1
|
Sharma V, Fernando V, Zheng X, Sweef O, Choi ES, Thomas V, Furuta S. Immunogenic shift of arginine metabolism triggers systemic metabolic and immunological reprogramming to prevent HER2+ breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619827. [PMID: 39484369 PMCID: PMC11527010 DOI: 10.1101/2024.10.23.619827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Arginine metabolism in tumors is often shunted into the pathway producing pro-tumor and immune suppressive polyamines (PAs), while downmodulating the alternative nitric oxide (NO) synthesis pathway. Aiming to correct arginine metabolism in tumors, arginine deprivation therapy and inhibitors of PA synthesis have been developed. Despite some therapeutic advantages, these approaches have often yielded severe side effects, making it necessary to explore an alternative strategy. We previously reported that supplementing SEP, the endogenous precursor of BH 4 (the essential NO synthase cofactor), could correct arginine metabolism in tumor cells and tumor-associated macrophages (TAMs) and induce their metabolic and phenotypic reprogramming. We saw that oral SEP treatment effectively suppressed the growth of HER2-positive mammary tumors in animals. SEP also has no reported dose-dependent toxicity in clinical trials for metabolic disorders. In the present study, we report that a long-term use of SEP in animals susceptible to HER2-positive mammary tumors effectively prevented tumor occurrence. These SEP-treated animals had undergone reprogramming of the systemic metabolism and immunity, elevating total T cell counts in the circulation and bone marrow. Given that bone marrow-resident T cells are mostly memory T cells, it is plausible that chronic SEP treatment promoted memory T cell formation, leading to a potent tumor prevention. These findings suggest the possible roles of the SEP/BH 4 /NO axis in promoting memory T cell formation and its potential therapeutic utility for preventing HER2-positive breast cancer.
Collapse
|
2
|
Park HJ, Shin MS, Shin JJ, Kim H, Kang B, Par-Young J, Unlu S, Afinogenova Y, Catanzaro J, Young J, Kim M, Lee SJ, Jeon S, You S, Racke MK, Bucala R, Kang I. IL-1 receptor 1 signaling shapes the development of viral antigen-specific CD4 + T cell responses following COVID-19 mRNA vaccination. EBioMedicine 2024; 103:105114. [PMID: 38640835 PMCID: PMC11041015 DOI: 10.1016/j.ebiom.2024.105114] [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: 11/06/2023] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND The innate immune cytokine interleukin (IL)-1 can affect T cell immunity, a critical factor in host defense. In a previous study, we identified a subset of human CD4+ T cells which express IL-1 receptor 1 (IL-1R1). However, the expression of such receptor by viral antigen-specific CD4+ T cells and its biological implication remain largely unexplored. This led us to investigate the implication of IL-1R1 in the development of viral antigen-specific CD4+ T cell responses in humans, including healthy individuals and patients with primary antibody deficiency (PAD), and animals. METHODS We characterized CD4+ T cells specific for SARS-CoV-2 spike (S) protein, influenza virus, and cytomegalovirus utilizing multiplexed single cell RNA-seq, mass cytometry and flow cytometry followed by an animal study. FINDINGS In healthy individuals, CD4+ T cells specific for viral antigens, including S protein, highly expressed IL-1R1. IL-1β promoted interferon (IFN)-γ expression by S protein-stimulated CD4+ T cells, supporting the functional implication of IL-1R1. Following the 2nd dose of COVID-19 mRNA vaccines, S protein-specific CD4+ T cells with high levels of IL-1R1 increased, likely reflecting repetitive antigenic stimulation. The expression levels of IL-1R1 by such cells correlated with the development of serum anti-S protein IgG antibody. A similar finding of increased expression of IL-1R1 by S protein-specific CD4+ T cells was also observed in patients with PAD following COVID-19 mRNA vaccination although the expression levels of IL-1R1 by such cells did not correlate with the levels of serum anti-S protein IgG antibody. In mice immunized with COVID-19 mRNA vaccine, neutralizing IL-1R1 decreased IFN-γ expression by S protein-specific CD4+ T cells and the development of anti-S protein IgG antibody. INTERPRETATION Our results demonstrate the significance of IL-1R1 expression in CD4+ T cells for the development of viral antigen-specific CD4+ T cell responses, contributing to humoral immunity. This provides an insight into the regulation of adaptive immune responses to viruses via the IL-1 and IL-1R1 interface. FUNDING Moderna to HJP, National Institutes of Health (NIH) 1R01AG056728 and R01AG055362 to IK and KL2 TR001862 to JJS, Quest Diagnostics to IK and RB, and the Mathers Foundation to RB.
Collapse
Affiliation(s)
- Hong-Jai Park
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Min Sun Shin
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Junghee J Shin
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Hyoungsu Kim
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA; Department of Internal Medicine, Hallym University School of Medicine, Chuncheon, Gangwon-do, 24252, South Korea
| | - Byunghyun Kang
- Mucosal Immunology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Jennefer Par-Young
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Serhan Unlu
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Yuliya Afinogenova
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jason Catanzaro
- Section of Pulmonary, Allergy, Immunology and Sleep Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Juan Young
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Minhyung Kim
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Sang Jin Lee
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA; Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, 41944, South Korea
| | - Sangchoon Jeon
- Yale University School of Nursing, West Haven, CT, 06516, USA
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | | | - Richard Bucala
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Insoo Kang
- Department of Internal Medicine (Section of Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
3
|
Chandran S, Tang Q. Impact of interleukin-6 on T cells in kidney transplant recipients. Am J Transplant 2022; 22 Suppl 4:18-27. [PMID: 36453710 DOI: 10.1111/ajt.17209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/23/2022] [Indexed: 12/02/2022]
Abstract
Interleukin-6 (IL-6), a multifunctional proinflammatory cytokine, plays a key role in T cell activation, survival, and differentiation. Acting as a switch that induces the differentiation of naïve T cells into Th17 cells and inhibits their development into regulatory T cells, IL-6 promotes rejection and abrogates tolerance. Therapies that target IL-6 signaling include antibodies to IL-6 and the IL-6 receptor and inhibitors of janus kinases; several of these therapeutics have demonstrated robust clinical efficacy in autoimmune and inflammatory diseases. Clinical trials of IL-6 inhibition in kidney transplantation have focused primarily on its effects on B cells, plasma cells, and HLA antibodies. In this review, we summarize the impact of IL-6 on T cells in experimental models of transplant and describe the effects of IL-6 inhibition on the T cell compartment in kidney transplant recipients.
Collapse
Affiliation(s)
- Sindhu Chandran
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Qizhi Tang
- Department of Surgery, Diabetes Center, Gladstone-UCSF Institute of Genome Immunology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
4
|
Romero-Martín L, Tarrés-Freixas F, Pedreño-López N, de la Concepción MLR, Cunyat F, Hartigan-O'Connor D, Carrillo J, Mothe B, Blanco J, Ruiz-Riol M, Brander C, Olvera A. T-Follicular-Like CD8 + T Cell Responses in Chronic HIV Infection Are Associated With Virus Control and Antibody Isotype Switching to IgG. Front Immunol 2022; 13:928039. [PMID: 35784304 PMCID: PMC9241491 DOI: 10.3389/fimmu.2022.928039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 01/26/2023] Open
Abstract
T cell responses are considered critical for the in vivo control of HIV, but the contribution of different T cell subsets to this control remains unclear. Using a boosted flow cytometric approach that is able to differentiate CD4+ and CD8+ T cell Th1/Tc1, Th2/Tc2, Th17/Tc17, Treg and Tfh/Tfc-like HIV-specific T cell populations, we identified CD8+ Tfc responses that were related to HIV plasma viral loads and associated with rate of antibody isotype class switching to IgG. This favorable balance towards IgG responses positively correlated with increased virus neutralization, higher avidity of neutralizing antibodies and more potent antibody-dependent cell cytotoxicity (ADCC) in PBMCs from HIV controllers compared to non-controllers. Our results identified the CD8+ Tfc-like T-cell response as a component of effective virus control which could possibly be exploited therapeutically.
