430 results on '"Dees C"'
Search Results
202. Purification of the Scrapie Agent by Density Gradient Centrifugation
- Author
-
Marsh, R. F., primary, Dees, C., additional, Castle, B. E., additional, Wade, W. F., additional, and German, T. L., additional
- Published
- 1984
- Full Text
- View/download PDF
203. Effects of Different Methods of Purification on Aggregation of Scrapie Infectivity
- Author
-
Castle, B. E., primary, Dees, C., additional, German, T. L., additional, and Marsh, R. F., additional
- Published
- 1987
- Full Text
- View/download PDF
204. Interactions of multilamellar phospholipid vesicles with bovine lymphocytes: Effects of α-tocopherol on lymphocyte blastogenesis
- Author
-
Fountain, M.W., primary, Dees, C., additional, Weete, J.D., additional, and Schultz, R.D., additional
- Published
- 1982
- Full Text
- View/download PDF
205. An ELISA test to detect antibody to
- Author
-
Dees, C., primary, Fountain, M.W., additional, Panangala, V.S., additional, Swango, L.J., additional, and Schultz, R.D., additional
- Published
- 1982
- Full Text
- View/download PDF
206. Inactivation of the Scrapie Agent by Ultraviolet Irradiation in the Presence of Chlorpromazine
- Author
-
Dees, C., primary, Wade, W. F., additional, German, T. L., additional, and Marsh, R. F., additional
- Published
- 1985
- Full Text
- View/download PDF
207. Incorporation of a follicle stimulating hormone used for embryo transfer in cattle into multilamellar liposomes
- Author
-
Dees, C., primary, Stringfellow, D., additional, and Schultz, R.D., additional
- Published
- 1984
- Full Text
- View/download PDF
208. Localization of quantitation of the ornithine lipid of Thiobacillus thiooxidans
- Author
-
Dees, C, primary and Shively, J M, additional
- Published
- 1982
- Full Text
- View/download PDF
209. Characterization of Proteins in Membrane Vesicles from Scrapie-infected Hamster Brain
- Author
-
Dees, C., primary, German, T. L., additional, Wade, W. F., additional, and Marsh, R. F., additional
- Published
- 1985
- Full Text
- View/download PDF
210. Enhanced intraphagocytic killing of in bovine mononuclear cells by liposomes-containing gentamicin
- Author
-
Dees, C., primary, Fountain, M.W., additional, Taylor, J.R., additional, and Schultz, R.D., additional
- Published
- 1985
- Full Text
- View/download PDF
211. Characterization of Lipids in Membrane Vesicles from Scrapie-infected Hamster Brain
- Author
-
Dees, C., primary, German, T. L., additional, Wade, W. F., additional, and Marsh, R. F., additional
- Published
- 1985
- Full Text
- View/download PDF
212. Enhanced intraphagocytic killing of [formula omitted] in bovine mononuclear cells by liposomes-containing gentamicin
- Author
-
Dees, C., Fountain, M.W., Taylor, J.R., and Schultz, R.D.
- Published
- 1985
- Full Text
- View/download PDF
213. Hyperphosphorylation of p53 induced by benzene, toluene, and chloroform
- Author
-
Dees, C. and Travis, C.
- Published
- 1994
- Full Text
- View/download PDF
214. Trichloroacetate stimulation of liver DNA synthesis in male and female mice
- Author
-
Dees, C. and Travis, C.
- Published
- 1994
- Full Text
- View/download PDF
215. The mitogenic potential of trichloroethylene in B6C3F1 mice
- Author
-
Dees, C. and Travis, C.
- Published
- 1993
- Full Text
- View/download PDF
216. Persistent cAMP-signals triggered by internalized G-protein-coupled receptors
- Author
-
Christian Dees, Martin J. Lohse, Davide Calebiro, Viacheslav O. Nikolaev, Tiziana de Filippis, Maria Cristina Gagliani, Carlo Tacchetti, Luca Persani, Calebiro, D., Nikolaev, V. O., Gagliani, M. C., DE FILIPPIS, T., Dees, C., Tacchetti, Carlo, Persani, L., and Lohse, M. J.
