1
|
McTaggart T, Lim JX, Smith KJ, Heaney B, McDonald D, Hulme G, Hussain R, Coxhead J, Degnan AE, Isaacs J, Pratt A, Amarnath S. Deep phenotyping of T regulatory cells in psoriatic arthritis highlights targetable mechanisms of disease. J Biol Chem 2024; 301:108059. [PMID: 39662827 DOI: 10.1016/j.jbc.2024.108059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/08/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Regulatory T cells (Tregs) are immune regulatory T cells that are vital for controlling inflammation. The role of Tregs in inflammatory diseases namely psoriatic arthritis (PsA) is still poorly understood. The underlying reason being a lack of robust unbiased analysis to test the immune regulatory phenotype of human Tregs. Here, we propose that checkpoint receptors can identify functional Tregs in PsA. Using unbiased BD Rhapsody single-cell analysis, we have analyzed the expression pattern of checkpoint receptors in Tregs and found that PsA Tregs are enriched in the expression of CTLA4, TIGIT, PD-1, and GITR while TIM3 was downregulated. Furthermore, PD-1+ Tregs in PsA had an increased type 1 phenotype and expressed the protease asparaginyl endopeptidase. By harnessing the PD-1 signaling pathway and inhibiting asparaginyl endopeptidase, PsA Treg function was significantly enhanced in in vitro suppressor assays. Next, we interrogated the cell interaction pathways of Tregs in PsA and found a diminished crosstalk with circulating osteoclast precursors through the CD244-CD48 coreceptor pathways. Therapeutically, PsA Treg function could be enhanced by modulating PD-1 and osteoclast interactions. Our study suggests that unconventional immune cell crosstalk with Tregs is severely diminished in PsA.
Collapse
Affiliation(s)
- Tegan McTaggart
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK; NIHR, Biomedical Research Centre, Newcastle Upon Tyne, UK
| | - Jing Xuan Lim
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK; NIHR, Biomedical Research Centre, Newcastle Upon Tyne, UK
| | - Katie J Smith
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK; NIHR, Biomedical Research Centre, Newcastle Upon Tyne, UK
| | - Bronagh Heaney
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK; NIHR, Biomedical Research Centre, Newcastle Upon Tyne, UK
| | - David McDonald
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Gillian Hulme
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Rafiqul Hussain
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Jonathan Coxhead
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Abbie Ea Degnan
- Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle Upon Tyne, UK; Department of Rheumatology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - John Isaacs
- Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle Upon Tyne, UK; Department of Rheumatology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Arthur Pratt
- Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle Upon Tyne, UK; Department of Rheumatology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Shoba Amarnath
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK; NIHR, Biomedical Research Centre, Newcastle Upon Tyne, UK.
| |
Collapse
|
2
|
Sumida TS, Cheru NT, Hafler DA. The regulation and differentiation of regulatory T cells and their dysfunction in autoimmune diseases. Nat Rev Immunol 2024; 24:503-517. [PMID: 38374298 PMCID: PMC11216899 DOI: 10.1038/s41577-024-00994-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2024] [Indexed: 02/21/2024]
Abstract
The discovery of FOXP3+ regulatory T (Treg) cells as a distinct cell lineage with a central role in regulating immune responses provided a deeper understanding of self-tolerance. The transcription factor FOXP3 serves a key role in Treg cell lineage determination and maintenance, but is not sufficient to enable the full potential of Treg cell suppression, indicating that other factors orchestrate the fine-tuning of Treg cell function. Moreover, FOXP3-independent mechanisms have recently been shown to contribute to Treg cell dysfunction. FOXP3 mutations in humans cause lethal fulminant systemic autoinflammation (IPEX syndrome). However, it remains unclear to what degree Treg cell dysfunction is contributing to the pathophysiology of common autoimmune diseases. In this Review, we discuss the origins of Treg cells in the periphery and the multilayered mechanisms by which Treg cells are induced, as well as the FOXP3-dependent and FOXP3-independent cellular programmes that maintain the suppressive function of Treg cells in humans and mice. Further, we examine evidence for Treg cell dysfunction in the context of common autoimmune diseases such as multiple sclerosis, inflammatory bowel disease, systemic lupus erythematosus and rheumatoid arthritis.
Collapse
Affiliation(s)
- Tomokazu S Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| | - Nardos T Cheru
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Sun D, Wang K, Chen Y, Zhang B, Tang J, Luo W, Liu J, Yu S. Immunological characteristics of CD103 +CD161 + T lymphocytes on chronic rhinosinusitis with nasal polyps. Cell Immunol 2024; 401-402:104842. [PMID: 38897020 DOI: 10.1016/j.cellimm.2024.104842] [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: 03/25/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNPs) is a heterogeneous disease characterized by local inflammation of the upper airway and sinus mucosa. T cell-mediated immune responses play irreplaceable roles in the pathogenesis of nasal polyps. CD161+ T cells have been implicated in the pathology of several diseases through cytokine production and cytotoxic activity. However, the immunological characteristics of CD161+ T cells in nasal mucosa are still not well understood, particularly in CRSwNPs. Our research revealed a notable enrichment of CD161+ T cells in nasal tissues compared to peripheral blood, with a significantly more infiltration of CD161+ T cells in CRSwNPs compared to control nasal samples. Phenotypical analysis found that CD161+ T cells predominantly co-expressed tissue-resident memory surface markers CD103, CD69, and CD45RO. CD161+CD103+ T cells demonstrated complicated effector functions, marked by elevated levels of PD-1, CTLA-4, IL-17, and IFN-γ and diminished expression of FoxP3 and CD25. Interestingly, despite CD161+ T cells was more abundant in polyp tissues compared to normal control tissues, and then further categorizing polyp samples into distinct groups based on clinical characteristics, only the recurrent CRSwNP group showed a significant reduction in CD161+CD8+ T cells compared to the primary CRSwNP group. This finding suggested the necessity for further research to comprehensively understand the underlying mechanisms and the broader significance of CD161+ T cells in the advancement and relapse of CRSwNPs.
Collapse
Affiliation(s)
- Danqi Sun
- Institute of Translational Medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, China; Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Kai Wang
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, China
| | - Youmou Chen
- The General Hospital of Western Theater Command, No. 270, Rongdu Avenue, Chengdu 610083, China
| | - Beiying Zhang
- Institute of Translational Medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, China
| | - Jun Tang
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, China
| | - Wei Luo
- Institute of Translational Medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, China
| | - Jia Liu
- Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Sifei Yu
- Institute of Translational Medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, China.
| |
Collapse
|
4
|
Sherri N, Assaf R, Bitar ER, Znait S, Borghol AH, Kassem A, Rahal EA. Epstein-Barr Virus DNA Exacerbates Arthritis in a Mouse Model via Toll-like Receptor 9. Int J Mol Sci 2024; 25:4661. [PMID: 38731877 PMCID: PMC11083462 DOI: 10.3390/ijms25094661] [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: 03/28/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Epstein-Barr virus (EBV) DNA is known to be shed upon reactivation of latent EBV. Based on our previous findings linking Toll-like receptor-9 (TLR9) to an EBV DNA-driven surge in IL-17A production, we aimed to examine the therapeutic potential of TLR9 inhibition in EBV DNA-exacerbated arthritis in a collagen-induced arthritis (CIA) mouse model. C57BL/6J mice were administered either collagen, EBV DNA + collagen, EBV DNA + collagen + TLR9 inhibitor, or only the TLR9 inhibitor. After 70 days, paw thicknesses, clinical scores, and gripping strength were recorded. Moreover, affected joints, footpads, and colons were histologically scored. Furthermore, the number of cells co-expressing IL-17A, IFN-γ, and FOXP3 in joint sections was determined by immunofluorescence assays. Significantly decreased paw thicknesses, clinical scores, and histological scores with a significantly increased gripping strength were observed in the group receiving EBV DNA + collagen + TLR9 inhibitor, compared to those receiving EBV DNA + collagen. Similarly, this group showed decreased IL-17A+ IFN-γ+, IL-17A+ FOXP3+, and IL-17A+ IFN-γ+ FOXP3+ foci counts in joints. We show that inhibiting TLR9 limits the exacerbation of arthritis induced by EBV DNA in a CIA mouse model, suggesting that TLR9 could be a potential therapeutic target for rheumatoid arthritis management in EBV-infected individuals.
Collapse
MESH Headings
- Animals
- Mice
- Arthritis, Experimental/virology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/metabolism
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/virology
- Disease Models, Animal
- DNA, Viral/genetics
- Epstein-Barr Virus Infections/virology
- Epstein-Barr Virus Infections/complications
- Epstein-Barr Virus Infections/pathology
- Herpesvirus 4, Human/physiology
- Interleukin-17/metabolism
- Mice, Inbred C57BL
- Toll-Like Receptor 9/metabolism
Collapse
Affiliation(s)
- Nour Sherri
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Rayan Assaf
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Elio R. Bitar
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Sabah Znait
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Abdul Hamid Borghol
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Aya Kassem
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Elias A. Rahal
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut 1107, Lebanon
| |
Collapse
|
5
|
Trujillo-Ochoa JL, Kazemian M, Afzali B. The role of transcription factors in shaping regulatory T cell identity. Nat Rev Immunol 2023; 23:842-856. [PMID: 37336954 PMCID: PMC10893967 DOI: 10.1038/s41577-023-00893-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/21/2023]
Abstract
Forkhead box protein 3-expressing (FOXP3+) regulatory T cells (Treg cells) suppress conventional T cells and are essential for immunological tolerance. FOXP3, the master transcription factor of Treg cells, controls the expression of multiples genes to guide Treg cell differentiation and function. However, only a small fraction (<10%) of Treg cell-associated genes are directly bound by FOXP3, and FOXP3 alone is insufficient to fully specify the Treg cell programme, indicating a role for other accessory transcription factors operating upstream, downstream and/or concurrently with FOXP3 to direct Treg cell specification and specialized functions. Indeed, the heterogeneity of Treg cells can be at least partially attributed to differential expression of transcription factors that fine-tune their trafficking, survival and functional properties, some of which are niche-specific. In this Review, we discuss the emerging roles of accessory transcription factors in controlling Treg cell identity. We specifically focus on members of the basic helix-loop-helix family (AHR), basic leucine zipper family (BACH2, NFIL3 and BATF), CUT homeobox family (SATB1), zinc-finger domain family (BLIMP1, Ikaros and BCL-11B) and interferon regulatory factor family (IRF4), as well as lineage-defining transcription factors (T-bet, GATA3, RORγt and BCL-6). Understanding the imprinting of Treg cell identity and specialized function will be key to unravelling basic mechanisms of autoimmunity and identifying novel targets for drug development.
Collapse
Affiliation(s)
- Jorge L Trujillo-Ochoa
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA.
| |
Collapse
|
6
|
Dudreuilh C, Basu S, Shaw O, Burton H, Mamode N, Harris F, Tree T, Nedyalko P, Terranova-Barberio M, Lombardi G, Scottà C, Dorling A. Highly sensitised individuals present a distinct Treg signature compared to unsensitised individuals on haemodialysis. FRONTIERS IN TRANSPLANTATION 2023; 2:1165320. [PMID: 38993845 PMCID: PMC11235238 DOI: 10.3389/frtra.2023.1165320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 09/20/2023] [Indexed: 07/13/2024]
Abstract
Introduction Highly sensitised (HS) patients represent up to 30% of patients on the kidney transplant waiting list. When they are transplanted, they have a high risk of acute/chronic rejection and long-term allograft loss. Regulatory T cells (Tregs) (CD4+CD25hiCD127lo) are T cells involved in the suppression of immune alloresponses. A particular subset, called T follicular regulatory T cells (Tfr, CXCR5+Bcl-6+), is involved in regulating interactions between T effectors and B cells within the germinal centre and can be found in peripheral blood. Therefore, we wanted to identify specific subsets of Tregs in the peripheral blood of HS individuals. Methods We recruited prospectively healthy volunteers (HV) (n = 9), non-sensitised patients on haemodialysis (HD) (n = 9) and HS individuals, all of whom were on haemodialysis (n = 15). Results We compared the Treg phenotypes of HV, HD and HS. HS patients had more CD161+ Tregs (p = 0.02) and more CD45RA-CCR7- T effectors (Teffs) (p = 0.04, memory Teffs able to home to the germinal centre) compared to HVs. HS patients had more Bcl-6+ Tregs (p < 0.05), fewer Th1-like Tregs, more Th2-like Tregs (p < 0.001) and more CD161+ (p < 0.05) Tregs compared to HD patients. This population has been described to be highly suppressive. HD had a deficiency in a Th17-like CD161+ effector Treg cluster (cluster iii., CCR6+CCR4+CXCR3- CD39+CD15s+ICOS-CCR7-CD161+) (p < 0.05). Discussion This is the first study presenting a deep Treg phenotype in HS patients. We confirmed that HS patients had more of a Th17-like CD161+ effector Treg from population III (CD4+CD25hiCD127loCD45RA-) compared to non-sensitised patients on HD. The clinical relevance of this highly suppressive Tregs population remains to be determined in the context of transplantation.