Collapse
Affiliation(s)
- Luis Romero-Martín
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- Departament de Biologia Cellular, de Fisiologia i d’Immunologia, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Ferran Tarrés-Freixas
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Núria Pedreño-López
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Maria L. Rodríguez de la Concepción
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Francesc Cunyat
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Dennis Hartigan-O'Connor
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Marta Ruiz-Riol
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- AELIX Therapeutics, Barcelona, Spain
| | - Alex Olvera
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| |
Collapse
|
5
|
Smith AM, Ramirez RM, Harper N, Jimenez F, Branum AP, Meunier JA, Pandranki L, Carrillo A, Winter C, Winter L, Rather CG, Ramirez DA, Andrews CP, Restrepo MI, Maselli DJ, Pugh JA, Clark RA, Lee GC, Moreira AG, Manoharan MS, Okulicz JF, Jacobs RL, Ahuja SK. Large-scale provocation studies identify maladaptive responses to ubiquitous aeroallergens as a correlate of severe allergic rhinoconjunctivitis and asthma. Allergy 2022; 77:1797-1814. [PMID: 34606106 PMCID: PMC9298287 DOI: 10.1111/all.15124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/24/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Allergic asthma (AA) and allergic rhinoconjunctivitis (ARC) are common comorbid environmentally triggered diseases. We hypothesized that severe AA/ARC reflects a maladaptive or unrestrained response to ubiquitous aeroallergens. METHODS We performed provocation studies wherein six separate cohorts of persons (total n = 217) with ARC, with or without AA, were challenged once or more with fixed concentrations of seasonal or perennial aeroallergens in an aeroallergen challenge chamber (ACC). RESULTS Aeroallergen challenges elicited fully or partially restrained vs. unrestrained evoked symptom responsiveness, corresponding to the resilient and adaptive vs. maladaptive AA/ARC phenotypes, respectively. The maladaptive phenotype was evoked more commonly during challenge with a non-endemic versus endemic seasonal aeroallergen. In an AA cohort, symptom responses evoked after house dust mite (HDM) challenges vs. recorded in the natural environment were more accurate and precise predictors of asthma severity and control, lung function (FEV1), and mechanistic correlates of maladaptation. Correlates included elevated levels of peripheral blood CD4+ and CD8+ T-cells, eosinophils, and T-cell activation, as well as gene expression proxies for ineffectual epithelial injury/repair responses. Evoked symptom severity after HDM challenge appeared to be more closely related to levels of CD4+ and CD8+ T-cells than eosinophils, neutrophils, or HDM-specific IgE. CONCLUSIONS Provocation studies support the concept that resilience, adaptation, and maladaptation to environmental disease triggers calibrate AA/ARC severity. Despite the ubiquity of aeroallergens, in response to these disease triggers in controlled settings (ie, ACC), most atopic persons manifest the resilient or adaptive phenotype. Thus, ARC/AA disease progression may reflect the failure to preserve the resilient or adaptive phenotype. The triangulation of CD8+ T-cell activation, airway epithelial injury/repair processes and maladaptation in mediating AA disease severity needs more investigation.
Collapse
|
6
|
Wu S, Wang A. Serum level and clinical significance of vitamin E in pregnant women with allergic rhinitis. J Chin Med Assoc 2022; 85:597-602. [PMID: 35324489 DOI: 10.1097/jcma.0000000000000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Allergic rhinitis is a frequent disorder during pregnancy, while in children it is triggered by significantly lower serum vitamin E level. This research aimed to investigate whether serum vitamin E level exhibited clinical significance in pregnant women with allergic rhinitis. METHODS In this study, 37 pregnant women with allergic rhinitis and 35 healthy pregnant women were recruited. Allergic rhinitis severity was analyzed by the Total Nasal Symptom Score (TNSS) questionnaire. Blood samples were collected to evaluate serum vitamin E, interleukin (IL), and total IgE levels. RESULTS In pregnant women with allergic rhinitis, serum level of vitamin E was significantly lower than in healthy pregnant women. Serum vitamin E level in pregnant women with allergic rhinitis showed a negative correlation with TNSS, IL-13, IL-4, and total IgE levels. CONCLUSION In conclusion, this research has demonstrated that pregnant women with allergic rhinitis showed significantly lower serum level of vitamin E. The decreased vitamin E showed a correlation with the pathogenesis of allergic rhinitis in pregnant women.
Collapse
Affiliation(s)
- Sihai Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Aiping Wang
- Department of Obstetrics and Gynecology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
7
|
Zhang H, Liu S, Li Y, Li J, Ni C, Yang M, Dong J, Wang Z, Qin Z. Dysfunction of S100A4 + effector memory CD8 + T cells aggravates asthma. Eur J Immunol 2022; 52:978-993. [PMID: 35340022 DOI: 10.1002/eji.202149572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/09/2022]
Abstract
Progressive loss of effector functions, especially IFN-γ secreting capability, in effector memory CD8+ T (CD8+ TEM ) cells plays a crucial role in asthma worsening. However, the mechanisms of CD8+ TEM cell dysfunction remain elusive. Here, we report that S100A4 drives CD8+ TEM cell dysfunction, impairing their protective memory response and promoting asthma worsening in an ovalbumin (OVA)-induced asthmatic murine model. We find that CD8+ TEM cells contain two subsets based on S100A4 expression. S100A4+ subsets exhibit dysfunctional effector phenotypes with increased proliferative capability, whereas S100A4- subsets retain effector function but are more inclined to apoptosis, giving rise a dysfunctional CD8+ TEM cell pool. Mechanistically, S100A4 upregulation of mitochondrial metabolism results in a decrease of acetyl-CoA levels, which impair the transcription of effector genes, especially ifn-γ, facilitating cell survival, tolerance and memory potential. Our findings thus reveal general insights into how S100A4 CD8+ TEM cells reprogram into dysfunctional and less protective phenotypes to aggravate asthma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Huilei Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuangqing Liu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanan Li
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianru Li
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Ni
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2300, Australia
| | - Jun Dong
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, 10117, Germany
| | - Zhaoqing Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
8
|
de Freitas DDN, Marinho Franceschina C, Muller D, Hilario GT, Gassen RB, Fazolo T, de Lima Kaminski V, Bogo Chies JA, Maito F, Antunes KH, Zanin RF, Rodrigues LC, Duarte de Souza AP. RvD1 treatment during primary infection modulates memory response increasing viral load during respiratory viral reinfection. Immunobiology 2021; 226:152151. [PMID: 34742024 DOI: 10.1016/j.imbio.2021.152151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 11/25/2022]
Abstract
Resolvin D1 (RvD1), which is biosynthesized from essential long-chain fatty acids, is involved in anti-inflammatory activity and modulation of T cell response. Memory CD8+ T cells are important for controlling tumor growth and viral infections. Exacerbated inflammation has been described as impairing memory CD8+ T cell differentiation. This study aimed to verify the effects of RvD1 on memory CD8+ T cells in vitro and in vivo in a respiratory virus infection model. Peripheral blood mononuclear cells were treated at different time points with RvD1 and stimulated with anti-CD3/anti-CD28 antibodies. Pre-treatment with RvD1 increases the expansion of memory CD8+ T cells. The IL-12 level, a cytokine described to control memory CD8+ T cells, was reduced with RvD1 pre-treatment. When the mTOR axis was inhibited, the IL-12 levels were restored. In a respiratory virus infection model, Balb/c mice were treated with RvD1 before infection or after 7 days after infection. RvD1 treatment after infection increased the frequency of memory CD8+ T cells in the lung expressing II4, II10, and Ifng. During reinfection, RvD1-treated and RSV-infected mice present a high viral load in the lung and lower antibody response in the serum. Our results show that RvD1 modulates the expansion and phenotype of memory CD8+ T cells but contributed to a non-protective response after RSV reinfection.
