23 results on '"A. Rius Rigau"'
Search Results
2. OP0166 SKIN RETICULARIZATION IN SYSTEMIC SCLEROSIS - POTENTIAL MEDIATORS
- Author
-
Chenguiti Fakhouri, S., primary, Zhu, H., additional, Ronicke, M., additional, Dees, C., additional, Konstantinidis, L., additional, Rius Rigau, A., additional, Matei, A. E., additional, LI, Y. N., additional, Eckstein, M., additional, Geppert, C., additional, Ludolph, I., additional, Kreuter, A., additional, Sticherling, M., additional, Berking, C., additional, Schett, G., additional, Horch, R., additional, Distler, J., additional, and Bergmann, C., additional
- Published
- 2024
- Full Text
- View/download PDF
3. OP0178 IMAGING MASS CYTOMETRY-BASED CHARACTERIZATION OF FIBROBLASTS SUBSETS, THEIR NETWORKS AND SPATIAL MICROENVIRONMENT IN SYSTEMIC SCLEROSIS
- Author
-
Rius Rigau, A., primary, Liang, M., additional, Filla, T., additional, Devakumar, V., additional, Matei, A. E., additional, Györfi, A. H., additional, Schett, G., additional, Distler, J., additional, and Li, Y. N., additional
- Published
- 2024
- Full Text
- View/download PDF
4. AB1141 THERAPEUTIC POTENTIAL OF INHIBITION OF JAK1 AND 2 BY RUXOLITINIB, OF ROCK2 BY BELUMOSUDIL, AND OF BTK BY IBRUTINIB IN SYSTEMIC SCLEROSIS
- Author
-
Hong, X., primary, Xiao, Y., additional, Györfi, A. H., additional, Xu, L., additional, Shen, L., additional, Neelagar, R., additional, Devakumar, V., additional, Filla, T., additional, Trinh-Minh, T., additional, Liang, M., additional, Rius Rigau, A., additional, Distler, J., additional, and Matei, A. E., additional
- Published
- 2024
- Full Text
- View/download PDF
5. Combined inhibition of IL-1, IL-33 and IL-36 signalling by targeting IL1RAP ameliorates skin and lung fibrosis in preclinical models of systemic sclerosis
- Author
-
Grönberg, Caitríona, primary, Rattik, Sara, additional, Tran-Manh, Cuong, additional, Zhou, Xiang, additional, Rius Rigau, Aleix, additional, Li, Yi-Nan, additional, Györfi, Andrea-Hermina, additional, Dickel, Nicholas, additional, Kunz, Meik, additional, Kreuter, Alexander, additional, Matei, Emil-Alexandru, additional, Zhu, Honglin, additional, Skoog, Petter, additional, Liberg, David, additional, Distler, Jörg HW, additional, and Trinh-Minh, Thuong, additional
- Published
- 2024
- Full Text
- View/download PDF
6. Characterization of Vascular Niche in Systemic Sclerosis by Spatial Proteomics
- Author
-
Rius Rigau, Aleix, primary, Li, Yi-Nan, additional, Matei, Alexandru-Emil, additional, Györfi, Andrea-Hermina, additional, Bruch, Peter-Martin, additional, Koziel, Sarah, additional, Devakumar, Veda, additional, Gabrielli, Armando, additional, Kreuter, Alexander, additional, Wang, Jiucun, additional, Dietrich, Sascha, additional, Schett, Georg, additional, Distler, Jörg H.W., additional, and Liang, Minrui, additional
- Published
- 2024
- Full Text
- View/download PDF
7. Mouse Models of Skin Fibrosis
- Author
-
Rius Rigau, Aleix, primary, Luber, Markus, additional, and Distler, Jörg H. W., additional
- Published
- 2021
- Full Text
- View/download PDF
8. OP0106 SYNOVIAL IMPRINTING OF SKIN-DERIVED IMMUNE CELLS INITIATES SPREADING OF INFLAMMATION FROM SKIN TO JOINT IN PSORIATIC ARTHRITIS
- Author
-
Raimondo, M. G., primary, Rauber, S., additional, Mohammadian, H., additional, Vogg, M., additional, Xu, C., additional, Rius Rigau, A., additional, Luber, M., additional, Labinsky, H., additional, Soare, A., additional, Distler, J., additional, Fearon, U., additional, Veale, D., additional, Sticherling, M., additional, Cañete, J. D. D., additional, Schett, G., additional, and Ramming, A., additional
- Published
- 2023
- Full Text
- View/download PDF
9. POS0450 TEMPORAL MIGRATION OF IMMUNE CELLS FROM PSORIATIC SKIN TO JOINTS INITIATING SYNOVIAL INFLAMMATION IN PSORIATIC ARTHRITIS
- Author
-
Raimondo, M. G., primary, Rauber, S., additional, Xu, C., additional, Mohammadian, H., additional, Vogg, M., additional, Anchang, C. G., additional, Rius Rigau, A., additional, Luber, M., additional, Labinsky, H., additional, Soare, A., additional, Distler, J. H. W., additional, Fearon, U., additional, Veale, D., additional, Sticherling, M., additional, Cañete, J. D. D., additional, Schett, G., additional, and Ramming, A., additional
- Published
- 2022
- Full Text
- View/download PDF
10. Mouse Models of Skin Fibrosis
- Author
-
Aleix, Rius Rigau, Markus, Luber, and Jörg H W, Distler
- Subjects
Bleomycin ,Disease Models, Animal ,Mice ,Scleroderma, Systemic ,Animals ,Graft vs Host Disease ,Humans ,Fibrosis ,Skin - Abstract
Systemic sclerosis (SSc) is a rare systemic autoimmune disease associated with a high mortality. The first histopathological hallmarks are vasculopathy and inflammation, followed by fibrosis of the skin and internal organs. The molecular and cellular mechanisms are incompletely understood. Rodent models provide important insights into the pathogenesis of SSc and are a mainstay for the development of novel targeted therapies. Here we describe the mechanistic insights of inducible and genetic models, and also discuss in detail the limitations and pitfalls of the most frequently used SSc mouse models. We also describe protocols for running the established bleomycin-induced scleroderma skin fibrosis model.
