9 results on '"T. Ashhurst"'
Search Results
2. Methotrexate treatment hampers induction of vaccine-specific CD4 T cell responses in patients with IMID.
- Author
-
Kummer LYL, Fernández Blanco L, Kreher C, Bos A, Kuijper LH, Verstegen NJM, van de Sandt CE, Konijn VAL, Duurland MC, Menage C, Jorritsma T, Steenhuis M, Hagen RR, van den Dijssel J, de Jongh R, Ashhurst T, van Gils MJ, Garcia-Vallejo JJ, Claireaux M, Stalman EW, van Dam KPJ, Wieske L, Boekel L, Wolbink G, Tas SW, Rispens T, Kuijpers TW, Eftimov F, van Ham SM, and Ten Brinke A
- Subjects
- Humans, Male, Middle Aged, Female, Aged, Adult, Antirheumatic Agents therapeutic use, Vaccination, Methotrexate therapeutic use, CD4-Positive T-Lymphocytes immunology, COVID-19 immunology, COVID-19 prevention & control, SARS-CoV-2 immunology, COVID-19 Vaccines immunology, COVID-19 Vaccines administration & dosage, Arthritis, Rheumatoid drug therapy, Arthritis, Rheumatoid immunology, B-Lymphocytes immunology, B-Lymphocytes drug effects
- Abstract
Objectives: Methotrexate (MTX) is one of the most commonly used medications to treat rheumatoid arthritis (RA). However, the effect of MTX treatment on cellular immune responses remains incompletely understood. This raises concerns about the vulnerability of these patients to emerging infections and following vaccination., Methods: In the current study, we investigated the impact of MTX treatment in patients with immune-mediated inflammatory disease on B and CD4 T cell SARS-CoV-2 vaccination responses. Eighteen patients with RA and two patients with psoriatic arthritis on MTX monotherapy were included, as well as 10 patients with RA without immunosuppressive treatment, and 29 healthy controls. CD4 T and B cell responses were analysed 7 days and 3-6 months after two SARS-CoV-2 messenger RNA vaccinations. High-dimensional flow cytometry analysis was used to analyse fresh whole blood, an activation-induced marker assay to measure antigen-specific CD4 T cells, and spike probes to study antigen-specific B cells., Results: Seven days following two SARS-CoV-2 vaccinations, total B and T cell counts were similar between MTX-treated patients and controls. In addition, spike-specific B cell frequencies were unaffected. Remarkably, the frequency of antigen-specific CD4 T cells was reduced in patients using MTX and correlated strongly with anti-RBD IgG antibodies. These results suggest that decreased CD4 T cell activity may result in slower vaccination antibody responses in MTX-treated patients., Conclusion: Taken together, MTX treatment reduces vaccine-induced CD4 T cell activation, which correlates with lower antibody responses., Trial Registration Number: NL8900., Competing Interests: Competing interests: FE, GW, SMvH and TWK report (governmental) grants from ZonMw to study immune response after SARS-Cov-2 vaccination in auto-immune diseases. FE also reports grants from Prinses Beatrix Spierfonds, CSL Behring, Kedrion, Terumo BCT, Grifols, Takeda Pharmaceutical and GBS-CIDP Foundation; consulting fees from UCB Pharma and CSl Behring; honoraria from Grifols. All other authors report no disclosures relevant to the manuscript., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
3. T cell activation markers CD38 and HLA-DR indicative of non-seroconversion in anti-CD20-treated patients with multiple sclerosis following SARS-CoV-2 mRNA vaccination.