Collapse
Affiliation(s)
- Deise do Nascimento de Freitas
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Caroline Marinho Franceschina
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Douglas Muller
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Gabriel T Hilario
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Rodrigo B Gassen
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tiago Fazolo
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Valéria de Lima Kaminski
- Applied Immunology Laboratory, Postgraduate Program in Biotechnology, Institute of Science and Technology - ICT, Federal University of São Paulo - UNIFESP, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunogenetics and Immunobiology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fábio Maito
- Laboratory of Histology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, RS, Brazil
| | - Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Infant Center, School of Medicine PUCRS, Porto Alegre, RS, Brazil
| | - Rafael F Zanin
- Department of Health and Human Development, La Salle University, Canoas, RS, Brazil
| | - Luiz Carlos Rodrigues
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Infant Center, School of Medicine PUCRS, Porto Alegre, RS, Brazil.
| |
Collapse
|
9
|
Repetitive aeroallergen challenges elucidate maladaptive epithelial and inflammatory traits that underpin allergic airway diseases. J Allergy Clin Immunol 2021; 148:533-549. [PMID: 33493557 PMCID: PMC8298629 DOI: 10.1016/j.jaci.2021.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/08/2021] [Accepted: 01/14/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Signifying the 2-compartments/1-disease paradigm, allergic rhinoconjunctivitis (ARC) and asthma (AA) are prevalent, comorbid conditions triggered by environmental factors (eg, house dust mites [HDMs]). However, despite the ubiquity of triggers, progression to severe ARC/AA is infrequent, suggesting either resilience or adaptation. OBJECTIVE We sought to determine whether ARC/AA severity relates to maladaptive responses to disease triggers. METHODS Adults with HDM-associated ARC were challenged repetitively with HDMs in an aeroallergen challenge chamber. Mechanistic traits associated with disease severity were identified. RESULTS HDM challenges evoked maladaptive (persistently higher ARC symptoms), adaptive (progressive symptom reduction), and resilient (resistance to symptom induction) phenotypes. Symptom severity in the natural environment was an imprecise correlate of the phenotypes. Nasal airway traits, defined by low inflammation-effectual epithelial integrity, moderate inflammation-effectual epithelial integrity, and higher inflammation-ineffectual epithelial integrity, were hallmarks of the resilient, adaptive, and maladaptive evoked phenotypes, respectively. Highlighting a crosstalk mechanism, peripheral blood inflammatory tone calibrated these traits: ineffectual epithelial integrity associated with CD8+ T cells, whereas airway inflammation associated with both CD8+ T cells and eosinophils. Hallmark peripheral blood maladaptive traits were increased natural killer and CD8+ T cells, lower CD4+ mucosal-associated invariant T cells, and deficiencies along the TLR-IRF-IFN antiviral pathway. Maladaptive traits tracking HDM-associated ARC also contributed to AA risk and severity models. CONCLUSIONS Repetitive challenges with HDMs revealed that maladaptation to disease triggers may underpin ARC/AA disease severity. A combinatorial therapeutic approach may involve reversal of loss-of-beneficial-function traits (ineffectual epithelial integrity, TLR-IRF-IFN deficiencies), mitigation of gain-of-adverse-function traits (inflammation), and blocking of a detrimental crosstalk between the peripheral blood and airway compartments.
Collapse
|
10
|
Pritzl CJ, Daniels MA, Teixeiro E. Interplay of Inflammatory, Antigen and Tissue-Derived Signals in the Development of Resident CD8 Memory T Cells. Front Immunol 2021; 12:636240. [PMID: 34234771 PMCID: PMC8255970 DOI: 10.3389/fimmu.2021.636240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/29/2021] [Indexed: 12/21/2022] Open
Abstract
CD8 positive, tissue resident memory T cells (TRM) are a specialized subset of CD8 memory T cells that surveil tissues and provide critical first-line protection against tumors and pathogen re-infection. Recently, much effort has been dedicated to understanding the function, phenotype and development of TRM. A myriad of signals is involved in the development and maintenance of resident memory T cells in tissue. Much of the initial research focused on the roles tissue-derived signals play in the development of TRM, including TGFß and IL-33 which are critical for the upregulation of CD69 and CD103. However, more recent data suggest further roles for antigenic and pro-inflammatory cytokines. This review will focus on the interplay of pro-inflammatory, tissue and antigenic signals in the establishment of resident memory T cells.
Collapse
Affiliation(s)
| | | | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
11
|
Infectious Complications Predict Premature CD8 + T-cell Senescence in CD40 Ligand-Deficient Patients. J Clin Immunol 2021; 41:795-806. [PMID: 33495902 DOI: 10.1007/s10875-021-00968-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/06/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE CD40 ligand (CD40L)-deficient patients display increased susceptibilities to infections that can be mitigated with effective prophylactic strategies including immunoglobulin G (IgG) replacement and prophylactic antibiotics. CD8+ T-cell senescence has been described in CD40L deficiency, but it is unclear if this is an intrinsic feature of the disease or secondary to infectious exposures. To address this question, we assessed CD8+ T-cell senescence and its relationship to clinical histories, including prophylaxis adherence and infections, in CD40L-deficient patients. METHODS Peripheral CD8+ T-cells from seven CD40L-deficient patients and healthy controls (HCs) were assessed for senescent features using T-cell receptor excision circle (TREC) analysis, flow cytometry, cytometry by time of flight (CyTOF) and in vitro functional determinations including CMV-specific proliferation and cytokine release assays. RESULTS Three patients (5, 28, and 34 years old) who were poorly adherent to immunoglobulin G replacement and Pneumocystis jirovecii pneumonia (PJP) prophylaxis and/or experienced multiple childhood pneumonias (patient group 1) had an expansion of effector memory CD8+ T-cells with the senescent phenotype when compared to HCs. Such changes were not observed in the patient group 2 (four patients, 16, 22, 24, and 33 years old) who were life-long adherents to prophylaxis and experienced few infectious complications. CyTOF analysis of CD8+ T-cells from the 5-year-old patient and older adult HCs showed similar expression patterns of senescence-associated molecules. CONCLUSIONS Our findings support that recurrent infections and non-adherence to prophylaxis promote early CD8+ T-cell senescence in CD40L deficiency. Premature senescence may increase malignant susceptibilities and further exacerbate infectious risk in CD40L-deficient patients.
Collapse
|
12
|
Kanno T, Adachi Y, Ohashi-Doi K, Matsuhara H, Hiratsuka R, Ishibashi KI, Yamanaka D, Ohno N. Latent 1,3-β-D-glucan acts as an adjuvant for allergen-specific IgE production induced by Japanese cedar pollen exposure. Allergol Int 2021; 70:105-113. [PMID: 32919904 DOI: 10.1016/j.alit.2020.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/06/2020] [Accepted: 08/13/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The pollen grains of several plant species contain 1,3-β-D-glucan (BG). BG activates dendritic cells (DCs) and subsequently regulates the innate immune responses. Within Japan, the most common disease associated with type-I hypersensitivity is Japanese cedar pollinosis. However, the role of BG in Japanese cedar pollen (JCP) remains unclear. This study examined the localization and immunological effects of BG in JCP. METHODS The localization of BG in JCP grain was determined by immunohistochemical staining using a soluble dectin-1 protein probe and a BG recognition protein (BGRP). The content of BG extracted from JCP was measured by a BGRP-based ELISA-like assay. The cytokine production by bone marrow-derived DCs (BMDCs) obtained from wild-type and BG receptor (dectin-1) knock-out mice was examined in vitro. The mice were intranasally administered JCP grains and the specific serum Ig levels were then quantified. RESULTS BG was detected in the exine and cell wall of the generative cell and tube cell of the JCP grain. Moreover, BG in the exine stimulated production of TNF-α and IL-6 in the BMDCs via a dectin-1-dependent mechanism. Meanwhile, JCP-specific IgE and IgG were detected in the serum of wild-type mice that had been intranasally administered with JCP grains. These mice also exhibited significantly enhanced sneezing behavior. However, dectin-1 knock-out mice exhibited significantly lower JCP-specific IgE and IgG levels compared to wild-type mice. CONCLUSIONS Latent BG in JCP can act as an adjuvant to induce JCP-specific antibody production via dectin-1.