- Subjects
Thyroid Gland ,Thyrotropin ,environment and public health ,Cell Biology/Cell Signaling ,Diabetes and Endocrinology/Thyroid ,Adenylyl cyclase ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cyclic AMP ,Biology (General) ,Organic Chemicals ,Internalization ,Receptor ,Cells, Cultured ,media_common ,0303 health sciences ,Microscopy, Confocal ,General Neuroscience ,Receptors, Thyrotropin ,GTP-Binding Protein alpha Subunits ,3. Good health ,Cell biology ,Second messenger system ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,General Agricultural and Biological Sciences ,hormones, hormone substitutes, and hormone antagonists ,Adenylyl Cyclases ,Signal Transduction ,Subcellular Fractions ,Research Article ,QH301-705.5 ,media_common.quotation_subject ,Mice, Transgenic ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Cell surface receptor ,Animals ,Humans ,Protein kinase A ,030304 developmental biology ,G protein-coupled receptor ,Fluorescent Dyes ,General Immunology and Microbiology ,Epithelial Cells ,chemistry ,030217 neurology & neurosurgery - Abstract
Real-time monitoring of G-protein-coupled receptor (GPCR) signaling in native cells suggests that the receptor for thyroid stimulating hormone remains active after internalization, challenging the current model for GPCR signaling., G-protein–coupled receptors (GPCRs) are generally thought to signal to second messengers like cyclic AMP (cAMP) from the cell surface and to become internalized upon repeated or prolonged stimulation. Once internalized, they are supposed to stop signaling to second messengers but may trigger nonclassical signals such as mitogen-activated protein kinase (MAPK) activation. Here, we show that a GPCR continues to stimulate cAMP production in a sustained manner after internalization. We generated transgenic mice with ubiquitous expression of a fluorescent sensor for cAMP and studied cAMP responses to thyroid-stimulating hormone (TSH) in native, 3-D thyroid follicles isolated from these mice. TSH stimulation caused internalization of the TSH receptors into a pre-Golgi compartment in close association with G-protein αs-subunits and adenylyl cyclase III. Receptors internalized together with TSH and produced downstream cellular responses that were distinct from those triggered by cell surface receptors. These data suggest that classical paradigms of GPCR signaling may need revision, as they indicate that cAMP signaling by GPCRs may occur both at the cell surface and from intracellular sites, but with different consequences for the cell., Author Summary Cells respond to many environmental cues through the activity of cell surface receptor proteins, which sense these cues and convey that information to signaling molecules inside the cell. G-protein–coupled receptors (GPCRs) form the largest eukaryotic family of plasma membrane receptors. They convert the information provided by extracellular stimuli into intracellular second messengers, like cyclic AMP (cAMP). After prolonged stimulation, they are internalized inside cells, an event that to date has been thought to terminate the production of second messengers. Though many of the key steps of GPCR signaling are known in detail, precisely how signaling and termination actually occur in time and space (i.e., in subcellular compartments or microdomains) is still largely unexplored. To observe GPCR signaling in living cells, we generated mice expressing a fluorescent sensor that allows monitoring the intracellular levels of cAMP with a microscope. We utilized this system to study, directly in native thyroid follicles, the signal sent by the receptor for thyroid-stimulating hormone (TSH). Our findings indicate that TSH receptors are internalized rapidly after activation but continue to stimulate cAMP production inside cells and that this sustained, cAMP production is apparently required for localized activation of downstream components. These data challenge the current model of the GPCR-cAMP pathway by suggesting the existence of previously unrecognized intracellular site(s) for cAMP generation and of differential signaling outcomes as a result of intracellular GPCR signaling. Such intracellular site(s) may provide specialized signaling platforms, thus contributing to the spatiotemporal regulation of cAMP production and to signaling specificity within the GPCR family.