- Author
-
Verstegen NJM, Hagen RR, Kreher C, Kuijper LH, Dijssel JVD, Ashhurst T, Kummer LYL, Palomares Cabeza V, Steenhuis M, Duurland MC, Jongh R, Schoot CEV, Konijn VAL, Mul E, Kedzierska K, van Dam KPJ, Stalman EW, Boekel L, Wolbink G, Tas SW, Killestein J, Rispens T, Wieske L, Kuijpers TW, Eftimov F, van Kempen ZLE, van Ham SM, Ten Brinke A, and van de Sandt CE
- Subjects
- Humans, Female, Male, Adult, Middle Aged, SARS-CoV-2 immunology, Lymphocyte Activation, Antibodies, Viral blood, mRNA Vaccines therapeutic use, Antigens, CD20 immunology, Vaccination, CD4-Positive T-Lymphocytes immunology, Membrane Glycoproteins, ADP-ribosyl Cyclase 1 immunology, Multiple Sclerosis immunology, Multiple Sclerosis drug therapy, COVID-19 Vaccines therapeutic use, COVID-19 Vaccines immunology, HLA-DR Antigens immunology, COVID-19 prevention & control, COVID-19 immunology, Antibodies, Monoclonal, Humanized therapeutic use
- Abstract
Background: Messenger RNA (mRNA) vaccines provide robust protection against SARS-CoV-2 in healthy individuals. However, immunity after vaccination of patients with multiple sclerosis (MS) treated with ocrelizumab (OCR), a B cell-depleting anti-CD20 monoclonal antibody, is not yet fully understood., Methods: In this study, deep immune profiling techniques were employed to investigate the immune response induced by SARS-CoV-2 mRNA vaccines in untreated patients with MS (n=21), OCR-treated patients with MS (n=57) and healthy individuals (n=30)., Results: Among OCR-treated patients with MS, 63% did not produce detectable levels of antibodies (non-seroconverted), and those who did have lower spike receptor-binding domain-specific IgG responses compared with healthy individuals and untreated patients with MS. Before vaccination, no discernible immunological differences were observed between non-seroconverted and seroconverted OCR-treated patients with MS. However, non-seroconverted patients received overall more OCR infusions, had shorter intervals since their last OCR infusion and displayed higher OCR serum concentrations at the time of their initial vaccination. Following two vaccinations, non-seroconverted patients displayed smaller B cell compartments but instead exhibited more robust activation of general CD4
+ and CD8+ T cell compartments, as indicated by upregulation of CD38 and HLA-DR surface expression, when compared with seroconverted patients., Conclusion: These findings highlight the importance of optimising treatment regimens when scheduling SARS-CoV-2 vaccination for OCR-treated patients with MS to maximise their humoral and cellular immune responses. This study provides valuable insights for optimising vaccination strategies in OCR-treated patients with MS, including the identification of CD38 and HLA-DR as potential markers to explore vaccine efficacy in non-seroconverting OCR-treated patients with MS., Competing Interests: Competing interests: JK received research grants for multicentre investigator-initiated trials (DOT-MS trial, ClinicalTrials.gov Identifier: NCT04260711 (ZonMw) and BLOOMS trial (ZonMw and Treatmeds), ClinicalTrials.gov Identifier: NCT05296161); received consulting fees from F Hoffmann-La Roche, Biogen, Teva, Merck, Novartis and Sanofi/Genzyme (all payments to institution); reports speaker relationships with F Hoffmann-La Roche, Biogen, Immunic, Teva, Merck, Novartis and Sanofi/Genzyme (all payments to institution); and adjudication committee of MS clinical trial of Immunic (payments to institution only)., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
