44 results on '"Göschl L"'
Search Results
2. AB0732 CELLULAR AND MOLECULAR HALLMARKS OF EARLY METHOTREXATE RESPONSE IN RA
- Author
-
Tosevska, A., primary, Preglej, T., additional, Brinkmann, M., additional, Schatzlmaier, P., additional, Simader, E., additional, Sieghart, D., additional, Aletaha, D., additional, Hofer, P., additional, Göschl, L., additional, and Bonelli, M., additional
- Published
- 2024
- Full Text
- View/download PDF
3. AB0056 CYTOKINE-DIRECTED CELLULAR CROSSTALK IMPRINTS SYNOVIAL PATHOTYPES IN RHEUMATOID ARTHRITIS
- Author
-
Kugler, M., primary, Dellinger, M., additional, Kartnig, F., additional, Müller, L., additional, Preglej, T., additional, Heinz, L., additional, Simader, E., additional, Göschl, L., additional, Puchner, S., additional, Weiss, S., additional, Superi-Furga, G., additional, Smolen, J. S., additional, Steiner, G., additional, Aletaha, D., additional, Kiener, H., additional, Tosevska, A., additional, Karonitsch, T. M., additional, and Bonelli, M., additional
- Published
- 2023
- Full Text
- View/download PDF
4. POS0559 ACCELERATED WANING OF HUMORAL IMMUNE RESPONSE TO A THIRD COVID-19 VACCINATION IN PATIENTS WITH IMMUNE-MEDIATED INFLAMMATORY DISEASES
- Author
-
Mrak, D., primary, Kartnig, F., additional, Sieghart, D., additional, Simader, E., additional, Radner, H., additional, Mandl, P., additional, Göschl, L., additional, Hofer, P., additional, Deimel, T., additional, Gessl, I., additional, Kain, R., additional, Winkler, S., additional, Smolen, J. S., additional, Perkmann, T., additional, Haslacher, H., additional, Aletaha, D., additional, Heinz, L., additional, and Bonelli, M., additional
- Published
- 2023
- Full Text
- View/download PDF
5. Abatacept (CTLA-4IG) treatment reduces the migratory capacity of monocytes in patients with rheumatoid arthritis
- Author
-
Bonelli, M., Ferner, E., Göschl, L., Blüml, S., Hladik, A., Karonitsch, T., Kiener, H. P., Byrne, R., Niederreiter, B., Steiner, C. W., Rath, E., Bergmann, M., Smolen, J. S., and Scheinecker, C.
- Published
- 2013
- Full Text
- View/download PDF
6. Histone deacetylases HDAC1 and HDAC2 control Cd8 silencing in CD4 lineage T cells: W17.007
- Author
-
Tschismarov, R., Boucheron, N., Lagger, S., Moser, M., Göschl, L., Taniuchi, I., Matthias, P., Seiser, C., and Ellmeier, W.
- Published
- 2012
7. Abatacept (CTLA-4Ig) therapy prevents activation induced cell death (AICD) of regulatory T cells and reduces the susceptibility of T cells to regulatory T cell suppression in patients with rheumatoid arthritis (RA)
- Author
-
Bonelli, M, Göschl, L, Blüml, S, Smolen, J S, and Scheinecker, C
- Published
- 2012
- Full Text
- View/download PDF
8. THU0084 Histone deacetylase 1 (HDAC1): a novel therapeutic target in patients with rheumatoid arthritis
- Author
-
Göschl, L., primary, Mueller, L., additional, Saferding, V., additional, Knapp, S., additional, Backlund, J., additional, Scheinecker, C., additional, Smolen, J., additional, Ellmeier, W., additional, Steiner, G., additional, and Bonelli, M., additional
- Published
- 2018
- Full Text
- View/download PDF
9. P116 Histone deacetylase 1: a novel therapeutic target for patients with rheumatoid arthritis
- Author
-
Göschl, L, primary, Müller, L, additional, Saferding, V, additional, Bäcklund, J, additional, Knapp, S, additional, Mathias, P, additional, Scheinecker, C, additional, Ellmeier, W, additional, Steiner, G, additional, and Bonelli, M, additional
- Published
- 2018
- Full Text
- View/download PDF
10. FRI0055 Histone deacetylase 1 (HDAC1): a novel therapeutic target in rheumatoid arthritis
- Author
-
Göschl, L, primary, Bonelli, M, additional, Saferding, V, additional, Preglej, T, additional, Seiser, C, additional, Knapp, S, additional, Backlund, J, additional, Bock, C, additional, Mathias, P, additional, Hirahara, K, additional, Scheinecker, C, additional, Steiner, G, additional, and Ellmeier, W, additional
- Published
- 2017
- Full Text
- View/download PDF
11. CYTOKINE-DIRECTED CELLULAR CROSSTALK IMPRINTS SYNOVIAL PATHOTYPES IN RHEUMATOID ARTHRITIS.
- Author
-
Kugler, M., Dellinger, M., Kartnig, F., Müller, L., Preglej, T., Heinz, L., Simader, E., Göschl, L., Puchner, S., Weiss, S., Superi-Furga, G., Smolen, J. S., Steiner, G., Aletaha, D., Kiener, H., Tosevska, A., Karonitsch, T. M., and Bonelli, M.
- Published
- 2023
- Full Text
- View/download PDF
12. ACCELERATED WANING OF HUMORAL IMMUNE RESPONSE TO A THIRD COVID-19 VACCINATION IN PATIENTS WITH IMMUNE-MEDIATED INFLAMMATORY DISEASES.
- Author
-
Mrak, D., Kartnig, F., Sieghart, D., Simader, E., Radner, H., Mandl, P., Göschl, L., Hofer, P., Deimel, T., Gessl, I., Kain, R., Winkler, S., Smolen, J. S., Perkmann, T., Haslacher, H., Aletaha, D., Heinz, L., and Bonelli, M.
- Published
- 2023
- Full Text
- View/download PDF
13. Response to letter regarding article, humoral and cellular response to the third COVID-19 vaccination in patients with inborn errors of immunity (IEI) or mannan-binding lectin (MBL) deficiency-A prospective controlled open-label trial: correspondence.
- Author
-
Vossen MG and Göschl L
- Abstract
Competing Interests: Conflict of interest: M.G. Vossen declares a grant from Pfizer in a not connected project. L. Göschl declares that she has no competing interests.
- Published
- 2025
- Full Text
- View/download PDF
14. Humoral and cellular response to the third COVID-19 vaccination in patients with inborn errors of immunity or mannose-binding lectin deficiency : A prospective controlled open-label trial.
- Author
-
Vossen MG, Kartnig F, Mrak D, Simader E, Stiasny K, Kain R, Perkmann T, Haslacher H, Aberle JH, Heinz LX, Sieghart D, Burgmann H, Aletaha D, Scheinecker C, Bonelli M, and Göschl L
- Subjects
- Humans, Male, Female, Prospective Studies, Adult, SARS-CoV-2 immunology, Middle Aged, Antibodies, Viral blood, Immunologic Deficiency Syndromes immunology, Mannose-Binding Lectin immunology, Mannose-Binding Lectin deficiency, Primary Immunodeficiency Diseases immunology, Immunization, Secondary, Metabolism, Inborn Errors, COVID-19 Vaccines immunology, COVID-19 Vaccines administration & dosage, COVID-19 prevention & control, COVID-19 immunology, Immunity, Humoral immunology, Immunity, Cellular immunology
- Abstract
Impaired immune response to COVID-19 (coronavirus disease 2019) vaccination has been reported in patients with inborn errors of immunity (IEI). Repetitive vaccinations are recommended for this vulnerable group. Due to the high diversity within IEI patients, additional safety and immunogenicity data are needed to better understand these aspects especially in less common immunodeficiency syndromes. In this prospective open-label clinical trial, we assessed the humoral immune response and the T‑cell response in patients with IEI or severe MBL (mannose-binding lectin) deficiency (IEI/MBLdef) after three vaccinations. A total of 16 patients and 16 matched healthy controls (HC) with suboptimal humoral response defined by anti-SARS-CoV‑2 RBD (severe acute respiratory syndrome coronavirus type 2 receptor binding domain) antibodies below 1500 BAU/ml (binding antibody units per ml) after the second COVID-19 vaccination were enrolled in this study and qualified for a third mRNA vaccine dose. After 4 weeks following vaccination, 100% of HC and 75% of IEI/MBLdef patients exhibited anti-SARS-CoV‑2 RBD antibodies > 1500 BAU/ml, although the difference was not statistically significant (75% vs. 100%; p = 0.109). Although post-vaccination IEI/MBLdef patients demonstrated significantly increased anti-SARS-CoV‑2 RBD antibodies and neutralizing antibodies compared to baseline, these responses were significantly lower in IEI/MBLdef patients compared to HCs. Notably, the third vaccination augmented the cellular immune response to both wild-type and omicron peptide stimulation. No serious adverse events were reported within the 4‑week follow-up period and, importantly, vaccination had little to no effect on the long-term disease activity and fatigue. This trial strongly supports the recommendation of repeated COVID-19 vaccinations for patients suffering from immunodeficiencies, especially when they exhibit an initially limited response to the vaccine., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. Cytokine-directed cellular cross-talk imprints synovial pathotypes in rheumatoid arthritis.