- Published
- 2021
11. La venda d'animals a la Rambla de Barcelona
- Author
-
Torres Roviralta, Marta., Ruiz Revelles, Alba., and Rius Rigau, Aleix.
- Subjects
Veterinaris Investigació Ètica professional ,Animals Indústria i comerç Investigació - Abstract
Treball presentat a la Facultat de Veterinària de la Universitat Autònoma de Barcelona. Treball presentat a l'assignatura de Deontologia i Veterinària Legal (21223)
- Published
- 2021
12. Mouse Models of Skin Fibrosis
- Author
-
Jörg H W Distler, Markus Luber, and Aleix Rius Rigau
- Subjects
030203 arthritis & rheumatology ,0301 basic medicine ,integumentary system ,business.industry ,Inflammation ,medicine.disease ,Bleomycin ,Scleroderma ,Systemic autoimmune disease ,Pathogenesis ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Fibrosis ,Immunology ,Genetic model ,medicine ,medicine.symptom ,skin and connective tissue diseases ,business - Abstract
Systemic sclerosis (SSc) is a rare systemic autoimmune disease associated with a high mortality. The first histopathological hallmarks are vasculopathy and inflammation, followed by fibrosis of the skin and internal organs. The molecular and cellular mechanisms are incompletely understood. Rodent models provide important insights into the pathogenesis of SSc and are a mainstay for the development of novel targeted therapies. Here we describe the mechanistic insights of inducible and genetic models, and also discuss in detail the limitations and pitfalls of the most frequently used SSc mouse models. We also describe protocols for running the established bleomycin-induced scleroderma skin fibrosis model.
- Published
- 2021
- Full Text
- View/download PDF
13. POS0450 TEMPORAL MIGRATION OF IMMUNE CELLS FROM PSORIATIC SKIN TO JOINTS INITIATING SYNOVIAL INFLAMMATION IN PSORIATIC ARTHRITIS
- Author
-
M. G. Raimondo, S. Rauber, C. Xu, H. Mohammadian, M. Vogg, C. G. Anchang, A. Rius Rigau, M. Luber, H. Labinsky, A. Soare, J. H. W. Distler, U. Fearon, D. Veale, M. Sticherling, J. D. D. Cañete, G. Schett, and A. Ramming
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundSpreading of inflammation from skin to joint is a key question behind the pathogenesis of psoriatic arthritis (PsA). Psoriasis (PsO), being one of the most prevalent skin diseases, usually anticipates joint manifestations, suggesting spreading of skin to joint disease, which happens in about 30% of the patients with psoriasis.1 To date, it is still obscure why the inflammatory process in some patients with PsO remains restrained to the skin, whereas in other patients it extents to tendons and joints.ObjectivesUsing a pre-clinical model of PsA, we aimed to unveil the skin-joint axis, i.e. the spreading of psoriatic inflammation from the skin to the joints.MethodsKAEDE transgenic mice expressing a photo-convertible fluorescent reporter were used to assess cell trafficking from inflamed skin to other organs in the mouse model of IL-23 overexpression (IL-23OE) induced PsA. Psoriatic skin lesions were irradiated with UV light to trigger the photoswitch from KAEDEGREEN to KAEDERED. Migration to different organs was determined by flow cytometry. Imaging flow cytometry was used to characterize the type of cells migrating from the skin to the joints. Migrating cells were further characterized by single-cell RNA-sequencing (scRNAseq) and functional analyses.ResultsMRI imaging and histological evaluation of IL-23OE mice revealed skin inflammation preceding joint inflammation in both wild-type and KAEDE-transgenic mice. Specific leukocyte migration from the skin to the joints started shortly after the onset of skin inflammation and before onset of inflammation within the joints of KAEDE transgenic mice. No migration was observed in healthy control animals. Other organs such as spleen or lymph nodes showed no model-dependent migration. Imaging flow cytometry revealed that the cells migrating to the joints were predominantly CD45+ CD11b+ cells. ScRNAseq analysis of sorted KAEDERED cells from inflamed joints confirmed that approximately 80% of the migrating cells were macrophages. Differential gene expression and pathway analysis revealed an imbalance between pro- and anti-inflammatory macrophages in the joints of experimental psoriatic arthritis.ConclusionWe describe IL-23-mediated migration of skin-derived macrophages from the skin to the joints during the onset of experimental psoriatic arthritis. This process may explain the spreading from psoriatic skin to joint disease as these cells foster the development by local cytokine production once arrived in the joints.References[1]Veale, D.J. & Fearon, U. The pathogenesis of psoriatic arthritis. Lancet391, 2273-2284 (2018).Disclosure of InterestsNone declared.
