1
|
Abbott V, Housden BE, Houldsworth A. Could immunotherapy and regulatory T cells be used therapeutically to slow the progression of Alzheimer's disease? Brain Commun 2025; 7:fcaf092. [PMID: 40078868 PMCID: PMC11896979 DOI: 10.1093/braincomms/fcaf092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease and other cognitive impairments are a growing problem in the healthcare world with the ageing population. There are currently no effective treatments available; however, it has been suggested that targeting neuroinflammation may be a successful approach in slowing the progression of neurodegeneration. Reducing the destructive hyperinflammatory pathology to maintain homeostasis in neural tissue is a promising option to consider. This review explores the mechanisms behind neuroinflammation and the effectiveness of immunotherapy in slowing the progression of cognitive decline in patients with Alzheimer's disease. The key components of neuroinflammation in Alzheimer's disease researched are microglia, astrocytes, cytokines and CD8+ effector T cells. The role of oxidative stress on modulating regulatory T cells and some of the limitations of regulatory T cell-based therapies are also explored. Increasing regulatory T cells can decrease activation of microglia, proinflammatory cytokines and astrocytes; however, it can also increase levels of inflammatory cytokines. There is a complex network of regulatory T cell interactions that reduce Alzheimer's disease pathology, which is not fully understood. Exploring the current literature, further research into the use of immunotherapy in Alzheimer's disease is vital to determine the potential of these techniques; however, there is sufficient evidence to suggest that increasing regulatory T cells count does prevent Alzheimer's disease symptoms and pathology in patients with Alzheimer's disease. Some exciting innovative therapies are muted to explore in the future. The function of regulatory T cells in the presence of reactive oxygen species and oxidative stress should be investigated further in patients with neurogenerative disorders to ascertain if combination therapies could reduce oxidative stress while also enhancing regulatory T cells function. Could methods of immunotherapy infuse exogenous functional Tregs or enhance the immune environment in favour of endogenous regulatory T cells differentiation, thus reducing neuroinflammation in neurodegenerative pathology, inhibiting the progression of Alzheimer's disease?
Collapse
Affiliation(s)
- Victoria Abbott
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| | - Benjamin E Housden
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Annwyne Houldsworth
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| |
Collapse
|
2
|
Dadey RE, Cui J, Rajasundaram D, Yano H, Liu C, Cohen JA, Liu AW, Kaplan DH, Workman CJ, Vignali DAA. Regulatory T cells in the tumor microenvironment display a unique chromatin accessibility profile. Immunohorizons 2025; 9:vlae014. [PMID: 39965167 PMCID: PMC11841976 DOI: 10.1093/immhor/vlae014] [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: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 02/20/2025] Open
Abstract
Regulatory T cells (Tregs) are a suppressive CD4+ T cell population that limit the antitumor immune response. In this study, we analyzed the chromatin accessibility of Tregs in the murine tumor microenvironment (TME) to identify tumor-specific accessible peaks and if these are altered over time in the tumor microenvironment, with or without anti-PD-1 immunotherapy. We found that despite little change in chromatin accessibility of Tregs in the tumor over time, Tregs have a distinct chromatin accessibility signature in the TME compared with Tregs in the periphery. This distinct tumor Treg chromatin accessibility profile highlights reduced accessibility at loci important for an CD4+ conventional T cell (CD4+ Foxp3-) effector phenotype. Analysis of chromatin accessibility in Tregs from B16 and MC38 tumor models indicated that Tregs from skin-resident tumors are most similar to naïve skin resident Tregs but still bear key differences attributable to the TME. We also found that Tregs do not alter their transcriptome or chromatin accessibility following immunotherapy. We conclude that although chromatin accessibility in Tregs is somewhat similar to their tissue residency, the TME may drive a unique chromatin accessibility profile. Treg chromatin accessibility in the tumor appears remarkably stable and unaltered by tumor type, over time, or following immunotherapy.
Collapse
Affiliation(s)
- Rebekah E Dadey
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jian Cui
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dhivyaa Rajasundaram
- Division of Health Informatics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Chang Liu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Jonathan A Cohen
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Andrew W Liu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Daniel H Kaplan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
3
|
Zhang M, Cui J, Chen H, Cheng Y, Chen Q, Zong F, Lu X, Qin L, Han Y, Kuai X, Zhang Y, Chu M, Wu S, Wu J. Increased SOAT2 expression in aged regulatory T cells is associated with altered cholesterol metabolism and reduced anti-tumor immunity. Nat Commun 2025; 16:630. [PMID: 39805872 PMCID: PMC11729894 DOI: 10.1038/s41467-025-56002-w] [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: 04/10/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Immune functions decline with aging, leading to increased susceptibility to various diseases including tumors. Exploring aging-related molecular targets in elderly patients with cancer is thus highly sought after. Here we find that an ER transmembrane enzyme, sterol O-acyltransferase 2 (SOAT2), is overexpressed in regulatory T (Treg) cells from elderly patients with lung squamous cell carcinoma (LSCC), while radiomics analysis of LSCC patients associates increased SOAT2 expression with reduced immune infiltration and poor prognosis. Mechanically, ex vivo human and mouse Treg cell data and in vivo mouse tumor models suggest that SOAT2 overexpression in Treg cells promotes cholesterol metabolism by activating the SREBP2-HMGCR-GGPP pathway, leading to enhanced Treg suppresser functions but reduced CD8+ T cell proliferation, migration, homeostasis and anti-tumor immunity. Our study thus identifies a potential mechanism responsible for altered Treg function in the context of immune aging, and also implicates SOAT2 as a potential target for tumor immunotherapy.
Collapse
Affiliation(s)
- Mingjiong Zhang
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiahua Cui
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, China
| | - Haoyan Chen
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Cheng
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiaoyu Chen
- Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Zong
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Lu
- Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
| | - Lang Qin
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Han
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xingwang Kuai
- Department of Pathology, Medical School, Nantong University, Nantong, China
| | - Yuxing Zhang
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Minjie Chu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, China.
| | - Shuangshuang Wu
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Jianqing Wu
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Granados ST, Yanushkevich S, Lok J, Concepcion AR. Analysis of Store-Operated Ca 2+ Entry in Primary T Cells. Methods Mol Biol 2025; 2904:91-113. [PMID: 40220228 DOI: 10.1007/978-1-0716-4414-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Calcium ions (Ca2+) are key second messengers for signal transduction in virtually all cells. In T cells, Ca2+ signals are generated upon T cell receptor (TCR) stimulation in a two-step integrated process known as Store-Operated Ca2+ Entry (SOCE), which involves the depletion of endoplasmic reticulum (ER) Ca2+ stores, followed by the influx of extracellular Ca2+ via Ca2+ release-activated Ca2+ (CRAC) channels. The Ca2+ influx generated by the opening of CRAC channels in T cells is essential for their metabolic reprogramming, proliferation, cytokine production, and adaptive immune response.In this book chapter, we review general concepts, discuss the rationale for using ratiometric Ca2+-sensitive chemical dyes to monitor SOCE in primary murine T cells, and weigh the advantages and disadvantages of the different methods that are currently available to detect cytosolic Ca2+ dynamics. We provide detailed protocols to measure SOCE in mouse T cells including flow cytometry, fluorescent microplate reader and single-cell imaging, and offer a general guideline on how to quantify SOCE in these cells. These protocols are easily adaptable to monitor cytosolic Ca2+ dynamics in human T cells and other cell types of interest.
Collapse
Affiliation(s)
- Sara T Granados
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Sergei Yanushkevich
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Jessica Lok
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Axel R Concepcion
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA.
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
5
|
Wang YH, Li W, McDermott M, Son GY, Maiti G, Zhou F, Tao AY, Raphael D, Moreira AL, Shen B, Vaeth M, Nadorp B, Chakravarti S, Lacruz RS, Feske S. IFN-γ-producing T H1 cells and dysfunctional regulatory T cells contribute to the pathogenesis of Sjögren's disease. Sci Transl Med 2024; 16:eado4856. [PMID: 39693412 DOI: 10.1126/scitranslmed.ado4856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/06/2024] [Accepted: 10/02/2024] [Indexed: 12/20/2024]
Abstract
Sjögren's disease (SjD) is an autoimmune disorder characterized by progressive salivary and lacrimal gland dysfunction, inflammation, and destruction, as well as extraglandular manifestations. SjD is associated with autoreactive B and T cells, but its pathophysiology remains incompletely understood. Abnormalities in regulatory T (Treg) cells occur in several autoimmune diseases, but their role in SjD is ambiguous. We had previously shown that the function and development of Treg cells depend on store-operated Ca2+ entry (SOCE), which is mediated by ORAI1 Ca2+ channels and stromal interaction protein 1 (STIM1) and STIM2. Here, we show that mice with a Foxp3+ Treg cell-specific deletion of Stim1 and Stim2 develop a phenotype that fulfills all classification criteria of human SjD. Mutant mice have salivary and lacrimal gland inflammation characterized by strong lymphocyte infiltration and transcriptional signatures dominated by T helper 1 (TH1) and interferon (IFN) signaling. CD4+ T cells from mutant mice are sufficient to induce SjD-like disease in an IFN-γ-dependent manner. Inhibition of IFN signaling with the JAK1/2 inhibitor baricitinib alleviated CD4+ T cell-induced SjD in mice. These findings are consistent with the transcriptional profiles of CD4+ T cells from patients with SjD, which indicate enhanced TH1 but reduced memory Treg cell function. Together, our study provides evidence for a critical role of dysfunctional Treg cells and IFN-γ-producing TH1 cells in the pathogenesis of SjD.
Collapse
Affiliation(s)
- Yin-Hu Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Wenyi Li
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maxwell McDermott
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ga-Yeon Son
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - George Maiti
- Department of Ophthalmology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Fang Zhou
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony Y Tao
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Dimitrius Raphael
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Andre L Moreira
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Boheng Shen
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Martin Vaeth
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Bettina Nadorp
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shukti Chakravarti
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Ophthalmology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Rodrigo S Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
6
|
Oktelik FB, Benamar M. Glycerolipids disrupt regulatory T-cells in asthma. Eur Respir J 2024; 64:2401386. [PMID: 39638361 DOI: 10.1183/13993003.01386-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/05/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Fatma Betul Oktelik
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine (Aziz Sancar DETAE), Istanbul University, Istanbul, Turkey
| | - Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Cruz de Casas P, Knöpper K, Dey Sarkar R, Kastenmüller W. Same yet different - how lymph node heterogeneity affects immune responses. Nat Rev Immunol 2024; 24:358-374. [PMID: 38097778 DOI: 10.1038/s41577-023-00965-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 05/04/2024]
Abstract
Lymph nodes are secondary lymphoid organs in which immune responses of the adaptive immune system are initiated and regulated. Distributed throughout the body and embedded in the lymphatic system, local lymph nodes are continuously informed about the state of the organs owing to a constant drainage of lymph. The tissue-derived lymph carries products of cell metabolism, proteins, carbohydrates, lipids, pathogens and circulating immune cells. Notably, there is a growing body of evidence that individual lymph nodes differ from each other in their capacity to generate immune responses. Here, we review the structure and function of the lymphatic system and then focus on the factors that lead to functional heterogeneity among different lymph nodes. We will discuss how lymph node heterogeneity impacts on cellular and humoral immune responses and the implications for vaccination, tumour development and tumour control by immunotherapy.