- Published
- 2009
217. CD19-targeting CAR T-cell therapy in patients with diffuse systemic sclerosis: a case series.
- Author
-
Auth J, Müller F, Völkl S, Bayerl N, Distler JHW, Tur C, Raimondo MG, Chenguiti Fakhouri S, Atzinger A, Coppers B, Eckstein M, Liphardt AM, Bäuerle T, Tascilar K, Aigner M, Kretschmann S, Wirsching A, Taubmann J, Hagen M, Györfi AH, Kharboutli S, Krickau T, Dees C, Spörl S, Rothe T, Harrer T, Bozec A, Grieshaber-Bouyer R, Fuchs F, Kuwert T, Berking C, Horch RE, Uder M, Mackensen A, Schett G, and Bergmann C
- Subjects
- Humans, Female, Middle Aged, Male, Adult, Treatment Outcome, Lung Diseases, Interstitial therapy, Lung Diseases, Interstitial immunology, Receptors, Chimeric Antigen immunology, Immunotherapy, Adoptive methods, Scleroderma, Diffuse therapy, Scleroderma, Diffuse immunology, Antigens, CD19 immunology
- Abstract
Background: CD19-targeting chimeric antigen receptor (CAR) T-cell therapy has shown remarkable outcomes in patients with systemic lupus erythematosus. The effects of CD19-targeting CAR T cells on organ manifestations in patients with systemic sclerosis have yet to be characterised. B cells have a central role in the pathogenesis of systemic sclerosis. We present a detailed analysis of the effects of CD19-targeting CAR T-cell therapy in patients with systemic sclerosis., Methods: Six patients with severe diffuse systemic sclerosis with an insufficient response to at least two treatments were consecutively recruited at the Department of Internal Medicine 3, University Hospital Erlangen (Erlangen, Germany) to receive CD19-targeting CAR T-cell treatment (1 × 10
6 CAR T cells per kg bodyweight). Events were predefined by progression of interstitial lung disease, onset of congestive heart failure, onset of renal failure, onset of arterial hypertension, or initiation of new immunosuppressive or antifibrotic therapy. Event-free time or treatment intensification after study entry was the primary outcome. Key secondary outcomes included changes in the modified Rodnan Skin Score (mRSS), imaging (a component of the assessment of lung fibrosis), laboratory assessments, patient-reported outcomes, and a modified version of the American College of Rheumatology Composite Response Index in Systemic Sclerosis (ACR-CRISS), assessed at baseline, 3 months, 6 months, 9 months, and 12 months., Findings: Between April 20, 2022, and Nov 8, 2023, six patients with severe diffuse systemic sclerosis (median age 42 years [IQR 36-53]; four men and two women; all White European) were recruited and received CD19-targeted CAR T-cell therapy. The median follow-up time was 487 days (IQR 342-585). No events occurred within the observational period. Probability of improvement in the ACR-CRISS score increased to a median of 100% (IQR 100-100) at 6 months. Median mRSS decreased by 31% (IQR 29-38), corresponding to a median of 8 points (IQR 7-13) within 100 days. The extent of disease on CT scan decreased by a median of 4% (IQR 3-4) due to reduction of ground-glass opacities while the reticular pattern remained stable. Forced vital capacity improved by a median of 195 mL (IQR 18-275) at the latest observational timepoint., Interpretation: We provide the first evidence that CD19-targeting CAR T-cell therapy might intercept with the progression of fibrotic organ manifestations in patients with systemic sclerosis., Funding: Deutsche Forschungsgemeinschaft, Deutsche Krebshilfe, ELAN-Foundation Erlangen, IZKF Erlangen, and Bundesministerium für Bildung und Forschung., Competing Interests: Declaration of interests JA and ChB received a travel grant from Kyverna therapeutics. ChB received a research grant from Boehringer Ingelheim and speaker fees from Novartis. CaB received consulting fees from Almirall, Delcath, BMS, Immunocore, Pierre Fabre, MSD, Novartis, Regeneron, and Sanofi; honoraria from Almirall, Leo Pharma, BMS, Pierre Fabre, and MSD; travel support from Pierre Fabre; and participates on the data safety monitoring or advisory board of Miltenyi Biotech and InflaRx. FM received a research grant from Kite/Gilead and consulting fees from AbbVie, ArgoBio, AstraZeneca, BMS, Crispr Therapeutics, Janssen, Kite, and Novartis; honoraria from AbbVie, ArgoBio, AstraZeneca, BMS, Crispr Therapeutics, Janssen, Kite, Kyverna, Miltenyi, Novartis, and Sobi; and participates on the BMS and Biontech data safety monitoring or advisory board. TKr received grants, consulting fees, honoraria, and travel support from Novartis and Pfizer, as well as consulting fees and honoraria from Kiowa Kirin, payment