- Published
- 2022
- Full Text
- View/download PDF
14. POS0328 ENGRAILED 1 COORDINATES CYTOSKELETAL ORGANIZATION TO PROMOTE MYOFIBROBLAST DIFFERENTIATION AND FIBROTIC TISSUE REMODELING
- Author
-
Györfi, A. H., primary, Matei, A. E., additional, Fuchs, M., additional, Rius Rigau, A., additional, Hong, X., additional, Honglin, Z., additional, Luber, M., additional, Bergmann, C., additional, Dees, C., additional, Ludolph, I., additional, Horch, R., additional, Distler, O., additional, Schett, G., additional, Kunz, M., additional, and Distler, J. H. W., additional
- Published
- 2021
- Full Text
- View/download PDF
15. Engrailed 1 coordinates cytoskeletal reorganization to induce myofibroblast differentiation
- Author
-
Bertram Bengsch, Raymund E. Horch, Meik Kunz, Markus Luber, Georg Schett, Maximilian Fuchs, Alexandru-Emil Matei, Aleix Rius Rigau, Xuezhi Hong, Ingo Ludolph, Jörg H W Distler, Christina Bergmann, Chunguang Liang, Oliver Distler, Honglin Zhu, Clara Dees, Andrea-Hermina Györfi, and Jiucun Wang
- Subjects
0301 basic medicine ,Adult ,Male ,Cytoskeleton organization ,Immunology ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Transforming Growth Factor beta ,Gene expression ,medicine ,Immunology and Allergy ,Animals ,Humans ,Cytoskeleton ,Fibroblast ,Myofibroblasts ,Aged ,Skin ,030203 arthritis & rheumatology ,Homeodomain Proteins ,Mice, Knockout ,rho-Associated Kinases ,Scleroderma, Systemic ,Chemistry ,Cell Differentiation ,Middle Aged ,Cell biology ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Case-Control Studies ,Female ,Signal transduction ,Myofibroblast ,Transforming growth factor - Abstract
Transforming growth factor-β (TGFβ) is a key mediator of fibroblast activation in fibrotic diseases, including systemic sclerosis. Here we show that Engrailed 1 (EN1) is reexpressed in multiple fibroblast subpopulations in the skin of SSc patients. We characterize EN1 as a molecular amplifier of TGFβ signaling in myofibroblast differentiation: TGFβ induces EN1 expression in a SMAD3-dependent manner, and in turn, EN1 mediates the profibrotic effects of TGFβ. RNA sequencing demonstrates that EN1 induces a profibrotic gene expression profile functionally related to cytoskeleton organization and ROCK activation. EN1 regulates gene expression by modulating the activity of SP1 and other SP transcription factors, as confirmed by ChIP-seq experiments for EN1 and SP1. Functional experiments confirm the coordinating role of EN1 on ROCK activity and the reorganization of cytoskeleton during myofibroblast differentiation, in both standard fibroblast culture systems and in vitro skin models. Consistently, mice with fibroblast-specific knockout of En1 demonstrate impaired fibroblast-to-myofibroblast transition and are partially protected from experimental skin fibrosis.