Collapse
Affiliation(s)
- Takashi Kanno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshiyuki Adachi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.
| | | | - Hiroki Matsuhara
- Research Laboratory, Torii Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Rie Hiratsuka
- Division of Biology, Department of Natural Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Ken-Ichi Ishibashi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Daisuke Yamanaka
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Naohito Ohno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
13
|
Qin Z, Wang PY, Wan JJ, Zhang Y, Wei J, Sun Y, Liu X. MicroRNA124-IL6R Mediates the Effect of Nicotine in Inflammatory Bowel Disease by Shifting Th1/Th2 Balance Toward Th1. Front Immunol 2020; 11:235. [PMID: 32153570 PMCID: PMC7050625 DOI: 10.3389/fimmu.2020.00235] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/28/2020] [Indexed: 12/20/2022] Open
Abstract
Epidemiological investigations have shown that smoking ameliorates ulcerative colitis (UC) but exacerbates Crohn's disease (CD), diseases that feature a Th2-mediated and Th1-mediated response, respectively. Cigarette extracts, especially nicotine, affect the Th1/Th2 balance. We previously reported that nicotine protects against mouse DSS colitis (similar to UC) by enhancing microRNA-124 (miR-124) expression. Intriguingly, elevation of miR-124 in CD is reported to aggravate the disease. Here we investigate the dual regulation of miR-124 in inflammatory bowel diseases (IBDs), which may explain the similar bidirectional regulation of tobacco. We found that overexpressed miR-124 protected against mouse DSS-induced colitis with a Th1 polarization in peripheral blood lymphocytes and colon tissues, which was also found in human peripheral blood lymphocytes. Conversely, miR-124 knockdown worsened DSS murine colitis with a Th2 polarization. Moreover, knockdown of miR-124 could eliminate the polarization toward Th1 after nicotine treatment, suggesting that miR-124 mediates the effect of nicotine on the Th1/Th2 balance. In addition, interference of IL-6R, which is a downstream target of miR-124, could remarkably weaken the Th1 polarization induced by miR-124. Taken together, these results suggest that nicotine shifts the balance of Th1/Th2 toward Th1 via a miR-124-mediated IL-6R pathway, which might explain its dual role in IBDs.
Collapse
Affiliation(s)
- Zhen Qin
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Peng-Yuan Wang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jing-Jing Wan
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yu Zhang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jie Wei
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yang Sun
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
14
|
Hinks TSC, Hoyle RD, Gelfand EW. CD8 + Tc2 cells: underappreciated contributors to severe asthma. Eur Respir Rev 2019; 28:28/154/190092. [PMID: 31748421 PMCID: PMC6887553 DOI: 10.1183/16000617.0092-2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 01/22/2023] Open
Abstract
The complexity of asthma is underscored by the number of cell types and mediators implicated in the pathogenesis of this heterogeneous syndrome. Type 2 CD4+ T-cells (Th2) and more recently, type 2 innate lymphoid cells dominate current descriptions of asthma pathogenesis. However, another important source of these type 2 cytokines, especially interleukin (IL)-5 and IL-13, are CD8+ T-cells, which are increasingly proposed to play an important role in asthma pathogenesis, because they are abundant and are comparatively insensitive to corticosteroids. Many common triggers of asthma exacerbations are mediated via corticosteroid-resistant pathways involving neutrophils and CD8+ T-cells. Extensive murine data reveal the plasticity of CD8+ T-cells and their capacity to enhance airway inflammation and airway dysfunction. In humans, Tc2 cells are predominant in fatal asthma, while in stable state, severe eosinophilic asthma is associated with greater numbers of Tc2 than Th2 cells in blood, bronchoalveolar lavage fluid and bronchial biopsies. Tc2 cells strongly express CRTH2, the receptor for prostaglandin D2, the cysteinyl leukotriene receptor 1 and the leukotriene B4 receptor. When activated, these elicit Tc2 cell chemotaxis and production of chemokines and type 2 and other cytokines, resulting directly or indirectly in eosinophil recruitment and survival. These factors position CD8+ Tc2 cells as important and underappreciated effector cells contributing to asthma pathogenesis. Here, we review recent advances and new insights in understanding the pro-asthmatic functions of CD8+ T-cells in eosinophilic asthma, especially corticosteroid-resistant asthma, and the molecular mechanisms underlying their pathologic effector function. Alongside Th2 and ILC2 cells, CD8+ T-cells are a cellular source of type 2 cytokines. We review recent findings and insights into the pathologic effector functions of type 2 CD8+ T-cells in eosinophilic asthma, especially steroid-resistant disease.http://bit.ly/2KbVGL2
Collapse
Affiliation(s)
- Timothy S C Hinks
- Respiratory Medicine Unit and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Nuffield Dept of Medicine Experimental Medicine, University of Oxford, Oxford, UK
| | - Ryan D Hoyle
- Respiratory Medicine Unit and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Nuffield Dept of Medicine Experimental Medicine, University of Oxford, Oxford, UK
| | - Erwin W Gelfand
- Division of Cell Biology, Dept of Pediatrics, National Jewish Health, Denver, CO, USA
| |
Collapse
|
15
|
Park H, Shin MS, Kim M, Bilsborrow JB, Mohanty S, Montgomery RR, Shaw AC, You S, Kang I. Transcriptomic analysis of human IL-7 receptor alpha low and high effector memory CD8 + T cells reveals an age-associated signature linked to influenza vaccine response in older adults. Aging Cell 2019; 18:e12960. [PMID: 31044512 PMCID: PMC6612637 DOI: 10.1111/acel.12960] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/10/2019] [Indexed: 12/20/2022] Open
Abstract
Here, we investigated the relationship of the age‐associated expansion of IL‐7 receptor alpha low (IL‐7Rαlow) effector memory (EM) CD8+ T cells with the global transcriptomic profile of peripheral blood cells in humans. We found 231 aging signature genes of IL‐7Rαlow EM CD8+ T cells that corresponded to 15% of the age‐associated genes (231/1,497) reported by a meta‐analysis study on human peripheral whole blood from approximately 15,000 individuals, having high correlation with chronological age. These aging signature genes were the target genes of several transcription factors including MYC, SATB1, and BATF, which also belonged to the 231 genes, supporting the upstream regulatory role of these transcription factors in altering the gene expression profile of peripheral blood cells with aging. We validated the differential expression of these transcription factors between IL‐7Rαlow and high EM CD8+ T cells as well as in peripheral blood mononuclear cells (PBMCs) of young and older adults. Finally, we found a significant association with influenza vaccine responses in older adults, suggesting the possible biological significance of the aging signature genes of IL‐7Rαlow EM CD8+ T cells. The results of our study support the relationship of the expansion of IL‐7Rαlow EM CD8+ T cells with the age‐associated changes in the gene expression profile of peripheral blood cells and its possible biological implications.