- Author
-
Kugler M, Dellinger M, Kartnig F, Müller L, Preglej T, Heinz LX, Simader E, Göschl L, Puchner SE, Weiss S, Shaw LE, Farlik M, Weninger W, Superti-Furga G, Smolen JS, Steiner G, Aletaha D, Kiener HP, Lewis MJ, Pitzalis C, Tosevska A, Karonitsch T, and Bonelli M
- Subjects
- Humans, Cytokines, Tumor Necrosis Factor-alpha pharmacology, Synovial Membrane pathology, Fibroblasts pathology, Cells, Cultured, Arthritis, Rheumatoid, Synoviocytes pathology
- Abstract
Introduction: Structural reorganisation of the synovium with expansion of fibroblast-like synoviocytes (FLS) and influx of immune cells is a hallmark of rheumatoid arthritis (RA). Activated FLS are increasingly recognised as a critical component driving synovial tissue remodelling by interacting with immune cells resulting in distinct synovial pathotypes of RA., Methods: Automated high-content fluorescence microscopy of co-cultured cytokine-activated FLS and autologous peripheral CD4
+ T cells from patients with RA was established to quantify cell-cell interactions. Phenotypic profiling of cytokine-treated FLS and co-cultured T cells was done by flow cytometry and RNA-Seq, which were integrated with publicly available transcriptomic data from patients with different histological synovial pathotypes. Computational prediction and knock-down experiments were performed in FLS to identify adhesion molecules for cell-cell interaction., Results: Cytokine stimulation, especially with TNF-α, led to enhanced FLS-T cell interaction resulting in cell-cell contact-dependent activation, proliferation and differentiation of T cells. Signatures of cytokine-activated FLS were significantly enriched in RA synovial tissues defined as lymphoid-rich or leucocyte-rich pathotypes, with the most prominent effects for TNF-α. FLS cytokine signatures correlated with the number of infiltrating CD4+ T cells in synovial tissue of patients with RA. Ligand-receptor pair interaction analysis identified ICAM1 on FLS as an important mediator in TNF-mediated FLS-T cell interaction. Both, ICAM1 and its receptors were overexpressed in TNF-treated FLS and co-cultured T cells. Knock-down of ICAM1 in FLS resulted in reduced TNF-mediated FLS-T cell interaction., Conclusion: Our study highlights the role of cytokine-activated FLS in orchestrating inflammation-associated synovial pathotypes providing novel insights into disease mechanisms of RA., Competing Interests: Competing interests: MB reports about personal fees from Eli-Lilly and received grants from Galapagos, DA received grants and consulting fees from AbbVie, Amgen, Lilly, Merck, Novartis, Pfizer, Roche and Sandozand is an editorial board member of Annals of the Rheumatic Diseases, JSS reports about grants, consulting and personal fees from AbbVie, Astra-Zeneca, Lilly, Novartis, Amgen, Astro, Bristol-Myers Squibb, Celgene, Celltrion, Chugai, Gilead, ILTOO, Janssen, Merck Sharp & Dohme, Novartis-Sandoz, Pfizer, Roche, Samsung and UCB and is editor-in-chief of Annals of the Rheumatic Diseases. ES reports speaker fees from Eli-Lilly and supports for attendance of meetings from Pfizer, Bristol-Myers Squibb, Boehringer Ingelheim and Astra Zeneca. CP and is an editorial board member of Annals of the Rheumatic Diseases. The authors CP and MJL are named inventors on a patent application (no. GB 2100821.4), submitted by Queen Mary University of London, that covers methods used to select treatments in rheumatoid arthritis. All other authors declare no competing interests., (© Author(s) (or their employer(s)) 2023. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
16. Lessons for the Clinical Nephrologist: Nephrotic syndrome associated with refractory Giardia duodenalis infection in a patient with acquired B cell depletion.
- Author
-
Eder M, Pimenov L, Böhmig GA, Kornek B, Göschl L, Bond G, Vossen MG, Pickl WF, Breuer M, Görzer I, Kozakowski N, Laferl H, and Winkler S
- Subjects
- Humans, Nephrologists, B-Lymphocytes, Feces, Genotype, Prevalence, Giardiasis complications, Giardiasis diagnosis, Giardiasis drug therapy, Nephrotic Syndrome complications, Nephrotic Syndrome diagnosis
- Published
- 2023
- Full Text
- View/download PDF
17. Advanced immunophenotyping: A powerful tool for immune profiling, drug screening, and a personalized treatment approach.
- Author
-
Preglej T, Brinkmann M, Steiner G, Aletaha D, Göschl L, and Bonelli M
- Subjects
- Humans, Immunophenotyping, Precision Medicine, Drug Evaluation, Preclinical, Leukocytes, Mononuclear, Autoimmune Diseases diagnosis, Autoimmune Diseases drug therapy
- Abstract
Various autoimmune diseases are characterized by distinct cell subset distributions and activation profiles of peripheral blood mononuclear cells (PBMCs). PBMCs can therefore serve as an ideal biomarker material, which is easily accessible and allows for screening of multiple cell types. A detailed understanding of the immune landscape is critical for the diagnosis of patients with autoimmune diseases, as well as for a personalized treatment approach. In our study, we investigate the potential of multi-parameter spectral flow cytometry for the identification of patients suffering from autoimmune diseases and its power as an evaluation tool for in vitro drug screening approaches (advanced immunophenotyping). We designed a combination of two 22-color immunophenotyping panels for profiling cell subset distribution and cell activation. Downstream bioinformatics analyses included percentages of individual cell populations and median fluorescent intensity of defined markers which were then visualized as heatmaps and in dimensionality reduction approaches. In vitro testing of epigenetic immunomodulatory drugs revealed an altered activation status upon treatment, which supports the use of spectral flow cytometry as a high-throughput drug screening tool. Advanced immunophenotyping might support the exploration of novel therapeutic drugs and contribute to future personalized treatment approaches in autoimmune diseases and beyond., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Preglej, Brinkmann, Steiner, Aletaha, Göschl and Bonelli.)
- Published
- 2023
- Full Text
- View/download PDF
18. Accelerated waning of immune responses to a third COVID-19 vaccination in patients with immune-mediated inflammatory diseases.
- Author
-
Mrak D, Kartnig F, Sieghart D, Simader E, Radner H, Mandl P, Göschl L, Hofer P, Deimel T, Gessl I, Kain R, Winkler S, Smolen JS, Perkmann T, Haslacher H, Aletaha D, Heinz LX, and Bonelli M
- Subjects
- Humans, COVID-19 Vaccines, Spike Glycoprotein, Coronavirus, SARS-CoV-2, Antibodies, Immunity, Humoral, Antibodies, Viral, Vaccination, COVID-19, Antirheumatic Agents
- Abstract
Background: A 3
rd COVID-19 vaccination is currently recommended for patients under immunosuppression. However, a fast decline of antibodies against the SARS-CoV-2 receptor-binding domain (RBD) of the spike protein has been observed. Currently it remains unclear whether immunosuppressive therapy affects kinetics of humoral and cellular immune responses., Methods: 50 patients under immunosuppression and 42 healthy controls (HCs) received a 3rd dose of an mRNA-based vaccine and were monitored over a 12-weeks period. Humoral immune response was assessed 4 and 12 weeks after 3rd dose. Antibodies were quantified using the Elecsys Anti-SARS-CoV-2 Spike immunoassay against the receptor-binding domain (RBD) of the spike protein. SARS-CoV-2-specific T cell responses were quantified by IFN-γ ELISpot assays. Adverse events, including SARS-CoV-2 infections, were monitored over a 12-week period., Results: At week 12, reduced anti-RBD antibody levels were observed in IMID patients as compared to HCs (median antibody level 5345 BAU/ml [1781-10,208] versus 9650 BAU/ml [6633-16,050], p < 0.001). Reduction in relative antibody levels was significantly higher in IMID patients as compared to HCs at week 12 (p < 0.001). Lowest anti-RBD antibody levels were detected in IMID patients who received biological disease-modifying anti-rheumatic drugs (DMARDs) or a combination therapy with conventional synthetic and biological DMARDs. Number of SARS-CoV-2-specific T cells against wildtype and Omicron variants remained stable over 12 weeks in IMID patients. No serious adverse events were reported., Conclusion: Due to a fast decline in anti-RBD antibodies in IMID patients an early 4th vaccination should be considered in this vulnerable group of patients., Competing Interests: Declaration of competing interest DM reports support for meeting attendances from Pfizer and consultation fees from AstraZeneca; JS reports about grants, consulting and personal fees from AbbVie, Amgen, AstraZeneca, Astro, Bristol-Myers Squibb, Celltrion, Gilead-Galapagos, Janssen, Lilly, Pfizer, R-Pharma, Samsung, Sanofi, Chugai, Merck Sharp & Dohme, Novartis-Sandoz Roche, Samsung and UCB and grants from Abbvie, AstraZeneca, Lilly, Novartis, and Roche; HR received speaker fees from Gilead, Merck Sharp and Pfizer and travel support from Janssen; HH received grants from Glock Health, BlueSky Immunotherapies and Neutrolis; DA received grants, speaker fees, or consultancy fees from Abbvie, Amgen, Galapagos, Lilly, Janssen, Merck, Novartis, Pfizer, Sandoz, and Sanofi; RK reports consulting fees from AstraZeneca, Takeda Pharma, MEDahead and Janssen Cilag and speaker fees from Otsuka; ES received travel support from Pfizer, Bristol-Myers Squibb and Boehringer-Ingelheim and speaker fees from Lilly; MB reports about personal fees from Eli-Lilly. All other authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
19. Safety and immunogenicity of a third COVID-19 vaccination in patients with immune-mediated inflammatory diseases compared with healthy controls.