for expert testimony from Novartis, and travel support from AbbVie. GS received honoraria from Cabaletta, Novartis, Janssen, and Kyverna. SoK received honoraria from BMS and Sobi, as well as travel support from Janssen, BMS, Sobi, Novartis, and Kite/Gilead. JHWD has consultancy relationships with and is part of the speaker or advisory board of AbbVie, Active Biotech, Anamar, ARXX, AstraZeneca, Bayer Pharma, Boehringer Ingelheim, Calliditas Therapeutics, Celgene, Galapagos, Genentech, GSK, Inventiva, Janssen, Novartis, Pfizer, Roche, and UCB; has received research funding from Anamar, Argenx, ARXX, BMS, Bayer Pharma, Boehringer Ingelheim, Cantargia, Celgene, CSL Behring, Galapagos, GSK, Inventiva, Kiniksa, Lassen, Sanofi-Aventis, RedX, and UCB; travel support from AbbVie and SOBI; and is Chief Executive Officer of 4D Science and Scientific Lead of FibroCure. AM received grants from Miltenyi Biomedicine and Kyverna, and consulting fees from BMS/Celgene, KITE/Gilead, Novartis, BioNTech, Miltenyi Biomedicine, and Century Therapeutics. AM received honoraria from BMS/Celgene, KITE/Gilead, Novartis, BioNTech, Miltenyi Biomedicine, and Century Therapeutics, and travel support from AbbVie and Janssen. RGB received grants from Kyverna and travel support from BMS and Novartis. MA received grants, honoraria, payment for expert testimony, travel support and equipment from Miltenyi Biotec; and received consulting fees, honoraria and travel support from Miltenyi Biomedicine. All other authors declare no competing interests., (Copyright © 2025 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
218. Phase II study of MEK inhibitor trametinib alone and in combination with AKT inhibitor GSK2141795/uprosertib in patients with metastatic triple negative breast cancer.
- Author
-
Prasath V, Boutrid H, Wesolowski R, Abdel-Rasoul M, Timmers C, Lustberg M, Layman RM, Macrae E, Mrozek E, Shapiro C, Glover K, Vater M, Budd GT, Harris L, Isaacs C, Dees C, Perou CM, Johnson GL, Poklepovic A, Chen H, Villalona-Calero M, Carson W, Stover DG, and Ramaswamy B
- Subjects
- Humans, Female, Middle Aged, Aged, Adult, Proto-Oncogene Proteins c-akt metabolism, Neoplasm Metastasis, Treatment Outcome, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors administration & dosage, Aged, 80 and over, Diamines, Pyrazoles, Pyridones therapeutic use, Pyridones administration & dosage, Pyrimidinones therapeutic use, Pyrimidinones administration & dosage, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms mortality, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
Purpose: While MEK inhibitors demonstrated activity in metastatic triple negative breast cancer (mTNBC) preclinical studies, preclinical, and clinical studies implicate rapid development of resistance limiting clinical benefit. The purpose of this study was to determine response rate for Trametinib alone and in combination with Uprosertib in patients with mTNBC previously treated with chemotherapy., Methods: This was an open-label, two-part, phase II, single-arm, multicenter study. Patients first received Trametinib monotherapy (2 mg daily; Part I) then at progression transitioned to Trametinib (1.5 mg) plus Uprosertib (50 mg; Part II)., Results: Between October 2013 and January 2017, 37 patients were enrolled to Part I. Subsequently, 19 patients entered Part II. Of the 37 patients receiving Trametinib monotherapy, 2 patients achieved partial response (PR) for an ORR of 5.4% (2/37) and an additional 6/37 (16.2%) achieved stable disease (SD). The clinical benefit rate (PR+SD) for patients receiving monotherapy was 21.6% (8/37). Of the 19 patients in Part II, 3 patients achieved PR for an ORR to Part II of 15.8% (3/19) and an additional 3 achieved SD. Median progression-free survival (PFS) was 7.7 weeks for Part I and 7.8 weeks for Part II. Circulating tumor DNA (ctDNA) clearance at C2D1 of Trametinib monotherapy was associated with improved PFS and overall survival., Conclusion: In patients with mTNBC, Trametinib monotherapy demonstrated limited efficacy and addition of Uprosertib was associated with numerically greater objective responses but no difference in PFS. Translational analyses suggest ctDNA clearance as a potential early biomarker of response., Competing Interests: Declarations. Conflict of interest: Authors have no conflicts of interest related to the current analyses., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
219. Combination therapies of porcupine inhibition with ruxolitinib, ibrutinib or belumosudil in murine sclerodermatous GvHD.