Collapse
Affiliation(s)
- Paulina Cruz de Casas
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Konrad Knöpper
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Rupak Dey Sarkar
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Wolfgang Kastenmüller
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
8
|
Karakus IS, Catak MC, Frohne A, Bayram Catak F, Yorgun Altunbas M, Babayeva R, Bal SK, Eltan SB, Yalcin Gungoren E, Esen F, Zemheri IE, Karakoc-Aydiner E, Ozen A, Caki-Kilic S, Kraakman MJ, Boztug K, Baris S. Rapamycin Controls Lymphoproliferation and Reverses T-Cell Responses in a Patient with a Novel STIM1 Loss-of-Function Deletion. J Clin Immunol 2024; 44:94. [PMID: 38578569 PMCID: PMC10997552 DOI: 10.1007/s10875-024-01682-0] [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: 11/06/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE Deficiency of stromal interaction molecule 1 (STIM1) results in combined immunodeficiency accompanied by extra-immunological findings like enamel defects and myopathy. We here studied a patient with a STIM1 loss-of-function mutation who presented with severe lymphoproliferation. We sought to explore the efficacy of the mTOR inhibitor rapamycin in controlling disease manifestations and reversing aberrant T-cell subsets and functions, which has never been used previously in this disorder. METHODS Clinical findings of the patient were collected over time. We performed immunological evaluations before and after initiation of rapamycin treatment, including detailed lymphocyte subset analyses, alterations in frequencies of circulating T follicular helper (cTFH) and regulatory T (Treg) cells and their subtypes as well as T cell activation and proliferation capacities. RESULTS A novel homozygous exon 2 deletion in STIM1 was detected in a 3-year-old girl with severe lymphoproliferation, recurrent infections, myopathy, iris hypoplasia, and enamel hypoplasia. Lymphoproliferation was associated with severe T-cell infiltrates. The deletion resulted in a complete loss of protein expression, associated with a lack of store-operated calcium entry response, defective T-cell activation, proliferation, and cytokine production. Interestingly, patient blood contained fewer cTFH and increased circulating follicular regulatory (cTFR) cells. Abnormal skewing towards TH2-like responses in certain T-cell subpopulations like cTFH, non-cTFH memory T-helper, and Treg cells was associated with increased eosinophil numbers and serum IgE levels. Treatment with rapamycin controlled lymphoproliferation, improved T-cell activation and proliferation capacities, reversed T-cell responses, and repressed high IgE levels and eosinophilia. CONCLUSIONS This study enhances our understanding of STIM1 deficiency by uncovering additional abnormal T-cell responses, and reveals for the first time the potential therapeutic utility of rapamycin for this disorder.
Collapse
Affiliation(s)
| | - Mehmet Cihangir Catak
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | | | - Feyza Bayram Catak
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Melek Yorgun Altunbas
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Royala Babayeva
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | | | - Sevgi Bilgic Eltan
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ezgi Yalcin Gungoren
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Fehim Esen
- Department of Ophthalmology, School of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Itir Ebru Zemheri
- Department of Pathology, Umraniye Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ahmet Ozen
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Suar Caki-Kilic
- Division of Pediatric Hematology, Umraniye Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | | | - Kaan Boztug
- Anna Children's Cancer Research Institute, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Anna Children's Hospital, Vienna, Austria
| | - Safa Baris
- Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik/Istanbul, Turkey.
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey.
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey.
| |
Collapse
|
9
|
Alvarez F, Liu Z, Bay A, Piccirillo CA. Deciphering the developmental trajectory of tissue-resident Foxp3 + regulatory T cells. Front Immunol 2024; 15:1331846. [PMID: 38605970 PMCID: PMC11007185 DOI: 10.3389/fimmu.2024.1331846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/14/2024] [Indexed: 04/13/2024] Open
Abstract
Foxp3+ TREG cells have been at the focus of intense investigation for their recognized roles in preventing autoimmunity, facilitating tissue recuperation following injury, and orchestrating a tolerance to innocuous non-self-antigens. To perform these critical tasks, TREG cells undergo deep epigenetic, transcriptional, and post-transcriptional changes that allow them to adapt to conditions found in tissues both at steady-state and during inflammation. The path leading TREG cells to express these tissue-specialized phenotypes begins during thymic development, and is further driven by epigenetic and transcriptional modifications following TCR engagement and polarizing signals in the periphery. However, this process is highly regulated and requires TREG cells to adopt strategies to avoid losing their regulatory program altogether. Here, we review the origins of tissue-resident TREG cells, from their thymic and peripheral development to the transcriptional regulators involved in their tissue residency program. In addition, we discuss the distinct signalling pathways that engage the inflammatory adaptation of tissue-resident TREG cells, and how they relate to their ability to recognize tissue and pathogen-derived danger signals.
Collapse
Affiliation(s)
- Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| | - Zhiyang Liu
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| | - Alexandre Bay
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| |
Collapse
|
10
|
Wang YH, Li W, McDermott M, Son GY, Maiti G, Zhou F, Tao A, Raphael D, Moreira AL, Shen B, Vaeth M, Nadorp B, Chakravarti S, Lacruz RS, Feske S. Regulatory T cells and IFN-γ-producing Th1 cells play a critical role in the pathogenesis of Sjögren's Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576314. [PMID: 38328096 PMCID: PMC10849570 DOI: 10.1101/2024.01.23.576314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Objectives Sjögren's Disease (SjD) is an autoimmune disorder characterized by progressive dysfunction, inflammation and destruction of salivary and lacrimal glands, and by extraglandular manifestations. Its etiology and pathophysiology remain incompletely understood, though a role for autoreactive B cells has been considered key. Here, we investigated the role of effector and regulatory T cells in the pathogenesis of SjD. Methods Histological analysis, RNA-sequencing and flow cytometry were conducted on glands, lungs, eyes and lymphoid tissues of mice with regulatory T cell-specific deletion of stromal interaction proteins (STIM) 1 and 2 ( Stim1/2 Foxp3 ), which play key roles in calcium signaling and T cell function. The pathogenicity of T cells from Stim1/2 Foxp3 mice was investigated through adoptively transfer into lymphopenic host mice. Additionally, single-cell transcriptomic analysis was performed on peripheral blood mononuclear cells (PBMCs) of patients with SjD and control subjects. Results Stim1/2 Foxp3 mice develop a severe SjD-like disorder including salivary gland (SG) and lacrimal gland (LG) inflammation and dysfunction, autoantibodies and extraglandular symptoms. SG inflammation in Stim1/2 Foxp3 mice is characterized by T and B cell infiltration, and transcriptionally by a Th1 immune response that correlates strongly with the dysregulation observed in patients with SjD. Adoptive transfer of effector T cells from Stim1/2 Foxp3 mice demonstrates that the SjD-like disease is driven by interferon (IFN)-γ producing autoreactive CD4 + T cells independently of B cells and autoantiboodies. scRNA-seq analysis identifies increased Th1 responses and attenuated memory Treg function in PBMCs of patients with SjD. Conclusions We report a more accurate mouse model of SjD while providing evidence for a critical role of Treg cells and IFN-γ producing Th1 cells in the pathogenesis of SjD, which may be effective targets for therapy.
Collapse
|
11
|
Abdelnaby AE, Trebak M. Store-Operated Ca 2+ Entry in Fibrosis and Tissue Remodeling. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241291374. [PMID: 39659877 PMCID: PMC11629433 DOI: 10.1177/25152564241291374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/29/2024] [Accepted: 09/27/2024] [Indexed: 12/12/2024]
Abstract
Fibrosis is a pathological condition characterized by excessive tissue deposition of extracellular matrix (ECM) components, leading to scarring and impaired function across multiple organ systems. This complex process is mediated by a dynamic interplay between cell types, including myofibroblasts, fibroblasts, immune cells, epithelial cells, and endothelial cells, each contributing distinctively through various signaling pathways. Critical to the regulatory mechanisms involved in fibrosis is store-operated calcium entry (SOCE), a calcium entry pathway into the cytosol active at the endoplasmic reticulum-plasma membrane contact sites and common to all cells. This review addresses the multifactorial nature of fibrosis with a focus on the pivotal roles of different cell types. We highlight the essential functions of myofibroblasts in ECM production, the transformation of fibroblasts, and the participation of immune cells in modulating the fibrotic landscape. We emphasize the contributions of SOCE in these different cell types to fibrosis, by exploring the involvement of SOCE in cellular functions such as proliferation, migration, secretion, and inflammatory responses. The examination of the cellular and molecular mechanisms of fibrosis and the role of SOCE in these mechanisms offers the potential of targeting SOCE as a therapeutic strategy for mitigating or reversing fibrosis.
Collapse
Affiliation(s)
- Ahmed Emam Abdelnaby
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Thatte AS, Hamilton AG, Nachod BE, Mukalel AJ, Billingsley MM, Palanki R, Swingle KL, Mitchell MJ. mRNA Lipid Nanoparticles for Ex Vivo Engineering of Immunosuppressive T Cells for Autoimmunity Therapies. NANO LETTERS 2023; 23:10179-10188. [PMID: 37906000 DOI: 10.1021/acs.nanolett.3c02573] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cell-based therapies for autoimmune diseases have gained significant traction, with several approaches centered around the regulatory T (Treg) cell─a well-known immunosuppressive cell characterized by its expression of the transcription factor Foxp3. Unfortunately, due to low numbers of Treg cells available in circulation, harvesting and culturing Treg cells remains a challenge. It has been reported that engineering Foxp3 expression in CD4+ T cells can result in a Treg-like phenotype; however, current methods result in the inefficient engineering of these cells. Here, we develop an ionizable lipid nanoparticle (LNP) platform to effectively deliver Foxp3 mRNA to CD4+ T cells. We successfully engineer CD4+ T cells into Foxp3-T (FP3T) cells that transiently exhibit an immunosuppressive phenotype and functionally suppress the proliferation of effector T cells. These results demonstrate the promise of an LNP platform for engineering immunosuppressive T cells with potential applications in autoimmunity therapies.