Collapse
Affiliation(s)
- C. Dudreuilh
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - S. Basu
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - O. Shaw
- Synnovis Clinical Transplantation Laboratory, Guy’s Hospital, London, United Kingdom
| | - H. Burton
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - N. Mamode
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - F. Harris
- Peter Gorer Department of Immunobiology, King’s College London, London, United Kingdom
| | - T. Tree
- Peter Gorer Department of Immunobiology, King’s College London, London, United Kingdom
| | - P. Nedyalko
- NIHR Guy’s and St Thomas’ Biomedical Research Centre at Guy’s and St Thomas NHS Foundation Trust, St Thomas’ Hospital, London, United Kingdom
| | - M. Terranova-Barberio
- NIHR Guy’s and St Thomas’ Biomedical Research Centre at Guy’s and St Thomas NHS Foundation Trust, St Thomas’ Hospital, London, United Kingdom
| | - G. Lombardi
- Peter Gorer Department of Immunobiology, King’s College London, London, United Kingdom
| | - C. Scottà
- Peter Gorer Department of Immunobiology, King’s College London, London, United Kingdom
| | - A. Dorling
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
7
|
Guo N, Lv L. Mechanistic insights into the role of probiotics in modulating immune cells in ulcerative colitis. Immun Inflamm Dis 2023; 11:e1045. [PMID: 37904683 PMCID: PMC10571014 DOI: 10.1002/iid3.1045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/15/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a persistent inflammatory disorder that affects the gastrointestinal tract, mainly the colon, which is defined by inflammatory responses and the formation of ulcers. Probiotics have been shown to directly impact various immune cells, including dendritic cells (DCs), macrophages, natural killer (NK) cells, and T and B cells. By interacting with cell surface receptors, they regulate immune cell activity, produce metabolites that influence immune responses, and control the release of cytokines and chemokines. METHODS This article is a comprehensive review wherein we conducted an exhaustive search across published literature, utilizing reputable databases like PubMed and Web of Science. Our focus centered on pertinent keywords, such as "UC," 'DSS," "TNBS," "immune cells," and "inflammatory cytokines," to compile the most current insights regarding the therapeutic potential of probiotics in managing UC. RESULTS This overview aims to provide readers with a comprehensive understanding of the effects of probiotics on immune cells in relation to UC. Probiotics have a crucial role in promoting the proliferation of regulatory T cells (Tregs), which are necessary for preserving immunological homeostasis and regulating inflammatory responses. They also decrease the activation of pro-inflammatory cells like T helper 1 (Th1) and Th17 cells, contributing to UC development. Thus, probiotics significantly impact both direct and indirect pathways of immune cell regulation in UC, promoting Treg differentiation, inhibiting pro-inflammatory cell activation, and regulating cytokine and chemokine release. CONCLUSION Probiotics demonstrate significant potential in modulating the immune reactions in UC. Their capacity to modulate different immune cells and inflammation-related processes makes them a promising therapeutic approach for managing UC. However, further studies are warranted to optimize their use and fully elucidate the molecular mechanisms underlying their beneficial effects in UC treatment.
Collapse
Affiliation(s)
- Ni Guo
- Department of GastroenterologyShengzhou People's Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch)ShengzhouZhejiang ProvinceChina
| | - Lu‐lu Lv
- Department of GastroenterologyShengzhou People's Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch)ShengzhouZhejiang ProvinceChina
| |
Collapse
|
8
|
Vaikunthanathan T, Landmann E, Correa DM, Romano M, Trevelin SC, Peng Q, Crespo E, Corrado M, Lozano JJ, Pearce EL, Perpinan E, Zoccarato A, Siew L, Edwards-Hicks J, Khan R, Luu NT, Thursz MR, Newsome PN, Martinez-Llordella M, Shah N, Lechler RI, Shah AM, Sanchez-Fueyo A, Lombardi G, Safinia N. Dysregulated anti-oxidant signalling and compromised mitochondrial integrity negatively influence regulatory T cell function and viability in liver disease. EBioMedicine 2023; 95:104778. [PMID: 37657135 PMCID: PMC10480539 DOI: 10.1016/j.ebiom.2023.104778] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Dysregulated inflammatory responses and oxidative stress are key pathogenic drivers of chronic inflammatory diseases such as liver cirrhosis (LC). Regulatory T cells (Tregs) are essential to prevent excessive immune activation and maintain tissue homeostasis. While inflammatory cues are well known to modulate the function and stability of Tregs, the extent to which Tregs are influenced by oxidative stress has not been fully explored. METHODS The phenotypic and functional properties of CD4+CD25+CD127lo/- Tregs isolated from patients with LC were compared to healthy controls (HC). Treg redox state was investigated by characterizing intracellular reactive oxygen species (ROS), NADPH oxidase-2 (Nox2) activity, mitochondrial function, morphology, and nuclear factor-erythroid 2-related factor (Nrf2) antioxidant signalling. The relevance of Nrf2 and its downstream target, Heme-oxygenase-1 (HO-1), in Treg function, stability, and survival, was further assessed using mouse models and CRISPR/Cas9-mediated HO-1 knock-out. FINDINGS Circulating Tregs from LC patients displayed a reduced suppressive function, correlating with liver disease severity, associated with phenotypic abnormalities and increased apoptosis. Mechanistically, this was linked to a dysregulated Nrf2 signalling with resultant lower levels of HO-1, enhanced Nox2 activation, and impaired mitochondrial respiration and integrity. The functional deficit in LC Tregs could be partially recapitulated by culturing control Tregs in patient sera. INTERPRETATION Our findings reveal that Tregs rely on functional redox homeostasis for their function, stability, and survival. Targeting Treg specific anti-oxidant pathways may have therapeutic potential to reverse the Treg impairment in conditions of oxidative damage such as advanced liver disease. FUNDING This study was funded by the Wellcome Trust (211113/A/18/Z).
Collapse
Affiliation(s)
- Trishan Vaikunthanathan
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Emmanuelle Landmann
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Diana Marin Correa
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Marco Romano
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Silvia Cellone Trevelin
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Qi Peng
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Elena Crespo
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Mauro Corrado
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Juan-José Lozano
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Joseph Stelzmannstrasse 26, 50931, Cologne, Germany.
| | - Erika L Pearce
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Calle Rossello 153 Bajos, O8036, Barcelona, Spain.
| | - Elena Perpinan
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Anna Zoccarato
- Department of Immunometabolism, Max Planck Institute of Immunobiology & Epigenetics, Stübeweg 51, 79108, Freiburg, Germany.
| | - Leonard Siew
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Joy Edwards-Hicks
- Centre for Liver and Gastroenterology Research and Birmingham National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.
| | - Reenam Khan
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, Liver Unit, 10th Floor QEQM Building, St Mary's Hospital, W2 1NY, London, United Kingdom.
| | - Nguyet-Thin Luu
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, Liver Unit, 10th Floor QEQM Building, St Mary's Hospital, W2 1NY, London, United Kingdom.
| | - Mark R Thursz
- Institute of Liver Sciences, King's College Hospital NHS Foundation Trust, London, SE5 9NU, United Kingdom.
| | - Philip N Newsome
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, Liver Unit, 10th Floor QEQM Building, St Mary's Hospital, W2 1NY, London, United Kingdom.
| | - Marc Martinez-Llordella
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Naina Shah
- James Black Centre, Department of Cardiovascular sciences, British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, SE5 9NU, United Kingdom.
| | - Robert I Lechler
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Ajay M Shah
- Department of Immunometabolism, Max Planck Institute of Immunobiology & Epigenetics, Stübeweg 51, 79108, Freiburg, Germany.
| | - Alberto Sanchez-Fueyo
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Niloufar Safinia
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| |
Collapse
|
9
|
Fadlallah S, Bitar ER, Hussein H, Jallad MA, Matar GM, Rahal EA. The interplay between Epstein-Barr virus DNA and gut microbiota in the development of arthritis in a mouse model. Microbiol Spectr 2023; 11:e0204223. [PMID: 37615438 PMCID: PMC10581075 DOI: 10.1128/spectrum.02042-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/02/2023] [Indexed: 08/25/2023] Open
Abstract
Epstein-Barr virus (EBV) DNA may influence the development of autoimmune diseases by increasing the production of proinflammatory cytokines. Such cytokines have been associated with inducing the dysbiosis of colonic microbiota, which, in turn, is a risk factor for autoimmune diseases such as rheumatoid arthritis (RA). Therefore, we investigated the role that EBV DNA may play in modulating the intestinal microbiota and consequent exacerbation of arthritis in a mouse model. Mice were treated with collagen (arthritis-inducing agent), EBV DNA and collagen, EBV DNA, or water. Fecal samples were collected from arthritic and control mice, and 16S rRNA sequencing was performed to determine the effect of EBV DNA on the composition of colonic microbiota. EBV DNA causes a change in the alpha diversity of the microbiota resulting in an increased Chao1 microbial richness and decreased Shannon diversity index in the RA mouse model. In addition, the abundance of particular genera/genus clusters was significantly altered among the various groups, with the EBV DNA-exacerbated arthritic group having the highest number of altered genera/genus cluster abundances. This group also had the highest number of cells co-expressing IL-17A, FOXP3, and IFNγ in the colons. Antimicrobial-cleared mice transplanted with fecal samples from EBV DNA-exacerbated arthritic mice showed a higher incidence and enhanced severity of RA compared to those transplanted with fecal samples from water or collagen-treated mice. IMPORTANCE Epstein-Barr virus (EBV) DNA alters the composition and diversity of the gut microbiota in a rheumatoid arthritis (RA) mouse model. These induced changes are associated with enhanced severity of symptoms. This better understanding of the various factors involved in the development of RA will possibly help in creating individualized treatments for RA patients including target mediators triggered by viral DNA. Given that a large swathe of the population harbors EBV, a significant proportion of subjects with arthritis may benefit from possible approaches that target EBV or mediators triggered by this virus.
Collapse
Affiliation(s)
- Sukayna Fadlallah
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Elio R. Bitar
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Hadi Hussein
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Mary-Ann Jallad
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Ghassan M. Matar
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Elias A. Rahal
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
10
|
Ni H, Chen Y. Differentiation, regulation and function of regulatory T cells in non-lymphoid tissues and tumors. Int Immunopharmacol 2023; 121:110429. [PMID: 37327512 DOI: 10.1016/j.intimp.2023.110429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023]
Abstract
Regulatory T cells (Tregs) play a substantial role in inhibiting excessive immune response. A large number of studies have focused on the tissue homeostasis maintenance and remodeling characteristics of Tregs in non-lymphoid tissues, such as the skin, colon, lung, brain, muscle, and adipose tissues. Herein, we overview the kinetics of Treg migration to non-lymphoid tissues and adaptation to the specific tissue microenvironment through the development of tissue-specific chemokine receptors, transcription factors, and phenotypes. Additionally, tumor-infiltrating Tregs (Ti-Tregs) play an important role in tumor generation and immunotherapy resistance. The phenotypes of Ti-Tregs are related to the histological location of the tumor and there is a large overlap between the transcripts of Ti-Tregs and those of tissue-specific Tregs. We recapitulate the molecular underpinnings of tissue-specific Tregs, which might shed new light on Treg-based therapeutic targets and biomarkers for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Hongbo Ni
- The First Clinical Medicine Faculty, China Medical University, Shenyang 110001, China
| | - Yinghan Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
11
|
Guardalupi F, Sorrentino C, Corradi G, Giancola R, Baldoni S, Ulbar F, Fabi B, Andres Ejarque R, Timms J, Restuccia F, Santarone S, Accorsi P, Sportoletti P, De Falco F, Rosati E, Carotti A, Falzetti F, Velardi A, Martelli MF, Kordasti S, Pierini A, Ruggeri L, Di Ianni M. A pro-inflammatory environment in bone marrow of Treg transplanted patients matches with graft-versus-leukemia effect. Leukemia 2023; 37:1572-1575. [PMID: 37286785 PMCID: PMC10317833 DOI: 10.1038/s41375-023-01932-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/03/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Affiliation(s)
- Francesco Guardalupi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Giulia Corradi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Chieti, Italy
| | | | - Stefano Baldoni
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Francesca Ulbar
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Bianca Fabi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Rosa Andres Ejarque
- System Cancer Immunology, Comprehensive Cancer Centre, King's College London, London, United Kingdom
| | - Jessica Timms
- System Cancer Immunology, Comprehensive Cancer Centre, King's College London, London, United Kingdom
| | | | - Stella Santarone
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Patrizia Accorsi
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Paolo Sportoletti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Filomena De Falco
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emanuela Rosati
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessandra Carotti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Franca Falzetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Shahram Kordasti
- System Cancer Immunology, Comprehensive Cancer Centre, King's College London, London, United Kingdom
| | - Antonio Pierini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Loredana Ruggeri
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Chieti, Italy.
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy.
| |
Collapse
|
12
|
Maeda K, Tanioka T, Takahashi R, Watanabe H, Sueki H, Takimoto M, Hashimoto SI, Ikeo K, Miwa Y, Kasama T, Iwamoto S. MCAM+CD161- Th17 Subset Expressing CD83 Enhances Tc17 Response in Psoriasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1867-1881. [PMID: 37186262 DOI: 10.4049/jimmunol.2200530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
Recent studies have highlighted the pathogenic roles of IL-17-producing CD8+ T cells (T-cytotoxic 17 [Tc17]) in psoriasis. However, the underlying mechanisms of Tc17 induction remain unclear. In this study, we focused on the pathogenic subsets of Th17 and their mechanism of promotion of Tc17 responses. We determined that the pathogenic Th17-enriched fraction expressed melanoma cell adhesion molecule (MCAM) and CCR6, but not CD161, because this subset produced IL-17A abundantly and the presence of these cells in the peripheral blood of patients has been correlated with the severity of psoriasis. Intriguingly, the serial analysis of gene expression revealed that CCR6+MCAM+CD161-CD4+ T cells displayed the gene profile for adaptive immune responses, including CD83, which is an activator for CD8+ T cells. Coculture assay with or without intercellular contact between CD4+ and CD8+ T cells showed that CCR6+MCAM+CD161-CD4+ T cells induced the proliferation of CD8+ T cells in a CD83-dependent manner. However, the production of IL-17A by CD8+ T cells required exogenous IL-17A, suggesting that intercellular contact via CD83 and the production of IL-17A from activated CD4+ T cells elicit Tc17 responses. Intriguingly, the CD83 expression was enhanced in the presence of IL-15, and CD83+ cells stimulated with IL-1β, IL-23, IL-15, and IL-15Rα did not express FOXP3. Furthermore, CCR6+MCAM+CD161-CD4+ T cells expressing CD83 were increased in the peripheral blood of patients, and the CD83+ Th17-type cells accumulated in the lesional skin of psoriasis. In conclusion, pathogenic MCAM+CD161- Th17 cells may be involved in the Tc17 responses via IL-17A and CD83 in psoriasis.