4. Faecal microbial transfer and complex carbohydrates mediate protection against COPD.
- Author
-
Budden KF, Shukla SD, Bowerman KL, Vaughan A, Gellatly SL, Wood DLA, Lachner N, Idrees S, Rehman SF, Faiz A, Patel VK, Donovan C, Alemao CA, Shen S, Amorim N, Majumder R, Vanka KS, Mason J, Haw TJ, Tillet B, Fricker M, Keely S, Hansbro N, Belz GT, Horvat J, Ashhurst T, van Vreden C, McGuire H, Fazekas de St Groth B, King NJC, Crossett B, Cordwell SJ, Bonaguro L, Schultze JL, Hamilton-Williams EE, Mann E, Forster SC, Cooper MA, Segal LN, Chotirmall SH, Collins P, Bowman R, Fong KM, Yang IA, Wark PAB, Dennis PG, Hugenholtz P, and Hansbro PM
- Subjects
- Humans, Mice, Animals, Lung metabolism, Lung pathology, Inflammation metabolism, Carbohydrates pharmacology, Pulmonary Disease, Chronic Obstructive etiology, Pneumonia etiology
- Abstract
Objective: Chronic obstructive pulmonary disease (COPD) is a major cause of global illness and death, most commonly caused by cigarette smoke. The mechanisms of pathogenesis remain poorly understood, limiting the development of effective therapies. The gastrointestinal microbiome has been implicated in chronic lung diseases via the gut-lung axis, but its role is unclear., Design: Using an in vivo mouse model of cigarette smoke (CS)-induced COPD and faecal microbial transfer (FMT), we characterised the faecal microbiota using metagenomics, proteomics and metabolomics. Findings were correlated with airway and systemic inflammation, lung and gut histopathology and lung function. Complex carbohydrates were assessed in mice using a high resistant starch diet, and in 16 patients with COPD using a randomised, double-blind, placebo-controlled pilot study of inulin supplementation., Results: FMT alleviated hallmark features of COPD (inflammation, alveolar destruction, impaired lung function), gastrointestinal pathology and systemic immune changes. Protective effects were additive to smoking cessation, and transfer of CS-associated microbiota after antibiotic-induced microbiome depletion was sufficient to increase lung inflammation while suppressing colonic immunity in the absence of CS exposure. Disease features correlated with the relative abundance of Muribaculaceae, Desulfovibrionaceae and Lachnospiraceae family members. Proteomics and metabolomics identified downregulation of glucose and starch metabolism in CS-associated microbiota, and supplementation of mice or human patients with complex carbohydrates improved disease outcomes., Conclusion: The gut microbiome contributes to COPD pathogenesis and can be targeted therapeutically., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
5. CD4 + T cell calibration of antigen-presenting cells optimizes antiviral CD8 + T cell immunity.
- Author
-
Gressier E, Schulte-Schrepping J, Petrov L, Brumhard S, Stubbemann P, Hiller A, Obermayer B, Spitzer J, Kostevc T, Whitney PG, Bachem A, Odainic A, van de Sandt C, Nguyen THO, Ashhurst T, Wilson K, Oates CVL, Gearing LJ, Meischel T, Hochheiser K, Greyer M, Clarke M, Kreutzenbeck M, Gabriel SS, Kastenmüller W, Kurts C, Londrigan SL, Kallies A, Kedzierska K, Hertzog PJ, Latz E, Chen YE, Radford KJ, Chopin M, Schroeder J, Kurth F, Gebhardt T, Sander LE, Sawitzki B, Schultze JL, Schmidt SV, and Bedoui S
- Subjects
- Humans, Calibration, Antigen-Presenting Cells, CD8-Positive T-Lymphocytes, CD40 Antigens, Interferon-alpha, CD4-Positive T-Lymphocytes, Antiviral Agents, COVID-19
- Abstract
Antiviral CD8
+ T cell immunity depends on the integration of various contextual cues, but how antigen-presenting cells (APCs) consolidate these signals for decoding by T cells remains unclear. Here, we describe gradual interferon-α/interferon-β (IFNα/β)-induced transcriptional adaptations that endow APCs with the capacity to rapidly activate the transcriptional regulators p65, IRF1 and FOS after CD4+ T cell-mediated CD40 stimulation. While these responses operate through broadly used signaling components, they induce a unique set of co-stimulatory molecules and soluble mediators that cannot be elicited by IFNα/β or CD40 alone. These responses are critical for the acquisition of antiviral CD8+ T cell effector function, and their activity in APCs from individuals infected with severe acute respiratory syndrome coronavirus 2 correlates with milder disease. These observations uncover a sequential integration process whereby APCs rely on CD4+ T cells to select the innate circuits that guide antiviral CD8+ T cell responses., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2023
- Full Text
- View/download PDF
6. Immune dynamics in SARS-CoV-2 experienced immunosuppressed rheumatoid arthritis or multiple sclerosis patients vaccinated with mRNA-1273.