- Author
-
Kartnig F, Mrak D, Simader E, Tobudic S, Radner H, Mandl P, Göschl L, Hommer N, Mayer M, Hofer P, Hummel T, Deimel T, Geßl I, Puchner A, Kerschbaumer A, Thalhammer R, Handisurya A, Kain R, Winkler S, Smolen JS, Stiasny K, Perkmann T, Haslacher H, Aberle JH, Aletaha D, Heinz LX, Sieghart D, and Bonelli M
- Subjects
- Humans, Antibodies, Viral, Antirheumatic Agents, COVID-19, Immunogenicity, Vaccine, Immunomodulating Agents, Vaccination, COVID-19 Vaccines adverse effects
- Abstract
Objectives: A third COVID-19 vaccination is recommended for immunosuppressed patients. However, data on immunogenicity and safety of a third COVID-19 vaccination in patients with immune-mediated inflammatory diseases (IMIDs) are sparse and therefore addressed within this clinical trial., Methods: 60 immunosuppressed patients and 48 healthy controls (HCs) received a third vaccination with an mRNA vaccine. The primary endpoint was defined as the presence of antibody levels against the receptor-binding domain (RBD)>1500 BAU/mL in patients with IMIDs versus HCs. Further endpoints included differences in neutralising antibodies and cellular immune responses after the third vaccination. Reactogenicity was recorded for 7 days, and safety was evaluated until week 4., Results: Rate of individuals with anti-RBD antibodies>1500 BAU/mL was not significantly different after the third vaccination between patients with IMIDs and HCs (91% vs 100% p=0.101). Anti-RBD and neutralising antibody levels were significantly lower in patients with IMIDs after the third vaccination than in HCs (p=0.002 and p=0.016, respectively). In contrast, fold increase in antibody levels between week 0 and 4 was higher in patients with IMIDs. Treatment with biological (b) disease-modifying anti-rheumatic drugs (DMARD) or combination of bDMARDs and conventional synthetic DMARDs was associated with reduced antibody levels. Enhanced cellular immune response to wild type and Omicron peptide stimulation was observed after the third vaccination. No serious adverse event was attributed to the third vaccination., Conclusion: Our clinical trial data support the immunogenicity and safety of a third COVID-19 vaccination in patients with IMIDs. However, effects of DMARD therapy on immunogenicity should be considered., Trial Registration Number: EudraCT No: 2021-002693-10., Competing Interests: Competing interests: DM reports support for meeting attendances from Pfizer. AK reports about contracts and personal fees from AbbVie, Bristol Myers Squibb, Celgene, Eli Lilly, Gilead, Merck Sharp and Dohme. PM reports speaker fees from AbbVie, Janssen and Novartis and research grants from AbbVie, BMS, Novartis, Janssen, MSD and UCB. JSS reports about grants, consulting and personal fees from AbbVie, Astra-Zeneca, Lilly, Novartis, Amgen, Astro, Bristol-Myers Squibb, Celgene, Celltrion, Chugai, Gilead, ILTOO, Janssen, Merck Sharp & Dohme, Novartis-Sandoz, Pfizer, Roche, Samsung and UCB. AH reports fees from MDGH. HH received grants from Glock Health, BlueSky Immunotherapies and Neutrolis. DA received grants and consulting fees from AbbVie, Amgen, Lilly, Merck, Novartis, Pfizer, Roche and Sandoz. MB reports about personal fees from Eli-Lilly. All other authors declare no competing interests., (© Author(s) (or their employer(s)) 2023. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
20. Immunogenicity and safety of a fourth COVID-19 vaccination in rituximab-treated patients: an open-label extension study.
- Author
-
Mrak D, Simader E, Sieghart D, Mandl P, Radner H, Perkmann T, Haslacher H, Mayer M, Koblischke M, Hofer P, Göschl L, Kartnig F, Deimel T, Kerschbaumer A, Hummel T, Kornek B, Thalhammer R, Stiasny K, Winkler S, Smolen JS, Aberle JH, Aletaha D, Heinz LX, and Bonelli M
- Subjects
- Humans, Rituximab adverse effects, Antibodies, Viral, SARS-CoV-2, BNT162 Vaccine, Vaccination, RNA, Messenger, Immunogenicity, Vaccine, COVID-19 Vaccines adverse effects, COVID-19 prevention & control
- Abstract
Objectives: Patients under rituximab therapy are at high risk for a severe COVID-19 disease course. Humoral immune responses to SARS-CoV-2 vaccination are vastly diminished in B-cell-depleted patients, even after a third vaccine dose. However, it remains unclear whether these patients benefit from a fourth vaccination and whether continued rituximab therapy affects antibody development., Methods: In this open-label extension trial, 37 rituximab-treated patients who received a third dose with either a vector or mRNA-based vaccine were vaccinated a fourth time with an mRNA-based vaccine (mRNA-1273 or BNT162b2). Key endpoints included the humoral and cellular immune response as well as safety after a fourth vaccination., Results: The number of patients who seroconverted increased from 12/36 (33%) to 21/36 (58%) following the fourth COVID-19 vaccination. In patients with detectable antibodies to the spike protein's receptor-binding domain (median: 8.0 binding antibody units (BAU)/mL (quartiles: 0.4; 13.8)), elevated levels were observed after the fourth vaccination (134.0 BAU/mL (quartiles: 25.5; 1026.0)). Seroconversion and antibody increase were strongly diminished in patients who received rituximab treatment between the third and the fourth vaccination. The cellular immune response declined 12 weeks after the third vaccination, but could only be slightly enhanced by a fourth vaccination. No unexpected safety signals were detected, one serious adverse event not related to vaccination occurred., Conclusions: A fourth vaccine dose is immunogenic in a fraction of rituximab-treated patients. Continuation of rituximab treatment reduced humoral immune response, suggesting that rituximab affects a second booster vaccination. It might therefore be considered to postpone rituximab treatment in clinically stable patients., Trial Registration Number: 2021-002348-57., Competing Interests: Competing interests: PM reports speaker fees from AbbVie, Janssen and Novartis and research grants from AbbVie, BMS, Novartis, Janssen, MSD and UCB. MB reports about personal fees from Eli-Lilly, DA received grants and consulting fees from AbbVie, Amgen, Lilly, Merck, Novartis, Pfizer, Roche and Sandoz. JSS reports about grants, consulting and personal fees from AbbVie, Astra-Zeneca, Lilly, Novartis, Amgen, Astro, Bristol-Myers Squibb, Celgene, Celltrion, Chugai, Gilead, ILTOO, Janssen, Merck Sharp & Dohme, Novartis-Sandoz, Pfizer, Roche, Samsung and UCB. DM received support for meeting attendances from Pfizer. HH received grants from Glock Health, BlueSky Immunotherapies and Neutrolis. AK reports about speaker and consulting fees from AbbVie, Amgen, Bristol Myers Squibb, Eli Lilly, Gilead, Janssen, Merck Sharp and Dohme, Novartis and Pfizer. All other authors declare no competing interests., (© Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
21. Reactogenicity and immunogenicity of the second COVID-19 vaccination in patients with inborn errors of immunity or mannan-binding lectin deficiency.