- Published
- 2020
16. SAT0302 INNATE LYMPHOID CELLS INDUCE A FIBROTIC PHENOTYPE OF FIBROBLASTS
- Author
-
Weber, S., primary, Anchang, C. G., additional, Rauber, S., additional, Luber, M., additional, Raimondo, M. G., additional, Ariza, Y., additional, Rius Rigau, A., additional, Kreuter, A., additional, Schett, G., additional, Distler, J., additional, and Ramming, A., additional
- Published
- 2020
- Full Text
- View/download PDF
17. SAT0354 TYPE 3 INNATE LYMPHOID CELLS ARE KEY DRIVERS OF PSORIATIC ARTHRITIS
- Author
-
Raimondo, M. G., primary, Rauber, S., additional, Luber, M., additional, Rius Rigau, A., additional, Weber, S., additional, Anchang, C. G., additional, Agarwal, R., additional, Soare, A., additional, Sticherling, M., additional, Rech, J., additional, Kleyer, A., additional, Distler, J., additional, Schett, G., additional, and Ramming, A., additional
- Published
- 2020
- Full Text
- View/download PDF
18. SAT0354 TYPE 3 INNATE LYMPHOID CELLS ARE KEY DRIVERS OF PSORIATIC ARTHRITIS
- Author
-
Andreas Ramming, Alina Soare, Michael Sticherling, Stefanie Weber, J. H. W. Distler, Markus Luber, Jürgen Rech, A. Rius Rigau, Simon Rauber, Maria Gabriella Raimondo, C. G. Anchang, Rahul Agarwal, Georg Schett, and A. Kleyer
- Subjects
medicine.medical_specialty ,business.industry ,Minimal disease ,Immunology ,Curve analysis ,High dimensional ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Peripheral blood ,Disease activity ,Psoriatic arthritis ,Rheumatology ,Internal medicine ,Healthy volunteers ,medicine ,Immunology and Allergy ,In patient ,business - Abstract
Background:Psoriasis (PsO) and psoriatic arthritis (PsA) are two types of chronic inflammatory diseases that share a similar cytokines profile. About 30% of PsO patients also develop a joint involvement, but the underlying mechanism is still unclear. Innate lymphoid cells (ILC) and specifically the type 3 ILCs (ILC3s) have raised increasing interest as possible player in the pathogenesis of both diseases, as they produce the pathological key cytokine IL-17A.Objectives:We addressed the contribution of ILC3s to the pathogenesis of PsO and PsA in patients as well as murinein vivomodels.Methods:130 patients satisfying the Classification Criteria for Psoriatic Arthritis (CASPAR), 40 patients with PsO and 35 healthy volunteers were enrolled in the study. Information regarding clinical features, laboratory parameters were collected and psoriasis area severity index (PASI), disease activity score 28 (DAS28), disease activity in psoriatic arthritis (DAPSA), minimal disease activity score (MDA) were calculated. Magnetic resonance imaging (MRI) and high-resolution peripheral CT (HR-pQCT) were taken and PsA MRI score (PsAMRIS) was assessed. Flow cytometric analysis was performed and IFNγ-producing ILC1s, IL-4/IL-5-producing ILC2s and IL-17/IL-22-producing ILC3s were identified among ILCs. Multivariate linear regression and Receiver-Operating Characteristic (ROC) Curve analysis was performed using the IBM SPSS Statistics software. Different in vivo models were used to assess functional implications of ILCs at different time points of the disease. Joint inflammation was assessed through MRI and H&E staining of ankle areas. Peripheral blood was obtained from mice of each group and flow cytometry analysis was performed. High dimensional analyses including RNA-seq was performed to identify phenotypic characteristics of ILCs implemented into the pathogenesis of the disease.Results:Total number of circulating ILCs were increased in PsA patients compared to PsO and healthy controls (pConclusion:ILC3s not only correlate with various facets of PsA manifestations but also functionally contribute to synovitis and enthesitis suggesting them as interesting target for upcoming treatment strategies in the near future.Disclosure of Interests:Maria Gabriella Raimondo Grant/research support from: Celgene, Partner Fellowship, Simon Rauber: None declared, Markus Luber: None declared, Aleix Rius Rigau: None declared, Stefanie Weber: None declared, Charles Gwellem Anchang: None declared, Rahul Agarwal: None declared, Alina Soare: None declared, Michael Sticherling Grant/research support from: Novartis, Consultant of: Advisory boards Abbvie, Celgene, Janssen Cilag, Lilly, Pfizer, MSD, Novartis, Amgen, Leo, Sanofi, UCB, Speakers bureau: Abbvie, Celgene, Janssen Cilag, Leo, MSD, Novartis, Pfizer, Jürgen Rech Consultant of: BMS, Celgene, Novartis, Roche, Chugai, Speakers bureau: AbbVie, Biogen, BMS, Celgene, MSD, Novartis, Roche, Chugai, Pfizer, Lilly, Arnd Kleyer Consultant of: Lilly, Gilead, Novartis,Abbvie, Speakers bureau: Novartis, Lilly, Jörg Distler Grant/research support from: Boehringer Ingelheim, Consultant of: Boehringer Ingelheim, Paid instructor for: Boehringer Ingelheim, Speakers bureau: Boehringer Ingelheim, Georg Schett Speakers bureau: AbbVie, BMS, Celgene, Janssen, Eli Lilly, Novartis, Roche and UCB, Andreas Ramming Grant/research support from: Pfizer, Novartis, Consultant of: Boehringer Ingelheim, Novartis, Gilead, Pfizer, Speakers bureau: Boehringer Ingelheim, Roche, Janssen
- Published
- 2020
- Full Text
- View/download PDF