Collapse
Affiliation(s)
- Ajay S Thatte
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alex G Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Benjamin E Nachod
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alvin J Mukalel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
13
|
Qiao H, Li H. PLP2 Could Be a Prognostic Biomarker and Potential Treatment Target in Glioblastoma Multiforme. Pharmgenomics Pers Med 2023; 16:991-1009. [PMID: 37964785 PMCID: PMC10642424 DOI: 10.2147/pgpm.s425251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Objective This study aimed to discern the association between PLP2 expression, its biological significance, and the extent of immune infiltration in human GBM. Methods Utilizing the GEPIA2 and TCGA databases, we contrasted the expression levels of PLP2 in GBM against normal tissue. We utilized GEPIA2 and LinkedOmics for survival analysis, recognized genes co-expressed with PLP2 via cBioPortal and GEPIA2, and implemented GO and KEGG analyses. The STRING database facilitated the construction of protein-protein interaction networks. We evaluated the relationship of PLP2 with tumor immune infiltrates using ssGSEA and the TIMER 2.0 database. An IHC assay assessed PLP2 and PDL-1 expression in GBM tissue, and the Drugbank database aided in identifying potential PLP2-targeting compounds. Molecular docking was accomplished using Autodock Vina 1.2.2. Results PLP2 expression was markedly higher in GBM tissues in comparison to normal tissues. High PLP2 expression correlated with a decrease in overall survival across two databases. Functional analyses highlighted a focus of PLP2 functions within leukocyte. Discrepancies in PLP2 expression were evident in immune infiltration, impacting CD4+ T cells, neutrophils, myeloid dendritic cells, and macrophages. There was a concomitant increase in PLP2 and PD-L1 expression in GBM tissues, revealing a link between the two. Molecular docking with ethosuximide and praziquantel yielded scores of -7.441 and -4.295 kcal/mol, correspondingly. Conclusion PLP2's upregulation in GBM may adversely influence the lifespan of GBM patients. The involvement of PLP2 in pathways linked to leukocyte function is suggested. The positive correlation between PLP2 and PD-L1 could provide insights into PLP2's role in glioma modulation. Our research hints at PLP2's potential as a therapeutic target for GBM, with ethosuximide and praziquantel emerging as potential treatment candidates, especially emphasizing the potential of these compounds in GBM treatment targeting PLP2.
Collapse
Affiliation(s)
- Hao Qiao
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Huanting Li
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| |
Collapse
|
14
|
Manolios N, Papaemmanouil J, Adams DJ. The role of ion channels in T cell function and disease. Front Immunol 2023; 14:1238171. [PMID: 37705981 PMCID: PMC10497217 DOI: 10.3389/fimmu.2023.1238171] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/21/2023] [Indexed: 09/15/2023] Open
Abstract
T lymphocytes (T cells) are an important sub-group of cells in our immune system responsible for cell-mediated adaptive responses and maintaining immune homeostasis. Abnormalities in T cell function, lead the way to the persistence of infection, impaired immunosurveillance, lack of suppression of cancer growth, and autoimmune diseases. Ion channels play a critical role in the regulation of T cell signaling and cellular function and are often overlooked and understudied. Little is known about the ion "channelome" and the interaction of ion channels in immune cells. This review aims to summarize the published data on the impact of ion channels on T cell function and disease. The importance of ion channels in health and disease plus the fact they are easily accessible by virtue of being expressed on the surface of plasma membranes makes them excellent drug targets.
Collapse
Affiliation(s)
- Nicholas Manolios
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Rheumatology, Westmead Hospital, Sydney, NSW, Australia
| | - John Papaemmanouil
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
15
|
Cao Y, Hou Y, Zhao L, Huang Y, Liu G. New insights into follicular regulatory T cells in the intestinal and tumor microenvironments. J Cell Physiol 2023. [PMID: 37210730 DOI: 10.1002/jcp.31039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/03/2023] [Accepted: 04/28/2023] [Indexed: 05/23/2023]
Abstract
Follicular regulatory T (Tfr) cells are a novel and unique subset of effector regulatory T (Treg) cells that are located in germinal centers (GCs). Tfr cells express transcription profiles that are characteristic of both follicular helper T (Tfh) cells and Treg cells and negatively regulate GC reactions, including Tfh cell activation and cytokine production, class switch recombination and B cell activation. Evidence also shows that Tfr cells have specific characteristics in different local immune microenvironments. This review focuses on the regulation of Tfr cell differentiation and function in unique local immune microenvironments, including the intestine and tumor.
Collapse
Affiliation(s)
- Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yueru Hou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Longhao Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yijin Huang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
16
|
Liu P, Kang C, Zhang J, Liu Y, Liu J, Hu T, Zeng X, Qiu S. The role of dendritic cells in allergic diseases. Int Immunopharmacol 2022; 113:109449. [DOI: 10.1016/j.intimp.2022.109449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022]
|
17
|
Giri PS, Bharti AH, Begum R, Dwivedi M. Calcium controlled NFATc1 activation enhances suppressive capacity of regulatory T cells isolated from generalized vitiligo patients. Immunology 2022; 167:314-327. [PMID: 35754117 DOI: 10.1111/imm.13538] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
NFATs and FOXP3 are linked with impaired regulatory T-cells (Tregs) in generalized vitiligo (GV). To elucidate calcium mediated NFATc1 signalling pathway and its effect on Treg suppressive capacity in GV. Calcium levels, calcineurin, NFATc1 and GSK-3β activity and cell proliferation were assessed in 52 GV patients and 50 controls by calcium assay kit, calcineurin phosphatase assay kit, TransAM NFATc1 kit, GSK-3β ELISA and BrdU cell proliferation assay. Transcripts (CNB, CAM, GSK3B, DYRK1A and calcium channel genes) and protein (IFN-γ, IL-10 and TGF-β) expressions were assessed by qPCR and ELISA, respectively. Reduced plasma and intracellular Tregs calcium levels and ORAI1 transcripts suggested altered calcium homeostasis in GV Tregs (p = 0.00387, p = 0.0048, p < 0.0001), which led to decreased calcineurin and NFATc1 activity in GV Tregs (p = 0.0299, p < 0.0001). CNB and CAM transcripts were reduced in GV Tregs (p < 0.0001, p = 0.0004). GSK-3β activity, GSK3B and DYRK1A transcripts significantly increased in GV Tregs (p = 0.0134, p < 0.0001 and p < 0.0001). Plasma (p = 0.0225, p = 0.032) and intracellular Treg (p = 0.0035, p = 0.005) calcium levels, calcineurin (p = 0.001) and NFATc1 (p = 0.001, p < 0.0001) activity and ORAI1 (p = 0.0093, p < 0.0001), CAM and CNB (p = 0.0214) transcripts significantly decreased in active vitiligo (AV) and severe GV (sGV) Tregs. Calcium treatment significantly increased intracellular calcium and ORAI1 transcripts in GV Tregs (p = 0.0042, p = 0.0035). Moreover, calcium treatment enhanced calcineurin and NFATc1 activity in GV Tregs (p = 0.0128, p < 0.0001). Remarkably, calcium treatment increased Treg mediated suppression of CD4+ and CD8+ T-cells (p = 0.015, p = 0.006) in GV and increased Tregs associated cytokines: IL-10 (p = 0.0323, p = 0.009), TGF-β (p = 0.0321, p = 0.01) and decreased IFN-γ production (p = 0.001, p = 0.016) by CD4+ and CD8+ T-cells. Intracellular calcium levels positively correlated with calcineurin (r = 0.83; p < 0.0001) and NFATc1 (r = 0.61; p < 0.0001) activity, suggesting the enhanced Treg immunosuppressive capacity after calcium treatment. Our study for the first time suggests that reduced plasma calcium and ORAI1 transcripts are linked to calcium uptake defects in Tregs, which leads to reduced calcineurin and NFATc1 activation, thereby contributing to decreased Tregs immunosuppressive capacity in GV. Elevated GSK-3β activity and GSKB and DYRK1A transcripts are involved in reduced NFATc1 activity in GV Tregs. Overall, the study suggests that calcium-NFATc1-signalling pathway is likely to be involved in defective Tregs function and can be implicated for development of effective Treg mediated therapeutics for GV.
Collapse
Affiliation(s)
- Prashant S Giri
- C. G. Bhakta Institute of Biotechnology, Faculty of Science, Uka Tarsadia University, Surat, Gujarat, India
| | - Ankit H Bharti
- Aura Skin Care, Laxmi Icon 2nd Floor, Vyara, Gujarat, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Mitesh Dwivedi
- C. G. Bhakta Institute of Biotechnology, Faculty of Science, Uka Tarsadia University, Surat, Gujarat, India
| |
Collapse
|
18
|
Wang YH, Noyer L, Kahlfuss S, Raphael D, Tao AY, Kaufmann U, Zhu J, Mitchell-Flack M, Sidhu I, Zhou F, Vaeth M, Thomas PG, Saunders SP, Stauderman K, Curotto de Lafaille MA, Feske S. Distinct roles of ORAI1 in T cell-mediated allergic airway inflammation and immunity to influenza A virus infection. SCIENCE ADVANCES 2022; 8:eabn6552. [PMID: 36206339 PMCID: PMC9544339 DOI: 10.1126/sciadv.abn6552] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 08/22/2022] [Indexed: 06/16/2023]
Abstract
T cell activation and function depend on Ca2+ signals mediated by store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI1 proteins. We here investigated how SOCE controls T cell function in pulmonary inflammation during a T helper 1 (TH1) cell-mediated response to influenza A virus (IAV) infection and TH2 cell-mediated allergic airway inflammation. T cell-specific deletion of Orai1 did not exacerbate pulmonary inflammation and viral burdens following IAV infection but protected mice from house dust mite-induced allergic airway inflammation. ORAI1 controlled the expression of genes including p53 and E2F transcription factors that regulate the cell cycle in TH2 cells in response to allergen stimulation and the expression of transcription factors and cytokines that regulate TH2 cell function. Systemic application of a CRAC channel blocker suppressed allergic airway inflammation without compromising immunity to IAV infection, suggesting that inhibition of SOCE is a potential treatment for allergic airway disease.
Collapse
Affiliation(s)
- Yin-Hu Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Lucile Noyer
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sascha Kahlfuss
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Dimitrius Raphael
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony Y. Tao
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ulrike Kaufmann
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jingjie Zhu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Marisa Mitchell-Flack
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ikjot Sidhu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Fang Zhou
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Martin Vaeth
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Paul G. Thomas
- St. Jude’s Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sean P. Saunders
- Division of Pulmonary, Critical Care and Sleep Medicine, Departments of Medicine and Cell Biology, New York University Grossman School of Medicine, NY 10016, USA
| | | | - Maria A. Curotto de Lafaille
- Division of Pulmonary, Critical Care and Sleep Medicine, Departments of Medicine and Cell Biology, New York University Grossman School of Medicine, NY 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
19
|
Quintana JF, Chandrasegaran P, Sinton MC, Briggs EM, Otto TD, Heslop R, Bentley-Abbot C, Loney C, de Lecea L, Mabbott NA, MacLeod A. Single cell and spatial transcriptomic analyses reveal microglia-plasma cell crosstalk in the brain during Trypanosoma brucei infection. Nat Commun 2022; 13:5752. [PMID: 36180478 PMCID: PMC9525673 DOI: 10.1038/s41467-022-33542-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/21/2022] [Indexed: 11/08/2022] Open
Abstract
Human African trypanosomiasis, or sleeping sickness, is caused by the protozoan parasite Trypanosoma brucei and induces profound reactivity of glial cells and neuroinflammation when the parasites colonise the central nervous system. However, the transcriptional and functional responses of the brain to chronic T. brucei infection remain poorly understood. By integrating single cell and spatial transcriptomics of the mouse brain, we identify that glial responses triggered by infection are readily detected in the proximity to the circumventricular organs, including the lateral and 3rd ventricle. This coincides with the spatial localisation of both slender and stumpy forms of T. brucei. Furthermore, in silico predictions and functional validations led us to identify a previously unknown crosstalk between homeostatic microglia and Cd138+ plasma cells mediated by IL-10 and B cell activating factor (BAFF) signalling. This study provides important insights and resources to improve understanding of the molecular and cellular responses in the brain during infection with African trypanosomes.