Collapse
Affiliation(s)
- Kohei Maeda
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Toshihiro Tanioka
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Rei Takahashi
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Hideaki Watanabe
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Hirohiko Sueki
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Masafumi Takimoto
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shin-Ichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuho Ikeo
- DNA Data Analysis Laboratory, National Institute of Genetics, Shizuoka, Japan
| | - Yusuke Miwa
- Department of Internal Medicine, Division of Rheumatology, Showa University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Kasama
- Department of Internal Medicine, Division of Rheumatology, Showa University School of Medicine, Tokyo, Japan
| | - Sanju Iwamoto
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| |
Collapse
|
13
|
Povoleri GAM, Durham LE, Gray EH, Lalnunhlimi S, Kannambath S, Pitcher MJ, Dhami P, Leeuw T, Ryan SE, Steel KJA, Kirkham BW, Taams LS. Psoriatic and rheumatoid arthritis joints differ in the composition of CD8+ tissue-resident memory T cell subsets. Cell Rep 2023; 42:112514. [PMID: 37195862 PMCID: PMC10790246 DOI: 10.1016/j.celrep.2023.112514] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/21/2023] [Accepted: 05/01/2023] [Indexed: 05/19/2023] Open
Abstract
CD69+CD103+ tissue-resident memory T (TRM) cells are important drivers of inflammation. To decipher their role in inflammatory arthritis, we apply single-cell, high-dimensional profiling to T cells from the joints of patients with psoriatic arthritis (PsA) or rheumatoid arthritis (RA). We identify three groups of synovial CD8+CD69+CD103+ TRM cells: cytotoxic and regulatory T (Treg)-like TRM cells are present in both PsA and RA, while CD161+CCR6+ type 17-like TRM cells with a pro-inflammatory cytokine profile (IL-17A+TNFα+IFNγ+) are specifically enriched in PsA. In contrast, only one population of CD4+CD69+CD103+ TRM cells is detected and at similarly low frequencies in both diseases. Type 17-like CD8+ TRM cells have a distinct transcriptomic signature and a polyclonal, but distinct, TCR repertoire. Type 17-like cells are also enriched in CD8+CD103- T cells in PsA compared with RA. These findings illustrate differences in the immunopathology of PsA and RA, with a particular enrichment for type 17 CD8+ T cells in the PsA joint.
Collapse
Affiliation(s)
- Giovanni A M Povoleri
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Lucy E Durham
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Elizabeth H Gray
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Sylvine Lalnunhlimi
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Shichina Kannambath
- BRC Genomics Core, NIHR Biomedical Research Center, Guy's and St Thomas' NHS Foundation Trust and King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Michael J Pitcher
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Pawan Dhami
- BRC Genomics Core, NIHR Biomedical Research Center, Guy's and St Thomas' NHS Foundation Trust and King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Thomas Leeuw
- Immunology & Inflammation Research TA, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Sarah E Ryan
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Kathryn J A Steel
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Bruce W Kirkham
- Rheumatology Department, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK.
| |
Collapse
|
14
|
Cheru N, Hafler DA, Sumida TS. Regulatory T cells in peripheral tissue tolerance and diseases. Front Immunol 2023; 14:1154575. [PMID: 37197653 PMCID: PMC10183596 DOI: 10.3389/fimmu.2023.1154575] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
Maintenance of peripheral tolerance by CD4+Foxp3+ regulatory T cells (Tregs) is essential for regulating autoreactive T cells. The loss of function of Foxp3 leads to autoimmune disease in both animals and humans. An example is the rare, X-linked recessive disorder known as IPEX (Immune Dysregulation, Polyendocrinopathy, Enteropathy X-linked) syndrome. In more common human autoimmune diseases, defects in Treg function are accompanied with aberrant effector cytokines such as IFNγ. It has recently become appreciated that Tregs plays an important role in not only maintaining immune homeostasis but also in establishing the tissue microenvironment and homeostasis of non-lymphoid tissues. Tissue resident Tregs show profiles that are unique to their local environments which are composed of both immune and non-immune cells. Core tissue-residence gene signatures are shared across different tissue Tregs and are crucial to homeostatic regulation and maintaining the tissue Treg pool in a steady state. Through interaction with immunocytes and non-immunocytes, tissue Tregs exert a suppressive function via conventional ways involving contact dependent and independent processes. In addition, tissue resident Tregs communicate with other tissue resident cells which allows Tregs to adopt to their local microenvironment. These bidirectional interactions are dependent on the specific tissue environment. Here, we summarize the recent advancements of tissue Treg studies in both human and mice, and discuss the molecular mechanisms that maintain tissue homeostasis and prevent pathogenesis.
Collapse
Affiliation(s)
- Nardos Cheru
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - David A. Hafler
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Tomokazu S. Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
15
|
Bian W, Li Y, Sun F, Sun X, Li R, Xia C, Fu J, Zhang Y, Chen S, Liu Y. Immune phenotype changes in IgG4-related disease: CD161 + Treg and Foxp3 + Treg. Clin Rheumatol 2023; 42:1113-1124. [PMID: 36567407 DOI: 10.1007/s10067-022-06445-z] [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: 07/24/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVE We aimed to characterize the alterations in the immune phenotypes and explore the potential relevance to pathogenesis in IgG4-RD. METHODS Forty-two IgG4-RD patients and thirty-eight healthy controls were recruited in this study. Peripheral immunocompetent cells including T cells, CD4 + T cells, CD8 + T cells, B cells, NK cells CD4 + CD45RA + T cells (naïve T cells), CD4 + CD25 - / + Foxp3 - T cells (Teff), CD4 + CD25hiCD127lowCD161 + T cells (CD161 + Treg), CD4 + CD25hiFoxp3 + T cells (Foxp3 + Treg), CD4 + CD4RA-CXCR5 + PD1 + CCR7low T cells (pTfh), T helper (Th) 1, Th2, and Th17 before and after treatment were immunophenotyped by flow cytometry. RESULTS Compared with healthy controls, IgG4-RD patients showed higher proportions of NK (20.1% vs 13.6%, p < 0.01), Th1 (CD4 + IFN-γ + : 17.9% vs 14.2%, p = 0.061; TNF-α: 43.7% vs 36.7%, p < 0.05), Th2 (CD4 + IL-4 + : 2.4% vs 1.3%, p < 0.0001), CD161 + Treg (14.9% vs 11.6%, p < 0.01), pTfh (3.2% vs 2.4%, p < 0.05), and Foxp3 + Treg (8.3% vs 7.0%, p < 0.01) and lower proportions of B lymphocytes (8.4% vs 13.1%, p < 0.001), Teff (91.6% vs 92.6%, p < 0.01), and naïve Th cells (19.9% vs 32.1%, p < 0.01) before treatment. Foxp3 + Treg percentage decreased significantly after treatment (8.6% vs 6.9%, p < 0.05). Both serum C3 (r = - 0.6374, p < 0.01) and C4 (r = - 0.6174, p < 0.01) levels were in negative correlation with CD161 + Treg. The eosinophil percentage was positively correlated with Foxp3 + Treg (r = 0.5435, p < 0.05). Serum IgE level was positively correlated with Th2 (r = 0.5545, p < 0.05). There was a positive correlation between CD161 + Treg and pTfh (r = 0.4974, p < 0.05) while a negative correlation between Th2 and B cells (r = - 0.4925, p < 0.05). CONCLUSION Immune phenotypes were altered in IgG4-RD. Treg/Teff balance was shifted toward Treg in IgG4-RD. CD161 + Treg was likely to be involved in the pathogenesis of IgG4-RD. Key Points •Immune phenotypes were altered in B cells, T cells, and NK cells in IgG4-RD. •Treg/Teff balance was shifted toward Treg in IgG4-RD. •CD161+ Treg maybe play a proinflammatory role in IgG4-RD.
Collapse
Affiliation(s)
- Wenjie Bian
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Yingni Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Feng Sun
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Ru Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Changsheng Xia
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Jiangnan Fu
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Yuxin Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Shuang Chen
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China
| | - Yanying Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11, Xizhimen South Street, Beijing, 100044, China.
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95, Yongan Road, Beijing, 100050, China.
| |
Collapse
|
16
|
Kou Y, Jiang Y, Liu S, Yang P, Lu Y, Liu H, Li M. Regulatory T cells showed characteristics of T helper-17(Th17) cells in mice periodontitis model. Oral Dis 2023; 29:1149-1162. [PMID: 34741371 DOI: 10.1111/odi.14072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/29/2021] [Accepted: 10/29/2021] [Indexed: 01/05/2023]
Abstract
OBJECTIVES This study aimed to clarify the regulatory role of Th17-Treg balance in periodontitis and further reveal Treg plasticity. MATERIALS AND METHODS An experimental periodontitis model was established by ligation and injection of Pg-LPS. Inflammatory factors were measured by ELISA and RT-PCR. Alveolar bone absorption was evaluated by micro-CT and histomorphology. Quantities of Treg and Th17 cell and their related gene expression were examined. Furthermore, after magnetic bead-sorting spleen Treg cells, Treg/Th17 characteristic genes were explored. Immunofluorescence double staining of Foxp3 and IL-17 was conducted to further reveal Treg plasticity. RESULTS Inflammatory cytokines in serum and gingival tissue increased significantly in periodontitis, which revealed obvious crestal bone loss. Further analysis showed that the number of Th17 cells and expression of related genes increased more significantly than Treg cells, demonstrating Treg-Th17 imbalance. Flow cytometry showed that the proportions of Treg cells in the blood and spleen were lower in periodontitis group. Furthermore, Foxp3 was downregulated, and Rorc/ IL-17A were increased in Treg cells of periodontitis group. Immunofluorescence double staining showed significantly increased number of IL-17+Foxp3+ cells in periodontitis. CONCLUSIONS These results provided evidence that Treg cells showed characteristics of Th17 cells in mice with periodontitis, although its mechanisms require further study.
Collapse
Affiliation(s)
- Yuying Kou
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Yujun Jiang
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Shanshan Liu
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Panpan Yang
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Yupu Lu
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Hongrui Liu
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Minqi Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| |
Collapse
|
17
|
de Sousa Palmeira PH, Peixoto RF, Csordas BG, de Medeiros IA, de Azevedo FDLAA, Veras RC, Janebro DI, Amaral IP, Keesen TSL. Differential regulatory T cell signature after recovery from mild COVID-19. Front Immunol 2023; 14:1078922. [PMID: 36969257 PMCID: PMC10030602 DOI: 10.3389/fimmu.2023.1078922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is characterized by a range of symptoms in which host immune response have been associated with disease progression. However, the putative role of regulatory T cells (Tregs) in determining COVID-19 outcomes has not been thoroughly investigated. Here, we compared peripheral Tregs between volunteers not previously infected with SARS-CoV-2 (healthy control [HC]) and volunteers who recovered from mild (Mild Recovered) and severe (Severe Recovered) COVID-19. Peripheral blood mononuclear cells (PBMC) were stimulated with SARS-CoV-2 synthetic peptides (Pool Spike CoV-2 and Pool CoV-2) or staphylococcal enterotoxin B (SEB). Results of a multicolor flow cytometric assay showed higher Treg frequency and expression of IL-10, IL-17, perforin, granzyme B, PD-1, and CD39/CD73 co-expression in Treg among the PBMC from the Mild Recovered group than in the Severe Recovered or HC groups for certain SARS-CoV-2 related stimulus. Moreover, Mild Recovered unstimulated samples presented a higher Tregs frequency and expression of IL-10 and granzyme B than did that of HC. Compared with Pool CoV-2 stimuli, Pool Spike CoV-2 reduced IL-10 expression and improved PD-1 expression in Tregs from volunteers in the Mild Recovered group. Interestingly, Pool Spike CoV-2 elicited a decrease in Treg IL-17+ frequency in the Severe Recovered group. In HC, the expression of latency-associated peptide (LAP) and cytotoxic granule co-expression by Tregs was higher in Pool CoV-2 stimulated samples. While Pool Spike CoV-2 stimulation reduced the frequency of IL-10+ and CTLA-4+ Tregs in PBMC from volunteers in the Mild Recovered group who had not experienced certain symptoms, higher levels of perforin and perforin+granzyme B+ co-expression by Tregs were found in the Mild Recovered group in volunteers who had experienced dyspnea. Finally, we found differential expression of CD39 and CD73 among volunteers in the Mild Recovered group between those who had and had not experienced musculoskeletal pain. Collectively, our study suggests that changes in the immunosuppressive repertoire of Tregs can influence the development of a distinct COVID-19 clinical profile, revealing that a possible modulation of Tregs exists among volunteers of the Mild Recovered group between those who did and did not develop certain symptoms, leading to mild disease.