Collapse
Affiliation(s)
- Hong‐Jai Park
- Department of Internal Medicine Yale University School of Medicine New Haven Connecticut
| | - Min Sun Shin
- Department of Internal Medicine Yale University School of Medicine New Haven Connecticut
| | - Minhyung Kim
- Departments of Surgery and Biomedical Sciences Cedars‐Sinai Medical Center Los Angeles California
| | - Joshua B. Bilsborrow
- Department of Internal Medicine Yale University School of Medicine New Haven Connecticut
| | - Subhasis Mohanty
- Department of Internal Medicine Yale University School of Medicine New Haven Connecticut
| | - Ruth R. Montgomery
- Department of Internal Medicine Yale University School of Medicine New Haven Connecticut
| | - Albert C. Shaw
- Department of Internal Medicine Yale University School of Medicine New Haven Connecticut
| | - Sungyong You
- Departments of Surgery and Biomedical Sciences Cedars‐Sinai Medical Center Los Angeles California
- Samuel Oschin Comprehensive Cancer Institute Cedars‐Sinai Medical Center Los Angeles California
| | - Insoo Kang
- Department of Internal Medicine Yale University School of Medicine New Haven Connecticut
| |
Collapse
|
16
|
Gelfand EW, Hinks TSC. Is there a role for type 2 CD8 + T cells in patients with steroid-resistant asthma? J Allergy Clin Immunol 2019; 144:648-650. [PMID: 31376407 DOI: 10.1016/j.jaci.2019.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/01/2022]
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo.
| | - Timothy S C Hinks
- Respiratory Medicine Unit and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Nuffield Department of Medicine Experimental Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Transcription factors gene expression in chronic rhinosinusitis with and without nasal polyps. Radiol Oncol 2019; 53:323-330. [PMID: 31326962 PMCID: PMC6765166 DOI: 10.2478/raon-2019-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022] Open
Abstract
Background Chronic rhinosinusitis (CRS) current therapeutic approaches still fail in some patients with severe persistent symptoms and recurrences after surgery. We aimed to evaluate the master transcription factors gene expression levels of T cell subtypes in chronic rhinosinusitis with nasal polyps (CRSwNP) and chronic rhinosinusitis without nasal polyps (CRSsNP) that could represent new, up-stream targets for topical DNAzyme treatment. Patients and methods Twenty-two newly diagnosed CRS patients (14 CRSwNP and 8 CRSsNP) were prospectively biopsied and examined histopathologically. Gene expression levels of T-box transcription factor (T-bet, TBX21), GATA binding protein 3 (GATA3), Retinoic acid-related orphan receptor C (RORC) and Forkhead box P3 (FOXP3) were analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). Results Eosinophilic CRSwNP was characterized by higher level of GATA3 gene expression compared to noneosinophilic CRSwNP, whereas there was no difference in T-bet, RORC and FOXP3 between eosinophilic and noneosinophilic CRSwNP. In CRSsNP, we found simultaneous upregulation of T-bet, GATA3 and RORC gene expression levels in comparison to CRSwNP; meanwhile, there was no difference in FOXP3 gene expression between CRSwNP and CRSsNP. Conclusions In eosinophilic CRSwNP, we confirmed the type 2 inflammation by elevated GATA3 gene expression level. In CRSsNP, we unexpectedly found simultaneous upregulation of T-bet and GATA3 that is currently unexplained; however, it might originate from activated CD8+ cells, abundant in nasal mucosa of CRSsNP patients. The elevated RORC in CRSsNP could be part of homeostatic nasal immune response that might be better preserved in CRSsNP patients compared to CRSwNP patients. Further data on transcription factors expression rates in CRS phenotypes are needed.
Collapse
|
18
|
Expansion of different subpopulations of CD26 -/low T cells in allergic and non-allergic asthmatics. Sci Rep 2019; 9:7556. [PMID: 31101830 PMCID: PMC6525268 DOI: 10.1038/s41598-019-43622-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
CD26 displays variable levels between effector (TH17 ≫ TH1 > TH2 > Treg) and naïve/memory (memory > naïve) CD4+ T lymphocytes. Besides, IL-6/IL−6R is associated with TH17-differentiation and asthma severity. Allergic/atopic asthma (AA) is dominated by TH2 responses, while TH17 immunity might either modulate the TH2-dependent inflammation in AA or be an important mechanism boosting non-allergic asthma (NAA). Therefore, in this work we have compared the expression of CD26 and CD126 (IL-6Rα) in lymphocytes from different groups of donors: allergic (AA) and non-allergic (NAA) asthma, rhinitis, and healthy subjects. For this purpose, flow cytometry, haematological/biochemical, and in vitro proliferation assays were performed. Our results show a strong CD26-CD126 correlation and an over-representation of CD26− subsets with a highly-differentiated effector phenotype in AA (CD4+CD26−/low T cells) and NAA (CD4−CD26− γδ-T cells). In addition, we found that circulating levels of CD26 (sCD26) were reduced in both AA and NAA, while loss of CD126 expression on different leukocytes correlated with higher disease severity. Finally, selective inhibition of CD26-mRNA translation led to enhanced T cell proliferation in vitro. These findings support that CD26 down-modulation could play a role in facilitating the expansion of highly-differentiated effector T cell subsets in asthma.
Collapse
|
19
|
Lacher MD, Bauer G, Fury B, Graeve S, Fledderman EL, Petrie TD, Coleal-Bergum DP, Hackett T, Perotti NH, Kong YY, Kwok WW, Wagner JP, Wiseman CL, Williams WV. SV-BR-1-GM, a Clinically Effective GM-CSF-Secreting Breast Cancer Cell Line, Expresses an Immune Signature and Directly Activates CD4 + T Lymphocytes. Front Immunol 2018; 9:776. [PMID: 29867922 PMCID: PMC5962696 DOI: 10.3389/fimmu.2018.00776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Targeted cancer immunotherapy with irradiated, granulocyte–macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic cancer cell lines has been an effective approach to reduce tumor burden in several patients. It is generally assumed that to be effective, these cell lines need to express immunogenic antigens coexpressed in patient tumor cells, and antigen-presenting cells need to take up such antigens then present them to patient T cells. We have previously reported that, in a phase I pilot study (ClinicalTrials.gov NCT00095862), a subject with stage IV breast cancer experienced substantial regression of breast, lung, and brain lesions following inoculation with clinical formulations of SV-BR-1-GM, a GM-CSF-secreting breast tumor cell line. To identify diagnostic features permitting the prospective identification of patients likely to benefit from SV-BR-1-GM, we conducted a molecular analysis of the SV-BR-1-GM cell line and of patient-derived blood, as well as a tumor specimen. Compared to normal human breast cells, SV-BR-1-GM cells overexpress genes encoding tumor-associated antigens (TAAs) such as PRAME, a cancer/testis antigen. Curiously, despite its presumptive breast epithelial origin, the cell line expresses major histocompatibility complex (MHC) class II genes (HLA-DRA, HLA-DRB3, HLA-DMA, HLA-DMB), in addition to several other factors known to play immunostimulatory roles. These factors include MHC class I components (B2M, HLA-A, HLA-B), ADA (encoding adenosine deaminase), ADGRE5 (CD97), CD58 (LFA3), CD74 (encoding invariant chain and CLIP), CD83, CXCL8 (IL8), CXCL16, HLA-F, IL6, IL18, and KITLG. Moreover, both SV-BR-1-GM cells and the responding study subject carried an HLA-DRB3*02:02 allele, raising the question of whether SV-BR-1-GM cells can directly present endogenous antigens to T cells, thereby inducing a tumor-directed immune response. In support of this, SV-BR-1-GM cells (which also carry the HLA-DRB3*01:01 allele) treated with yellow fever virus (YFV) envelope (Env) 43–59 peptides reactivated YFV-DRB3*01:01-specific CD4+ T cells. Thus, the partial HLA allele match between SV-BR-1-GM and the clinical responder might have enabled patient T lymphocytes to directly recognize SV-BR-1-GM TAAs as presented on SV-BR-1-GM MHCs. Taken together, our findings are consistent with a potentially unique mechanism of action by which SV-BR-1-GM cells can act as APCs for previously primed CD4+ T cells.