19. POS0328 ENGRAILED 1 COORDINATES CYTOSKELETAL ORGANIZATION TO PROMOTE MYOFIBROBLAST DIFFERENTIATION AND FIBROTIC TISSUE REMODELING
- Author
-
X. Hong, Georg Schett, A. E. Matei, Clara Dees, Zhu Honglin, O. Distler, J. H. W. Distler, A. Rius Rigau, Andrea-Hermina Györfi, Christina Bergmann, Raymund E. Horch, Markus Luber, Ingo Ludolph, Meik Kunz, and Maximilian Fuchs
- Subjects
Gene knockdown ,Cytoskeleton organization ,business.industry ,Immunology ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Microtubule polymerization ,Extracellular matrix ,medicine.anatomical_structure ,Rheumatology ,Fibrosis ,medicine ,Cancer research ,Immunology and Allergy ,Skin equivalent ,business ,Fibroblast ,Myofibroblast - Abstract
Background:Engrailed 1 (EN1) is a homeodomain-containing transcription factor with essential roles in embryonic development. In most cell types, the expression of EN1 is restricted to embryonic development. However, under pathological conditions, EN1 can be re-expressed to promote phenotypical adaptation. En1 is transiently expressed in the developing dermis of murine embryos in a distinct fibroblast lineage and silenced before birth (1). Former EN1-expressing cells give rise to a subpopulation of fibroblasts that has a high capacity for extracellular matrix production in adult murine skin. The role of EN1 in systemic sclerosis (SSc) was previously not explored.Objectives:To study the role of EN1 in the pathological activation of fibroblasts in tissue fibrosis.Methods:Bulk RNA-Seq and EN1 or SP1 ChIP-Seq were performed from cultured human dermal fibroblasts. The expression of EN1 was inhibited by siRNA. Cytoskeletal drugs paclitaxel, vinblastin and ROCK inhibitor (Y27632) were used to modulate the cytoskeleton in EN1 knockdown or overexpressing dermal fibroblasts. The role of EN1 in fibroblast activation was evaluated by functional experiments with EN1 knockdown or overexpression in standard 2D culture systems as well as in 3D skin equivalent models. The role of EN1 in skin fibrosis was further studied in En1fl/fl X Col6Cre mice, with fibroblast-specific knockout of En1 in three complementary mouse models: overexpression of a constitutively active TGFß-receptor I (TBRICA), bleomycin-induced skin fibrosis and TSK1 mice.Results:Pathologically activated dermal fibroblasts from SSc patients express higher levels of EN1 compared with age and sex matched healthy individuals in the skin and in vitro. TGFβ induces EN1 expression in fibroblasts in a SMAD3-dependent manner both in cultured fibroblasts and in murine skin. Knockdown of EN1 prevents TGFβ-induced fibroblast activation, whereas overexpression of EN1 fosters the pro-fibrotic effects of TGFβ with increased expression of αSMA, stress fibers and collagen. RNA sequencing demonstrates that EN1 induces a pro-fibrotic gene expression profile functionally related to cytoskeleton organization and ROCK activation. In silico analyses of the promoters of En1 target genes coupled with siRNA-mediated knockdown demonstrated that EN1 regulates these pro-fibrotic target genes by modulating the activity of regulatory modules that contain transcription factors of the specificity protein (SP) family. Functional experiments with selective modulators of ROCK and of microtubule polymerization confirm the coordinating role of EN1 on ROCK activity and the re-organization of cytoskeleton during myofibroblast differentiation in both conventional culture systems and 3D skin equivalents. Consistently, mice with fibroblast-specific knockout of En1 demonstrate impaired fibroblast-to-myofibroblast transition, reduced dermal thickening and impaired collagen deposition in the TBRICA, bleomycin-induced and TSK1 models.Conclusion:We characterize the homeodomain transcription factor EN1 as a molecular amplifier of TGFβ signaling in myofibroblast differentiation that coordinates cytoskeletal organization in a SP-dependent manner. EN1 might thus be a novel candidate for molecular targeted therapies to interfere with myofibroblast differentiation in fibrotic diseases.References:[1]Rinkevich Y, Walmsley GG, Hu MS, Maan ZN, Newman AM, Drukker M, et al. Skin fibrosis. Identification and isolation of a dermal lineage with intrinsic fibrogenic potential. Science. 2015;348(6232):aaa2151.Disclosure of Interests:Andrea-Hermina Györfi: None declared, Alexandru-Emil Matei: None declared, Maximilian Fuchs: None declared, Aleix Rius Rigau: None declared, Xuezhi Hong: None declared, ZHU Honglin: None declared, Markus Luber: None declared, Christina Bergmann: None declared, Clara Dees: None declared, Ingo Ludolph: None declared, Raymund Horch: None declared, Oliver Distler Consultant of: Actellion, AbbVie, Acceleron Pharma, Anamar, Amgen, Blade Therapeutics, CSL Behring, ChemomAb, Ergonex, Glenmark Pharma, GSK, Inventiva, Italfarmaco, iQvia, Medac, Medscape, Lilly, Sanofi, Target BioScience, UCB, Bayer, Boehringer Ingelheim, Catenion, iQone, Menarini, Mepha, Novartis, Mitsubishi, MSD, Roche, Pfizer, Georg Schett: None declared, Meik Kunz: None declared, Jörg H.W. Distler Consultant of: Actelion, Active Biotech, Anamar, ARXX, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, JB Therapeutics, Medac, Pfizer, RuiYi and UCB., Grant/research support from: Anamar, Active Biotech, Array Biopharma, aTyr, BMS, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, Novartis, Sanofi-Aventis, RedX, UCB