- Author
-
Hong X, Xiao Y, Xu L, Shen L, Neelagar R, Devakumar V, Trinh-Minh T, Liang M, Rius Rigau A, Zhang Y, Li YN, Dees C, Györfi AH, Wolff D, Herr W, Schett G, Distler JHW, and Matei AE
- Published
- 2025
- Full Text
- View/download PDF
220. Mutual Amplification of GLI2/Hedgehog and Transcription Factor JUN/AP-1 Signaling in Fibroblasts in Systemic Sclerosis: Potential Implications for Combined Therapies.
- Author
-
Bergmann C, Chenguiti Fakhouri S, Trinh-Minh T, Filla T, Rius Rigau A, Ekici AB, Merlevede B, Hallenberger L, Zhu H, Dees C, Matei AE, Auth J, Györfi AH, Zhou X, Rauber S, Bozec A, Dickel N, Liang C, Kunz M, Schett G, and Distler JHW
- Subjects
- Humans, Animals, Mice, Skin metabolism, Up-Regulation, Cells, Cultured, Female, Smoothened Receptor genetics, Smoothened Receptor metabolism, Case-Control Studies, Nuclear Proteins, Fibroblasts metabolism, Zinc Finger Protein Gli2 genetics, Zinc Finger Protein Gli2 metabolism, Scleroderma, Systemic metabolism, Scleroderma, Systemic genetics, Scleroderma, Systemic pathology, Hedgehog Proteins metabolism, Signal Transduction physiology, Transcription Factor AP-1 metabolism, Proto-Oncogene Proteins c-jun metabolism
- Abstract
Objective: 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., Methods: 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., Results: 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 Smo
ACT 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)., Conclusion: 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., (© 2024 The Author(s). Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)- Published
- 2025
- Full Text
- View/download PDF
221. Disturbed spatial WNT activation - a potential driver of the reticularized skin phenotype in Systemic Sclerosis (SSc).