- Author
-
Verstegen NJM, Hagen RR, van den Dijssel J, Kuijper LH, Kreher C, Ashhurst T, Kummer LYL, Steenhuis M, Duurland M, de Jongh R, de Jong N, van der Schoot CE, Bos AV, Mul E, Kedzierska K, van Dam KPJ, Stalman EW, Boekel L, Wolbink G, Tas SW, Killestein J, van Kempen ZLE, Wieske L, Kuijpers TW, Eftimov F, Rispens T, van Ham SM, Ten Brinke A, and van de Sandt CE
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Antibodies, Viral, CD8-Positive T-Lymphocytes, Humans, Immunosuppressive Agents therapeutic use, SARS-CoV-2, Vaccination, Arthritis, Rheumatoid drug therapy, COVID-19 prevention & control, Multiple Sclerosis drug therapy, Viral Vaccines genetics
- Abstract
Background: Patients affected by different types of autoimmune diseases, including common conditions such as multiple sclerosis (MS) and rheumatoid arthritis (RA), are often treated with immunosuppressants to suppress disease activity. It is not fully understood how the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific humoral and cellular immunity induced by infection and/or upon vaccination is affected by immunosuppressants., Methods: The dynamics of cellular immune reactivation upon vaccination of SARS-CoV-2 experienced MS patients treated with the humanized anti-CD20 monoclonal antibody ocrelizumab (OCR) and RA patients treated with methotrexate (MTX) monotherapy were analyzed at great depth via high-dimensional flow cytometry of whole blood samples upon vaccination with the SARS-CoV-2 mRNA-1273 (Moderna) vaccine. Longitudinal B and T cell immune responses were compared to SARS-CoV-2 experienced healthy controls (HCs) before and 7 days after the first and second vaccination., Results: OCR-treated MS patients exhibit a preserved recall response of CD8
+ T central memory cells following first vaccination compared to HCs and a similar CD4+ circulating T follicular helper 1 and T helper 1 dynamics, whereas humoral and B cell responses were strongly impaired resulting in absence of SARS-CoV-2-specific humoral immunity. MTX treatment significantly delayed antibody levels and B reactivation following the first vaccination, including sustained inhibition of overall reactivation marker dynamics of the responding CD4+ and CD8+ T cells., Conclusions: Together, these findings indicate that SARS-CoV-2 experienced MS-OCR patients may still benefit from vaccination by inducing a broad CD8+ T cell response which has been associated with milder disease outcome. The delayed vaccine-induced IgG kinetics in RA-MTX patients indicate an increased risk after the first vaccination, which might require additional shielding or alternative strategies such as treatment interruptions in vulnerable patients., Funding: This research project was supported by ZonMw (The Netherlands Organization for Health Research and Development, #10430072010007), the European Union's Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement (#792532 and #860003), the European Commission (SUPPORT-E, #101015756) and by PPOC (#20_21 L2506), the NHMRC Leadership Investigator Grant (#1173871)., Competing Interests: NV, RH, Jv, LK, CK, TA, LK, MS, MD, Rd, Nd, Cv, AB, EM, KK, Kv, ES, LB, GW, ST, Zv, LW, TK, FE, TR, Sv, At, Cv No competing interests declared, JK has speaking relationships with Merck, Biogen, TEVA, Sanofi, Genzyme, Roche and Novartis. AmsterdamUMC, location VUmc, MS Center Amsterdam has received financial support for research activities from Merck, Celgene, Biogen, GlaxoSmithKline, Immunic, Roche, Teva, Sanofi, Genzyme, and Novartis, (© 2022, Verstegen et al.)- Published