Collapse
Affiliation(s)
- Pedro Henrique de Sousa Palmeira
- Postgraduate program in Physiology Science, Immunology Laboratory of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Rephany Fonseca Peixoto
- Postgraduate program in Physiology Science, Immunology Laboratory of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Bárbara Guimarães Csordas
- Postgraduate program in Natural and Synthetic Bioactive Products, Immunology Laboratory of Infectious Diseases, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Isac Almeida de Medeiros
- Research Institute for Drugs and Medicines, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | | | - Robson Cavalcante Veras
- Research Institute for Drugs and Medicines, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Daniele Idalino Janebro
- Research Institute for Drugs and Medicines, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Ian P.G. Amaral
- Biotechnology Graduation Program, Immunology Laboratory of Infectious Diseases, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Tatjana Souza Lima Keesen
- Immunology Laboratory of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| |
Collapse
|
18
|
Malviya V, Yshii L, Junius S, Garg AD, Humblet-Baron S, Schlenner SM. Regulatory T-cell stability and functional plasticity in health and disease. Immunol Cell Biol 2023; 101:112-129. [PMID: 36479949 DOI: 10.1111/imcb.12613] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
FOXP3-expressing regulatory T cells (Treg ) are indispensable for immune homeostasis and tolerance, and in addition tissue-resident Treg have been found to perform noncanonical, tissue-specific functions. For optimal tolerogenic function during inflammatory disease, Treg are equipped with mechanisms that assure lineage stability. Treg lineage stability is closely linked to the installation and maintenance of a lineage-specific epigenetic landscape, specifically a Treg -specific DNA demethylation pattern. At the same time, for local and directed immune regulation Treg must possess a level of functional plasticity that requires them to partially acquire T helper cell (TH ) transcriptional programs-then referred to as TH -like Treg . Unleashing TH programs in Treg , however, is not without risk and may threaten the epigenetic stability of Treg with consequently pathogenic ex-Treg contributing to (auto-) inflammatory conditions. Here, we review how the Treg -stabilizing epigenetic landscape is installed and maintained, and further discuss the development, necessity and lineage instability risks of TH 1-, TH 2-, TH 17-like Treg and follicular Treg .
Collapse
Affiliation(s)
- Vanshika Malviya
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| | - Lidia Yshii
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Steffie Junius
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stephanie Humblet-Baron
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| | - Susan M Schlenner
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Povoleri GAM, Fleskens V, Taams LS. Isolation and Functional Characterization of Regulatory CD4+ T Cells from the Inflamed Joints of Patients with Rheumatoid Arthritis. Methods Mol Biol 2023; 2559:189-203. [PMID: 36180634 DOI: 10.1007/978-1-0716-2647-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Regulatory T cells play a critical role in maintaining immune homeostasis and in preventing and controlling unwanted immune activation. These cells are often studied in the context of human peripheral blood, but can also be isolated from other biofluids. Here we describe methods for the isolation and functional characterization of human CD4+ CD25hi CD127low regulatory T cells from the synovial fluid of patients with inflammatory arthritis.
Collapse
Affiliation(s)
- Giovanni A M Povoleri
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Veerle Fleskens
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK.
| |
Collapse
|
20
|
Zhang M, Zhang X. T cells in ocular autoimmune uveitis: Pathways and therapeutic approaches. Int Immunopharmacol 2023; 114:109565. [PMID: 36535124 DOI: 10.1016/j.intimp.2022.109565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Autoimmune uveitis is a non-infectious intraocular condition that affects the uveal tract of the eye and threatens vision if not treated properly. Increasing evidence suggests that activated CD4+ T cells are associated with progressive and permanent destruction of photoreceptors in ocular autoimmune diseases. As such, the purpose of this review is to offer an overview of the role of CD4+ T cells in autoimmune uveitis as well as a justification for the current development and assessment of innovative autoimmune uveitis medications targeting CD4+ T cells. With an emphasis on T helper (Th)17, Th1, and Th2 cells, follicular helper CD4+ T cells, and regulatory T cells, this review presents a summary of recent research related to the pathways and signaling that encourage CD4+ T cells to develop into specialized effector cells. We also describe immunotherapeutic approaches based on CD4+ T cell subsets and their potential as therapeutic agents for autoimmune disorders.
Collapse
Affiliation(s)
- Mi Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.
| |
Collapse
|
21
|
Chandran S, Tang Q. Impact of interleukin-6 on T cells in kidney transplant recipients. Am J Transplant 2022; 22 Suppl 4:18-27. [PMID: 36453710 DOI: 10.1111/ajt.17209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/23/2022] [Indexed: 12/02/2022]
Abstract
Interleukin-6 (IL-6), a multifunctional proinflammatory cytokine, plays a key role in T cell activation, survival, and differentiation. Acting as a switch that induces the differentiation of naïve T cells into Th17 cells and inhibits their development into regulatory T cells, IL-6 promotes rejection and abrogates tolerance. Therapies that target IL-6 signaling include antibodies to IL-6 and the IL-6 receptor and inhibitors of janus kinases; several of these therapeutics have demonstrated robust clinical efficacy in autoimmune and inflammatory diseases. Clinical trials of IL-6 inhibition in kidney transplantation have focused primarily on its effects on B cells, plasma cells, and HLA antibodies. In this review, we summarize the impact of IL-6 on T cells in experimental models of transplant and describe the effects of IL-6 inhibition on the T cell compartment in kidney transplant recipients.
Collapse
Affiliation(s)
- Sindhu Chandran
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Qizhi Tang
- Department of Surgery, Diabetes Center, Gladstone-UCSF Institute of Genome Immunology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
22
|
Pouw JNJ, Nordkamp MAMMO, van Kempen TT, Concepcion ANA, van Laar JMJ, van Wijk FF, Spierings JJ, Leijten EFAE, Boes MM. Regulatory T cells in psoriatic arthritis: an IL-17A-producing, Foxp3 intCD161 + RORγt + ICOS + phenotype, that associates with the presence of ADAMTSL5 autoantibodies. Sci Rep 2022; 12:20675. [PMID: 36450783 PMCID: PMC9712434 DOI: 10.1038/s41598-022-24924-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
In psoriatic arthritis (PsA), predisposing class I HLA alleles, the presence of synovial clonally proliferated CD8 + T cells and autoantibodies all point towards the loss of immune tolerance. However, the key mechanisms that lead to immune dysregulation are not fully understood. In other types of inflammatory arthritis, T regulatory cell (Treg) dysfunction and plasticity at sites of inflammation were suggested to negatively affect peripheral tolerance. We here addressed if Treg variances associate with psoriatic disease. We collected clinical data, sera and peripheral blood mononuclear cells from 13 healthy controls, 21 psoriasis and 21 PsA patients. In addition, we obtained synovial fluid mononuclear cells from 6 PsA patients. We studied characteristics of CD4 + CD25 + CD127loFoxp3 + Tregs by flow cytometry and used ELISA to quantify antibodies against ADAMTSL5, a recently discovered autoantigen in psoriatic disease. In comparison with their circulating counterparts, Tregs from inflamed joints express increased levels of ICOS, CTLA-4 and TIGIT. Furthermore, synovial fluid-derived Tregs have a distinct phenotype, characterized by IL-17A production and upregulation of CD161 and RORγt. We identified a subset of Tregs with intermediate Foxp3 expression as the major cytokine producer. Furthermore, ICOS + Tregs associate with PsA disease activity as measured by PASDAS. Lastly, we observed that presence of the Foxp3int Tregs associates with an increased abundance of anti-ADAMTSL5 autoantibodies. Tregs derived from the inflammatory environment of inflamed PsA joints exhibit a distinct phenotype, which associates with loss of peripheral immune tolerance in psoriatic disease.
Collapse
Affiliation(s)
- J. N. Juliëtte Pouw
- grid.5477.10000000120346234Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, H03.103, P.O. Box 85500, 3508 GA Utrecht, The Netherlands ,grid.5477.10000000120346234Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3508 AB Utrecht, The Netherlands
| | - M. A. M. Michel Olde Nordkamp
- grid.5477.10000000120346234Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3508 AB Utrecht, The Netherlands
| | - T. Tessa van Kempen
- grid.440506.30000 0000 9631 4629Biomedical Laboratory Sciences, Avans University of Applied Sciences, 4800 RA Breda, The Netherlands
| | - A. N. Arno Concepcion
- grid.5477.10000000120346234Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, H03.103, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - J. M. Jacob van Laar
- grid.5477.10000000120346234Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, H03.103, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - F. Femke van Wijk
- grid.5477.10000000120346234Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3508 AB Utrecht, The Netherlands
| | - J. Julia Spierings
- grid.5477.10000000120346234Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, H03.103, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - E. F. A. Emmerik Leijten
- grid.452818.20000 0004 0444 9307Department of Rheumatology, Sint Maartenskliniek, 6500 GM Nijmegen, The Netherlands
| | - M. Marianne Boes
- grid.5477.10000000120346234Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3508 AB Utrecht, The Netherlands ,grid.5477.10000000120346234Department of Pediatric Immunology, Wilhelmina Children’s Hospital, Utrecht University, 3508 AB Utrecht, The Netherlands
| |
Collapse
|
23
|
Bláha J, Skálová T, Kalousková B, Skořepa O, Cmunt D, Grobárová V, Pazicky S, Poláchová E, Abreu C, Stránský J, Kovaľ T, Dušková J, Zhao Y, Harlos K, Hašek J, Dohnálek J, Vaněk O. Structure of the human NK cell NKR-P1:LLT1 receptor:ligand complex reveals clustering in the immune synapse. Nat Commun 2022; 13:5022. [PMID: 36028489 PMCID: PMC9418145 DOI: 10.1038/s41467-022-32577-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Signaling by the human C-type lectin-like receptor, natural killer (NK) cell inhibitory receptor NKR-P1, has a critical role in many immune-related diseases and cancer. C-type lectin-like receptors have weak affinities to their ligands; therefore, setting up a comprehensive model of NKR-P1-LLT1 interactions that considers the natural state of the receptor on the cell surface is necessary to understand its functions. Here we report the crystal structures of the NKR-P1 and NKR-P1:LLT1 complexes, which provides evidence that NKR-P1 forms homodimers in an unexpected arrangement to enable LLT1 binding in two modes, bridging two LLT1 molecules. These interaction clusters are suggestive of an inhibitory immune synapse. By observing the formation of these clusters in solution using SEC-SAXS analysis, by dSTORM super-resolution microscopy on the cell surface, and by following their role in receptor signaling with freshly isolated NK cells, we show that only the ligation of both LLT1 binding interfaces leads to effective NKR-P1 inhibitory signaling. In summary, our findings collectively support a model of NKR-P1:LLT1 clustering, which allows the interacting proteins to overcome weak ligand-receptor affinity and to trigger signal transduction upon cellular contact in the immune synapse. NKR-P1 is an inhibitory receptor on the surface of natural killer cells, and its engagement with the ligand LLT1 on activated monocytes and B cells triggers NK cell self-tolerance and other immunological processes. Here authors set up a comprehensive, structure-based model of NKR-P1-LLT1 interaction that involves NKR-P1 homodimer formation and subsequent bridging of two LLT1 molecules.
Collapse
Affiliation(s)
- Jan Bláha
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic.,EMBL, Hamburg Unit c/o DESY, Notkestrasse 85, 22607, Hamburg, Germany
| | - Tereza Skálová
- Institute of Biotechnology, The Czech Academy of Sciences, BIOCEV Centre, Průmyslová 595, 25250, Vestec, Czech Republic
| | - Barbora Kalousková
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic.,Institute of Applied Physics - Biophysics group, TU Wien, Getreidemarkt 9, 1060, Vienna, Austria
| | - Ondřej Skořepa
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic
| | - Denis Cmunt
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic.,Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Valéria Grobárová
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 12800, Prague, Czech Republic
| | - Samuel Pazicky
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic.,School of Biological Sciences, Nanyang Technological University, Nanyang Drive 60, 637551, Singapore, Singapore
| | - Edita Poláchová
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic
| | - Celeste Abreu
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic
| | - Jan Stránský
- Institute of Biotechnology, The Czech Academy of Sciences, BIOCEV Centre, Průmyslová 595, 25250, Vestec, Czech Republic
| | - Tomáš Kovaľ
- Institute of Biotechnology, The Czech Academy of Sciences, BIOCEV Centre, Průmyslová 595, 25250, Vestec, Czech Republic
| | - Jarmila Dušková
- Institute of Biotechnology, The Czech Academy of Sciences, BIOCEV Centre, Průmyslová 595, 25250, Vestec, Czech Republic
| | - Yuguang Zhao
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, OX3 7BN, Oxford, UK
| | - Karl Harlos
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, OX3 7BN, Oxford, UK
| | - Jindřich Hašek
- Institute of Biotechnology, The Czech Academy of Sciences, BIOCEV Centre, Průmyslová 595, 25250, Vestec, Czech Republic
| | - Jan Dohnálek
- Institute of Biotechnology, The Czech Academy of Sciences, BIOCEV Centre, Průmyslová 595, 25250, Vestec, Czech Republic
| | - Ondřej Vaněk
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12800, Prague, Czech Republic.
| |
Collapse
|
24
|
Rajendeeran A, Tenbrock K. Regulatory T cell function in autoimmune disease. J Transl Autoimmun 2022; 4:100130. [PMID: 35005594 PMCID: PMC8716637 DOI: 10.1016/j.jtauto.2021.100130] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022] Open
Abstract
Autoimmune diseases are characterized by a failure of tolerance to own body components resulting in tissue damage. Regulatory T cells are gatekeepers of tolerance. This review focusses on the function and pathophysiology of regulatory T cells in the context of autoimmune diseases including rheumatoid and juvenile idiopathic arthritis as well as systemic lupus erythematosus with an overview over current and future therapeutic options to boost Treg function. Regulatory T cells are critical mediators of immune tolerance and critically depend on external IL-2. Tregs are expanded during inflammation, where the local milieu enhances resistance to suppression in T effector cells. Human Tregs are characterized by different markers, which hampers the comparability of studies in patients with autoimmunity.