Collapse
Affiliation(s)
- Juan F Quintana
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK.
| | - Praveena Chandrasegaran
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Matthew C Sinton
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Emma M Briggs
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Thomas D Otto
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Infection and Immunity, MVLS, University of Glasgow, Glasgow, UK
| | - Rhiannon Heslop
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Calum Bentley-Abbot
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Colin Loney
- School of Infection and Immunity, MVLS, University of Glasgow, Glasgow, UK
- MRC Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Luis de Lecea
- Stanford University School of Medicine, Stanford, CA, USA
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| |
Collapse
|
20
|
McGee MC, Zhang T, Magazine N, Islam R, Carossino M, Huang W. PD-1 and ICOS counter-regulate tissue resident regulatory T cell development and IL-10 production during flu. Front Immunol 2022; 13:984476. [PMID: 36159872 PMCID: PMC9492985 DOI: 10.3389/fimmu.2022.984476] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/19/2022] [Indexed: 11/18/2022] Open
Abstract
Regulatory T cells that express the transcription factor Foxp3 (Treg cells) are a highly heterogenous population of immunoregulatory cells critical for maintaining immune homeostasis and preventing immunopathology during infections. Tissue resident Treg (TR-Treg) cells are maintained within nonlymphoid tissues and have been shown to suppress proinflammatory tissue resident T cell responses and promote tissue repair. Human populations are repetitively exposed to influenza infections and lung tissue resident effector T cell responses are associated with flu-induced long-term pulmonary sequelae. The kinetics of TR-Treg cell development and molecular features of TR-Treg cells during repeated and/or long-term flu infections are unclear. Utilizing a Foxp3RFP/IL-10GFP dual reporter mouse model along with intravascular fluorescent in vivo labeling, we characterized the TR-Treg cell responses to repetitive heterosubtypic influenza infections. We found lung tissue resident Treg cells accumulated and expressed high levels of co-inhibitory and co-stimulatory receptors post primary and secondary infections. Blockade of PD-1 or ICOS signaling reveals that PD-1 and ICOS signaling pathways counter-regulate TR-Treg cell expansion and IL-10 production, during secondary influenza infection. Furthermore, the virus-specific TR-Treg cell response displayed distinct kinetics, when compared to conventional CD4+ tissue resident memory T cells, during secondary flu infection. Our results provide insight into the tissue resident Foxp3+ regulatory T cell response during repetitive flu infections, which may be applicable to other respiratory infectious diseases such as tuberculosis and COVID.
Collapse
Affiliation(s)
- Michael C. McGee
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Tianyi Zhang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Nicholas Magazine
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Rezwanul Islam
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
- *Correspondence: Weishan Huang,
| |
Collapse
|
21
|
Differentiation and homeostasis of effector Treg cells are regulated by inositol polyphosphates modulating Ca 2+ influx. Proc Natl Acad Sci U S A 2022; 119:e2121520119. [PMID: 35776543 PMCID: PMC9271192 DOI: 10.1073/pnas.2121520119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Activated Foxp3+ regulatory T (Treg) cells differentiate into effector Treg (eTreg) cells to maintain peripheral immune homeostasis and tolerance. T cell receptor (TCR)-mediated induction and regulation of store-operated Ca2+ entry (SOCE) is essential for eTreg cell differentiation and function. However, SOCE regulation in Treg cells remains unclear. Here, we show that inositol polyphosphate multikinase (IPMK), which generates inositol tetrakisphosphate and inositol pentakisphosphate, is a pivotal regulator of Treg cell differentiation downstream of TCR signaling. IPMK is highly expressed in TCR-stimulated Treg cells and promotes a TCR-induced Treg cell program. IPMK-deficient Treg cells display aberrant T cell activation and impaired differentiation into RORγt+ Treg cells and tissue-resident Treg cells. Mechanistically, IPMK controls the generation of higher-order inositol phosphates, thereby promoting Ca2+ mobilization and Treg cell effector functions. Our findings identify IPMK as a critical regulator of TCR-mediated Ca2+ influx and highlight the importance of IPMK in Treg cell-mediated immune homeostasis.
Collapse
|
22
|
Gu W, Madrid DMC, Joyce S, Driver JP. A single-cell analysis of thymopoiesis and thymic iNKT cell development in pigs. Cell Rep 2022; 40:111050. [PMID: 35793622 PMCID: PMC9704770 DOI: 10.1016/j.celrep.2022.111050] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/26/2022] [Accepted: 06/13/2022] [Indexed: 12/13/2022] Open
Abstract
Many aspects of the porcine immune system remain poorly characterized, which poses a barrier to improving swine health and utilizing pigs as preclinical models. Here, we employ single-cell RNA sequencing (scRNA-seq) to create a cell atlas of the early-adolescent pig thymus. Our data show conserved features as well as species-specific differences in cell states and cell types compared with human thymocytes. We also describe several unconventional T cell types with gene expression profiles associated with innate effector functions. This includes a cell census of more than 11,000 differentiating invariant natural killer T (iNKT) cells, which reveals that the functional diversity of pig iNKT cells differs substantially from the iNKT0/1/2/17 subset differentiation paradigm established in mice. Our data characterize key differentiation events in porcine thymopoiesis and iNKT cell maturation and provide important insights into pig T cell development.
Collapse
Affiliation(s)
- Weihong Gu
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
| | | | - Sebastian Joyce
- Department of Veterans Affairs, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institution for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John P Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
23
|
Liu Y, Wang H, Taylor M, Cook C, Martínez-Berdeja A, North JP, Harirchian P, Hailer AA, Zhao Z, Ghadially R, Ricardo-Gonzalez RR, Grekin RC, Mauro TM, Kim E, Choi J, Purdom E, Cho RJ, Cheng JB. Classification of human chronic inflammatory skin disease based on single-cell immune profiling. Sci Immunol 2022; 7:eabl9165. [PMID: 35427179 PMCID: PMC9301819 DOI: 10.1126/sciimmunol.abl9165] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Inflammatory conditions represent the largest class of chronic skin disease, but the molecular dysregulation underlying many individual cases remains unclear. Single-cell RNA sequencing (scRNA-seq) has increased precision in dissecting the complex mixture of immune and stromal cell perturbations in inflammatory skin disease states. We single-cell-profiled CD45+ immune cell transcriptomes from skin samples of 31 patients (7 atopic dermatitis, 8 psoriasis vulgaris, 2 lichen planus (LP), 1 bullous pemphigoid (BP), 6 clinical/histopathologically indeterminate rashes, and 7 healthy controls). Our data revealed active proliferative expansion of the Treg and Trm components and universal T cell exhaustion in human rashes, with a relative attenuation of antigen-presenting cells. Skin-resident memory T cells showed the greatest transcriptional dysregulation in both atopic dermatitis and psoriasis, whereas atopic dermatitis also demonstrated recurrent abnormalities in ILC and CD8+ cytotoxic lymphocytes. Transcript signatures differentiating these rash types included genes previously implicated in T helper cell (TH2)/TH17 diatheses, segregated in unbiased functional networks, and accurately identified disease class in untrained validation data sets. These gene signatures were able to classify clinicopathologically ambiguous rashes with diagnoses consistent with therapeutic response. Thus, we have defined major classes of human inflammatory skin disease at the molecular level and described a quantitative method to classify indeterminate instances of pathologic inflammation. To make this approach accessible to the scientific community, we created a proof-of-principle web interface (RashX), where scientists and clinicians can visualize their patient-level rash scRNA-seq-derived data in the context of our TH2/TH17 transcriptional framework.
Collapse
Affiliation(s)
- Yale Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi 710004, P. R. China
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Hao Wang
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mark Taylor
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Christopher Cook
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | | | - Jeffrey P North
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Paymann Harirchian
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Ashley A Hailer
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Zijun Zhao
- Santa Clara Valley Medical Center, Santa Clara, CA 95128, USA
| | - Ruby Ghadially
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Roberto R Ricardo-Gonzalez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Roy C Grekin
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Theodora M Mauro
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Esther Kim
- Department of Plastic Surgery, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Jaehyuk Choi
- Department of Dermatology, Northwestern School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth Purdom
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Raymond J Cho
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Jeffrey B Cheng
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| |
Collapse
|
24
|
The glucose transporter GLUT3 controls T helper 17 cell responses through glycolytic-epigenetic reprogramming. Cell Metab 2022; 34:516-532.e11. [PMID: 35316657 PMCID: PMC9019065 DOI: 10.1016/j.cmet.2022.02.015] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/04/2022] [Accepted: 02/23/2022] [Indexed: 12/16/2022]
Abstract
Metabolic reprogramming is a hallmark of activated T cells. The switch from oxidative phosphorylation to aerobic glycolysis provides energy and intermediary metabolites for the biosynthesis of macromolecules to support clonal expansion and effector function. Here, we show that glycolytic reprogramming additionally controls inflammatory gene expression via epigenetic remodeling. We found that the glucose transporter GLUT3 is essential for the effector functions of Th17 cells in models of autoimmune colitis and encephalomyelitis. At the molecular level, we show that GLUT3-dependent glucose uptake controls a metabolic-transcriptional circuit that regulates the pathogenicity of Th17 cells. Metabolomic, epigenetic, and transcriptomic analyses linked GLUT3 to mitochondrial glucose oxidation and ACLY-dependent acetyl-CoA generation as a rate-limiting step in the epigenetic regulation of inflammatory gene expression. Our findings are also important from a translational perspective because inhibiting GLUT3-dependent acetyl-CoA generation is a promising metabolic checkpoint to mitigate Th17-cell-mediated inflammatory diseases.
Collapse
|
25
|
Ishihara S, Sato T, Fujikado N, Miyazaki H, Yoshimoto T, Yamamoto H, Fukuda S, Katagiri K. Rap1 prevents colitogenic Th17 cell expansion and facilitates Treg cell differentiation and distal TCR signaling. Commun Biol 2022; 5:206. [PMID: 35246619 PMCID: PMC8897436 DOI: 10.1038/s42003-022-03129-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 02/08/2022] [Indexed: 11/25/2022] Open
Abstract
T-cell-specific Rap1 deletion causes spontaneous colitis in mice. In the present study, we revealed that Rap1 deficiency in T cells impaired the preceding induction of intestinal RORγt+ Treg cells. In the large intestinal lamina propria (LILP) of T-cell-specific Rap1-knockout mice (Rap1KO mice), Th17 cells were found to increase in a microbiota-dependent manner, and the inhibition of IL-17A production prevented the development of colitis. In the LILP of Rap1KO mice, RORγt+ Treg cells were scarcely induced by 4 weeks of age. The expression of CTLA-4 on Rap1-deficient Treg cells was reduced and the expression of CD80 and CD86 on dendritic cells was consequently elevated in Rap1KO mice. When cultured under each polarizing condition, Rap1-deficient naïve CD4+ T cells did not show biased differentiation into Th17 cells; their differentiation into Treg cells as well as Th1 and Th2 cells was lesser than that of wild-type cells. Rap1-deficient naïve CD4+ T cells were found to exhibit the defective nuclear translocation of NFAT and formation of actin foci in response to TCR engagement. These data suggest that Rap1 amplifies the TCR signaling required for Treg-mediated control of intestinal colitogenic Th17 responses.