- Author
-
Göschl L, Mrak D, Grabmeier-Pfistershammer K, Stiasny K, Haslacher H, Schneider L, Deimel T, Kartnig F, Tobudic S, Aletaha D, Burgmann H, Bonelli M, Pickl WF, Förster-Waldl E, Scheinecker C, and Vossen MG
- Subjects
- Antibodies, Viral, Humans, Interferon-gamma, SARS-CoV-2, Vaccination, COVID-19 prevention & control, COVID-19 Vaccines adverse effects, Mannose-Binding Lectin
- Abstract
Background: Patients with inborn errors of immunity (IEI) are at increased risk for severe courses of SARS-CoV-2 infection. COVID-19 vaccination provides effective protection in healthy individuals. However, it remains unclear whether vaccination is efficient and safe in patients with constitutional dysfunctions of the immune system. Thus, we analyzed the humoral response, adverse reactions and assessed the disease activity of the underlying disease after COVID-19 vaccination in a cohort of patients suffering from IEIs or mannan-binding lectin deficiency (MBLdef)., Methods: Vaccination response was assessed after basic immunization using the Elecsys anti-SARS-CoV-2 S immunoassay and via Vero E6 cell based assay to detect neutralization capabilities. Phenotyping of lymphocytes was performed by flow cytometry. Patient charts were reviewed for disease activity, autoimmune phenomena as well as immunization status and reactogenicity of the vaccination. Activity of the underlying disease was assessed using a patient global numeric rating scale (NRS)., Results: Our cohort included 11 individuals with common variable immunodeficiency (CVID), one patient with warts hypogammaglobulinemia immunodeficiency myelokathexis (WHIM) syndrome, two patients with X-linked agammaglobulinemia (XLA), one patient with Muckle Wells syndrome, two patients with cryopyrin-associated periodic syndrome, one patient with Interferon-gamma (IFN-gamma) receptor defect, one patient with selective deficiency in pneumococcal antibody response combined with a low MBL level and seven patients with severe MBL deficiency. COVID-19 vaccination was generally well tolerated with little to no triggering of autoimmune phenomena. 20 out of 26 patients developed an adequate humoral vaccine response. 9 out of 11 patients developed a T cell response comparable to healthy control subjects. Tested immunoglobulin replacement therapy (IgRT) preparations contained Anti-SARS-CoV-2 S antibodies implicating additional protection through IgRT., Summary: In summary the data support the efficacy and safety of a COVID-19 vaccination in patients with IEIs/MBLdef. We recommend evaluation of the humoral immune response and testing for virus neutralization after vaccination in this cohort., Competing Interests: DA is a member of the editorial board of “Annals of the Rheumatic diseases”. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Göschl, Mrak, Grabmeier-Pfistershammer, Stiasny, Haslacher, Schneider, Deimel, Kartnig, Tobudic, Aletaha, Burgmann, Bonelli, Pickl, Förster-Waldl, Scheinecker and Vossen.)
- Published
- 2022
- Full Text
- View/download PDF
22. Heterologous vector versus homologous mRNA COVID-19 booster vaccination in non-seroconverted immunosuppressed patients: a randomized controlled trial.
- Author
-
Mrak D, Sieghart D, Simader E, Tobudic S, Radner H, Mandl P, Göschl L, Koblischke M, Hommer N, Wagner A, Mayer M, Schubert L, Hartl L, Kozbial K, Hofer P, Kartnig F, Hummel T, Kerschbaumer A, Deimel T, Puchner A, Gudipati V, Thalhammer R, Munda P, Uyanik-Ünal K, Zuckermann A, Novacek G, Reiberger T, Garner-Spitzer E, Reindl-Schwaighofer R, Kain R, Winkler S, Smolen JS, Stiasny K, Fischer GF, Perkmann T, Haslacher H, Zeitlinger M, Wiedermann U, Aberle JH, Aletaha D, Heinz LX, and Bonelli M
- Subjects
- Antibodies, Viral, ChAdOx1 nCoV-19, Humans, Immunization, Secondary, RNA, Messenger, SARS-CoV-2 genetics, Vaccination, Vaccines, Synthetic, mRNA Vaccines, BNT162 Vaccine, COVID-19 prevention & control, COVID-19 Vaccines adverse effects
- Abstract
Impaired response to COVID-19 vaccination is of particular concern in immunosuppressed patients. To determine the best vaccination strategy for this vulnerable group we performed a single center, 1:1 randomized blinded clinical trial. Patients who failed to seroconvert upon two mRNA vaccinations (BNT162b2 or mRNA-1273) are randomized to receive either a third dose of the same mRNA or the vector vaccine ChAdOx1 nCoV-19. Primary endpoint is the difference in SARS-CoV-2 spike antibody seroconversion rate between vector and mRNA vaccinated patients four weeks after the third dose. Secondary outcomes include cellular immune responses. Seroconversion rates at week four are significantly higher in the mRNA (homologous vaccination, 15/24, 63%) as compared to the vector vaccine group (heterologous vaccination, 4/22, 18%). SARS-CoV-2-specific T-cell responses are reduced but could be increased after a third dose of either vector or mRNA vaccine. In a multivariable logistic regression analysis, patient age and vaccine type are associated with seroconversion. No serious adverse event is attributed to COVID-19 booster vaccination. Efficacy and safety data underline the importance of a booster vaccination and support the use of a homologous mRNA booster vaccination in immunosuppressed patients.Trial registration: EudraCT No.: 2021-002693-10., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
23. Segmental endobronchial valve therapy for a vasculitis-induced emphysema.
- Author
-
Bal C, Göschl L, Milos RI, Gerstbrein K, Kerschbaumer A, Idzko M, and Gompelmann D
- Abstract
A 53-year old female patient with history of hypocomplementaemic urticarial vasculitis syndrome (HUVS) and polyarteritis nodosa presented with progressive dyspnoea on exertion due to emphysema. Lung function revealed a severe obstructive ventilator disorder with a forced expiratory volume in 1 second of 22% of predicted, and a significant hyperinflation with a residual volume of 321% of predicted. Multi-detector computed tomography (MDCT) scan and quantitative CT analysis (StratX software) confirmed a lower lobe predominant emphysema. Considering the young age, the very severely impaired lung function, the relatively low nicotine abuse, the exclusion of alpha-1 antitrypsin deficiency, together with the known diagnosis of HUVS, the emphysema was more likely due to the vasculitis than to a typical chronic obstructive lung disease. MDCT scan showed that particularly the segment 8 of the right lower lobe was severely emphysematous destroyed and hyperinflated. Invasive Chartis® measurement revealed no significant collateral ventilation of the isolated segment 8 of the right lower lobe, so that an endobronchial valve placement was performed. Three months following intervention, the MDCT scan revealed a complete collapse of the segment 8 on the right, which was associated with a significant clinical benefit and a mild reduction of the hyperinflation in the lung function test., (© 2022 The Authors. Published by Elsevier Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
24. Detection of DHCMT long-term metabolite glucuronides with LC-MSMS as an alternative approach to conventional GC-MSMS analysis.
- Author
-
Göschl L, Gmeiner G, Gärtner P, Steinacher M, and Forsdahl G
- Subjects
- Gas Chromatography-Mass Spectrometry methods, Glucuronides urine, Humans, Substance Abuse Detection methods, Anabolic Agents chemistry, Doping in Sports
- Abstract
Dehydrochloromethyltestosterone (DHCMT) is one of the most detected illicit used anabolic-androgenic steroids in professional sports. Therefore, a fast and accurate analysis of this substance is of great importance for a constructive fight against doping abuse. The conventional method for the analysis of this drug, GC-MSMS, is very sensitive and selective but also very time- and resource-consuming. With the presented work, a new approach for simple detection with LC-HRMSMS without any sample preparation is introduced. The method is based on the direct analysis of two newly described phase-II metabolites of the DHCMT long-term metabolite 4-chloro-18-nor-17β-hydroxymethyl-17α-methyl-5β-androst-13-en-3α-ol (M3). LC-HRMSMS, GC-MSMS, fractionation and derivatization experiments are combined to identify and characterize for the first time two different glucuronide-acid conjugates of this metabolite in positive human urine samples. In addition, a third glucuronide metabolite was identified, however without isomeric structure determination. The detection of these metabolites is particularly interesting for confirmation analyses, as the method is rapid and requires little sample material., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
25. Stanozolol-N-glucuronide metabolites in human urine samples as suitable targets in terms of routine anti-doping analysis.
- Author
-
Göschl L, Gmeiner G, Gärtner P, Stadler G, Enev V, Thevis M, Schänzer W, Guddat S, and Forsdahl G
- Subjects
- Anabolic Agents metabolism, Anabolic Agents urine, Female, Glucuronides analysis, Glucuronides urine, Humans, Limit of Detection, Male, Solid Phase Extraction methods, Stanozolol metabolism, Stanozolol urine, Time Factors, Anabolic Agents analysis, Doping in Sports prevention & control, Stanozolol analysis, Substance Abuse Detection methods
- Abstract
The exogenous anabolic-androgenic steroid (AAS) stanozolol stays one of the most detected substances in professional sports. Its detection is a fundamental part of doping analysis, and the analysis of this steroid has been intensively investigated for a long time. This contribution to the detection of stanozolol doping describes for the first time the unambiguous proof for the existence of 17-epistanozolol-1'N-glucuronide and 17-epistanozolol-2'N-glucuronide in stanozolol-positive human urine samples due to the access to high-quality reference standards. Examination of excretion study samples shows large detection windows for the phase-II metabolites stanozolol-1'N-glucuronide and 17-epistanozolol-1'N-glucuronide up to 12 days and respectively up to almost 28 days. In addition, we present appropriate validation parameters for the analysis of these metabolites using a fully automatic method online solid-phase extraction (SPE) method already published before. Limits of identification (LOIs) as low as 100 pg/ml and other validation parameters like accuracy, precision, sensitivity, robustness, and linearity are given., (© 2021 The Authors. Drug Testing and Analysis published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
26. Immunologisch relevante Aspekte der neuen COVID-19-Impfstoffe : Ein Positionspapier der Österreichischen Gesellschaft für Allergologie und Immunologie (ÖGAI) und des Ärzteverbands Deutscher Allergologen (AeDA).