Collapse
Affiliation(s)
- Anandi Rajendeeran
- RWTH Aachen University, Department of Pediatrics, Pediatric Rheumatology, Pauwelsstr 30, 52074, Aachen, Germany
| | - Klaus Tenbrock
- RWTH Aachen University, Department of Pediatrics, Pediatric Rheumatology, Pauwelsstr 30, 52074, Aachen, Germany
| |
Collapse
|
25
|
Hannan R, Mohamad O, Diaz de Leon A, Manna S, Pop LM, Zhang Z, Mannala S, Christie A, Christley S, Monson N, Ishihara D, Hsu EJ, Ahn C, Kapur P, Chen M, Arriaga Y, Courtney K, Cantarel B, Wakeland EK, Fu YX, Pedrosa I, Cowell L, Wang T, Margulis V, Choy H, Timmerman RD, Brugarolas J. Outcome and Immune Correlates of a Phase II Trial of High-Dose Interleukin-2 and Stereotactic Ablative Radiotherapy for Metastatic Renal Cell Carcinoma. Clin Cancer Res 2021; 27:6716-6725. [PMID: 34551906 PMCID: PMC9924935 DOI: 10.1158/1078-0432.ccr-21-2083] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/13/2021] [Accepted: 09/20/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE This phase II clinical trial evaluated whether the addition of stereotactic ablative radiotherapy (SAbR), which may promote tumor antigen presentation, improves the overall response rate (ORR) to high-dose IL2 (HD IL2) in metastatic renal cell carcinoma (mRCC). PATIENTS AND METHODS Patients with pathologic evidence of clear cell renal cell carcinoma (RCC) and radiographic evidence of metastasis were enrolled in this single-arm trial and were treated with SAbR, followed by HD IL2. ORR was assessed based on nonirradiated metastases. Secondary endpoints included overall survival (OS), progression-free survival (PFS), toxicity, and treatment-related tumor-specific immune response. Correlative studies involved whole-exome and transcriptome sequencing, T-cell receptor sequencing, cytokine analysis, and mass cytometry on patient samples. RESULTS Thirty ethnically diverse mRCC patients were enrolled. A median of two metastases were treated with SAbR. Among 25 patients evaluable by RECIST v1.1, ORR was 16% with 8% complete responses. Median OS was 37 months. Treatment-related adverse events (AE) included 22 grade ≥3 events that were not dissimilar from HD IL2 alone. There were no grade 5 AEs. A correlation was observed between SAbR to lung metastases and improved PFS (P = 0.0165). Clinical benefit correlated with frameshift mutational load, mast cell tumor infiltration, decreased circulating tumor-associated T-cell clones, and T-cell clonal expansion. Higher regulatory/CD8+ T-cell ratios at baseline in the tumor and periphery correlated with no clinical benefit. CONCLUSIONS Adding SAbR did not improve the response rate to HD IL2 in patients with mRCC in this study. Tissue analyses suggest a possible correlation between frameshift mutation load as well as tumor immune infiltrates and clinical outcomes.
Collapse
Affiliation(s)
- Raquibul Hannan
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Osama Mohamad
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Alberto Diaz de Leon
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Subrata Manna
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Laurentiu M Pop
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ze Zhang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Samantha Mannala
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alana Christie
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Scott Christley
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nancy Monson
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dan Ishihara
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Eric J Hsu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chul Ahn
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yull Arriaga
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kevin Courtney
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Brandi Cantarel
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ivan Pedrosa
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lindsay Cowell
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tao Wang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vitaly Margulis
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hak Choy
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Robert D Timmerman
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Brugarolas
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
26
|
Singh RP, Bischoff DS, Hahn BH. CD8 + T regulatory cells in lupus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:147-156. [PMID: 35880241 PMCID: PMC9242525 DOI: 10.2478/rir-2021-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 04/11/2023]
Abstract
T regulatory cells (Tregs) have a key role in the maintenance of immune homeostasis and the regulation of immune tolerance by preventing the inflammation and suppressing the autoimmune responses. Numerical and functional deficits of these cells have been reported in systemic lupus erythematosus (SLE) patients and mouse models of SLE, where their imbalance and dysregulated activities have been reported to significantly influence the disease pathogenesis, progression and outcomes. Most studies in SLE have focused on CD4+ Tregs and it has become clear that a critical role in the control of immune tolerance after the breakdown of self-tolerance is provided by CD8+ Tregs. Here we review the role, cellular and molecular phenotypes, and mechanisms of action of CD8+ Tregs in SLE, including ways to induce these cells for immunotherapeutic modulation in SLE.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bevra H. Hahn
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
27
|
Wyrożemski Ł, Qiao SW. Immunobiology and conflicting roles of the human CD161 receptor in T cells. Scand J Immunol 2021; 94:e13090. [PMID: 35611672 DOI: 10.1111/sji.13090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/29/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022]
Abstract
Human C-type lectin-like CD161 is a type-II transmembrane protein expressed on the surface of various lymphocytes across innate and adaptive immune systems. CD161+ T cells displayed enhanced ability to produce cytokines and were shown to be enriched in the gut. Independently of function, CD161 was used as marker of innate-like T cells and marker of IL-17-producing cells. The function of CD161 is still not fully understood. In T cells, CD161 was proposed to act as co-signalling receptor that influence T-cell receptor-dependent responses. However, conflicting studies were published demonstrating lack of agreement over the role of CD161 during T-cell activation. In this review, we outline phenotypical and functional consequences of CD161 expression in T cells. We provide critical discussion over the most pressing issues including in depth evaluation of the literature concerning CD161 putative co-signalling properties.
Collapse
Affiliation(s)
- Łukasz Wyrożemski
- K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway.,Department of Immunology, University of Oslo, Oslo, Norway
| | - Shuo-Wang Qiao
- K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway.,Department of Immunology, University of Oslo, Oslo, Norway.,Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
28
|
PD-L1 signaling on human memory CD4+ T cells induces a regulatory phenotype. PLoS Biol 2021; 19:e3001199. [PMID: 33901179 PMCID: PMC8101994 DOI: 10.1371/journal.pbio.3001199] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 05/06/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) is expressed on T cells upon T cell receptor (TCR) stimulation. PD-1 ligand 1 (PD-L1) is expressed in most tumor environments, and its binding to PD-1 on T cells drives them to apoptosis or into a regulatory phenotype. The fact that PD-L1 itself is also expressed on T cells upon activation has been largely neglected. Here, we demonstrate that PD-L1 ligation on human CD25-depleted CD4+ T cells, combined with CD3/TCR stimulation, induces their conversion into highly suppressive T cells. Furthermore, this effect was most prominent in memory (CD45RA−CD45RO+) T cells. PD-L1 engagement on T cells resulted in reduced ERK phosphorylation and decreased AKT/mTOR/S6 signaling. Importantly, T cells from rheumatoid arthritis patients exhibited high basal levels of phosphorylated ERK and following PD-L1 cross-linking both ERK signaling and the AKT/mTOR/S6 pathway failed to be down modulated, making them refractory to the acquisition of a regulatory phenotype. Altogether, our results suggest that PD-L1 signaling on memory T cells could play an important role in resolving inflammatory responses; maintaining a tolerogenic environment and its failure could contribute to ongoing autoimmunity. This study shows that programmed death cell receptor ligand 1 (PD-L1) signaling in memory CD4+ T cells from healthy individuals induces a regulatory phenotype; this mechanism seems to be defective in equivalent T cells from rheumatoid arthritis patients and could be in part responsible for the pathology.
Collapse
|
29
|
Opstelten R, Amsen D. Separating the wheat from the chaff: Making sense of Treg heterogeneity for better adoptive cellular therapy. Immunol Lett 2021; 239:96-112. [PMID: 33676975 DOI: 10.1016/j.imlet.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Regulatory T (Treg) cells are essential for immunological tolerance and can be used to suppress unwanted or excessive immune responses through adoptive cellular therapy. It is increasingly clear that many subsets of Treg cells exist, which have different functions and reside in different locations. Treg cell therapies may benefit from tailoring the selected subset to the tissue that must be protected as well as to characteristics of the immune response that must be suppressed, but little attention is given to this topic in current therapies. Here, we will discuss how three major axes of heterogeneity can be discerned among the Treg cell population, which determine function and lineage fidelity. A first axis relates to the developmental route, as Treg cells can be generated from immature T cells in the thymus or from already mature Tconv cells in the immunological periphery. Heterogeneity furthermore stems from activation history (naïve or effector) and location (lymphoid or peripheral tissues). Each of these axes bestows specific properties on Treg cells, which are further refined by additional processes leading to yet further variation. A critical aspect impacting on Treg cell heterogeneity is TCR specificity, which determines when and where Treg cells are generated as well as where they exhibit their effector functions. We will discuss the implications of this heterogeneity and the role of the TCR for the design of next generation adoptive cellular therapy with Treg cells.
Collapse
Affiliation(s)
- Rianne Opstelten
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Derk Amsen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
30
|
Yang R, Peng Y, Pi J, Liu Y, Yang E, Shen X, Yao L, Shen L, Modlin RL, Shen H, Sha W, Chen ZW. A CD4+CD161+ T-Cell Subset Present in Unexposed Humans, Not Tb Patients, Are Fast Acting Cells That Inhibit the Growth of Intracellular Mycobacteria Involving CD161 Pathway, Perforin, and IFN-γ/Autophagy. Front Immunol 2021; 12:599641. [PMID: 33732233 PMCID: PMC7959736 DOI: 10.3389/fimmu.2021.599641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/07/2021] [Indexed: 12/30/2022] Open
Abstract
It remains undefined whether a subset of CD4+ T cells can function as fast-acting cells to control Mycobacterium tuberculosis (Mtb) infection. Here we show that the primary CD4+CD161+ T-cell subset, not CD4+CD161-, in unexposed healthy humans fast acted as unconventional T cells capable of inhibiting intracellular Mtb and BCG growth upon exposure to infected autologous and allogeneic macrophages or lung epithelial A549 cells. Such inhibition coincided with the ability of primary CD4+CD161+ T cells to rapidly express/secrete anti-TB cytokines including IFN-γ, TNF-α, IL-17, and perforin upon exposure to Mtb. Mechanistically, blockades of CD161 pathway, perforin or IFN-γ by blocking mAbs abrogated the ability of CD4+CD161+ T cells to inhibit intracellular mycobacterial growth. Pre-treatment of infected macrophages with inhibitors of autophagy also blocked the CD4+CD161+ T cell-mediated growth inhibition of mycobacteria. Furthermore, adoptive transfer of human CD4+CD161+ T cells conferred protective immunity against mycobacterial infection in SCID mice. Surprisingly, CD4+CD161+ T cells in TB patients exhibited a loss or reduction of their capabilities to produce perforin/IFN-γ and to inhibit intracellular growth of mycobacteria in infected macrophages. These immune dysfunctions were consistent with PD1/Tim3 up-regulation on CD4+CD161+ T cells in active tuberculosis patients, and the blockade of PD1/Tim3 on this subset cells enhanced the inhibition of intracellular mycobacteria survival. Thus, these findings suggest that a fast-acting primary CD4+CD161+T-cell subset in unexposed humans employs the CD161 pathway, perforin, and IFN-γ/autophagy to inhibit the growth of intracellular mycobacteria, thereby distinguishing them from the slow adaptive responses of conventional CD4+ T cells. The presence of fast-acting CD4+CD161+ T-cell that inhibit mycobacterial growth in unexposed humans but not TB patients also implicates the role of these cells in protective immunity against initial Mtb infection.