Collapse
Affiliation(s)
- Sayaka Ishihara
- Department of Biosciences, School of Science, Kitasato University, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0344, Japan
| | - Tsuyoshi Sato
- Department of Biosciences, School of Science, Kitasato University, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0344, Japan
| | - Noriyuki Fujikado
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, CA, 92121, USA
| | - Haruka Miyazaki
- Department of Biosciences, School of Science, Kitasato University, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0344, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Hiromitsu Yamamoto
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, 3-25-13 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
- Transborder Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Koko Katagiri
- Department of Biosciences, School of Science, Kitasato University, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0344, Japan.
| |
Collapse
|
26
|
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ signaling pathway that is evolutionarily conserved across eukaryotes. SOCE is triggered physiologically when the endoplasmic reticulum (ER) Ca2+ stores are emptied through activation of inositol 1,4,5-trisphosphate receptors. SOCE is mediated by the Ca2+ release-activated Ca2+ (CRAC) channels, which are highly Ca2+ selective. Upon store depletion, the ER Ca2+-sensing STIM proteins aggregate and gain extended conformations spanning the ER-plasma membrane junctional space to bind and activate Orai, the pore-forming proteins of hexameric CRAC channels. In recent years, studies on STIM and Orai tissue-specific knockout mice and gain- and loss-of-function mutations in humans have shed light on the physiological functions of SOCE in various tissues. Here, we describe recent findings on the composition of native CRAC channels and their physiological functions in immune, muscle, secretory, and neuronal systems to draw lessons from transgenic mice and human diseases caused by altered CRAC channel activity.
Collapse
Affiliation(s)
- Scott M Emrich
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA;
| | - Ryan E Yoast
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA;
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA;
- Department of Pharmacology and Chemical Biology and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
27
|
Ribeiro F, Perucha E, Graca L. T follicular cells: the regulators of germinal centre homeostasis. Immunol Lett 2022; 244:1-11. [DOI: 10.1016/j.imlet.2022.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 01/05/2023]
|
28
|
Adriawan IR, Atschekzei F, Dittrich-Breiholz O, Garantziotis P, Hirsch S, Risser LM, Kosanke M, Schmidt RE, Witte T, Sogkas G. Novel aspects of regulatory T cell dysfunction as a therapeutic target in giant cell arteritis. Ann Rheum Dis 2022; 81:124-131. [PMID: 34583923 PMCID: PMC8762021 DOI: 10.1136/annrheumdis-2021-220955] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/15/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Giant cell arteritis (GCA) is the most common primary vasculitis, preferentially affecting the aorta and its large-calibre branches. An imbalance between proinflammatory CD4+ T helper cell subsets and regulatory T cells (Tregs) is thought to be involved in the pathogenesis of GCA and Treg dysfunction has been associated with active disease. Our work aims to explore the aetiology of Treg dysfunction and the way it is affected by remission-inducing immunomodulatory regimens. METHODS A total of 41 GCA patients were classified into active disease (n=14) and disease in remission (n=27). GCA patients' and healthy blood donors' (HD) Tregs were sorted and subjected to transcriptome and phenotypic analysis. RESULTS Transcriptome analysis revealed 27 genes, which were differentially regulated between GCA-derived and HD-derived Tregs. Among those, we identified transcription factors, glycolytic enzymes and IL-2 signalling mediators. We confirmed the downregulation of forkhead box P3 (FOXP3) and interferon regulatory factor 4 (IRF4) at protein level and identified the ineffective induction of glycoprotein A repetitions predominant (GARP) and CD25 as well as the reduced T cell receptor (TCR)-induced calcium influx as correlates of Treg dysfunction in GCA. Inhibition of glycolysis in HD-derived Tregs recapitulated most identified dysfunctions of GCA Tregs, suggesting the central pathogenic role of the downregulation of the glycolytic enzymes. Separate analysis of the subgroup of tocilizumab-treated patients identified the recovery of the TCR-induced calcium influx and the Treg suppressive function to associate with disease remission. CONCLUSIONS Our findings suggest that low glycolysis and calcium signalling account for Treg dysfunction and inflammation in GCA.
Collapse
Affiliation(s)
- Ignatius Ryan Adriawan
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | - Faranaz Atschekzei
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | | | | | - Stefanie Hirsch
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | | | - Maike Kosanke
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Reinhold Ernst Schmidt
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | - Torsten Witte
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | - Georgios Sogkas
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| |
Collapse
|
29
|
SantaCruz-Calvo S, Bharath L, Pugh G, SantaCruz-Calvo L, Lenin RR, Lutshumba J, Liu R, Bachstetter AD, Zhu B, Nikolajczyk BS. Adaptive immune cells shape obesity-associated type 2 diabetes mellitus and less prominent comorbidities. Nat Rev Endocrinol 2022; 18:23-42. [PMID: 34703027 PMCID: PMC11005058 DOI: 10.1038/s41574-021-00575-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are increasing in prevalence owing to decreases in physical activity levels and a shift to diets that include addictive and/or high-calorie foods. These changes are associated with the adoption of modern lifestyles and the presence of an obesogenic environment, which have resulted in alterations to metabolism, adaptive immunity and endocrine regulation. The size and quality of adipose tissue depots in obesity, including the adipose tissue immune compartment, are critical determinants of overall health. In obesity, chronic low-grade inflammation can occur in adipose tissue that can progress to systemic inflammation; this inflammation contributes to the development of insulin resistance, T2DM and other comorbidities. An improved understanding of adaptive immune cell dysregulation that occurs during obesity and its associated metabolic comorbidities, with an appreciation of sex differences, will be critical for repurposing or developing immunomodulatory therapies to treat obesity and/or T2DM-associated inflammation. This Review critically discusses how activation and metabolic reprogramming of lymphocytes, that is, T cells and B cells, triggers the onset, development and progression of obesity and T2DM. We also consider the role of immunity in under-appreciated comorbidities of obesity and/or T2DM, such as oral cavity inflammation, neuroinflammation in Alzheimer disease and gut microbiome dysbiosis. Finally, we discuss previous clinical trials of anti-inflammatory medications in T2DM and consider the path forward.
Collapse
Affiliation(s)
- Sara SantaCruz-Calvo
- Department of Pharmacology and Nutritional Sciences and the Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA.
| | - Leena Bharath
- Department of Nutrition and Public Health, Merrimack College, North Andover, MA, USA
| | - Gabriella Pugh
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Lucia SantaCruz-Calvo
- Department of Chemistry and Food Technology, Technical University of Madrid, Madrid, Spain
| | - Raji Rajesh Lenin
- Department of Pharmacology and Nutritional Sciences and the Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Jenny Lutshumba
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Beibei Zhu
- Department of Pharmacology and Nutritional Sciences and the Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Sciences and the Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
30
|
Imam S, Dar P, Aziz SW, Zahid ZA, Sarwar H, Karim T, Faisal S, Haseeb I, Naqvi AS, Shah R, Haque A, Salim N, Jaume JC. Immune Cell Plasticity Allows for Resetting of Phenotype From Effector to Regulator With Combined Inhibition of Notch/eIF5A Pathways. Front Cell Dev Biol 2021; 9:777805. [PMID: 34881246 PMCID: PMC8645838 DOI: 10.3389/fcell.2021.777805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/04/2021] [Indexed: 01/23/2023] Open
Abstract
Type 1 diabetes (T1D) results from the destruction of pancreatic β-cells caused by an altered immune balance in the pancreatic microenvironment. In humans as well as in mouse models, T cells are well recognized as key orchestrators of T1D, which is characterized by T helper (Th) 1 and Th17 cell bias and/or low/defective T-regulatory cells (Treg), and culminates in cytotoxic T-cell (CTL)-mediated destruction of β-cells. Refitting of immune cells toward the non-inflammatory phenotype in the pancreas may represent a way to prevent/treat T1D. Recently we developed a unique spontaneous humanized mouse model of type 1 diabetes, wherein mouse MHC-II molecules were replaced by human DQ8, and β-cells were made to express human glutamic acid decarboxylase (GAD) 65 auto-antigen. The mice spontaneously developed T1D resembling the human disease. Humanized T1D mice showed hyperglycemic (250-300 mg/dl) symptoms by the 4th week of life. The diabetogenic T cells (CD4, CD8) present in our model are GAD65 antigen-specific in nature. Intermolecular antigen spreading recorded during 3rd-6th week of age is like that observed in the human preclinical period of T1D. In this paper, we tested our hypothesis in our spontaneous humanized T1D mouse model. We targeted two cell-signaling pathways and their inhibitions: eIF5A pathway inhibition influences T helper cell dynamics toward the non-inflammatory phenotype and Notch signaling inhibition enrich Tregs and targets auto-reactive CTLs, rescues the pancreatic islet structure, and increases the functionality of β-cells in terms of insulin production. We report that inhibition of (eIF5A + Notch) signaling mediates suppression of diabetogenic T cells by inducing plasticity in CD4 + T cells co-expressing IL-17 and IFNγ (IL-17 + IFNγ +) toward the Treg cells phenotype.
Collapse
Affiliation(s)
- Shahnawaz Imam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Pervaiz Dar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States.,Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, India
| | - Saba Wasim Aziz
- Department of Internal Medicine, Division of Endocrinology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Zeeshan A Zahid
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Haider Sarwar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States.,Windsor University School of Medicine, Cayon, West Indies
| | - Tamanna Karim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Sarah Faisal
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,College of Art and Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Ibrahim Haseeb
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Ahmed S Naqvi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Ottawa Hills High School, Ottawa, OH, United States
| | - Rayyan Shah
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Sylvania Northview High School, Toledo, OH, United States
| | - Amna Haque
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Austin College, Sherman, TX, United States
| | - Nancy Salim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Juan C Jaume
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| |
Collapse
|
31
|
Grover P, Goel PN, Greene MI. Regulatory T Cells: Regulation of Identity and Function. Front Immunol 2021; 12:750542. [PMID: 34675933 PMCID: PMC8524049 DOI: 10.3389/fimmu.2021.750542] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/14/2021] [Indexed: 12/22/2022] Open
Abstract
T regulatory cells suppress a variety of immune responses to self-antigens and play a role in peripheral tolerance maintenance by limiting autoimmune disorders, and other pathological immune responses such as limiting immune reactivity to oncoprotein encoded antigens. Forkhead box P3 (FOXP3) expression is required for Treg stability and affects functional activity. Mutations in the master regulator FOXP3 and related components have been linked to autoimmune diseases in humans, such as IPEX, and a scurfy-like phenotype in mice. Several lines of evidence indicate that Treg use a variety of immunosuppressive mechanisms to limit an immune response by targeting effector cells, including secretion of immunoregulatory cytokines, granzyme/perforin-mediated cell cytolysis, metabolic perturbation, directing the maturation and function of antigen-presenting cells (APC) and secretion of extracellular vesicles for the development of immunological tolerance. In this review, several regulatory mechanisms have been highlighted and discussed.