- 2022
- Full Text
- View/download PDF
7. Immune responses in COVID-19 respiratory tract and blood reveal mechanisms of disease severity.
- Author
-
Zhang W, Chua B, Selva K, Kedzierski L, Ashhurst T, Haycroft E, Shoffner S, Hensen L, Boyd D, James F, Mouhtouris E, Kwong J, Chua K, Drewett G, Copaescu A, Dobson J, Rowntree L, Habel J, Allen L, Koay HF, Neil J, Gartner M, Lee C, Andersson P, Seemann T, Sherry N, Amanat F, Krammer F, Londrigan S, Wakim L, King N, Godfrey D, Mackay L, Thomas P, Nicholson S, Arnold K, Chung A, Holmes N, Smibert O, Trubiano J, Gordon C, Nguyen T, and Kedzierska K
- Abstract
Although the respiratory tract is the primary site of SARS-CoV-2 infection and the ensuing immunopathology, respiratory immune responses are understudied and urgently needed to understand mechanisms underlying COVID-19 disease pathogenesis. We collected paired longitudinal blood and respiratory tract samples (endotracheal aspirate, sputum or pleural fluid) from hospitalized COVID-19 patients and non-COVID-19 controls. Cellular, humoral and cytokine responses were analysed and correlated with clinical data. SARS-CoV-2-specific IgM, IgG and IgA antibodies were detected using ELISA and multiplex assay in both the respiratory tract and blood of COVID-19 patients, although a higher receptor binding domain (RBD)-specific IgM and IgG seroconversion level was found in respiratory specimens. SARS-CoV-2 neutralization activity in respiratory samples was detected only when high levels of RBD-specific antibodies were present. Strikingly, cytokine/chemokine levels and profiles greatly differed between respiratory samples and plasma, indicating that inflammation needs to be assessed in respiratory specimens for the accurate assessment of SARS-CoV-2 immunopathology. Diverse immune cell subsets were detected in respiratory samples, albeit dominated by neutrophils. Importantly, we also showed that dexamethasone and/or remdesivir treatment did not affect humoral responses in blood of COVID-19 patients. Overall, our study unveils stark differences in innate and adaptive immune responses between respiratory samples and blood and provides important insights into effect of drug therapy on immune responses in COVID-19 patients.
- Published
- 2021
- Full Text
- View/download PDF
8. IgG 3 + B cells are associated with the development of multiple sclerosis.
- Author
-
Marsh-Wakefield F, Ashhurst T, Trend S, McGuire HM, Juillard P, Zinger A, Jones AP, Kermode AG, Hawke S, Grau GE, Hart PH, and Byrne SN
- Abstract
Objectives: Disease-modifying therapies (DMTs) targeting B cells are amongst the most effective for preventing multiple sclerosis (MS) progression. IgG
3 antibodies and their uncharacterised B-cell clones are predicted to play a pathogenic role in MS. Identifying subsets of IgG3 + B cells involved in MS progression could improve diagnosis, could inform timely disease intervention and may lead to new DMTs that target B cells more specifically., Methods: We designed a 31-parameter B-cell-focused mass cytometry panel to interrogate the role of peripheral blood IgG3 + B cells in MS progression of two different patient cohorts: one to investigate the B-cell subsets involved in conversion from clinically isolated syndrome (CIS) to MS; and another to compare MS patients with inactive or active stages of disease. Each independent cohort included a group of non-MS controls., Results: Nine distinct CD20+ IgD- IgG3 + B-cell subsets were identified. Significant changes in the proportion of CD21+ CD24+ CD27- CD38- and CD27+ CD38hi CD71hi memory B-cell subsets correlated with changes in serum IgG3 levels and time to conversion from CIS to MS. The same CD38- double-negative B-cell subset was significantly elevated in MS patients with active forms of the disease. A third CD21+ CD24+ CD27+ CD38- subset was elevated in patients with active MS, whilst narrowband UVB significantly reduced the proportion of this switched-memory B-cell subset., Conclusion: We have identified previously uncharacterised subsets of IgG3 + B cells and shown them to correlate with autoimmune attacks on the central nervous system (CNS). These results highlight the potential for therapies that specifically target IgG3 + B cells to impact MS progression., Competing Interests: The authors declare no conflict of interest., (© 2020 The Authors. Clinical & Translational Immunology published by John Wiley & Sons Australia, Ltd on behalf of Australian and New Zealand Society for Immunology, Inc.)- Published
- 2020
- Full Text
- View/download PDF
9. IRF9 Prevents CD8 + T Cell Exhaustion in an Extrinsic Manner during Acute Lymphocytic Choriomeningitis Virus Infection.