Collapse
Affiliation(s)
| | - Gerhard Bauer
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Brian Fury
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Sanne Graeve
- BriaCell Therapeutics Corp., Berkeley, CA, United States
| | - Emily L Fledderman
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tye D Petrie
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Dane P Coleal-Bergum
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tia Hackett
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Nicholas H Perotti
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Ying Y Kong
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | | | | | | |
Collapse
|
20
|
Suwanpradid J, Shih M, Pontius L, Yang B, Birukova A, Guttman-Yassky E, Corcoran DL, Que LG, Tighe RM, MacLeod AS. Arginase1 Deficiency in Monocytes/Macrophages Upregulates Inducible Nitric Oxide Synthase To Promote Cutaneous Contact Hypersensitivity. THE JOURNAL OF IMMUNOLOGY 2017; 199:1827-1834. [PMID: 28747341 DOI: 10.4049/jimmunol.1700739] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/18/2017] [Indexed: 12/24/2022]
Abstract
The innate immune components that modulate allergic contact hypersensitivity (CHS) responses are poorly defined. Using human skin from contact dermatitis patients and a mouse model of CHS, we find that hapten allergens disrupt the Arginase1 (Arg1) and inducible NO synthase (iNOS) dynamic in monocytes/macrophages (mono/MΦ), which renders those cells ineffectual in suppressing skin inflammation. Mice lacking Arg1 in MΦ develop increased CHS characterized by elevated ear thickening, mono/MΦ-dominated dermal inflammation, and increased iNOS and IL-6 expression compared with control mice. Treatment of Arg1flox/flox; LysMCre+/- mice with a selective NOS inhibitor or knockout of Nos2, encoding iNOS, significantly ameliorates CHS. Our findings suggest a critical role for Arg1 in mono/MΦ in suppressing CHS through dampening Nos2 expression. These results support that increasing Arg1 may be a potential therapeutic avenue in treating allergic contact dermatitis.
Collapse
Affiliation(s)
| | - Michael Shih
- Department of Dermatology, Duke University, Durham, NC 27710
| | - Lauren Pontius
- Department of Dermatology, Duke University, Durham, NC 27710
| | - Bin Yang
- Department of Dermatology, Duke University, Durham, NC 27710
| | | | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine, Mount Sinai Medical Center, New York, NY 10029
| | - David L Corcoran
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708; and
| | - Loretta G Que
- Department of Medicine, Duke University, Durham, NC 27710
| | - Robert M Tighe
- Department of Medicine, Duke University, Durham, NC 27710
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC 27710; .,Duke Immunology, Duke University, Durham, NC 27710
| |
Collapse
|
21
|
Fu SH, Yeh LT, Chu CC, Yen BLJ, Sytwu HK. New insights into Blimp-1 in T lymphocytes: a divergent regulator of cell destiny and effector function. J Biomed Sci 2017; 24:49. [PMID: 28732506 PMCID: PMC5520377 DOI: 10.1186/s12929-017-0354-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/12/2017] [Indexed: 12/14/2022] Open
Abstract
B lymphocyte-induced maturation protein-1 (Blimp-1) serves as a master regulator of the development and function of antibody-producing B cells. Given that its function in T lymphocytes has been identified within the past decade, we review recent findings with emphasis on its role in coordinated control of gene expression during the development, differentiation, and function of T cells. Expression of Blimp-1 is mainly confined to activated T cells and is essential for the production of interleukin (IL)-10 by a subset of forkhead box (Fox)p3+ regulatory T cells with an effector phenotype. Blimp-1 is also required to induce cell elimination in the thymus and critically modulates peripheral T cell activation and proliferation. In addition, Blimp-1 promotes T helper (Th) 2 lineage commitment and limits Th1, Th17 and follicular helper T cell differentiation. Furthermore, Blimp-1 coordinates with other transcription factors to regulate expression of IL-2, IL-21 and IL-10 in effector T lymphocytes. In CD8+ T cells, Blimp-1 expression is distinct in heterogeneous populations at the stages of clonal expansion, differentiation, contraction and memory formation when they encounter antigens. Moreover, Blimp-1 plays a fundamental role in coordinating cytokine receptor signaling networks and transcriptional programs to regulate diverse aspects of the formation and function of effector and memory CD8+ T cells and their exhaustion. Blimp-1 also functions as a gatekeeper of T cell activation and suppression to prevent or dampen autoimmune disease, antiviral responses and antitumor immunity. In this review, we discuss the emerging roles of Blimp-1 in the complex regulation of gene networks that regulate the destiny and effector function of T cells and provide a Blimp-1-dominated transcriptional framework for T lymphocyte homeostasis.
Collapse
Affiliation(s)
- Shin-Huei Fu
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, 161, Section 6, Min-Chuan East Road, Neihu District, Taipei, 11490, Taiwan
| | - Li-Tzu Yeh
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, 161, Section 6, Min-Chuan East Road, Neihu District, Taipei, 11490, Taiwan
| | - Chin-Chen Chu
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, 71104, Taiwan. .,Department of Recreation and Health-Care Management, Chia Nan University of Pharmacy and Science, Tainan, 71104, Taiwan.
| | - B Lin-Ju Yen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Huey-Kang Sytwu
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, 161, Section 6, Min-Chuan East Road, Neihu District, Taipei, 11490, Taiwan.
| |
Collapse
|
22
|
Gelfand EW, Joetham A, Wang M, Takeda K, Schedel M. Spectrum of T-lymphocyte activities regulating allergic lung inflammation. Immunol Rev 2017; 278:63-86. [PMID: 28658551 PMCID: PMC5501488 DOI: 10.1111/imr.12561] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite advances in the treatment of asthma, optimization of symptom control remains an unmet need in many patients. These patients, labeled severe asthma, are responsible for a substantial fraction of the disease burden. In these patients, research is needed to define the cellular and molecular pathways contributing to disease which in large part are refractory to corticosteroid treatment. The causes of steroid-resistant asthma are multifactorial and result from complex interactions of genetics, environmental factors, and innate and adaptive immunity. Adaptive immunity, addressed here, integrates the activities of distinct T-cell subsets and by definition is dynamic and responsive to an ever-changing environment and the influences of epigenetic modifications. These T-cell subsets exhibit different susceptibilities to the actions of corticosteroids and, in some, corticosteroids enhance their functional activation. Moreover, these subsets are not fixed in lineage differentiation but can undergo transcriptional reprogramming in a bidirectional manner between protective and pathogenic effector states. Together, these factors contribute to asthma heterogeneity between patients but also in the same patient at different stages of their disease. Only by carefully defining mechanistic pathways, delineating their sensitivity to corticosteroids, and determining the balance between regulatory and effector pathways will precision medicine become a reality with selective and effective application of targeted therapies.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Meiqin Wang
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| |
Collapse
|
23
|
Samji T, Khanna KM. Understanding memory CD8 + T cells. Immunol Lett 2017; 185:32-39. [PMID: 28274794 PMCID: PMC5508124 DOI: 10.1016/j.imlet.2017.02.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/09/2017] [Accepted: 02/20/2017] [Indexed: 12/28/2022]
Abstract
Memory CD8+ T cells were originally thought to exist as two populations (effector and central memory). In recent years, a third population called resident memory T cells has been discovered and further to this these populations are being divided into different subtypes. Understanding the function and developmental pathways of memory CD8+ T cells is key to developing effective therapies against cancer and infectious diseases. Here we have reviewed what is currently known about all three subsets of memory CD8+ T populations and as to how each population was originally discovered and the developmental pathways of each subpopulation. Each memory population appears to play a distinct role in adaptive immune responses but we are still a long way from understanding how the populations are generated and what roles they play in protection against invading pathogens and if they contribute to the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Tasleem Samji
- Department of Immunology, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Kamal M Khanna
- Department of Immunology, University of Connecticut Health, Farmington, CT 06030, United States of America; Department of Pediatrics, University of Connecticut Health, Farmington, CT 06030, United States of America.