Collapse
Affiliation(s)
- Payal Grover
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Peeyush N Goel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark I Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
32
|
Zhao SJ, Jia H, Xu XL, Bu WB, Zhang Q, Chen X, Ji J, Sun JF. Identification of the Role of Wnt/β-Catenin Pathway Through Integrated Analyses and in vivo Experiments in Vitiligo. Clin Cosmet Investig Dermatol 2021; 14:1089-1103. [PMID: 34511958 PMCID: PMC8423189 DOI: 10.2147/ccid.s319061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/14/2021] [Indexed: 01/18/2023]
Abstract
Purpose Vitiligo is an acquired depigmentation skin disease, which affects an average of 1% of the world’s population. The purpose of this study is to identify the key genes and pathways responsible for vitiligo and find new therapeutic targets. Methods The datasets GSE65127, GSE53146, and GSE75819 were downloaded from the Gene Expression Omnibus (GEO) database. R language was used to identify the differentially expressed genes (DEGs) between lesional skin of vitiligo and non-lesional skin. Next, the key pathways were obtained by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. The protein–protein interaction (PPI) networks were conducted by STRING database and Cytoscape software. Subsequently, module analysis was performed by Cytoscape. Among these results, the Wnt/β-catenin pathway and melanogenesis pathway caught our attention. The expression level of β-catenin, microphthalmia-associated transcription factor (MITF) and tyrosinase (TYR) was detected by immunofluorescence in vitiligo lesions and healthy skin. Moreover, zebrafish was treated with XAV-939, an inhibitor of the Wnt/β-catenin pathway. After that, the area of melanin granules as a percentage of the head area was measured. The mRNA expression of β-catenin, lymphoid-enhancing factor 1(lef1), tyr and mitf were detected by q-PCR (quantitative polymerase chain reaction) in zebrafish (Danio rerio). Results A total of 2442 DEGs were identified, including 1068 upregulated and 1374 downregulated DEGs. The key pathways were identified by GO and KEGG analyses, such as “NOD-like receptor signaling pathway”, “Wnt signaling pathway”, “Melanogenesis”, “mTOR signaling pathway”, “PI3K-Akt signaling pathway”, “Calcium signaling pathway” and “Rap1 signaling pathway”. The immunofluorescence results showed that the level of β-catenin, MITF and TYR was significantly downregulated in vitiligo lesional skin. In zebrafish, the mean percentage area of melanin granules and the expression of β-catenin, lef1, tyr and mitf were decreased after treated with XAV-939. Conclusion The present study identified key genes and signaling pathways associated with the pathophysiology of vitiligo. Among them, the Wnt/β-catenin pathway played an essential role in pigmentation and could be a breakthrough point in vitiligo treatment.
Collapse
Affiliation(s)
- Si-Jia Zhao
- Department of Pathology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, People's Republic of China
| | - Hong Jia
- Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, People's Republic of China
| | - Xiu-Lian Xu
- Department of Pathology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, People's Republic of China
| | - Wen-Bo Bu
- Department of Dermatologic Surgery, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, People's Republic of China
| | - Qian Zhang
- Department of Dermatologic Surgery, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, People's Republic of China
| | - Xi Chen
- Department of Medicine 3, Universitätsklinikum Erlangen, Friedrich Alexander University Erlangen Nuremberg, Erlangen, Bavaria, Germany
| | - Juan Ji
- Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, People's Republic of China
| | - Jian-Fang Sun
- Department of Pathology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
33
|
Zissler UM, Jakwerth CA, Guerth F, Lewitan L, Rothkirch S, Davidovic M, Ulrich M, Oelsner M, Garn H, Schmidt‐Weber CB, Chaker AM. Allergen-specific immunotherapy induces the suppressive secretoglobin 1A1 in cells of the lower airways. Allergy 2021; 76:2461-2474. [PMID: 33528894 DOI: 10.1111/all.14756] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/29/2020] [Accepted: 01/03/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND While several systemic immunomodulatory effects of allergen-specific immunotherapy (AIT) have been discovered, local anti-inflammatory mechanisms in the respiratory tract are largely unknown. We sought to elucidate local and epithelial mechanisms underlying allergen-specific immunotherapy in a genome-wide approach. METHODS We induced sputum in hay fever patients and healthy controls during the pollen peak season and stratified patients by effective allergen immunotherapy or as untreated. Sputum was directly processed after induction and subjected to whole transcriptome RNA microarray analysis. Nasal secretions were analyzed for Secretoglobin1A1 (SCGB1A1) and IL-24 protein levels in an additional validation cohort at three defined time points during the 3-year course of AIT. Subsequently, RNA was extracted and subjected to an array-based whole transcriptome analysis. RESULTS Allergen-specific immunotherapy inhibited pro-inflammatory CXCL8, IL24, and CCL26mRNA expression, while SCGB1A1, IL7, CCL5, CCL23, and WNT5BmRNAs were induced independently of the asthma status and allergen season. In our validation cohort, local increase of SCGB1A1 occurred concomitantly with the reduction of local IL-24 in upper airways during the course of AIT. Additionally, SCGB1A1 was identified as a suppressor of epithelial gene expression. CONCLUSIONS Allergen-specific immunotherapy induces a yet unknown local gene expression footprint in the lower airways that on one hand appears to be a result of multiple regulatory pathways and on the other hand reveals SCGB1A1 as novel anti-inflammatory mediator of long-term allergen-specific therapeutic intervention in the local environment.
Collapse
Affiliation(s)
- Ulrich M. Zissler
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
- Member of the Helmholtz I&I Initiative Munich Germany
| | - Constanze A. Jakwerth
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
| | - Ferdinand Guerth
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
| | - Larissa Lewitan
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
- Department of Otorhinolaryngology and Head and Neck Surgery Medical School Technical University of Munich Munich Germany
| | - Sandra Rothkirch
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
- Department of Otorhinolaryngology and Head and Neck Surgery Medical School Technical University of Munich Munich Germany
| | - Miodrag Davidovic
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
- Department of Otorhinolaryngology and Head and Neck Surgery Medical School Technical University of Munich Munich Germany
| | - Moritz Ulrich
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
- Department of Otorhinolaryngology and Head and Neck Surgery Medical School Technical University of Munich Munich Germany
| | - Madlen Oelsner
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry Philipps University MarburgMedical FacultyMember of the German Center of Lung Research Marburg Germany
| | - Carsten B. Schmidt‐Weber
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
- Member of the Helmholtz I&I Initiative Munich Germany
| | - Adam M. Chaker
- Center of Allergy & Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
- Member of the German Center for Lung Research (DZL) Munich Germany
- Department of Otorhinolaryngology and Head and Neck Surgery Medical School Technical University of Munich Munich Germany
| |
Collapse
|
34
|
Prenek L, Litvai T, Balázs N, Kugyelka R, Boldizsár F, Najbauer J, Németh P, Berki T. Regulatory T cells are less sensitive to glucocorticoid hormone induced apoptosis than CD4 + T cells. Apoptosis 2021; 25:715-729. [PMID: 32737651 PMCID: PMC7527366 DOI: 10.1007/s10495-020-01629-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Earlier we have reported that thymic regulatory T cells (Treg) are resistant to in vivo glucocorticoid hormone (GC)-induced apoptosis, while the most GC-sensitive DP thymocytes died through the activation of mitochondrial apoptotic pathway. Here we analyzed the apoptosis-inducing effect of high dose (10-6 M) in vitro dexamethasone (DX) treatment in mouse thymic- and splenic Tregs and CD4+ T cells. Activation of both extrinsic and intrinsic apoptotic pathways started after 2 h of DX treatment in CD4 SP thymocytes and was 3 × higher than in CD4+ splenocytes, while in Treg cells, weak activation of the extrinsic apoptotic pathway started only after 3 h. We also investigated the expression of 21 apoptosis-related molecules using a protein array and found higher level of both pro-and anti-apoptotic molecules in Tregs compared to CD4+ T cells. 4 h in vitro DX treatment induced upregulation of most apoptosis-related molecules both in Tregs and CD4+ T cells, except for the decrease of Bcl-2 expression in CD4+ T cells. We found high basal cytosolic Ca2+ levels in untreated Treg cells, which further increased after DX treatment, while the specific TCR-induced Ca2+ signal was lower in Tregs than in CD4+ T cells. Our results suggest that in the background of the relative apoptosis resistance of Treg cells to GCs might be their high basal cytosolic Ca2+ level and upregulated Bcl-2 expression. In contrast, downregulation of Bcl-2 expression in CD4+ T cells can explain their higher, DX-induced apoptosis sensitivity.
Collapse
Affiliation(s)
- Lilla Prenek
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
| | - Tímea Litvai
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
| | - Noémi Balázs
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
| | - Réka Kugyelka
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
| | - Ferenc Boldizsár
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
| | - József Najbauer
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
| | - Péter Németh
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
| | - Timea Berki
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.
| |
Collapse
|
35
|
Mijnheer G, Lutter L, Mokry M, van der Wal M, Scholman R, Fleskens V, Pandit A, Tao W, Wekking M, Vervoort S, Roberts C, Petrelli A, Peeters JGC, Knijff M, de Roock S, Vastert S, Taams LS, van Loosdregt J, van Wijk F. Conserved human effector Treg cell transcriptomic and epigenetic signature in arthritic joint inflammation. Nat Commun 2021; 12:2710. [PMID: 33976194 PMCID: PMC8113485 DOI: 10.1038/s41467-021-22975-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
Treg cells are critical regulators of immune homeostasis, and environment-driven Treg cell differentiation into effector (e)Treg cells is crucial for optimal functioning. However, human Treg cell programming in inflammation is unclear. Here, we combine transcriptional and epigenetic profiling to identify a human eTreg cell signature. Inflammation-derived functional Treg cells have a transcriptional profile characterized by upregulation of both a core Treg cell (FOXP3, CTLA4, TIGIT) and effector program (GITR, BLIMP-1, BATF). We identify a specific human eTreg cell signature that includes the vitamin D receptor (VDR) as a predicted regulator in eTreg cell differentiation. H3K27ac/H3K4me1 occupancy indicates an altered (super-)enhancer landscape, including enrichment of the VDR and BATF binding motifs. The Treg cell profile has striking overlap with tumor-infiltrating Treg cells. Our data demonstrate that human inflammation-derived Treg cells acquire a conserved and specific eTreg cell profile guided by epigenetic changes, and fine-tuned by environment-specific adaptations.