- Author
-
Huber M, Suprunenko T, Ashhurst T, Marbach F, Raifer H, Wolff S, Strecker T, Viengkhou B, Jung SR, Obermann HL, Bauer S, Xu HC, Lang PA, Tom A, Lang KS, King NJC, Campbell IL, and Hofer MJ
- Subjects
- Acute Disease, Animals, CD8-Positive T-Lymphocytes pathology, Chronic Disease, Interferon Regulatory Factor-7, Interferon Type I genetics, Interferon Type I immunology, Interferon-Stimulated Gene Factor 3, gamma Subunit genetics, Lymphocytic Choriomeningitis genetics, Lymphocytic Choriomeningitis pathology, Lymphocytic choriomeningitis virus genetics, Mice, Mice, Knockout, Receptor, Interferon alpha-beta genetics, Receptor, Interferon alpha-beta immunology, Signal Transduction genetics, CD8-Positive T-Lymphocytes immunology, Interferon-Stimulated Gene Factor 3, gamma Subunit immunology, Lymphocytic Choriomeningitis immunology, Lymphocytic choriomeningitis virus immunology, Signal Transduction immunology
- Abstract
Effective CD8
+ T cell responses play an important role in determining the course of a viral infection. Overwhelming antigen exposure can result in suboptimal CD8+ T cell responses, leading to chronic infection. This altered CD8+ T cell differentiation state, termed exhaustion, is characterized by reduced effector function, upregulation of inhibitory receptors, and altered expression of transcription factors. Prevention of overwhelming antigen exposure to limit CD8+ T cell exhaustion is of significant interest for the control of chronic infection. The transcription factor interferon regulatory factor 9 (IRF9) is a component of type I interferon (IFN-I) signaling downstream of the IFN-I receptor (IFNAR). Using acute infection of mice with lymphocytic choriomeningitis virus (LCMV) strain Armstrong, we show here that IRF9 limited early LCMV replication by regulating expression of interferon-stimulated genes and IFN-I and by controlling levels of IRF7, a transcription factor essential for IFN-I production. Infection of IRF9- or IFNAR-deficient mice led to a loss of early restriction of viral replication and impaired antiviral responses in dendritic cells, resulting in CD8+ T cell exhaustion and chronic infection. Differences in the antiviral activities of IRF9- and IFNAR-deficient mice and dendritic cells provided further evidence of IRF9-independent IFN-I signaling. Thus, our findings illustrate a CD8+ T cell-extrinsic function for IRF9, as a signaling factor downstream of IFNAR, in preventing overwhelming antigen exposure resulting in CD8+ T cell exhaustion and, ultimately, chronic infection. IMPORTANCE During early viral infection, overwhelming antigen exposure can cause functional exhaustion of CD8+ T cells and lead to chronic infection. Here we show that the transcription factor interferon regulatory factor 9 (IRF9) plays a decisive role in preventing CD8+ T cell exhaustion. Using acute infection of mice with LCMV strain Armstrong, we found that IRF9 limited early LCMV replication by regulating expression of interferon-stimulated genes and Irf7 , encoding a transcription factor crucial for type I interferon (IFN-I) production, as well as by controlling the levels of IFN-I. Infection of IRF9-deficient mice led to a chronic infection that was accompanied by CD8+ T cell exhaustion due to defects extrinsic to T cells. Our findings illustrate an essential role for IRF9, as a mediator downstream of IFNAR, in preventing overwhelming antigen exposure causing CD8+ T cell exhaustion and leading to chronic viral infection., (Copyright © 2017 American Society for Microbiology.)- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.