| |
Collapse
|
24
|
Lourenço O, Fonseca AM, Taborda-Barata L. Human CD8+ T Cells in Asthma: Possible Pathways and Roles for NK-Like Subtypes. Front Immunol 2016; 7:638. [PMID: 28066445 PMCID: PMC5179570 DOI: 10.3389/fimmu.2016.00638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/12/2016] [Indexed: 12/22/2022] Open
Abstract
Asthma affects approximately 300 million people worldwide and is the most common chronic lung disease, which usually is associated with bronchial inflammation. Most research has focused upon the role of CD4+ T cells, and relatively few studies have addressed the phenotypic and functional roles of CD8+ T cell types and subtypes. Human NK-like CD8+ T cells may involve cells that have been described as CD8+CD28−, CD8+CD28−CD57+, CD8+CD27−, or CD8+ effector memory (TEM) cells, among other. However, most of the data that are available regarding these various cell types were obtained in murine models did not thoroughly characterize these cells with phenotypically or functionally or did not involve asthma-related settings. Nevertheless, one may conceptualize three principal roles for human NK-like CD8+ T cells in asthma: disease-promoting, regulatory, and/or tissue repair. Although evidence for some of these roles is scarce, it is possible to extrapolate some data from overlapping or related CD8+ T cell phenotypes, with caution. Clearly, further research is warranted, namely in terms of thorough functional and phenotypic characterization of human NK-like CD8+ T cells in human asthma of varying severity.
Collapse
Affiliation(s)
- Olga Lourenço
- CICS - UBI, Health Sciences Research Centre, University of Beira Interior , Covilhã , Portugal
| | - Ana Mafalda Fonseca
- CICS - UBI, Health Sciences Research Centre, University of Beira Interior , Covilhã , Portugal
| | - Luis Taborda-Barata
- CICS - UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; Department of Allergy and Clinical Immunology, Cova da Beira Hospital Centre, Covilhã, Portugal
| |
Collapse
|
25
|
Lan F, Zhang N, Gevaert E, Zhang L, Bachert C. Viruses and bacteria in Th2-biased allergic airway disease. Allergy 2016; 71:1381-92. [PMID: 27188632 DOI: 10.1111/all.12934] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2016] [Indexed: 01/24/2023]
Abstract
Allergic airway diseases are typically characterized by a type 2-biased inflammation. Multiple distinct viruses and bacteria have been detected in the airways. Recently, it has been confirmed that the microbiome of allergic individuals differs from that of healthy subjects, showing a close relationship with the type 2 response in allergic airway disease. In this review, we summarize the recent findings on the prevalence of viruses and bacteria in type 2-biased airway diseases and on the mechanisms employed by viruses and bacteria in propagating type 2 responses. The understanding of the microbial composition and postinfectious immune programming is critical for the reconstruction of the normal microflora and immune status in allergic airway diseases.
Collapse
Affiliation(s)
- F. Lan
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - N. Zhang
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - E. Gevaert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - L. Zhang
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - C. Bachert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Division of ENT Diseases; Clintec; Karolinska Institute; Stockholm Sweden
| |
Collapse
|
26
|
Abstract
Asthma is the most common chronic disease of childhood, affecting millions of children in the United States and worldwide. Prematurity is a risk factor for asthma, and certain ethnic or racial minorities such as Puerto Ricans and non-Hispanic blacks are disproportionately affected by both prematurity and asthma. In this review, we examine current evidence to support maternal psychosocial stress as a putative link between prematurity and asthma, while also focusing on disruption of the hypothalamic-pituitary-adrenal (HPA) axis and immune responses as potential underlying mechanisms for stress-induced "premature asthma." Prenatal stress may cause not only abnormalities in the HPA axis but also epigenetic changes in the fetal glucocorticoid receptor gene (NR3C1), leading to impaired glucocorticoid metabolism. Moreover, maternal stress can alter fetal cytokine balance, favoring TH2 (allergic) immune responses characteristic of atopic asthma: interleukin 6 (IL-6), which has been associated with premature labor, can promote TH2 responses by stimulating production of IL-4 and IL-13. Given a link among stress, prematurity, and asthma, future research should include birth cohorts aimed at confirming and better characterizing "premature asthma." If confirmed, clinical trials of prenatal maternal stress reduction would be warranted to reduce the burden of these common comorbidities. While awaiting the results of such studies, sound policies to prevent domestic and community violence (eg, from firearms) are justified, not only by public safety but also by growing evidence of detrimental effects of violence-induced stress on psychiatric and somatic health.
Collapse
|
27
|
May RD, Fung M. Strategies targeting the IL-4/IL-13 axes in disease. Cytokine 2016; 75:89-116. [PMID: 26255210 DOI: 10.1016/j.cyto.2015.05.018] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
Abstract
IL-4 and IL-13 are pleiotropic Th2 cytokines produced by a wide variety of different cell types and responsible for a broad range of biology and functions. Physiologically, Th2 cytokines are known to mediate host defense against parasites but they can also trigger disease if their activities are dysregulated. In this review we discuss the rationale for targeting the IL-4/IL-13 axes in asthma, atopic dermatitis, allergic rhinitis, COPD, cancer, inflammatory bowel disease, autoimmune disease and fibrotic disease as well as evaluating the associated clinical data derived from blocking IL-4, IL-13 or IL-4 and IL-13 together.
Collapse
|
28
|
den Otter I, Willems LNA, van Schadewijk A, van Wijngaarden S, Janssen K, de Jeu RC, Sont JK, Sterk PJ, Hiemstra PS. Lung function decline in asthma patients with elevated bronchial CD8, CD4 and CD3 cells. Eur Respir J 2016; 48:393-402. [PMID: 27230446 DOI: 10.1183/13993003.01525-2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 04/04/2016] [Indexed: 11/05/2022]
Abstract
Which inflammatory markers in the bronchial mucosa of asthma patients are associated with decline of lung function during 14 years of prospective follow-up?To address this question, 19 mild-to-moderate, atopic asthmatic patients underwent spirometry and bronchoscopy at baseline and after 14 years of follow-up (t=14). Baseline bronchial biopsies were analysed for reticular layer thickness, eosinophil cationic protein (EG2), mast cell tryptase (AA1), CD3, CD4 and CD8. Follow-up biopsies were stained for EG2, AA1, neutrophil elastase, CD3, CD4, CD8, CD20, granzyme B, CD68, DC-SIGN, Ki67 and mucins.Decline in forced expiratory volume in 1 s (FEV1) % predicted was highest in patients with high CD8 (p=0.01, both pre- and post-bronchodilator) or high CD4 counts at baseline (p=0.04 pre-bronchodilator, p=0.03 post-bronchodilator). Patients with high CD8, CD3 or granzyme B counts at t=14 also exhibited faster decline in FEV1 (p=0.00 CD8 pre-bronchodilator, p=0.04 CD8 post-bronchodilator, p=0.01 granzyme B pre-bronchodilator, and p<0.01 CD3 pre-bronchodilator).Long-term lung function decline in asthma is associated with elevation of bronchial CD8 and CD4 at baseline, and CD8, CD3 and granzyme B at follow-up. This suggests that high-risk groups can be identified on the basis of inflammatory phenotypes.