Collapse
MESH Headings
- Adolescent
- Arthritis, Juvenile/genetics
- Arthritis, Juvenile/immunology
- Arthritis, Juvenile/pathology
- Base Sequence
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/immunology
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Case-Control Studies
- Cell Differentiation
- Child
- Child, Preschool
- Epigenesis, Genetic
- Female
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression Profiling
- Gene Regulatory Networks
- Glucocorticoid-Induced TNFR-Related Protein/genetics
- Glucocorticoid-Induced TNFR-Related Protein/immunology
- Histones/genetics
- Histones/immunology
- Humans
- Joints/immunology
- Joints/pathology
- Male
- Metabolic Networks and Pathways/genetics
- Metabolic Networks and Pathways/immunology
- Positive Regulatory Domain I-Binding Factor 1/genetics
- Positive Regulatory Domain I-Binding Factor 1/immunology
- Primary Cell Culture
- Receptors, Calcitriol/genetics
- Receptors, Calcitriol/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Transcriptome
- Young Adult
Collapse
Affiliation(s)
- Gerdien Mijnheer
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lisanne Lutter
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Michal Mokry
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Department of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
- Epigenomics facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marlot van der Wal
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rianne Scholman
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Veerle Fleskens
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Aridaman Pandit
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Weiyang Tao
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mark Wekking
- Epigenomics facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stephin Vervoort
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ceri Roberts
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Alessandra Petrelli
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Janneke G C Peeters
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marthe Knijff
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sytze de Roock
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sebastiaan Vastert
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Jorg van Loosdregt
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, Pediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Ye Y, Wang M, Huang H. Follicular regulatory T cell biology and its role in immune-mediated diseases. J Leukoc Biol 2021; 110:239-255. [PMID: 33938586 DOI: 10.1002/jlb.1mr0321-601rr] [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] [Indexed: 12/24/2022] Open
Abstract
Follicular regulatory T (Tfr) cells are recently found to be a special subgroup of regulatory T (Treg) cells. Tfr cells play an important role in regulating the germinal center (GC) response, especially modulating follicular helper T (Tfh) cells and GC-B cells, thereby affecting the production of antibodies. Tfr cells are involved in the generation and development of many immune-related and inflammatory diseases. This article summarizes the advances in several aspects of Tfr cell biology, with special focus on definition and phenotype, development and differentiation, regulatory factors, functions, and interactions with T/B cells and molecules involved in performance and regulation of Tfr function. Finally, we highlight the current understanding of Tfr cells involvement in autoimmunity and alloreactivity, and describe some drugs targeting Tfr cells. These latest studies have answered some basic questions in Tfr cell biology and explored the roles of Tfr cells in immune-mediated diseases.
Collapse
Affiliation(s)
- Yishan Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Mowang Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| |
Collapse
|
37
|
Yu F, Agrebi N, Mackeh R, Abouhazima K, KhudaBakhsh K, Adeli M, Lo B, Hassan A, Machaca K. Novel ORAI1 Mutation Disrupts Channel Trafficking Resulting in Combined Immunodeficiency. J Clin Immunol 2021; 41:1004-1015. [PMID: 33650027 PMCID: PMC8249264 DOI: 10.1007/s10875-021-01004-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/19/2021] [Indexed: 11/23/2022]
Abstract
Store-operated Ca2+ entry (SOCE) represents a predominant Ca2+ influx pathway in non-excitable cells. SOCE is required for immune cell activation and is mediated by the plasma membrane (PM) channel ORAI1 and the endoplasmic reticulum (ER) Ca2+ sensor STIM1. Mutations in the Orai1 or STIM1 genes abolish SOCE leading to combined immunodeficiency (CID), muscular hypotonia, and anhidrotic ectodermal dysplasia. Here, we identify a novel autosomal recessive mutation in ORAI1 in a child with CID. The patient is homozygous for p.C126R mutation in the second transmembrane domain (TM2) of ORAI1, a region with no previous loss-of-function mutations. SOCE is suppressed in the patient’s lymphocytes, which is associated with impaired T cell proliferation and cytokine production. Functional analyses demonstrate that the p.C126R mutation does not alter protein expression but disrupts ORAI1 trafficking. Orai1-C126R does not insert properly into the bilayer resulting in ER retention. Insertion of an Arg on the opposite face of TM2 (L135R) also results in defective folding and trafficking. We conclude that positive side chains within ORAI1 TM2 are not tolerated and result in misfolding, defective bilayer insertion, and channel trafficking thus abolishing SOCE and resulting in CID.
Collapse
Affiliation(s)
- Fang Yu
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar.,Calcium Signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Nourhen Agrebi
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
| | - Rafah Mackeh
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
| | - Khaled Abouhazima
- Pediatric Gastroenterology, Sidra Medicine, Education City, Doha, Qatar
| | | | - Mehdi Adeli
- Pediatric Allergy and Immunology Department, Sidra Medicine, Education City, Doha, Qatar
| | - Bernice Lo
- Translational Medicine Department, Sidra Medicine, Doha, Qatar. .,College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| | - Amel Hassan
- Pediatric Allergy and Immunology Department, Sidra Medicine, Education City, Doha, Qatar.
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar. .,Calcium Signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar.
| |
Collapse
|
38
|
Metabolic profiles of regulatory T cells in the tumour microenvironment. Cancer Immunol Immunother 2021; 70:2417-2427. [DOI: 10.1007/s00262-021-02881-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/01/2021] [Indexed: 12/27/2022]
|
39
|
Hofschröer V, Najder K, Rugi M, Bouazzi R, Cozzolino M, Arcangeli A, Panyi G, Schwab A. Ion Channels Orchestrate Pancreatic Ductal Adenocarcinoma Progression and Therapy. Front Pharmacol 2021; 11:586599. [PMID: 33841132 PMCID: PMC8025202 DOI: 10.3389/fphar.2020.586599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease with a dismal prognosis. Therapeutic interventions are largely ineffective. A better understanding of the pathophysiology is required. Ion channels contribute substantially to the "hallmarks of cancer." Their expression is dysregulated in cancer, and they are "misused" to drive cancer progression, but the underlying mechanisms are unclear. Ion channels are located in the cell membrane at the interface between the intracellular and extracellular space. They sense and modify the tumor microenvironment which in itself is a driver of PDAC aggressiveness. Ion channels detect, for example, locally altered proton and electrolyte concentrations or mechanical stimuli and transduce signals triggered by these microenvironmental cues through association with intracellular signaling cascades. While these concepts have been firmly established for other cancers, evidence has emerged only recently that ion channels are drivers of PDAC aggressiveness. Particularly, they appear to contribute to two of the characteristic PDAC features: the massive fibrosis of the tumor stroma (desmoplasia) and the efficient immune evasion. Our critical review of the literature clearly shows that there is still a remarkable lack of knowledge with respect to the contribution of ion channels to these two typical PDAC properties. Yet, we can draw parallels from ion channel research in other fibrotic and inflammatory diseases. Evidence is accumulating that pancreatic stellate cells express the same "profibrotic" ion channels. Similarly, it is at least in part known which major ion channels are expressed in those innate and adaptive immune cells that populate the PDAC microenvironment. We explore potential therapeutic avenues derived thereof. Since drugs targeting PDAC-relevant ion channels are already in clinical use, we propose to repurpose those in PDAC. The quest for ion channel targets is both motivated and complicated by the fact that some of the relevant channels, for example, KCa3.1, are functionally expressed in the cancer, stroma, and immune cells. Only in vivo studies will reveal which arm of the balance we should put our weights on when developing channel-targeting PDAC therapies. The time is up to explore the efficacy of ion channel targeting in (transgenic) murine PDAC models before launching clinical trials with repurposed drugs.
Collapse
Affiliation(s)
| | - Karolina Najder
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Micol Rugi
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Rayhana Bouazzi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Marco Cozzolino
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|
40
|
Bayati F, Mohammadi M, Valadi M, Jamshidi S, Foma AM, Sharif-Paghaleh E. The Therapeutic Potential of Regulatory T Cells: Challenges and Opportunities. Front Immunol 2021; 11:585819. [PMID: 33519807 PMCID: PMC7844143 DOI: 10.3389/fimmu.2020.585819] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) are an immunosuppressive subgroup of CD4+ T cells which are identified by the expression of forkhead box protein P3 (Foxp3). The modulation capacity of these immune cells holds an important role in both transplantation and the development of autoimmune diseases. These cells are the main mediators of self-tolerance and are essential for avoiding excessive immune reactions. Tregs play a key role in the induction of peripheral tolerance that can prevent autoimmunity, by protecting self-reactive lymphocytes from the immune reaction. In contrast to autoimmune responses, tumor cells exploit Tregs in order to prevent immune cell recognition and anti-tumor immune response during the carcinogenesis process. Recently, numerous studies have focused on unraveling the biological functions and principles of Tregs and their primary suppressive mechanisms. Due to the promising and outstanding results, Tregs have been widely investigated as an alternative tool in preventing graft rejection and treating autoimmune diseases. On the other hand, targeting Tregs for the purpose of improving cancer immunotherapy is being intensively evaluated as a desirable and effective method. The purpose of this review is to point out the characteristic function and therapeutic potential of Tregs in regulatory immune mechanisms in transplantation tolerance, autoimmune diseases, cancer therapy, and also to discuss that how the manipulation of these mechanisms may increase the therapeutic options.
Collapse
Affiliation(s)
- Fatemeh Bayati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Maryam Valadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Jamshidi
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Arron Munggela Foma
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
41
|
Abstract
T cells are an essential component of the immune system that provide antigen-specific acute and long lasting immune responses to infections and tumors, ascertain the maintenance of immunological tolerance and, on the flipside, mediate autoimmunity in a variety of diseases. The activation of T cells through antigen recognition by the T cell receptor (TCR) results in transient and sustained Ca2+ signals that are shaped by the opening of Ca2+ channels in the plasma membrane and cellular organelles. The dynamic regulation of intracellular Ca2+ concentrations controls a variety of T cell functions on the timescale of seconds to days after signal initiation. Among the more recently identified roles of Ca2+ signaling in T cells is the regulation of metabolic pathways that control the function of many T cell subsets. In this review, we discuss how Ca2+ regulates several metabolic programs in T cells such as the activation of AMPK and the PI3K-AKT-mTORC1 pathway, aerobic glycolysis, mitochondrial metabolism including tricarboxylic acid (TCA) cycle function and oxidative phosphorylation (OXPHOS), as well as lipid metabolism.
Collapse
Affiliation(s)
- Yinhu Wang
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Anthony Tao
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Martin Vaeth
- Institute of Systems Immunology, Julius Maximilians University of Würzburg, Würzburg, Germany
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
42
|
Seeing is believing: Visualizing immune cells and calcium signals at different stages of neuroinflammation. Proc Natl Acad Sci U S A 2020; 117:20360-20362. [PMID: 32769207 DOI: 10.1073/pnas.2013377117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
43
|
Vaeth M, Kahlfuss S, Feske S. CRAC Channels and Calcium Signaling in T Cell-Mediated Immunity. Trends Immunol 2020; 41:878-901. [PMID: 32711944 DOI: 10.1016/j.it.2020.06.012] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022]
Abstract
Calcium (Ca2+) signals play fundamental roles in immune cell function. The main sources of Ca2+ influx in mammalian lymphocytes following antigen receptor stimulation are Ca2+ release-activated Ca2+ (CRAC) channels. These are formed by ORAI proteins in the plasma membrane and are activated by stromal interaction molecules (STIM) located in the endoplasmic reticulum (ER). Human loss-of-function (LOF) mutations in ORAI1 and STIM1 that abolish Ca2+ influx cause a unique disease syndrome called CRAC channelopathy that is characterized by immunodeficiency autoimmunity and non-immunological symptoms. Studies in mice lacking Stim and Orai genes have illuminated many cellular and molecular mechanisms by which these molecules control lymphocyte function. CRAC channels are required for the differentiation and function of several T lymphocyte subsets that provide immunity to infection, mediate inflammation and prevent autoimmunity. This review examines new insights into how CRAC channels control T cell-mediated immunity.