- Published
- 2021
- Full Text
- View/download PDF
20. SAT0302 INNATE LYMPHOID CELLS INDUCE A FIBROTIC PHENOTYPE OF FIBROBLASTS
- Author
-
Y. Ariza, Stefanie Weber, Andreas Ramming, J. H. W. Distler, Simon Rauber, Markus Luber, Maria Gabriella Raimondo, Alexander Kreuter, A. Rius Rigau, Georg Schett, and C. G. Anchang
- Subjects
JAG1 ,business.industry ,Immunology ,Innate lymphoid cell ,Notch signaling pathway ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Idiopathic pulmonary fibrosis ,medicine.anatomical_structure ,Rheumatology ,Fibrosis ,medicine ,Immunology and Allergy ,Progenitor cell ,Fibroblast ,business ,Tissue homeostasis - Abstract
Background:Fibrotic diseases are characterized by excessive extracellular matrix production as a result of immune-mediated permanent fibroblast activation. Innate lymphoid cells type II (ILC2) are an only recently discovered cell type involved in barrier integrity and tissue homeostasis. There is upcoming evidence that ILC2s play a central role in mediating fibrotic diseases.Objectives:The aim of the study was to further elucidate the role of ILC2s in fibrotic tissue remodeling and fibroblast activation.Methods:Skin biopsies of patients with systemic sclerosis (SSc) or sclerodermatous chronic graft versus host disease (scGvHD) as well as lung biopsies of patients with idiopathic pulmonary fibrosis (IPF) were analyzed by immunofluorescence (IF) staining. Single cell RNA-sequencing (scRNA-seq) was performed on ILCs from fibrotic skin and lung of bleomycin-challenged mice. Further characterization of ILC2 phenotypes in fibrosis models was done by flow cytometry.In vitroculture of fibroblasts and ILC2s was used to study cellular interaction and fibrotic activation. Quantitative realtime-PCR, western blot, IF staining and ELISA were used as readouts.Results:Two different subtypes of ILC2s were found in skin of SSc and scGvHD patients as well as in lungs of IPF patients with one subpopulation being particularly increased in fibrotic tissue. Single cell RNA-sequencing confirmed the existence of two major populations of ILC2s in experimental fibrosis. One subtype showed features of immature ILC2 progenitors and was actively recruited from the bone marrow during fibrotic tissue remodeling. The other ILC2 subset was highly activated and expressed pro-fibrotic cytokines. These profibrotic ILC2s directly interacted with fibroblasts in a cell contact dependent manner. Semaphorin 4A (SEMA4A) expressed by ILC2s bound to Plexin D1 (PLXND1) on fibroblasts. This interaction resulted into fibrotic imprinting with high expression levels of the transcription factor PU.1 which was recently described as central regulator of the pro-fibrotic gene expression program (Wohlfahrt et al. 2019). Signaling through Jagged 1 (JAG1) and Notch receptor 2 (NOTCH2) was identified as a second mechanism of interaction between fibroblasts and ILC2s. JAG1 expressed by fibroblasts activated NOTCH2 signaling in ILC2s which emphazised the secretion of pro-fibrotic cytokines.Conclusion:We identified a bidirectional interaction between ILCs and fibroblasts incorporating a vicious circle of fibrotic tissue remodelling. As ILCs are still not accessible as therapeutic targets these results might contribute to the development of new strategies for anti-fibrotic therapies.References:[1]Wohlfahrt, Thomas, Simon Rauber, Steffen Uebe, Markus Luber, Alina Soare, Arif Ekici, Stefanie Weber, Alexandru-Emil Matei, Chih-Wei Chen, Christiane Maier, Emmanuel Karouzakis, Hans P. Kiener, Elena Pachera, Clara Dees, Christian Beyer, Christoph Daniel, Kolja Gelse, Andreas E. Kremer, Elisabeth Naschberger, Michael Stürzl, Falk Butter, Michael Sticherling, Susetta Finotto, Alexander Kreuter, Mark H. Kaplan, Astrid Jüngel, Steffen Gay, Stephen L. Nutt, David W. Boykin, Gregory M. K. Poon, Oliver Distler, Georg Schett, Jörg H. W. Distler, and Andreas Ramming. 2019. ‘PU.1 controls fibroblast polarization and tissue fibrosis’,Nature, 566: 344-49.Disclosure of Interests:Stefanie Weber: None declared, Charles Gwellem Anchang: None declared, Simon Rauber: None declared, Markus Luber: None declared, Maria Gabriella Raimondo Grant/research support from: Celgene, Partner Fellowship, Yuko Ariza Employee of: Ono Pharmaceutical Co., Ltd., Aleix Rius Rigau: None declared, Alexander Kreuter: None declared, Georg Schett Speakers bureau: AbbVie, BMS, Celgene, Janssen, Eli Lilly, Novartis, Roche and UCB, Jörg Distler Grant/research support from: Boehringer Ingelheim, Consultant of: Boehringer Ingelheim, Paid instructor for: Boehringer Ingelheim, Speakers bureau: Boehringer Ingelheim, Andreas Ramming Grant/research support from: Pfizer, Novartis, Consultant of: Boehringer Ingelheim, Novartis, Gilead, Pfizer, Speakers bureau: Boehringer Ingelheim, Roche, Janssen
- Published
- 2020
- Full Text
- View/download PDF
21. Mutual Amplification of GLI2/Hedgehog and Transcription Factor JUN/AP‐1 Signaling in Fibroblasts in Systemic Sclerosis: Potential Implications for Combined Therapies.