Collapse
Affiliation(s)
- Irene den Otter
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Luuk N A Willems
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Kirsten Janssen
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ronald C de Jeu
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacob K Sont
- Dept of Medical Decision Making, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter J Sterk
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands Dept of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
Expansion of CD8(+) T cells lacking the IL-6 receptor α chain in patients with coronary artery diseases (CAD). Atherosclerosis 2016; 249:44-51. [PMID: 27062409 DOI: 10.1016/j.atherosclerosis.2016.03.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 02/29/2016] [Accepted: 03/31/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND AIMS The pathogenesis of coronary artery disease (CAD) is closely associated with chronic inflammatory processes. CD8(+) T cells are a key participant in the pathogenesis of atherosclerosis, the major cause of CAD; however, it remains unclear which CD8(+) T-cell subset is responsible. We investigated the immunological features of CD8(+) T cells expressing low and high levels of the IL-6 receptor α chain (IL-6Rα), a cytokine known to play a key role in cardiovascular diseases. METHODS The expression of IL-6Rα on CD8(+) T cells and its association with plasma levels of soluble components of the IL-6/IL-6Rs as well as with clinical parameters were analyzed using FACS analysis and ELISA of CAD patients and age-matched healthy controls (HCs). Immunological characteristics of CD8(+) T cells expressing low and high levels of IL-6Rα (CD8(+)IL-6Rα(low or high)) were examined by in vitro culture and intracellular FACS analysis. RESULTS CAD patients had higher frequencies of circulating CD8(+)IL-6Rα(low) effector memory (EM) T cells compared with HCs (median frequency; 74.59% vs. 60.09%, p = 0.0158). Expanded CD8(+)IL-6Rα(low) T cells positively correlated with the frequency of senescent, cytotoxic CD8(+)CD57(+) T cells (r = 0.6655, p < 0.0001) and plasma IL-6 level (r = 0.3995, p = 0.0432) in CAD patients. Loss of IL-6Rα expression on CD8(+) T cells was induced by the combination of IL-6 and IL-15 with accompanying TCR-independent proliferation (p = 0.0101). Moreover, these CD8(+)IL-6Rα(low) T cells had features of type 1 cytotoxic CD8(+) T cells. CONCLUSIONS Our findings suggest the possible involvement of expanded CD8(+)IL-6Rα(low) EM T cells in CAD through their pro-inflammatory and highly cytotoxic capacities.
Collapse
|
30
|
Zissler UM, Esser-von Bieren J, Jakwerth CA, Chaker AM, Schmidt-Weber CB. Current and future biomarkers in allergic asthma. Allergy 2016; 71:475-94. [PMID: 26706728 DOI: 10.1111/all.12828] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 12/12/2022]
Abstract
Diagnosis early in life, sensitization, asthma endotypes, monitoring of disease and treatment progression are key motivations for the exploration of biomarkers for allergic rhinitis and allergic asthma. The number of genes related to allergic rhinitis and allergic asthma increases steadily; however, prognostic genes have not yet entered clinical application. We hypothesize that the combination of multiple genes may generate biomarkers with prognostic potential. The current review attempts to group more than 161 different potential biomarkers involved in respiratory inflammation to pave the way for future classifiers. The potential biomarkers are categorized into either epithelial or infiltrate-derived or mixed origin, epithelial biomarkers. Furthermore, surface markers were grouped into cell-type-specific categories. The current literature provides multiple biomarkers for potential asthma endotypes that are related to T-cell phenotypes such as Th1, Th2, Th9, Th17, Th22 and Tregs and their lead cytokines. Eosinophilic and neutrophilic asthma endotypes are also classified by epithelium-derived CCL-26 and osteopontin, respectively. There are currently about 20 epithelium-derived biomarkers exclusively derived from epithelium, which are likely to innovate biomarker panels as they are easy to sample. This article systematically reviews and categorizes genes and collects current evidence that may promote these biomarkers to become part of allergic rhinitis or allergic asthma classifiers with high prognostic value.
Collapse
Affiliation(s)
- U. M. Zissler
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| | - J. Esser-von Bieren
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| | - C. A. Jakwerth
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| | - A. M. Chaker
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health 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
| | - C. B. Schmidt-Weber
- Center of Allergy & Environment (ZAUM); Technical University of Munich and Helmholtz Center Munich; German Research Center for Environmental Health member of the German Center for Lung Research (DZL); Munich Germany
| |
Collapse
|
31
|
Affiliation(s)
- Tara F Carr
- Department of Internal Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Monica Kraft
- Department of Internal Medicine, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
32
|
Shin MS, You S, Kang Y, Lee N, Yoo SA, Park K, Kang KS, Kim SH, Mohanty S, Shaw AC, Montgomery RR, Hwang D, Kang I. DNA Methylation Regulates the Differential Expression of CX3CR1 on Human IL-7Rαlow and IL-7Rαhigh Effector Memory CD8+ T Cells with Distinct Migratory Capacities to the Fractalkine. THE JOURNAL OF IMMUNOLOGY 2015; 195:2861-9. [PMID: 26276874 DOI: 10.4049/jimmunol.1500877] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/22/2015] [Indexed: 11/19/2022]
Abstract
DNA methylation is an epigenetic mechanism that modulates gene expression in mammalian cells including T cells. Memory T cells are heterogeneous populations. Human effector memory (EM) CD8(+) T cells in peripheral blood contain two cell subsets with distinct traits that express low and high levels of the IL-7Rα. However, epigenetic mechanisms involved in defining such cellular traits are largely unknown. In this study, we use genome-wide DNA methylation and individual gene expression to show the possible role of DNA methylation in conferring distinct traits of chemotaxis and inflammatory responses in human IL-7Rα(low) and IL-7Rα(high) EM CD8(+) T cells. In particular, IL-7Rα(low) EM CD8(+) T cells had increased expression of CX3CR1 along with decreased DNA methylation in the CX3CR1 gene promoter compared with IL-7Rα(high) EM CD8(+) T cells. Altering the DNA methylation status of the CX3CR1 gene promoter changed its activity and gene expression. IL-7Rα(low) EM CD8(+) T cells had an increased migratory capacity to the CX3CR1 ligand fractalkine compared with IL-7Rα(high) EM CD8(+) T cells, suggesting an important biological outcome of the differential expression of CX3CR1. Moreover, IL-7Rα(low) EM CD8(+) T cells induced fractalkine expression on endothelial cells by producing IFN-γ and TNF-α, forming an autocrine amplification loop. Overall, our study shows the role of DNA methylation in generating unique cellular traits in human IL-7Rα(low) and IL-7Rα(high) EM CD8(+) T cells, including differential expression of CX3CR1, as well as potential biological implications of this differential expression.
Collapse
Affiliation(s)
- Min Sun Shin
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; Division of Cancer Biology and Therapeutics, Department of Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; Division of Cancer Biology and Therapeutics, Department of Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Youna Kang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Naeun Lee
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Seung-Ah Yoo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Kieyoung Park
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520; Department of Pediatrics, College of Medicine, Ulsan University, Ulsan 680-749, Republic of Korea
| | - Ki Soo Kang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520; Department of Pediatrics, Jeju National University School of Medicine, Jeju 690-756, Republic of Korea
| | - Sang Hyun Kim
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520; Department of Microbiology, College of Medicine, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Subhasis Mohanty
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Albert C Shaw
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang 790-784, Republic of Korea; and Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 711-873, Republic of Korea
| | - Insoo Kang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520;
| |
Collapse
|