Collapse
Affiliation(s)
- Rui Yang
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China.,Wuhan YZY Biopharma Co., Ltd, Biolake, Wuhan, China
| | - Ying Peng
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Jiang Pi
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, United States
| | - Yidian Liu
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Enzhuo Yang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, United States
| | - Xiaona Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Lan Yao
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Ling Shen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, United States
| | - Robert L Modlin
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Wei Sha
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Zheng W Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, United States
| |
Collapse
|
31
|
Kim DH, Kim HY, Cho S, Yoo SJ, Kim WJ, Yeon HR, Choi K, Choi JM, Kang SW, Lee WW. Induction of the IL-1RII decoy receptor by NFAT/FOXP3 blocks IL-1β-dependent response of Th17 cells. eLife 2021; 10:61841. [PMID: 33507149 PMCID: PMC7872515 DOI: 10.7554/elife.61841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/28/2021] [Indexed: 12/29/2022] Open
Abstract
Derived from a common precursor cell, the balance between Th17 and Treg cells must be maintained within immune system to prevent autoimmune diseases. IL-1β-mediated IL-1 receptor (IL-1R) signaling is essential for Th17-cell biology. Fine-tuning of IL-1R signaling is controlled by two receptors, IL-1RI and IL-RII, IL-1R accessory protein, and IL-1R antagonist. We demonstrate that the decoy receptor, IL-1RII, is important for regulating IL-17 responses in TCR-stimulated CD4+ T cells expressing functional IL-1RI via limiting IL-1β responsiveness. IL-1RII expression is regulated by NFAT via its interaction with Foxp3. The NFAT/FOXP3 complex binds to the IL-1RII promoter and is critical for its transcription. Additionally, IL-1RII expression is dysregulated in CD4+ T cells from patients with rheumatoid arthritis. Thus, differential expression of IL-1Rs on activated CD4+ T cells defines unique immunological features and a novel molecular mechanism underlies IL-1RII expression. These findings shed light on the modulatory effects of IL-1RII on Th17 responses.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hee Young Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute and Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sunjung Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Su-Jin Yoo
- Department of Internal Medicine, Chungnam National University School of Medicine, 282 Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | - Won-Ju Kim
- Department of Life Science, College of Natural Sciences and Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Hye Ran Yeon
- Department of Biochemistry and Molecular Biology, Department of Biomedical Sciences, and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyungho Choi
- Department of Biochemistry and Molecular Biology, Department of Biomedical Sciences, and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences and Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Seong Wook Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, 282 Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute and Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine; Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| |
Collapse
|
32
|
Shevyrev D, Tereshchenko V, Kozlov V, Sizikov A, Chumasova O, Koksharova V. T-regulatory cells from patients with rheumatoid arthritis retain suppressor functions in vitro. Exp Ther Med 2021; 21:209. [PMID: 33500700 PMCID: PMC7818555 DOI: 10.3892/etm.2021.9641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic disease of connective tissue caused by intolerance to self-antigens. Regulatory T cells (Tregs) are key players in maintaining autotolerance through a variety of suppressor mechanisms. RA is generally believed to develop due to disorders in Tregs; however, there is no consensus on this issue. Thus, the present study focused on phenotypical analysis of Treg cells and their ability to suppress CD4+ and CD8+ cell proliferation. The present study used peripheral blood samples from 21 patients with RA and 22 healthy donors. The CD25+FoxP3+ subpopulation of Tregs was analyzed using flow cytometry to evaluate the expression of CTLA-4, PD-L1, HLA-DR, CCR4, CD86 and RORyt. Tregs suppressor activity was calculated in terms of suppression of the proliferation of CD4+ and CD8+ lymphocytes in vitro. Suppressor activity of the total Treg population did not differ between patients with RA and healthy donors. However, the patients had elevated CD25loFoxP3+ levels and lower CD25hiFoxP3+ levels; in addition, they had more activated Tregs expressing PD-L1, HLA-DR, CCR4 and CD86. The surface expression of CTLA-4 was below the reference level. The patients also had transitional FoxP3+RORyt+ cells and elevated CD4+RORyt+ levels, which were highly correlated with disease activity. These results show that in RA, Treg cells are activated and have an immunosuppressive activity. However, it is the transitional FoxP3+RORyt+ cells and increased CD4+RORyt+ percentages in peripheral blood that appear to be associated with the pathological conversion of some Treg cells into Th-17. This process appears to be key in RA pathogenesis.
Collapse
Affiliation(s)
- Daniil Shevyrev
- Laboratory of Clinical Immunopathology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia
| | - Valeriy Tereshchenko
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia
| | - Vladimir Kozlov
- Laboratory of Clinical Immunopathology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia
| | - Alexey Sizikov
- Rheumatology Department, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia
| | - Oksana Chumasova
- Rheumatology Department, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia
| | - Veroniсa Koksharova
- Rheumatology Department, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia
| |
Collapse
|
33
|
Janssens I, Cools N. Regulating the regulators: Is introduction of an antigen-specific approach in regulatory T cells the next step to treat autoimmunity? Cell Immunol 2020; 358:104236. [PMID: 33137651 DOI: 10.1016/j.cellimm.2020.104236] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/22/2022]
Abstract
In autoimmunity, the important and fragile balance between immunity and tolerance is disturbed, resulting in abnormal immune responses to the body's own tissues and cells. CD4+CD25hiFoxP3+ regulatory T cells (Tregs) induce peripheral tolerance in vivo by means of direct cell-cell contact and release of soluble factors, or indirectly through antigen-presenting cells (APC), thereby controlling auto-reactive effector T cells. Based on these unique capacities of Tregs, preclinical studies delivered proof-of-principle for the clinical use of Tregs for the treatment of autoimmune diseases. To date, the first clinical trials using ex vivo expanded polyclonal Tregs have been completed. These pioneering studies demonstrate the feasibility of generating large numbers of polyclonal Tregs in a good manufacturing practices (GMP)-compliant manner, and that infusion of Tregs is well tolerated by patients with no evidence of general immunosuppression. Nonetheless, only modest clinical results were observed, arguing that a more antigen-specific approach might be needed to foster a durable patient-specific clinical cell therapy without the risk for general immunosuppression. In this review, we discuss current knowledge, applications and future goals of adoptive immune-modulatory Treg therapy for the treatment of autoimmune disease and transplant rejection. We describe the key advances and prospects of the potential use of T cell receptor (TCR)- and chimeric antigen receptor (CAR)-engineered Tregs in future clinical applications. These approaches could deliver the long-awaited breakthrough in stopping undesired autoimmune responses and transplant rejections.
Collapse
Affiliation(s)
- Ibo Janssens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
34
|
Yi G, Zhao Y, Xie F, Zhu F, Wan Z, Wang J, Wang X, Gao K, Cao L, Li X, Chen C, Kuang Y, Qiu X, Yang H, Wang J, Su B, Chen L, Zhang W, Hou Y, Xu X, He Y, Tsun A, Liu X, Li B. Single-cell RNA-seq unveils critical regulators of human FOXP3 + regulatory T cell stability. Sci Bull (Beijing) 2020; 65:1114-1124. [PMID: 36659163 DOI: 10.1016/j.scib.2020.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/09/2019] [Accepted: 10/10/2019] [Indexed: 01/21/2023]
Abstract
The heterogeneity and plasticity of T lymphocytes is critical for determining immune response outcomes. Functional regulatory T (Treg) cells are commonly characterized by stable FOXP3 expression and have reported to exhibit heterogeneous phenotypes under inflammatory conditions. However, the interplay between inflammation and Treg cell suppressive activity still remains elusive. Here, we utilized single-cell RNA sequencing to investigate how human Treg cells respond to the pro-inflammatory cytokine interleukin-6 (IL-6). We observed that Treg cells divided into two subpopulations after IL-6 stimulation. TIGIT- unstable Treg cells lost FOXP3 expression and gained an effector-like T cell phenotype, whereas TIGIT+ Treg cells retained robust suppressive function. Single cell transcriptome analysis revealed a spectrum of cellular states of IL-6-stimulated Treg cells and how cytochrome P450 family 1 subfamily A member 1 (CYP1A1) is a crucial regulator of Treg cell suppressive capability and stability. CYP1A1-deficient human Treg cells developed a Th17-like phenotype after IL-6 stimulation. Our findings implicate CYP1A1 as a previously unidentified regulator of Treg cells that may have target potential for clinical application for biotherapies.
Collapse
Affiliation(s)
- Gang Yi
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Biotheus Inc., Zhuhai 519080, China
| | - Yi Zhao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; BGI-Shenzhen, Shenzhen 518083, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Feng Xie
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fuxiang Zhu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ziyun Wan
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Xie Wang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Kai Gao
- BGI-Shenzhen, Shenzhen 518083, China
| | - Lixia Cao
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Chen Chen
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yashu Kuang
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510006, China
| | - Xiu Qiu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510006, China
| | | | - Jian Wang
- James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Bing Su
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Chen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei Zhang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yong Hou
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yinyan He
- Department of Obstetrics and Gynaecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | | | - Xiao Liu
- BGI-Shenzhen, Shenzhen 518083, China.
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
35
|
Opstelten R, de Kivit S, Slot MC, van den Biggelaar M, Iwaszkiewicz-Grześ D, Gliwiński M, Scott AM, Blom B, Trzonkowski P, Borst J, Cuadrado E, Amsen D. GPA33: A Marker to Identify Stable Human Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:3139-3148. [DOI: 10.4049/jimmunol.1901250] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/16/2020] [Indexed: 12/16/2022]
|
36
|
Zhu L, Song H, Zhang L, Meng H. Characterization of IL-17-producing Treg cells in type 2 diabetes patients. Immunol Res 2020; 67:443-449. [PMID: 31713831 DOI: 10.1007/s12026-019-09095-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The association between type 2 diabetes (T2D) pathogenesis and immune-mediated tissue damage and insulin resistance suggests that T2D patients might benefit from the suppression of pathogenic inflammation. Foxp3+ Treg cells are crucial suppressors of inflammation, but the differentiation of Foxp3+ Treg cells is not static and is subject to conversion into IL-17-producing Th17-like cells upon receiving external signals. In this study, we examined the production of IL-17 by Treg cells. Compared to non-T2D controls, T2D patients presented significantly higher levels of IL-17-expressing cells in both Foxp3- CD4 T cells and Foxp3+ Treg cells. The frequencies of IL-17-nonexpressing Foxp3+ Treg cells, on the other hand, were not changed. Interestingly, IL-17-expressing Foxp3+ Treg cells were mutually exclusive from IL-10-expressing and TGF-β-expressing Foxp3+ Treg cells, suggesting that multiple subpopulations exist within the Foxp3+ Treg cells from T2D patients. In T2D patients, the frequencies of IL-17-expressing Foxp3+ Treg cells were positively correlated with the body mass index (BMI) and the HbA1c levels of T2D patients. The frequencies of IL-10-expressing Treg cells, on the other hand, were inversely associated with the BMI of both non-T2D controls and T2D patients. In addition, the suppressive activity of Treg cells was significantly lower in T2D patients than in non-T2D controls. Together, our study uncovered a dysregulation in Foxp3+ Treg cells from T2D patients, characterized by high IL-17 expression and low suppression activity.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Endocrinology, Shandong Provincial Third Hospital, 11 Wuyingshan Middle Road, Jinan, Shandong, China.
| | - Haihan Song
- DICAT Biomedical Computation Centre, Vancouver, British Columbia, Canada
| | - Li Zhang
- Department of Endocrinology, Shandong Provincial Third Hospital, 11 Wuyingshan Middle Road, Jinan, Shandong, China
| | - Haiyan Meng
- Department of Cardiology, Shandong Provincial Third Hospital, 11 Wuyingshan Middle Road, Jinan, Shandong, China.
| |
Collapse
|
37
|
Suwandi JS, Laban S, Vass K, Joosten A, van Unen V, Lelieveldt BP, Höllt T, Zwaginga JJ, Nikolic T, Roep BO. Multidimensional analyses of proinsulin peptide-specific regulatory T cells induced by tolerogenic dendritic cells. J Autoimmun 2020; 107:102361. [DOI: 10.1016/j.jaut.2019.102361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/06/2019] [Accepted: 11/06/2019] [Indexed: 12/18/2022]
|
38
|
Jia L, Wu R, Han N, Fu J, Luo Z, Guo L, Su Y, Du J, Liu Y. Porphyromonas gingivalis and Lactobacillus rhamnosus GG regulate the Th17/Treg balance in colitis via TLR4 and TLR2. Clin Transl Immunology 2020; 9:e1213. [PMID: 33282294 PMCID: PMC7685903 DOI: 10.1002/cti2.1213] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 03/13/2020] [Accepted: 10/21/2020] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES CD4+ T cells are the key to many immune-inflammatory diseases mediated by microbial disorders, especially inflammatory bowel disease (IBD). The purpose of this study was to explore how pathogenic and probiotic bacteria directly affect the T helper (Th)17 and T regulatory (Treg) cell balance among CD4+ T cells to regulate inflammation. METHODS Porphyromonas gingivalis (Pg; ATCC 33277) and Lactobacillus rhamnosus GG (LGG; CICC 6141) were selected as representative pathogenic and probiotic bacteria, respectively. Bacterial extracts were obtained via ultrasonication and ultracentrifugation. Flow cytometry, RT-qPCR, ELISAs, immunofluorescence and a Quantibody cytokine array were used. The dextran sodium sulphate (DSS)-induced colitis model was selected for verification. RESULTS The Pg ultrasonicate induced the apoptosis of CD4+ T cells and upregulated the expression of the Th17-associated transcription factor RoRγt and the production of the proinflammatory cytokines IL-17 and IL-6, but downregulated the expression of the essential Treg transcription factor Foxp3 and the production of the anti-inflammatory factors TGF-β and IL-10 via the TLR4 pathway. However, LGG extract maintained Th17/Treg homeostasis by decreasing the IL-17+ Th17 proportion and increasing the CD25+ Foxp3+ Treg proportion via the TLR2 pathway. In vivo, Pg-stimulated CD4+ T cells aggravated DSS-induced colitis by increasing the Th17/Treg ratio in the colon and lamina propria lymphocytes (LPLs), and Pg + LGG-stimulated CD4+ T cells relieved colitis by decreasing the Th17/Treg ratio via the JAK-STAT signalling pathway. CONCLUSIONS Our findings suggest that pathogenic Pg and probiotic LGG can directly regulate the Th17/Treg balance via different TLRs.
