potential pathogens. On the other hand, the endothelium contributes to the blood-retinal barrier that protects the retina by excluding circulating molecular toxins, microorganisms, and pro-inflammatory leukocytes. Features required to fulfill these functions may also predispose to disease processes, such as retinal vascular leakage and neovascularization, and trafficking of microbes and inflammatory cells. Thus, the retinal endothelial cell is a key participant in retinal ischemic vasculopathies that include diabetic retinopathy and retinopathy of prematurity, and retinal inflammation or infection, as occurs in posterior uveitis. Using gene expression and proteomic profiling, it has been possible to explore the molecular phenotype of the human retinal endothelial cell and contribute to understanding of the pathogenesis of these diseases. In addition to providing support for the involvement of well-characterized endothelial molecules, profiling has the power to identify new players in retinal pathologies. Findings may have implications for the design of new biological therapies. Additional progress in this field is anticipated as other technologies, including epigenetic profiling methods, whole transcriptome shotgun sequencing, and metabolomics, are used to study the human retinal endothelial cell.
Collapse
Affiliation(s)
| | | | - Phillip A. Wilmarth
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University
| | - Yuzhen Pan
- Casey Eye Institute, Oregon Health & Science University
| | | | | | | | | | - Dongseok Choi
- Department of Public Health and Preventive Medicine, Oregon Health & Science University
| | - Larry L. David
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University
| | - Justine R. Smith
- Casey Eye Institute, Oregon Health & Science University
- Department of Cell & Developmental Biology, Oregon Health & Science University
| |
Collapse
|