- Author
-
Untersmayr E, Förster-Waldl E, Bonelli M, Boztug K, Brunner PM, Eiwegger T, Eller K, Göschl L, Grabmeier-Pfistershammer K, Hötzenecker W, Jordakieva G, Moschen AR, Pfaller B, Pickl W, Reinisch W, Wiedermann U, Klimek L, Bergmann KC, Brehler R, Novak N, Merk HF, Rabe U, Schlenter WW, Ring J, Wehrmann W, Mülleneisen NK, Wrede H, Fuchs T, and Jensen-Jarolim E
- Published
- 2021
- Full Text
- View/download PDF
27. Immunologically relevant aspects of the new COVID-19 vaccines-an ÖGAI (Austrian Society for Allergology and Immunology) and AeDA (German Society for Applied Allergology) position paper.
- Author
-
Untersmayr E, Förster-Waldl E, Bonelli M, Boztug K, Brunner PM, Eiwegger T, Eller K, Göschl L, Grabmeier-Pfistershammer K, Hötzenecker W, Jordakieva G, Moschen AR, Pfaller B, Pickl W, Reinisch W, Wiedermann U, Klimek L, Bergmann KC, Brehler R, Pfützner W, Novak N, Merk H, Rabe U, Schlenter W, Ring J, Wehrmann W, Mülleneisen N, Wrede H, Fuchs T, and Jensen-Jarolim E
- Abstract
Background: The vaccines against the coronavirus disease 2019 (COVID-19) approved in the European Union represent a decisive step in the fight against the pandemic. The application of these available vaccines to patients with pre-existing immunological conditions leads to a multitude of questions regarding efficacy, side effects and the necessary patient information., Results: This review article provides insight into mechanisms of action of the currently available severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines and summarises the current state of science as well as expert recommendations regarding tolerability of the vaccines. In addition, the potential to develop protective immune responses is determined. A special focus is given on patients under immunosuppression or in treatment with immunomodulatory drugs. Special groups of the population such as children, pregnant women and the elderly are also considered., Conclusion: Despite the need for a patient-specific risk-benefit assessment, the consensus among experts is that patients with immunological diseases in particular benefit from the induced immune protection after COVID-19 vaccination and do not have an increased risk of side effects., Competing Interests: Conflict of interestP.M. Brunner is an employee of the Medical University of Vienna and has received personal fees from LEO Pharma, Pfizer, Sanofi Genzyme, Eli Lilly, Novartis, Celgene, UCB Pharma, Biotest, Boehringer Ingelheim, AbbVie, Amgen and Arena Pharmaceuticals. He also works as an investigator on behalf of Novartis and has received grants for his institute. T. Eiwegger serves as principal investigator for DBV’s company-sponsored studies and as a secondary investigator (sub-investigator) for Regeneron. He also receives grants from Innovation Fund Denmark and the Canadian Institutes of Health Research (CIHR) outside the submitted work. He is co-investigator or scientific lead in three investigator-initiated oral immunotherapy studies supported by the Food Allergy and Anaphylaxis Program‘SickKids’. In addition, serves as Associate Editor for Allergy. He/his laboratory has received unconditional in-kind support from Macro Array Diagnostics and ALK. He also serves as an Advisory Board Member for ALK. W. Reinisch has acted in an advisory capacity for: Abbvie, Algernon, Amgen, AM Pharma, AMT, AOP Orphan, Arena Pharmaceuticals, Astellas, Astra Zeneca, Avaxia, Roland Berger GmBH, Bioclinica, Biogen IDEC, Boehringer-Ingelheim, Bristol-Myers Squibb, Cellerix, Chemocentryx, Celgene, Centocor, Celltrion, Covance, Danone Austria, DSM, Elan, Eli Lilly, Ernest & Young, Falk Pharma GmbH, Ferring, Galapagos, Gatehouse Bio Inc., Genentech, Gilead, Grünenthal, ICON, Index Pharma, Inova, Intrinsic Imaging, Janssen, Johnson & Johnson, Kyowa Hakko Kirin Pharma, Lipid Therapeutics, LivaNova, Mallinckrodt, Medahead, MedImmune, Millenium, Mitsubishi Tanabe Pharma Corporation, MSD, Nash Pharmaceuticals, Nestle, Nippon Kayaku, Novartis, Ocera, OMass, Otsuka, Parexel, PDL, Periconsulting, Pharmacosmos, Philip Morris Institute, Pfizer, Procter & Gamble, Prometheus, Protagonist, Provention, Quell Therapeutics, Robarts Clinical Trial, Sandoz, Schering-Plough, Second Genome, Seres Therapeutics, Setpointmedical, Sigmoid, Sublimity, Takeda, Therakos, Theravance, Tigenix, UCB, Vifor, Zealand, Zyngenia and 4SC. E. Untersmayr, E. Förster-Waldl, M. Bonelli,K. Boztug, K. Eller, L. Göschl, K. Grabmeier-Pfistershammer, W. Hötzenecker, G. Jordakieva, A.R. Moschen, B. Pfaller, W. Pickl, U. Wiedermann, L. Klimek, K.-C. Bergmann, R. Brehler, W. Pfützner, N. Novak, H. Merk, U. Rabe, W. Schlenter, J. Ring, W. Wehrmann, N. Mülleneisen, H. Wrede, T. Fuchs and E. Jensen-Jarolim declare that they have no competing interests., (© The Author(s) 2021.)
- Published
- 2021
- Full Text
- View/download PDF
28. Development and validation of a simple online-SPE method coupled to high-resolution mass spectrometry for the analysis of stanozolol-N-glucuronides in urine samples.
- Author
-
Göschl L, Gmeiner G, Enev V, Kratena N, Gärtner P, and Forsdahl G
- Subjects
- Anabolic Agents urine, Doping in Sports prevention & control, Female, Humans, Limit of Detection, Male, Reproducibility of Results, Solid Phase Extraction, Tandem Mass Spectrometry, Chromatography, High Pressure Liquid methods, Glucuronides urine, Stanozolol urine, Substance Abuse Detection methods
- Abstract
Stanozolol is still the most commonly used illicit anabolic-androgenic steroid (AAS) in professional sports. Therefore, accurate and fast analysis and long detection windows are of great interest in the field of antidoping analysis. In this work, a very simple, fast, and highly sensitive online solid-phase extraction method coupled with liquid chromatography-high-resolution tandem mass spectrometry (HPLC-HRMSMS) for the analysis of stanozolol-N-glucuronides was developed. This fully validated procedure is characterized by only a few manual steps (dilution and addition of internal standard) in the sample preparation. A limit of identification (LOI) of 75 pg/mL, high accuracy (87.1%-102.1%), precision (3.1%-7.8%), and sensitivity was achieved. Furthermore, good linearity (> 0.99) and robustness, as well as no carry-over effects, could be observed. In addition to excellent confirmation analysis performance, this method shows sufficient potential for the identification and characterization of unknown metabolites. Using this method, it was possible to unambiguously confirm the presence of 1'N- and 2'N-stanozolol-glucuronide in human urine for the first time due to the access to reference material., (© 2020 The Authors. Drug Testing and Analysis published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
29. Treg cells in health and autoimmune diseases: New insights from single cell analysis.
- Author
-
Scheinecker C, Göschl L, and Bonelli M
- Subjects
- Animals, Autoimmune Diseases diagnosis, Autoimmune Diseases therapy, Biomarkers, Disease Management, Epigenesis, Genetic, Epigenomics, Gene Expression Regulation, Homeostasis, Humans, Single-Cell Analysis, Autoimmune Diseases etiology, Autoimmune Diseases metabolism, Autoimmunity, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism
- Abstract
Autoimmune diseases, such as Systemic Lupus Erythematosus (SLE) or Rheumatoid Arthritis (RA) are characterized by the breakdown of immunological tolerance. Defects of regulatory T cells have been described among the various mechanisms, that are important for the development of autoimmune diseases, due to their critical role as regulators of peripheral immune tolerance and homeostasis. Initially T suppressor cells have been described as one population of peripheral T cells. Based on new technological advances a new understanding of the heterogeneity of different Treg cell populations in the lymphoid and non-lymphoid tissue has evolved over the last years. While initially Foxp3 has been defined as the main master regulator of Treg cells, we have learned that Treg cells from various tissue can be identified by a specific transcriptomic and epigenetic signature. Epigenetic mechanisms allow Treg cell stability, but we have also learned that certain Treg subsets are plastic and can under specific circumstances even enhance autoimmunity and inflammatory processes. Quantitative and functional defects of Treg cells have been observed in a variety of autoimmune diseases. Due to our understanding of the nature of this cell population, Treg cells have been a target of new Treg based therapies, such as low-dose IL-2. In addition, ongoing clinical trials aim to test safety and efficacy of transferred, in vitro expanded Treg cells in patients with autoimmune diseases and transplant patients., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