Collapse
Affiliation(s)
- Lu Jia
- Laboratory of Tissue Regeneration and Immunology and Department of PeriodonticsBeijing Key Laboratory of Tooth Regeneration and Function ReconstructionSchool of StomatologyCapital Medical UniversityBeijingChina
| | - Ruiqing Wu
- Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Nannan Han
- Laboratory of Tissue Regeneration and Immunology and Department of PeriodonticsBeijing Key Laboratory of Tooth Regeneration and Function ReconstructionSchool of StomatologyCapital Medical UniversityBeijingChina
| | - Jingfei Fu
- Laboratory of Tissue Regeneration and Immunology and Department of PeriodonticsBeijing Key Laboratory of Tooth Regeneration and Function ReconstructionSchool of StomatologyCapital Medical UniversityBeijingChina
| | - Zhenhua Luo
- Laboratory of Tissue Regeneration and Immunology and Department of PeriodonticsBeijing Key Laboratory of Tooth Regeneration and Function ReconstructionSchool of StomatologyCapital Medical UniversityBeijingChina
| | - Lijia Guo
- Department of OrthodonticsSchool of StomatologyCapital Medical UniversityBeijingChina
| | - Yingying Su
- Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of PeriodonticsBeijing Key Laboratory of Tooth Regeneration and Function ReconstructionSchool of StomatologyCapital Medical UniversityBeijingChina
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of PeriodonticsBeijing Key Laboratory of Tooth Regeneration and Function ReconstructionSchool of StomatologyCapital Medical UniversityBeijingChina
| |
Collapse
|
39
|
Salmani A, Mohammadi M, Farid Hosseini R, Tavakol Afshari J, Fouladvand A, Dehnavi S, Khoshkhooi M, Jabbari Azad F. A significant increase in expression of FOXP3 and IL-17 genes in patients with allergic rhinitis underwent accelerated rush immunotherapy. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:989-996. [PMID: 31807241 PMCID: PMC6880522 DOI: 10.22038/ijbms.2019.32979.7878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Objective(s): Allergic rhinitis (AR) is a common hypersensitivity disease worldwide. Immunotherapy has been performed as the best treatment for years. This study aimed to study the gene expression pattern of immune system cells following an accelerated rush immunotherapy protocol (ARIT) in patients with AR. Materials and Methods: Fifteen patients with AR (15–55 years old) resident in Mashhad, Iran, with positive prick test to regional aeroallergens (weed mix, grass mix, tree mix, and Salsola) enrolled in this study. All patients were treated for three months with 3-day ARIT protocol between July 2015 and August 2016. Clinical symptoms and quality of life were recorded by two questioners. The expression levels of FOXP3, TGF-β, IL-10, IL-17, IL-4, and IFN-γ genes in patient’s peripheral blood mononuclear cells were evaluated by SYBR Green real-time RT-PCR technique. Results: The severity of disease and quality of life showed significant improvement following ARIT (P-value<0.05). Gene expression of IFN-γ and IL-10 was increased whereas TGF-β and IL-4 down-regulated, following ARIT, but these changes were not significant. However, gene expression of FOXP3 and IL-17 was significantly increased after intervention when compared with the baseline (P-value< 0.002). Conclusion: Significant up-regulation of FOXP3 and IL-17 genes, additionally, a significant improvement in the clinical signs following ARIT might be related to increases in HLA-DR- and FOXP3+ Treg population at the initiation phase of ARIT. Employing the flow cytometry technique to study the phenotype of these cells is suggested for future studies.
Collapse
Affiliation(s)
- Amirabbas Salmani
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Farid Hosseini
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Fouladvand
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Dehnavi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Khoshkhooi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
40
|
Chen Q, Lv J, Yang W, Xu B, Wang Z, Yu Z, Wu J, Yang Y, Han Y. Targeted inhibition of STAT3 as a potential treatment strategy for atherosclerosis. Theranostics 2019; 9:6424-6442. [PMID: 31588227 PMCID: PMC6771242 DOI: 10.7150/thno.35528] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is the main pathological basis of ischemic cardiovascular and cerebrovascular diseases and has attracted more attention in recent years. Multiple studies have demonstrated that the signal transducer and activator of transcription 3 (STAT3) plays essential roles in the process of atherosclerosis. Moreover, aberrant STAT3 activation has been shown to contribute to the occurrence and development of atherosclerosis. Therefore, the study of STAT3 inhibitors has gradually become a focal research topic. In this review, we describe the crucial roles of STAT3 in endothelial cell dysfunction, macrophage polarization, inflammation, and immunity during atherosclerosis. STAT3 in mitochondria is mentioned as well. Then, we present a summary and classification of STAT3 inhibitors, which could offer potential treatment strategies for atherosclerosis. Furthermore, we enumerate some of the problems that have interfered with the development of mature therapies utilizing STAT3 inhibitors to treat atherosclerosis. Finally, we propose ideas that may help to solve these problems to some extent. Collectively, this review may be useful for developing future STAT3 inhibitor therapies for atherosclerosis.
Collapse
|
41
|
Ma F, Li S, Gao X, Zhou J, Zhu X, Wang D, Cai Y, Li F, Yang Q, Gu X, Ge W, Liu H, Xiao X, Hao H. Interleukin-6-mediated CCR9 + interleukin-17-producing regulatory T cells polarization increases the severity of necrotizing enterocolitis. EBioMedicine 2019; 44:71-85. [PMID: 31129099 PMCID: PMC6604880 DOI: 10.1016/j.ebiom.2019.05.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 01/03/2023] Open
Abstract
Background Increased frequency of CCR9+ CD4+ T cells in peripheral blood is linked to several gastrointestinal inflammatory diseases; however, its relationship with necrotizing enterocolitis (NEC) is not understood. We investigated whether the frequencies of CCR9+ CD4+ T cells and related subsets were increased in peripheral blood of both patients and mice with NEC. Methods CCR9+ CD4+ T cells and related subsets were evaluated by flow cytometry in peripheral blood collected from both patients and mice with NEC and controls. The suppressive function of CCR9+ regulatory T (Treg) cells in NEC was assessed via in vitro suppression assay. An in vitro T cell polarization assay was performed to investigate the role of proinflammatory cytokines in Treg cell polarization. In vivo Treg cell polarization analysis was performed using NEC mice treated with anti-interleukin-6 (IL-6) receptor antibody. Findings A higher proportion of CCR9+ CD4+ T cells occurred in peripheral blood of both patients and mice with NEC than in controls. Elevated CCR9+ CD4+ T cells were primarily CCR9+ IL-17-producing Treg cells, possessing features of conventional Treg cells, but their suppressive activity was seriously impaired and negatively correlated with the severity of intestinal tissue injury. IL-6 promoted polarization of CCR9+ Treg cells to CCR9+ IL-17-producing Treg cells, and blocking IL-6 signalling inhibited this conversion in vitro and ameliorated experimental NEC in vivo. Interpretation Collectively, these data suggested that CCR9+ IL-17-producing Treg cells may be a biomarker of severity and highlight the possibility that antibodies targeting IL-6R could ameliorate NEC by modulating lymphocyte balance. Fund This work was supported by the Science and Technology Planning Project of Guangdong Province, China (2017A020215100), the Science and Technology Foundation of Guangzhou, China (201704020086 and 201604020154), the Medical Scientific Research Foundation of Guangdong Province, China (A2017304 and A2014704), and the Social Science and Technology Development Foundation of Dongguan, China (2016108101037).
Collapse
Affiliation(s)
- Fei Ma
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sitao Li
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Gao
- Department of Neonatology, the Foshan Women and Children Hospital, Foshan, China
| | - Jialiang Zhou
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xiaochun Zhu
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Desheng Wang
- Department of Neonatology, the Fifth People's Hospital of Dongguan, Dongguan, China
| | - Yao Cai
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fei Li
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiuping Yang
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xia Gu
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wuping Ge
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Huanliang Liu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Xin Xiao
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Hu Hao
- Department of Neonatology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
42
|
Goldberg R, Scotta C, Cooper D, Nissim-Eliraz E, Nir E, Tasker S, Irving PM, Sanderson J, Lavender P, Ibrahim F, Corcoran J, Prevost T, Shpigel NY, Marelli-Berg F, Lombardi G, Lord GM. Correction of Defective T-Regulatory Cells From Patients With Crohn's Disease by Ex Vivo Ligation of Retinoic Acid Receptor-α. Gastroenterology 2019; 156:1775-1787. [PMID: 30710527 DOI: 10.1053/j.gastro.2019.01.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/07/2019] [Accepted: 01/11/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Crohn's disease (CD) is characterized by an imbalance of effector and regulatory T cells in the intestinal mucosa. The efficacy of anti-adhesion therapies led us to investigate whether impaired trafficking of T-regulatory (Treg) cells contributes to the pathogenesis of CD. We also investigated whether proper function could be restored to Treg cells by ex vivo expansion in the presence of factors that activate their regulatory activities. METHODS We measured levels of the integrin α4β7 on Treg cells isolated from peripheral blood or lamina propria of patients with CD and healthy individuals (controls). Treg cells were expanded ex vivo and incubated with rapamycin with or without agonists of the retinoic acid receptor-α (RARA), and their gene expression profiles were analyzed. We also studied the cells in cytokine challenge, suppression, and flow chamber assays and in SCID mice with human intestinal xenografts. RESULTS We found that Treg cells from patients with CD express lower levels of the integrin α4β7 than Treg cells from control patients. The pathway that regulates the expression of integrin subunit α is induced by retinoic acid (RA). Treg cells from patients with CD incubated with rapamycin and an agonist of RARA (RAR568) expressed high levels of integrin α4β7, as well as CD62L and FOXP3, compared with cells incubated with rapamycin or rapamycin and all-trans retinoic acid. These Treg cells had increased suppressive activities in assays and migrated under conditions of shear flow; they did not produce inflammatory cytokines, and RAR568 had no effect on cell stability or lineage commitment. Fluorescently labeled Treg cells incubated with RAR568 were significantly more likely to traffic to intestinal xenografts than Treg cells expanded in control medium. CONCLUSIONS Treg cells from patients with CD express lower levels of the integrin α4β7 than Treg cells from control patients. Incubation of patients' ex vivo expanded Treg cells with rapamycin and an RARA agonist induced expression of α4β7 and had suppressive and migratory activities in culture and in intestinal xenografts in mice. These cells might be developed for treatment of CD. ClinicalTrials.gov, Number: NCT03185000.
Collapse
Affiliation(s)
- Rimma Goldberg
- Inflammatory Bowel Disease Unit, Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, UK; School of Immunology and Microbial Sciences, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, Guy's and St Thomas' NHS Trust and King's College London, London, UK
| | - Cristiano Scotta
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Dianne Cooper
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Einat Nissim-Eliraz
- Department of Respiratory Medicine and Allergy, King's College London, London, UK
| | - Eilam Nir
- Department of Respiratory Medicine and Allergy, King's College London, London, UK
| | - Scott Tasker
- School of Immunology and Microbial Sciences, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, Guy's and St Thomas' NHS Trust and King's College London, London, UK
| | - Peter M Irving
- Inflammatory Bowel Disease Unit, Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jeremy Sanderson
- Inflammatory Bowel Disease Unit, Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Paul Lavender
- Department of Respiratory Medicine and Allergy, King's College London, London, UK
| | - Fowzia Ibrahim
- Department of Rheumatology, King's College London School of Medicine, Weston Education Centre, King's College London, London, UK
| | - Jonathan Corcoran
- Wolfson Centre for Age Related Diseases, King's College London, London, UK
| | - Toby Prevost
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - Nahum Y Shpigel
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | | | - Giovanna Lombardi
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, Guy's and St Thomas' NHS Trust and King's College London, London, UK.
| |
Collapse
|
43
|
Néel A, Bucchia M, Néel M, Tilly G, Caristan A, Yap M, Rimbert M, Bruneau S, Cadoux M, Agard C, Hourmant M, Godmer P, Brouard S, Bressollette C, Hamidou M, Josien R, Fakhouri F, Degauque N. Dampening of CD8+ T Cell Response by B Cell Depletion Therapy in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Rheumatol 2019; 71:641-650. [PMID: 30375745 DOI: 10.1002/art.40766] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/25/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To compare the effects of rituximab (RTX) and conventional immunosuppressants (CIs) on CD4+ T cells, Treg cells, and CD8+ T cells in antineutrophil cytoplasmic antibody-associated vasculitis (AAV). METHODS A thorough immunophenotype analysis of CD4+, Treg, and CD8+ cells from 51 patients with AAV was performed. The production of cytokines and chemokines by CD8+ T cells stimulated in vitro was assessed using a multiplex immunoassay. The impact of AAV B cells on CD8+ T cell response was assessed using autologous and heterologous cocultures. RESULTS CD4+ and Treg cell subsets were comparable among RTX-treated and CI-treated patients. In contrast, within the CD8+ T cell compartment, RTX, but not CIS, reduced CD45RA+CCR7- (TEMRA) cell frequency (from a median of 39% before RTX treatment to 10% after RTX treatment [P < 0.01]) and efficiently dampened cytokine/chemokine production (e.g., the median macrophage inflammatory protein 1α level was 815 pg/ml in patients treated with RTX versus 985 pg/ml in patients treated with CIs versus 970 pg/ml in those with active untreated AAV [P < 0.01]). CD8+ T cell subsets cocultured with autologous B cells produced more proinflammatory cytokines in AAV patients than in controls (e.g., for tumor necrosis factor-producing effector memory CD8+ T cells: 14% in AAV patients versus 9.2% in controls [P < 0.05]). In vitro disruption of AAV B cell-CD8+ T cell cross-talk reduced CD8+ T cell cytokine production, mirroring the reduced CD8+ response observed ex vivo after RTX treatment. CONCLUSION The disruption of a pathogenic B cell/CD8+ T cell axis may contribute to the efficacy of RTX in AAV. Further studies are needed to determine the value of CD8+ T cell immunomonitoring in B cell-targeted therapies.