- Author
-
Bergmann, Christina, Chenguiti Fakhouri, Sara, Trinh‐Minh, Thuong, Filla, Tim, Rius Rigau, Aleix, Ekici, Arif B., Merlevede, Benita, Hallenberger, Ludwig, Zhu, Honglin, Dees, Clara, Matei, Alexandru‐Emil, Auth, Janina, Györfi, Andrea‐Hermina, Zhou, Xiang, Rauber, Simon, Bozec, Aline, Dickel, Nicholas, Liang, Chunguang, Kunz, Meik, and Schett, Georg
- Abstract
Objective Methods Results Conclusion Deregulation of the cJUN/AP‐1 and hedgehog/GLI2 signaling pathways has been implicated in fibroblast activation in systemic sclerosis (SSc). However, the consequences of their concomitant up‐regulation are unknown. Here, we tested the hypothesis that mutual amplification of both pathways might drive persistent fibroblast activation.Cultured fibroblasts and skin sections of patients with diffuse SSc and healthy volunteers were analyzed. cJUN/AP‐1 signaling and hedgehog/GLI2 signaling were inhibited using knockdown and pharmacologic approaches. Hedgehog signaling was activated in mice by fibroblast‐specific overexpression of constitutively active Smoothened.cJUN and GLI2 are concomitantly up‐regulated and colocalize in fibroblasts of patients with SSc compared to healthy controls. Activation of hedgehog/GLI2 signaling induces the expression of cJUN in vitro and in vivo, whereas inactivation of GLI2 inhibits cJUN expression. Likewise, inactivation of cJUN impairs the expression of GLI2. This mutual regulation occurs at the level of transcription with binding of cJUN and GLI2 to specific binding motifs. Interference with this mutual amplification of cJUN signaling and GLI2 signaling inhibits fibroblast activation and collagen release: Inhibition of cJUN/AP‐1 signaling ameliorates hedgehog‐induced fibroblast activation and skin fibrosis in SmoACT mice with a reduction of skin thickness of 103% (P = 0.0043) in the treatment group compared to the fibrotic control group. Moreover, combined pharmacologic inhibition of cJUN/AP‐1 and hedgehog/GLI2 exerts additive antifibrotic effects in a model of TGFβ‐driven experimental fibrosis (TBRACT mice).The transcription factors cJUN and GLI2 reinforce each other's activity to promote fibroblast activation in SSc. Interruption of this crosstalk by combined inhibition of both pathways exerts additive antifibrotic effects at well‐tolerated doses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Imaging mass cytometry-based characterisation of fibroblast subsets and their cellular niches in systemic sclerosis.