30. Metabolism of steroids and sport drug testing.
- Author
-
Stojanovic BJ, Göschl L, Forsdahl G, and Günter G
- Subjects
- Anabolic Agents metabolism, Chromatography, Gas, Chromatography, High Pressure Liquid, Humans, Steroids analysis, Tandem Mass Spectrometry, Anabolic Agents analysis, Doping in Sports, Steroids metabolism, Substance Abuse Detection
- Published
- 2020
- Full Text
- View/download PDF
31. Histone deacetylase 1 (HDAC1): A key player of T cell-mediated arthritis.
- Author
-
Göschl L, Preglej T, Boucheron N, Saferding V, Müller L, Platzer A, Hirahara K, Shih HY, Backlund J, Matthias P, Niederreiter B, Hladik A, Kugler M, Gualdoni GA, Scheinecker C, Knapp S, Seiser C, Holmdahl R, Tillmann K, Plasenzotti R, Podesser B, Aletaha D, Smolen JS, Karonitsch T, Steiner G, Ellmeier W, and Bonelli M
- Subjects
- Animals, Arthritis, Rheumatoid pathology, Biomarkers, Collagen adverse effects, Cytokines metabolism, Disease Models, Animal, Gene Expression Regulation, Histone Deacetylase 1 genetics, Humans, Inflammation Mediators metabolism, Mice, Mice, Knockout, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Arthritis, Rheumatoid etiology, Arthritis, Rheumatoid metabolism, Disease Susceptibility, Histone Deacetylase 1 metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Rheumatoid Arthritis (RA) represents a chronic T cell-mediated inflammatory autoimmune disease. Studies have shown that epigenetic mechanisms contribute to the pathogenesis of RA. Histone deacetylases (HDACs) represent one important group of epigenetic regulators. However, the role of individual HDAC members for the pathogenesis of arthritis is still unknown. In this study we demonstrate that mice with a T cell-specific deletion of HDAC1 (HDAC1-cKO) are resistant to the development of Collagen-induced arthritis (CIA), whereas the antibody response to collagen type II was undisturbed, indicating an unaltered T cell-mediated B cell activation. The inflammatory cytokines IL-17 and IL-6 were significantly decreased in sera of HDAC1-cKO mice. IL-6 treated HDAC1-deficient CD4
+ T cells showed an impaired upregulation of CCR6. Selective inhibition of class I HDACs with the HDAC inhibitor MS-275 under Th17-skewing conditions inhibited the upregulation of chemokine receptor 6 (CCR6) in mouse and human CD4+ T cells. Accordingly, analysis of human RNA-sequencing (RNA-seq) data and histological analysis of synovial tissue samples from human RA patients revealed the existence of CD4+ CCR6+ cells with enhanced HDAC1 expression. Our data indicate a key role for HDAC1 for the pathogenesis of CIA and suggest that HDAC1 and other class I HDACs might be promising targets of selective HDAC inhibitors (HDACi) for the treatment of RA., Competing Interests: Declaration of competing interest None. This research has received support from the Innovative Medicines Initiative (IMI) Joint Undertaking under grant agreement n°115142 (BTCure), and grant agreement n(o)777357 (RTCure) resources of which are composed of financial contribution from the European Union's Framework Programmes and EFPIA companies' in kind contribution. The work has been supported by the Austrian Science Fund (FWF) project F26193 (to WE) and the FWF special research program SFB F70 (subproject F7003 to MB, F7004 to NB and F7005 to WE)., (Copyright © 2019. Published by Elsevier Ltd.)- Published
- 2020
- Full Text
- View/download PDF
32. Histone deacetylases 1 and 2 restrain CD4+ cytotoxic T lymphocyte differentiation.
- Author
-
Preglej T, Hamminger P, Luu M, Bulat T, Andersen L, Göschl L, Stolz V, Rica R, Sandner L, Waltenberger D, Tschismarov R, Faux T, Boenke T, Laiho A, Elo LL, Sakaguchi S, Steiner G, Decker T, Bohle B, Visekruna A, Bock C, Strobl B, Seiser C, Boucheron N, and Ellmeier W
- Subjects
- Animals, CD4-Positive T-Lymphocytes drug effects, Fatty Acids pharmacology, Histone Deacetylase 1 genetics, Histone Deacetylase 2 genetics, Humans, Mice, Mice, Knockout, Signal Transduction physiology, T-Lymphocytes, Cytotoxic drug effects, Up-Regulation drug effects, Up-Regulation physiology, CD4-Positive T-Lymphocytes cytology, Cell Differentiation physiology, Histone Deacetylase 1 physiology, Histone Deacetylase 2 physiology, T-Lymphocytes, Cytotoxic physiology
- Abstract
Some effector CD4+ T cell subsets display cytotoxic activity, thus breaking the functional dichotomy of CD4+ helper and CD8+ cytotoxic T lymphocytes. However, molecular mechanisms regulating CD4+ cytotoxic T lymphocyte (CD4+ CTL) differentiation are poorly understood. Here we show that levels of histone deacetylases 1 and 2 (HDAC1-HDAC2) are key determinants of CD4+ CTL differentiation. Deletions of both Hdac1 and 1 Hdac2 alleles (HDAC1cKO-HDAC2HET) in CD4+ T cells induced a T helper cytotoxic program that was controlled by IFN-γ-JAK1/2-STAT1 signaling. In vitro, activated HDAC1cKO-HDAC2HET CD4+ T cells acquired cytolytic activity and displayed enrichment of gene signatures characteristic of effector CD8+ T cells and human CD4+ CTLs. In vivo, murine cytomegalovirus-infected HDAC1cKO-HDAC2HET mice displayed a stronger induction of CD4+ CTL features compared with infected WT mice. Finally, murine and human CD4+ T cells treated with short-chain fatty acids, which are commensal-produced metabolites acting as HDAC inhibitors, upregulated CTL genes. Our data demonstrate that HDAC1-HDAC2 restrain CD4+ CTL differentiation. Thus, HDAC1-HDAC2 might be targets for the therapeutic induction of CD4+ CTLs.
- Published
- 2020
- Full Text
- View/download PDF
33. Unreported Missense Mutation in the Dimerization Domain of ADA2 Leads to ADA2 Deficiency Associated with Severe Oral Ulcers and Neutropenia in a Female Somalian Patient-Addendum to the Genotype-Phenotype Puzzle.
- Author
-
Göschl L, Winkler S, Dmytrus J, Heredia RJ, Lagler H, Ramharter M, Scheinecker C, Bonelli M, Schmetterer K, Pickl WF, Grabmeier-Pfistershammer K, Hershfield MS, Boztug K, Förster-Waldl E, and Gualdoni GA
- Subjects
- Agammaglobulinemia genetics, Dimerization, Female, Genotype, Humans, Phenotype, Severe Combined Immunodeficiency genetics, Adenosine Deaminase deficiency, Adenosine Deaminase genetics, Intercellular Signaling Peptides and Proteins deficiency, Intercellular Signaling Peptides and Proteins genetics, Mutation, Missense genetics, Neutropenia genetics, Oral Ulcer genetics
- Published
- 2020
- Full Text
- View/download PDF
34. Synthesis of a human long-term oxymetholone metabolite.
- Author
-
Kratena N, Biedermann N, Stojanovic B, Göschl L, Weil M, Enev VS, Gmeiner G, and Gärtner P
- Subjects
- Crystallography, X-Ray, Humans, Models, Molecular, Molecular Conformation, Oxymetholone chemistry, Oxymetholone chemical synthesis, Oxymetholone metabolism
- Abstract
A long-term metabolite of the doping agent oxymetholone (OXM-M2, 17β-hydroxymethyl-2,17α-methyl-18-norandrost-13-en-3-one) which has been identified by GC-MS/MS was synthesized from commercially available materials. Two efficient synthetic routes to access both C-17 epimers of tentative metabolites were developed. The identity and molecular configuration of the in vivo metabolite: 17β-hydroxymethyl-2α,17α-methyl-18-norandrost-13-en-3-one was confirmed by single crystal X-ray diffraction., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
35. Treg cells in autoimmunity: from identification to Treg-based therapies.
- Author
-
Göschl L, Scheinecker C, and Bonelli M
- Subjects
- Animals, Autoimmune Diseases etiology, Autoimmune Diseases immunology, Autoimmune Diseases metabolism, Autoimmune Diseases therapy, Cell Differentiation immunology, Cell Plasticity immunology, Epigenesis, Genetic, Humans, Immunomodulation, Immunotherapy, Adoptive, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocytes, Regulatory metabolism, Autoimmunity, T-Lymphocytes, Regulatory immunology
- Abstract
Regulatory (Treg) cells are key regulators of inflammation and important for immune tolerance and homeostasis. A major progress has been made in the identification and classification of Treg cells. Due to technological advances, we have gained deep insights in the epigenetic regulation of Treg cells. The use of fate reporter mice allowed addressing the functional consequences of loss of Foxp3 expression. Depending on the environment Treg cells gain effector functions upon loss of Foxp3 expression. However, the traditional view that Treg cells become necessarily pathogenic by gaining effector functions was challenged by recent findings and supports the notion of Treg cell lineage plasticity. Treg cell stability is also a major issue for Treg cell therapies. Clinical trials are designed to use polyclonal Treg cells as therapeutic tools. Here, we summarize the role of Treg cells in selected autoimmune diseases and recent advances in the field of Treg targeted therapies.