Collapse
Affiliation(s)
- Antoine Néel
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Marie Bucchia
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Mélanie Néel
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Gaelle Tilly
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Aurélie Caristan
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | | | - Marie Rimbert
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Sarah Bruneau
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Marion Cadoux
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | | | - Maryvonne Hourmant
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes France, and Centre Hospitalier Bretagne Atlantique, Vannes, France
| | - Pascal Godmer
- Centre Hospitalier Bretagne Atlantique, Vannes, France
| | - Sophie Brouard
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | | | - Mohamed Hamidou
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Regis Josien
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Fadi Fakhouri
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| | - Nicolas Degauque
- INSERM UMR1064, Université de Nantes and CHU Nantes, Nantes, France
| |
Collapse
|
44
|
A clinically meaningful metric of immune age derived from high-dimensional longitudinal monitoring. Nat Med 2019; 25:487-495. [PMID: 30842675 DOI: 10.1038/s41591-019-0381-y] [Citation(s) in RCA: 262] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 01/07/2019] [Indexed: 01/08/2023]
Abstract
Immune responses generally decline with age. However, the dynamics of this process at the individual level have not been characterized, hindering quantification of an individual's immune age. Here, we use multiple 'omics' technologies to capture population- and individual-level changes in the human immune system of 135 healthy adult individuals of different ages sampled longitudinally over a nine-year period. We observed high inter-individual variability in the rates of change of cellular frequencies that was dictated by their baseline values, allowing identification of steady-state levels toward which a cell subset converged and the ordered convergence of multiple cell subsets toward an older adult homeostasis. These data form a high-dimensional trajectory of immune aging (IMM-AGE) that describes a person's immune status better than chronological age. We show that the IMM-AGE score predicted all-cause mortality beyond well-established risk factors in the Framingham Heart Study, establishing its potential use in clinics for identification of patients at risk.
Collapse
|
45
|
Viisanen T, Gazali AM, Ihantola EL, Ekman I, Näntö-Salonen K, Veijola R, Toppari J, Knip M, Ilonen J, Kinnunen T. FOXP3+ Regulatory T Cell Compartment Is Altered in Children With Newly Diagnosed Type 1 Diabetes but Not in Autoantibody-Positive at-Risk Children. Front Immunol 2019; 10:19. [PMID: 30723474 PMCID: PMC6349758 DOI: 10.3389/fimmu.2019.00019] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/04/2019] [Indexed: 01/11/2023] Open
Abstract
The dysfunction of FOXP3-positive regulatory T cells (Tregs) plays a key role in the pathogenesis of autoimmune diseases, including type 1 diabetes (T1D). However, previous studies analyzing the peripheral blood Treg compartment in patients with T1D have yielded partially conflicting results. Moreover, the phenotypic complexity of peripheral blood Tregs during the development of human T1D has not been comprehensively analyzed. Here, we used multi-color flow cytometry to analyze the frequency of distinct Treg subsets in blood samples from a large cohort comprising of 74 children with newly diagnosed T1D, 76 autoantibody-positive children at-risk for T1D and 180 age- and HLA-matched control children. The frequency of CD4+CD25+CD127lowFOXP3+ Tregs was higher in children with T1D compared to control children, and this change was attributable to a higher proportion of naïve Tregs in these subjects. Further longitudinal analyses demonstrated that the increase in Treg frequency correlated with disease onset. The frequencies of the minor subsets of CD25+FOXP3low memory Tregs as well as CD25lowCD127lowFOXP3+ Tregs were also increased in children with T1D. Moreover, the ratio of CCR6-CXCR3+ and CCR6+CXCR3- memory Tregs was altered and the frequency of proliferating Ki67-positive and IFN-γ producing memory Tregs was decreased in children with T1D. The frequency of CXCR5+FOXP3+ circulating follicular T regulatory cells was not altered in children with T1D. Importantly, none of the alterations observed in children with T1D were observed in autoantibody-positive at-risk children. In conclusion, our study reveals multiple alterations in the peripheral blood Treg compartment at the diagnosis of T1D that appear not to be features of early islet autoimmunity.
Collapse
Affiliation(s)
- Tyyne Viisanen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ahmad M Gazali
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Emmi-Leena Ihantola
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ilse Ekman
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | | | - Riitta Veijola
- PEDEGO Research Unit, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, Turku, Finland.,Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland.,Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland.,Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Eastern Finland Laboratory Centre (ISLAB), Kuopio, Finland
| |
Collapse
|
46
|
Human retinoic acid-regulated CD161 + regulatory T cells support wound repair in intestinal mucosa. Nat Immunol 2018; 19:1403-1414. [PMID: 30397350 DOI: 10.1038/s41590-018-0230-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 09/07/2018] [Indexed: 01/08/2023]
Abstract
Repair of tissue damaged during inflammatory processes is key to the return of local homeostasis and restoration of epithelial integrity. Here we describe CD161+ regulatory T (Treg) cells as a distinct, highly suppressive population of Treg cells that mediate wound healing. These Treg cells were enriched in intestinal lamina propria, particularly in Crohn's disease. CD161+ Treg cells had an all-trans retinoic acid (ATRA)-regulated gene signature, and CD161 expression on Treg cells was induced by ATRA, which directly regulated the CD161 gene. CD161 was co-stimulatory, and ligation with the T cell antigen receptor induced cytokines that accelerated the wound healing of intestinal epithelial cells. We identified a transcription-factor network, including BACH2, RORγt, FOSL2, AP-1 and RUNX1, that controlled expression of the wound-healing program, and found a CD161+ Treg cell signature in Crohn's disease mucosa associated with reduced inflammation. These findings identify CD161+ Treg cells as a population involved in controlling the balance between inflammation and epithelial barrier healing in the gut.
Collapse
|
47
|
Atif M, Warner S, Oo YH. Linking the gut and liver: crosstalk between regulatory T cells and mucosa-associated invariant T cells. Hepatol Int 2018; 12:305-314. [PMID: 30027532 PMCID: PMC6097019 DOI: 10.1007/s12072-018-9882-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022]
Abstract
The gut–liver axis is increasingly considered to play a vital part in the progression of chronic inflammatory gut and liver diseases. Hence, a detailed understanding of the local and systemic regulatory mechanisms is crucial to develop novel therapeutic approaches. In this review, we discuss in-depth the roles of regulatory T cells (Tregs) and mucosal-associated invariant T cells (MAITs) within the context of inflammatory bowel disease, primary sclerosing cholangitis, and non-alcoholic steatohepatitis. Tregs are crucial in maintaining peripheral tolerance and preventing autoimmunity. MAIT cells have a unique ability to rapidly recognize microbial metabolites and mount a local immune response and act as a ‘biliary firewall’ at the gut and biliary epithelial barrier. We also outline how current knowledge can be exploited to develop novel therapies to control the propagation of chronic gut- and liver-related inflammatory and autoimmune conditions. We specifically focus on the nature of the Tregs’ cell therapy product and outline an adjunctive role for low-dose IL-2. All in all, it is clear that translational immunology is at crucial crossroads. The success of ongoing clinical trials in cellular therapies for inflammatory gut and liver conditions could revolutionize the treatment of these conditions and the lives of our patients in the coming years.
Collapse
Affiliation(s)
- Muhammad Atif
- Centre for Liver Research and National Institute of Health Research Liver Biomedical Research Centre Birmingham, Institute of Immunology and lmmunotherapy, University of Birmingham, Birmingham, UK.,Academic Department of Surgery, University of Birmingham, Birmingham, UK
| | - Suz Warner
- Centre for Liver Research and National Institute of Health Research Liver Biomedical Research Centre Birmingham, Institute of Immunology and lmmunotherapy, University of Birmingham, Birmingham, UK
| | - Ye H Oo
- Centre for Liver Research and National Institute of Health Research Liver Biomedical Research Centre Birmingham, Institute of Immunology and lmmunotherapy, University of Birmingham, Birmingham, UK. .,Liver Transplant and Hepatobiliary Unit, University Hospital of Birmingham NHS Foundation Trust, Birmingham, UK.
| |
Collapse
|
48
|
van Eden W. Immune tolerance therapies for autoimmune diseases based on heat shock protein T-cell epitopes. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0531. [PMID: 29203716 DOI: 10.1098/rstb.2016.0531] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2017] [Indexed: 12/11/2022] Open
Abstract
Experimental models of autoimmune diseases have revealed the disease protective role of heat shock proteins (HSPs). Both the administration of exogenous extracellular, mostly recombinant, HSP and the experimental co-induction of endogenous intracellular HSP in models have been shown to lead to production of disease protective regulatory T cells (Tregs). Similar to HSP taken up from extracellular bodily fluids, due to stress-related autophagy upregulated HSP also from intracellular sources is a major provider for the major histocompatibility class II (MHCII) ligandome; therefore, both extracellular and intracellular HSP can be prominent targets of Treg. The development of therapeutic peptide vaccines for the restoration of immune tolerance in inflammatory diseases is an area of intensive research. In this area, HSPs are a target for tolerance-inducing T-cell therapy, because of their wide expression in inflamed tissues. In humans, in whom the actual disease trigger is frequently unknown, HSP peptides offer chances for tolerance-promoting interventions through induction of HSP-specific Treg. Recently, we have shown the ability of a bacterial HSP70-derived peptide, HSP70-B29, to induce HSP-specific Tregs that suppressed arthritis by cross-recognition of their mammalian HSP70 homologues, abundantly present in the MHCII ligandome of stressed mouse and human antigen-presenting cells in inflamed tissues.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Yalelaan 1, 3584CL Utrecht, The Netherlands
| |
Collapse
|
49
|
Ohl K, Nickel H, Moncrieffe H, Klemm P, Scheufen A, Föll D, Wixler V, Schippers A, Wagner N, Wedderburn LR, Tenbrock K. The transcription factor CREM drives an inflammatory phenotype of T cells in oligoarticular juvenile idiopathic arthritis. Pediatr Rheumatol Online J 2018; 16:39. [PMID: 29925386 PMCID: PMC6011589 DOI: 10.1186/s12969-018-0253-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Inflammatory effector T cells trigger inflammation despite increased numbers of Treg cells in the synovial joint of patients suffering from juvenile idiopathic arthritis (JIA). The cAMP response element (CREM)α is known to play a major role in regulation of T cells in SLE, colitis, and EAE. However, its role in regulation of effector T cells within the inflammatory joint is unknown. METHODS CREM expression was analyzed in synovial fluid cells from oligoarticular JIA patients by flow cytometry. Peripheral blood mononuclear cells were incubated with synovial fluid and analyzed in the presence and absence of CREM using siRNA experiments for T cell phenotypes. To validate the role of CREM in vivo, ovalbumin-induced T cell dependent arthritis experiments were performed. RESULTS CREM is highly expressed in synovial fluid T cells and its expression can be induced by treating healthy control PBMCs with synovial fluid. Specifically, CREM is more abundant in CD161+ subsets, than CD161- subsets, of T cells and contributes to cytokine expression by these cells. Finally, development of ovalbumin-induced experimental arthritis is ameliorated in mice with adoptively transferred CREM-/- T cells. CONCLUSION In conclusion, our study reveals that beyond its role in SLE T cells CREM also drives an inflammatory phenotype of T cells in JIA.
Collapse
Affiliation(s)
- Kim Ohl
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Helge Nickel
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Halima Moncrieffe
- 0000 0000 9025 8099grid.239573.9Center for Autoimmune Genomics & Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,0000 0001 2179 9593grid.24827.3bDepartment of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH USA
| | - Patricia Klemm
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Anja Scheufen
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Dirk Föll
- 0000 0004 0551 4246grid.16149.3bDepartment of Pediatric Rheumatology and Immunology, University Hospital Muenster, Muenster, Germany
| | - Viktor Wixler
- 0000 0001 2172 9288grid.5949.1Institute of Virology, Westfaelische Wilhelms University, 48149 Muenster, Germany
| | - Angela Schippers
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Norbert Wagner
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Lucy R. Wedderburn
- Arthritis Research UK Centre for Adolescent Rheumatology at UCL UCLH and GOSH, London, UK ,0000000121901201grid.83440.3bUCL GOS Institute of Child Health, University College London, London, UK ,0000 0001 2116 3923grid.451056.3NIHR- Great Ormond Street Hospital Biomedical Research Centre (BRC), London, UK
| | - Klaus Tenbrock
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074, Aachen, Germany.
| |
Collapse
|
50
|
Wu X, Tian J, Wang S. Insight Into Non-Pathogenic Th17 Cells in Autoimmune Diseases. Front Immunol 2018; 9:1112. [PMID: 29892286 PMCID: PMC5985293 DOI: 10.3389/fimmu.2018.01112] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022] Open
Abstract
Th17 cells are generally considered to be positive regulators of immune responses because they produce pro-inflammatory cytokines, including IL-17A, IL-17F, and IL-22. Cytokine production not only promotes accumulation of immune cells, such as macrophages, neutrophils and lymphocytes, at inflammatory sites but can also cause tissue pathologies. Conversely, certain Th17 cells can also negatively regulate immune responses by secreting immunosuppressive factors, such as IL-10; these cells are termed non-pathogenic Th17 cells. In this review, we summarize recent advances in the development and regulatory functions of non-pathogenic Th17 cells in autoimmune diseases.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|