Collapse
Affiliation(s)
- Martin Vaeth
- Institute of Systems Immunology, Julius-Maximilians University of Würzburg, Würzburg, Germany; Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Sascha Kahlfuss
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
44
|
Dhande IS, Zhu Y, Kneedler SC, Joshi AS, Hicks MJ, Wenderfer SE, Braun MC, Doris PA. Stim1 Polymorphism Disrupts Immune Signaling and Creates Renal Injury in Hypertension. J Am Heart Assoc 2020; 9:e014142. [PMID: 32075490 PMCID: PMC7335582 DOI: 10.1161/jaha.119.014142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Spontaneously hypertensive rats of the stroke‐prone line (SHR‐A3) develop hypertensive renal disease as a result of naturally occurring genetic variation. Our prior work identified a single‐nucleotide polymorphism unique to SHR‐A3 that results in truncation of the carboxy terminus of STIM1. The SHR‐B2 line, which is also hypertensive but resists hypertensive renal injury, expresses the wild‐type STIM1. STIM1 plays a central role in lymphocyte calcium signaling that directs immune effector responses. Here we show that major defects in lymphocyte function affecting calcium signaling, nuclear factor of activated T cells activation, cytokine production, proliferation, apoptosis, and regulatory T‐cell development are present in SHR‐A3 and attributable to STIM1. Methods and Results To assess the role of Stim1 variation in susceptibility to hypertensive renal injury, we created a Stim1 congenic line, SHR‐A3(Stim1‐B2), and STIM1 function was rescued in SHR‐A3. We found that Stim1 gene rescue restores disturbed lymphocyte function in SHR‐A3. Hypertensive renal injury was compared in SHR‐A3 and the SHR‐A3(Stim1‐B2) congenic line. Histologically assessed renal injury was markedly reduced in SHR‐A3(Stim1‐B2), as were renal injury biomarker levels measured in urine. Stim1 deficiency has been linked to the emergence of antibody‐mediated autoimmunity. Renal glomerular immunoglobulin deposition was greater in SHR‐A3 than SHR‐B2 and was reduced by Stim1 congenic substitution. Serum anti–double‐stranded DNA antibody titers in SHR‐A3 were elevated compared with SHR‐B2 and were reduced in SHR‐A3(Stim1‐B2). Conclusions Stim1 deficiency in lymphocyte function originating from Stim1 truncation in SHR‐A3 combines with hypertension to create end organ disease and may do so as a result of antibody formation.
Collapse
Affiliation(s)
- Isha S Dhande
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - Yaming Zhu
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - Sterling C Kneedler
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - Aniket S Joshi
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - M John Hicks
- Department of Pathology and Immunology Baylor College of Medicine and Texas Children's Hospital Houston TX
| | - Scott E Wenderfer
- Department of Pediatrics Baylor College of Medicine and Texas Children's Hospital Houston TX
| | - Michael C Braun
- Department of Pediatrics Baylor College of Medicine and Texas Children's Hospital Houston TX
| | - Peter A Doris
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| |
Collapse
|
45
|
Huang Y, Chen Z, Wang H, Ba X, Shen P, Lin W, Wang Y, Qin K, Huang Y, Tu S. Follicular regulatory T cells: a novel target for immunotherapy? Clin Transl Immunology 2020; 9:e1106. [PMID: 32082569 PMCID: PMC7019198 DOI: 10.1002/cti2.1106] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/22/2019] [Accepted: 01/05/2020] [Indexed: 12/15/2022] Open
Abstract
High‐affinity antibodies are produced during multiple processes in germinal centres (GCs), where follicular helper T (Tfh) cells interact closely with B cells to support B‐cell survival, differentiation and proliferation. Recent studies have revealed that a specialised subset of regulatory T cells, follicular regulatory T (Tfr) cells, especially fine‐tune Tfh cells and GC B cells, ultimately regulating GC reactions. Alterations in frequencies or function of Tfr cells may result in multiple autoantibody‐mediated or autoantibody‐associated diseases. This review discusses recent insights into the physiology and pathology of Tfr cells, with a special emphasis on their potential roles in human diseases. Discrepancies are common among studies, reflecting the limited understanding of Tfr cells. Further exploration of the mechanisms of Tfr cells in these diseases and thus targeting Tfr cells may help reinstate immune homeostasis and provide novel immunotherapy.
Collapse
Affiliation(s)
- Yao Huang
- Institute of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Zhe Chen
- Department of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Hui Wang
- Institute of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xin Ba
- Institute of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Pan Shen
- Institute of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Weiji Lin
- Institute of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yu Wang
- Department of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ying Huang
- Department of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shenghao Tu
- Institute of Integrated Traditional Chinese and Western Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
46
|
Bardos J, Fiorentino D, Longman RE, Paidas M. Immunological Role of the Maternal Uterine Microbiome in Pregnancy: Pregnancies Pathologies and Alterated Microbiota. Front Immunol 2020; 10:2823. [PMID: 31969875 PMCID: PMC6960114 DOI: 10.3389/fimmu.2019.02823] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
Understanding what happens at the time of embryo implantation has been the subject of significant research. Investigators from many differing fields including maternal fetal medicine, microbiology, genetics, reproductive endocrinology and immunology have all been studying the moment the embryo interacts with the maternal endometrium. A perfect relationship between the uterus and the embryo, mediated by a tightly controlled interaction between the embryo and the endometrium, is required for successful implantation. Any factors affecting this communication, such as altered microbiome may lead to poor reproductive outcomes. Current theories suggest that altered microbiota may trigger an inflammatory response in the endometrium that affects the success of embryo implantation, as inflammatory mediators are tightly regulated during the adhesion of the blastocyst to the epithelial endometrial wall. In this review, we will highlight the various microbiome found during the periconceptual period, the microbiomes interaction with immunological responses surrounding the time of implantation, its effect on implantation, placentation and ultimately maternal and neonatal outcomes.
Collapse
Affiliation(s)
- Jonah Bardos
- Department of Obstetrics and Gynecology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Division of Clinical and Translational Genetics, Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Desiree Fiorentino
- Department of Obstetrics and Gynecology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Division of Clinical and Translational Genetics, Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ryan E Longman
- Department of Obstetrics and Gynecology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Division of Clinical and Translational Genetics, Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Michael Paidas
- Department of Obstetrics and Gynecology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
47
|
Scheinecker C, Göschl L, Bonelli M. Treg cells in health and autoimmune diseases: New insights from single cell analysis. J Autoimmun 2019; 110:102376. [PMID: 31862128 DOI: 10.1016/j.jaut.2019.102376] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases, such as Systemic Lupus Erythematosus (SLE) or Rheumatoid Arthritis (RA) are characterized by the breakdown of immunological tolerance. Defects of regulatory T cells have been described among the various mechanisms, that are important for the development of autoimmune diseases, due to their critical role as regulators of peripheral immune tolerance and homeostasis. Initially T suppressor cells have been described as one population of peripheral T cells. Based on new technological advances a new understanding of the heterogeneity of different Treg cell populations in the lymphoid and non-lymphoid tissue has evolved over the last years. While initially Foxp3 has been defined as the main master regulator of Treg cells, we have learned that Treg cells from various tissue can be identified by a specific transcriptomic and epigenetic signature. Epigenetic mechanisms allow Treg cell stability, but we have also learned that certain Treg subsets are plastic and can under specific circumstances even enhance autoimmunity and inflammatory processes. Quantitative and functional defects of Treg cells have been observed in a variety of autoimmune diseases. Due to our understanding of the nature of this cell population, Treg cells have been a target of new Treg based therapies, such as low-dose IL-2. In addition, ongoing clinical trials aim to test safety and efficacy of transferred, in vitro expanded Treg cells in patients with autoimmune diseases and transplant patients.
Collapse
Affiliation(s)
- Clemens Scheinecker
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Lisa Göschl
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Michael Bonelli
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
48
|
Aloulou M, Fazilleau N. Regulation of B cell responses by distinct populations of CD4 T cells. Biomed J 2019; 42:243-251. [PMID: 31627866 PMCID: PMC6818157 DOI: 10.1016/j.bj.2019.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 01/28/2023] Open
Abstract
Maturation of B cells in Germinal Centers (GC) is a hallmark in adaptive immunity and the basis of successful vaccines that protect us against lethal infections. Nonetheless, vaccination efficacy is very much reduced in aged population and against highly mutagenic viruses. Therefore, it is key to understand how B cell selection takes place in GC in order to develop new and fully protective vaccines. The cellular mechanisms that control selection of GC B cells are performed by different T cell populations. On one side, cognate entanglement of B cells with T follicular helper (Tfh) cells through cytokines and co-stimulatory signals promotes survival, proliferation, mutagenesis and terminal differentiation of GC B cells. On the other hand, regulatory T cells have also been reported within GC and interfere with T cell help for antibody production. These cells have been classified as a distinct T cell sub-population called T Follicular regulatory cells (Tfr). In this review, we investigate the phenotype, function and differentiation of these two cell populations. In addition, based on the different functions of these cell subsets, we highlight the open questions surrounding their heterogeneity.
Collapse
Affiliation(s)
- Meryem Aloulou
- Center for Pathophysiology of Toulouse Purpan, Toulouse, France; INSERM U1043, Toulouse, France; CNRS UMR5282, Toulouse, France; University of Toulouse III, Toulouse, France
| | - Nicolas Fazilleau
- Center for Pathophysiology of Toulouse Purpan, Toulouse, France; INSERM U1043, Toulouse, France; CNRS UMR5282, Toulouse, France; University of Toulouse III, Toulouse, France.
| |
Collapse
|
49
|
Abstract
Regulatory T (Treg) cells expressing the transcription factor forkhead box P3 (Foxp3) play a requisite role in the maintenance of immunological homeostasis and prevention of peripheral self-tolerance breakdown. Although Foxp3 by itself is neither necessary nor sufficient to specify many aspects of the Treg cell phenotype, its sustained expression in Treg cells is indispensable for their phenotypic stability, metabolic fitness, and regulatory function. In this review, we summarize recent advances in Treg cell biology, with a particular emphasis on the role of Foxp3 as a transcriptional modulator and metabolic gatekeeper essential to an effective immune regulatory response. We discuss these findings in the context of human inborn errors of immune dysregulation, with a focus on FOXP3 mutations, leading to Treg cell deficiency. We also highlight emerging concepts of therapeutic Treg cell reprogramming to restore tolerance in the settings of immune dysregulatory disorders.
Collapse
|