- Published
- 2019
- Full Text
- View/download PDF
36. Response to letter to the editor: "Comments on Unambiguous identification and characterization of a long-term human metabolite of dehydrochloromethyltestosterone".
- Author
-
Forsdahl G, Geisendorfer T, Göschl L, Pfeffer S, Gärtner P, Thevis M, and Gmeiner G
- Subjects
- Humans, Amines, Methyltestosterone metabolism
- Published
- 2019
- Full Text
- View/download PDF
37. CCR6 controls autoimmune but not innate immunity-driven experimental arthritis.
- Author
-
Bonelli M, Puchner A, Göschl L, Hayer S, Niederreiter B, Steiner G, Tillmann K, Plasenzotti R, Podesser B, Georgel P, Smolen J, Scheinecker C, and Blüml S
- Subjects
- Animals, Arthritis, Experimental immunology, Arthritis, Experimental pathology, Arthritis, Rheumatoid immunology, Arthritis, Rheumatoid pathology, Autoimmune Diseases immunology, Autoimmune Diseases pathology, Chemokine CCL20 immunology, Humans, Immunity, Innate genetics, Mice, Receptors, CCR6 immunology, Synovial Membrane immunology, Synovial Membrane pathology, Arthritis, Experimental genetics, Arthritis, Rheumatoid genetics, Autoimmune Diseases genetics, Chemokine CCL20 genetics, Receptors, CCR6 genetics
- Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease, characterized by synovial infiltration of various inflammatory cells. Chemokines are involved in controlling the recruitment of different cell types into the synovial membrane. The role of CCR6 in the development of arthritis so far remains unclear. In this study, we investigated the role of CCR6 in the pathogenesis of arthritis using three different murine arthritis models. Compared to WT animals, CCR6
-/- mice developed less clinical signs of arthritis in the collagen-induced arthritis model but not in the K/BxN serum transfer arthritis model and in the human tumour necrosis factor transgenic arthritis model, suggesting a defect in adaptive effector functions but intact innate effector functions in the development of arthritis in CCR6-/- animals. In line with this, anti-collagen antibody levels were significantly reduced in CCR6-/- mice compared with WT mice. Moreover, we demonstrate enhanced osteoclastogenesis in vitro in CCR6-/- mice compared with WT mice. However, we did not detect differences in bone mass under steady state conditions in vivo between WT and CCR6-deficient mice. These data suggest that CCR6 is crucially involved in adaptive but not in innate immunity-driven arthritis. CCR6 or its chemokine ligand CCL20 might represent a possible new target for the treatment of RA., (© 2018 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.)- Published
- 2018
- Full Text
- View/download PDF
38. Unambiguous identification and characterization of a long-term human metabolite of dehydrochloromethyltestosterone.
- Author
-
Forsdahl G, Geisendorfer T, Göschl L, Pfeffer S, Gärtner P, Thevis M, and Gmeiner G
- Abstract
In doping control analysis, the characterization of urinary steroid metabolites is of high interest for a targeted and long-term detection of prohibited anabolic androgenic steroids (AAS). In this work, the structure of a long-term metabolite of dehydrochloromethyltestosterone (DHCMT) was elucidated. Altogether, 8 possible metabolites with a 17α-methyl-17β-hydroxymethyl - structures were synthesized and compared to a major DHCMT long-term metabolite detected in reference urine excretion samples. The confirmed structure of the metabolite was 4α-chloro-18-nor-17β-hydroxymethyl-17α-methyl-5α-androst-13-en-3α-ol., (Copyright © 2018 John Wiley & Sons, Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
39. A T cell-specific deletion of HDAC1 protects against experimental autoimmune encephalomyelitis.
- Author
-
Göschl L, Preglej T, Hamminger P, Bonelli M, Andersen L, Boucheron N, Gülich AF, Müller L, Saferding V, Mufazalov IA, Hirahara K, Seiser C, Matthias P, Penz T, Schuster M, Bock C, Waisman A, Steiner G, and Ellmeier W
- Subjects
- Animals, Cell Movement, Cells, Cultured, Chimera, Disease Models, Animal, Encephalomyelitis, Autoimmune, Experimental immunology, Histone Deacetylase 1 genetics, Humans, Mice, Mice, Knockout, Multiple Sclerosis immunology, Receptors, CCR4 metabolism, Receptors, CCR6 metabolism, STAT1 Transcription Factor genetics, Encephalomyelitis, Autoimmune, Experimental metabolism, Histone Deacetylase 1 metabolism, Multiple Sclerosis metabolism, STAT1 Transcription Factor metabolism, Th17 Cells physiology
- Abstract
Multiple sclerosis (MS) is a human neurodegenerative disease characterized by the invasion of autoreactive T cells from the periphery into the CNS. Application of pan-histone deacetylase inhibitors (HDACi) ameliorates experimental autoimmune encephalomyelitis (EAE), an animal model for MS, suggesting that HDACi might be a potential therapeutic strategy for MS. However, the function of individual HDAC members in the pathogenesis of EAE is not known. In this study we report that mice with a T cell-specific deletion of HDAC1 (using the Cd4-Cre deleter strain; HDAC1-cKO) were completely resistant to EAE despite the ability of HDAC1cKO CD4
+ T cells to differentiate into Th17 cells. RNA sequencing revealed STAT1 as a prominent upstream regulator of differentially expressed genes in activated HDAC1-cKO CD4+ T cells and this was accompanied by a strong increase in phosphorylated STAT1 (pSTAT1). This suggests that HDAC1 controls STAT1 activity in activated CD4+ T cells. Increased pSTAT1 levels correlated with a reduced expression of the chemokine receptors Ccr4 and Ccr6, which are important for the migration of T cells into the CNS. Finally, EAE susceptibility was restored in WT:HDAC1-cKO mixed BM chimeric mice, indicating a cell-autonomous defect. Our data demonstrate a novel pathophysiological role for HDAC1 in EAE and provide evidence that selective inhibition of HDAC1 might be a promising strategy for the treatment of MS., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
40. The AMP analog AICAR modulates the Treg/Th17 axis through enhancement of fatty acid oxidation.
- Author
-
Gualdoni GA, Mayer KA, Göschl L, Boucheron N, Ellmeier W, and Zlabinger GJ
- Subjects
- Adenosine Monophosphate metabolism, Aminoimidazole Carboxamide pharmacology, Animals, Cell Cycle drug effects, Cell Proliferation drug effects, Hypoglycemic Agents pharmacology, Interleukin-2 metabolism, Metabolic Networks and Pathways drug effects, Metformin pharmacology, Oxidation-Reduction, T-Lymphocytes, Regulatory metabolism, Th17 Cells metabolism, Aminoimidazole Carboxamide analogs & derivatives, Fatty Acids metabolism, Lipid Metabolism drug effects, Ribonucleotides pharmacology, T-Lymphocytes, Regulatory drug effects, Th17 Cells drug effects
- Abstract
T cells must tightly regulate their metabolic processes to cope with varying bioenergetic demands depending on their state of differentiation. The metabolic sensor AMPK is activated in states of low energy supply and modulates cellular metabolism toward a catabolic state. Although this enzyme is known to be particularly active in regulatory T (T
reg ) cells, its impact on T helper (Th )-cell differentiation is poorly understood. We investigated the impact of several AMPK activators on Treg -cell differentiation and found that the direct activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide), but not the indirect activators metformin and 2-deoxyglucose, strongly enhanced Treg -cell induction by specifically enhancing Treg -cell expansion. Conversely, Th 17 generation was impaired by the agent. Further investigation of the metabolic background of our observations revealed that AICAR enhanced both cellular mitochondrogenesis and fatty acid uptake. Consistently, increased Treg induction was entirely reversible on inhibition of fatty acid oxidation, thus confirming the dependence of AICAR's effects on metabolic pathways alterations. Translating our findings to an in vivo model, we found that the substance enhanced Treg cell generation on IL-2 complex-induced immune stimulation. We provide a previously unrecognized insight into the delicate interplay between immune cell function and metabolism and delineate a potential novel strategy for metabolism-targeting immunotherapy.-Gualdoni, G. A., Mayer, K. A., Göschl, L., Boucheron, N., Ellmeier, W., Zlabinger, G. J. The AMP analog AICAR modulates the Treg /Th 17 axis through enhancement of fatty acid oxidation., (© FASEB.)- Published
- 2016
- Full Text
- View/download PDF
41. Abatacept (CTLA-4Ig) treatment reduces T cell apoptosis and regulatory T cell suppression in patients with rheumatoid arthritis.