- Author
-
Rius Rigau A, Liang M, Devakumar V, Neelagar R, Matei AE, Györfi AH, Bergmann C, Filla T, Fedorchenko V, Schett G, Distler JHW, and Li YN
- Abstract
Objectives: Transcriptomic data demonstrated that fibroblasts are heterogeneous with functionally diverse subpopulations. Although fibroblasts are key effector cells of fibrotic diseases such as systemic sclerosis (SSc), they have not yet been characterised spatially at the cellular level. Here, we aimed to investigate fibroblast subpopulations using imaging mass cytometry (IMC) as a proteomic-based, spatially resolved omics approach., Methods: We applied IMC to deconvolute the heterogeneity of 49 969 cells including 6501 fibroblasts at the single-cell level, to analyse their spatial distribution and to characterise their cellular niches in skin sections of patients with SSc and controls in situ., Results: We identified 13 different subpopulations of fibroblasts in SSc and control skin, the proportion increases in five fibroblast subpopulations (myofibroblasts, FAP
high , S1PR+ , Thy1+ ;ADAM12high ;PU.1high and ADAM12+ ;GLI1+ fibroblasts) and decreases in three subpopulations (TFAMhigh , PI16+ ;FAP+ and Thy1+ ;ADAM12low fibroblasts). Several fibroblast subpopulations demonstrated spatial enrichment and altered cellular interactions in SSc. The proportion of S1PR+ -fibroblast positively correlated with more extensive skin fibrosis, whereas high numbers of PI16+ ;FAP- -fibroblasts were associated with milder skin fibrosis. The frequency of aberrant cellular interaction between S1PR+ and ADAM12+ ;GLI1+ -fibroblasts also positively associated with the extent of skin fibrosis in SSc., Conclusion: Using IMC, we demonstrated profound changes in composition and localisation of the majority of fibroblast subpopulations in SSc skin. These findings may provide a rationale for specific targeting of deregulated fibroblast subpopulations in SSc. Quantification of S1PR+ -fibroblast and PI16+ ;FAP- -fibroblasts may offer potential for patient stratification according to severity of skin fibrosis., Competing Interests: Competing interests: A-HG received lecture fees from Boehringer Ingelheim and Abbvie. JHWD has consultancy relationships and/or has received research funding from AbbVie, Actelion, BMS, Celgene, Bayer Pharma, Boehringer Ingelheim, JB Therapeutics, Sanofi-Aventis, Novartis, UCB, GSK, Array Biopharma and Active Biotech in the area of potential treatments of SSc, is CEO of 4D Science and Scientific lead/Co-CEO of FibroCure. A-HG has received research founding from Boehringer Ingelheim. The project was supported by an unrestricted grant from the Hiller Foundation., (© 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 on behalf of EULAR.)- Published
- 2024
- Full Text
- View/download PDF
23. CD19-CAR T-cell therapy induces deep tissue depletion of B cells.
- Author
-
Tur C, Eckstein M, Velden J, Rauber S, Bergmann C, Auth J, Bucci L, Corte G, Hagen M, Wirsching A, Grieshaber-Bouyer R, Reis P, Kittan N, Wacker J, Rius Rigau A, Ramming A, D'Agostino MA, Hartmann A, Müller F, Mackensen A, Bozec A, Schett G, and Raimondo MG
- Abstract
Objectives: CD19-targeting chimeric antigen receptor (CAR) T-cell therapy can induce long-term drug-free remission in patients with autoimmune diseases (AIDs). The efficacy of CD19-CAR T-cell therapy is presumably based on deep tissue depletion of B cells; however, such effect has not been proven in humans in vivo., Methods: Sequential ultrasound-guided inguinal lymph node biopsies were performed at baseline and after CD19-CAR T-cell therapy in patients with AIDs. Results were compared with lymph node biopsies from rituximab (RTX)-treated AID patients with absence of peripheral B cells. Conventional and immunohistochemistry staining were performed on lymph node tissue to assess architecture as well the number of B cells, follicular dendritic cells (FDCs), plasma cells, T cells and macrophages., Results: Sequential lymph node biopsies were analysed from five patients with AID before and after CD19-CAR T-cell therapy and from five patients with AID after RTX treatment. In addition, non-lymphoid organ biopsies (colon, kidney and gallbladder) from three additional patients with AID after CD19-CAR T-cell therapy were analysed. CD19
+ and CD20+ B cells were completely depleted in the lymph nodes after CD19-CAR T-cell therapy, but not after RTX treatment. Plasma cells, T cells and macrophages in the lymph nodes remained unchanged. Follicular structures were disrupted and FDCs were depleted in the lymph nodes after CD19-CAR T-cell therapy, but not after RTX. Non-lymphoid organs were completely depleted of B cells., Discussion: This study demonstrates complete B-cell depletion in secondary lymphoid tissues of patients with AIDs following CD19-CAR T-cell therapy combined with standard lymphodepleting therapy., Competing Interests: Competing interests: GS has received speaker honoraria from BMS, Cabaletta, Janssen, Kyverna, Miltenyi and Novartis. AM has received speaker honoraria and consulting fees from BMS/Celgene, Kite/Gilead, Novartis, BioNTech, Miltenyi Biomedicine, Century Therapeutics. MADA has received grants or contracts from Amgen, Abbvie, UCB, Pfizer, J&J and Galapagos and speaker honoraria and consulting fees from Abbvie, Amgen, Novartis, BMS, UCB, J&J, Biogen, MSD, Lilly and Galapagos. FM has received speaker honoraria and consulting fees from AstraZeneca, Kite/Gilead, Novartis, Sobi, BMS, Miltenyi, Janssen, BNT., (© 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 on behalf of EULAR.)- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.