- Author
-
Bonelli M, Göschl L, Blüml S, Karonitsch T, Hirahara K, Ferner E, Steiner CW, Steiner G, Smolen JS, and Scheinecker C
- Subjects
- Abatacept therapeutic use, Antirheumatic Agents therapeutic use, Apoptosis immunology, Arthritis, Rheumatoid drug therapy, CD4-Positive T-Lymphocytes drug effects, Cells, Cultured, Female, Humans, Immune Tolerance drug effects, Immunophenotyping, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Male, Middle Aged, fas Receptor immunology, Abatacept pharmacology, Antirheumatic Agents pharmacology, Apoptosis drug effects, Arthritis, Rheumatoid immunology, T-Lymphocyte Subsets drug effects, T-Lymphocytes, Regulatory drug effects
- Abstract
Objective: Abatacept (CTLA-4Ig) blocks CD28-mediated T cell activation by binding to the costimulatory B7 ligands CD80/CD86 on antigen presenting cells. Costimulatory molecules, however, can also be expressed on T cells upon activation. Therefore, the aim of our study was to investigate direct effects of CTLA-4Ig on distinct T cell subsets in RA patients., Methods: Phenotypic and functional analyses of CD4(+) T cells, including CD4(+) FoxP3(+) CD25(+) regulatory T cells (Treg), from RA patients were performed before and during CTLA-4Ig therapy. In addition T cells from healthy volunteers were analysed on in vitro culture with CTLA-4Ig or anti-CD80 and anti-CD86 antibodies. Apoptotic DNA fragmentation in CD4(+) and CD4(+) FoxP3(+) T cells was measured by TUNEL staining., Results: We observed an increase in T cells, including Treg cells, after initiation of CTLA-4Ig therapy, which was linked to a downregulation of activation-associated marker molecules and CD95 on CD4(+) T cells and Treg cells. CTLA-4Ig decreased CD95-mediated cell death in vitro in a dose-dependent manner. Functional analysis of isolated Treg cells from RA patients further revealed a diminished suppression of responder T cell proliferation. This was found to be due to CTLA-4Ig-mediated blocking of CD80 and CD86 on responder T cells that led to a diminished susceptibility for Treg cell suppression., Conclusion: CTLA-4Ig therapy in RA patients exerts effects beyond the suppression of T cell activation, which has to be taken into account as an additional mechanism of CTLA-4Ig treatment., (© The Author 2015. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
42. Rapid multiplex analysis of lipid raft components with single-cell resolution.
- Author
-
Schatzlmaier P, Supper V, Göschl L, Zwirzitz A, Eckerstorfer P, Ellmeier W, Huppa JB, and Stockinger H
- Subjects
- Humans, Jurkat Cells, Membrane Microdomains genetics, Membrane Proteins genetics, Cell Differentiation, Flow Cytometry methods, Membrane Microdomains metabolism, Membrane Proteins metabolism, Mutation, Signal Transduction
- Abstract
Lipid rafts, a distinct class of highly dynamic cell membrane microdomains, are integral to cell homeostasis, differentiation, and signaling. However, their quantitative examination is challenging when working with rare cells, developmentally heterogeneous cell populations, or molecules that only associate weakly with lipid rafts. We present a fast biochemical method, which is based on lipid raft components associating with the nucleus upon partial lysis during centrifugation through nonionic detergent. Requiring little starting material or effort, our protocol enabled the multidimensional flow cytometric quantitation of raft-resident proteins with single-cell resolution, thereby assessing the membrane components from a few cells in complex cell populations, as well as their dynamics resulting from cell signaling, differentiation, or genetic mutation., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
43. HDAC1 controls CD8+ T cell homeostasis and antiviral response.
- Author
-
Tschismarov R, Firner S, Gil-Cruz C, Göschl L, Boucheron N, Steiner G, Matthias P, Seiser C, Ludewig B, and Ellmeier W
- Subjects
- Animals, Antiviral Agents therapeutic use, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Proliferation, Hyaluronan Receptors metabolism, Interferon-gamma metabolism, Interleukin-2 metabolism, Ionomycin pharmacology, Lymphocytic Choriomeningitis drug therapy, Lymphocytic Choriomeningitis immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Tumor Necrosis Factor-alpha metabolism, Antiviral Agents pharmacology, CD8-Positive T-Lymphocytes drug effects, Histone Deacetylase 1 metabolism
- Abstract
Reversible lysine acetylation plays an important role in the regulation of T cell responses. HDAC1 has been shown to control peripheral T helper cells, however the role of HDAC1 in CD8+ T cell function remains elusive. By using conditional gene targeting approaches, we show that LckCre-mediated deletion of HDAC1 led to reduced numbers of thymocytes as well as peripheral T cells, and to an increased fraction of CD8+CD4- cells within the CD3/TCRβlo population, indicating that HDAC1 is essential for the efficient progression of immature CD8+CD4- cells to the DP stage. Moreover, CD44hi effector CD8+ T cells were enhanced in mice with a T cell-specific deletion of HDAC1 under homeostatic conditions and HDAC1-deficient CD44hi CD8+ T cells produced more IFNγ upon ex vivo PMA/ionomycin stimulation in comparison to wild-type cells. Naïve (CD44l°CD62L+) HDAC1-null CD8+ T cells displayed a normal proliferative response, produced similar amounts of IL-2 and TNFα, slightly enhanced amounts of IFNγ, and their in vivo cytotoxicity was normal in the absence of HDAC1. However, T cell-specific loss of HDAC1 led to a reduced anti-viral CD8+ T cell response upon LCMV infection and impaired expansion of virus-specific CD8+ T cells. Taken together, our data indicate that HDAC1 is required for the efficient generation of thymocytes and peripheral T cells, for proper CD8+ T cell homeostasis and for an efficient in vivo expansion and activation of CD8+ T cells in response to LCMV infection.
- Published
- 2014
- Full Text
- View/download PDF
44. CD4⁺CD25⁻Foxp3⁺ T cells: a marker for lupus nephritis?
- Author
-
Bonelli M, Göschl L, Blüml S, Karonitsch T, Steiner CW, Steiner G, Smolen JS, and Scheinecker C
- Subjects
- Adult, Female, Flow Cytometry, Forkhead Transcription Factors immunology, Humans, Immunophenotyping, Male, Middle Aged, Phenotype, Biomarkers blood, CD4-Positive T-Lymphocytes immunology, Lupus Nephritis immunology, T-Lymphocyte Subsets immunology
- Abstract
Introduction: Systemic lupus erythematosus (SLE) is a heterogenous autoimmune disease, which can affect different organs. Increased proportions of CD4⁺CD25-Foxp3⁺ T cells have been described in SLE patients. The exact role of this cell population in SLE patients still remains unclear. We therefore analyzed this T cell subset in a large cohort of SLE patients with different organ manifestations., Methods: Phenotypic analyses, proportions and absolute cell numbers of CD4⁺CD25-Foxp3⁺ T cells were determined by flow cytometry (FACS) in healthy controls (HC) (n = 36) and SLE patients (n = 61) with different organ manifestations. CD4⁺CD25⁻Foxp3⁺ T cells were correlated with clinical data, the immunosuppressive therapy and different disease activity indices. In patients with active glomerulonephritis, CD4⁺CD25⁻Foxp3⁺ T cells were analyzed in urine sediment samples. Time course analyses of CD4⁺CD25⁻Foxp3⁺ T cells were performed in patients with active disease activity before and after treatment with cyclophosphamide and prednisone., Results: CD4⁺CD25⁻Foxp3⁺ T cells were significantly increased in active SLE patients and the majority expressed Helios. Detailed analysis of this patient cohort revealed increased proportions of CD4⁺CD25⁻Foxp3⁺ T cells in SLE patients with renal involvement. CD4⁺CD25⁻Foxp3⁺ T cells were also detected in urine sediment samples of patients with active glomerulonephritis and correlated with the extent of proteinuria., Conclusion: CD4⁺CD25⁻Foxp3⁺ T cells resemble regulatory rather than activated T cells. Comparative analysis of CD4⁺CD25⁻Foxp3⁺ T cells in SLE patients revealed a significant association of this newly described cell population with active nephritis. Therefore CD4⁺CD25⁻Foxp3⁺ T cells might serve as an important tool to recognize and monitor SLE patients with renal involvement.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.