- Author
-
Chenguiti Fakhouri S, Zhu H, Li YN, Ronicke M, Rigau AR, Dees C, Konstantinidis L, Schmid R, Matei AE, Eckstein M, Geppert C, Ludolph I, Kreuter A, Sticherling M, Berking C, Horch RE, Schett G, Distler JHW, and Bergmann C
- Abstract
Objectives: Little is known on the mechanisms necessary to maintain the physiological adult human skin integrity. This study aims to quantitatively describe anatomical changes in systemic sclerosis (SSc)-skin compared to controls and investigate the underlying mechanisms., Methods: Skin morphology was histologically assessed in twenty-three SSc-patients, eighteen controls and fifteen patients with hypertrophic scars. Spatial WNT/β-catenin-activation was analyzed by RNAscope and immunofluorescence-staining. Enrichment of reticular marker genes in predefined fibroblast subpopulations was done using Gene Ontology enrichment and gene set enrichment analysis., Results: SSc-skin showed a decrease in number (p<0.0001/p=0.0004), area (p<0.0001) and height (p<0.0001) of papillae compared to controls and hypertrophic scars, respectively. The expression of papillary/reticular marker genes shifted towards a reticular expression profile in SSc. On the level of previously defined fibroblast populations, the increase of reticular marker genes was particularly pronounced in the PI16+- and SFRP4+-populations (p<0.0001, respectively). Mechanistically, the expression of the WNT/β-catenin target AXIN2 and the number of fibroblasts with nuclear β-catenin-staining-pattern increased in the papillary compared to the reticular dermis in healthy skin. This polarization was lost in SSc with a two-fold increase in β-catenin-positive fibroblasts and AXIN2-expressing fibroblasts throughout the dermis (p=0.0095). Enrichment of genes related to WNT/β-catenin-regulation was found in the PI16+-population that also relocates from the reticular to the papillary dermis in SSc., Conclusions: We demonstrate an association of the "reticularized" skin phenotype in SSc with a profound loss of physiological spatial WNT/β-catenin-activation. Rescuing the spatial WNT/β-catenin-activation might help restore the physiological skin organization in future therapeutic approaches of fibrosing disorders., (This article is protected by copyright. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
222. Noncanonical WNT5A controls the activation of latent TGF-β to drive fibroblast activation and tissue fibrosis.
- Author
-
Trinh-Minh T, Chen CW, Tran Manh C, Li YN, Zhu H, Zhou X, Chakraborty D, Zhang Y, Rauber S, Dees C, Lin NY, Kah D, Gerum R, Bergmann C, Kreuter A, Reuter C, Groeber-Becker F, Eckes B, Distler O, Fabry B, Ramming A, Schambony A, Schett G, and Distler JH
- Subjects
- Fibrosis, Scleroderma, Systemic metabolism, Scleroderma, Systemic pathology, Graft vs Host Disease metabolism, Graft vs Host Disease pathology, Idiopathic Pulmonary Fibrosis metabolism, Idiopathic Pulmonary Fibrosis pathology, MAP Kinase Kinase 4 metabolism, rho-Associated Kinases metabolism, Ligands, Skin metabolism, Skin pathology, Humans, Animals, Mice, Cells, Cultured, Up-Regulation, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, Myofibroblasts metabolism, Myofibroblasts pathology, Transforming Growth Factor beta metabolism, Wnt-5a Protein metabolism, Wnt-5a Protein pharmacology, Cell Transdifferentiation
- Abstract
Transforming growth factor β (TGF-β) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-β remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-β in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-β. The activation of latent TGF-β requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-β, rebalanced TGF-β signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-β in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.
- Published
- 2024
- Full Text
- View/download PDF
223. Assessment of myocardial fibrosis in patients with systemic sclerosis using [ 68 Ga]Ga-FAPI-04-PET-CT.
- Author
-
Treutlein C, Distler JHW, Tascilar K, Fakhouri SC, Györfi AH, Atzinger A, Matei AE, Dees C, Büttner-Herold M, Kuwert T, Prante O, Bäuerle T, Uder M, Schett G, Schmidkonz C, and Bergmann C
- Subjects
- Humans, Positron Emission Tomography Computed Tomography, Contrast Media, Gadolinium, Fibrosis, Gallium Radioisotopes, Scleroderma, Systemic complications, Scleroderma, Systemic diagnostic imaging
- Abstract
Purpose: Myocardial fibrosis (MF) is a factor of poor prognosis in systemic sclerosis (SSc). Direct in-vivo visualization of fibroblast activation as early readout of MF has not been feasible to date. Here, we characterize
68 Gallium-labeled-Fibroblast-Activation-Inhibitor-04 ([68 Ga]Ga-FAPI-04)-PET-CT as a diagnostic tool in SSc-related MF., Methods: In this proof-of-concept trial, six SSc patients with and eight without MF of the EUSTAR cohort Erlangen underwent [68 Ga]Ga-FAPI-04-PET-CT and cardiac MRI (cMRI) and clinical and serologic investigations just before baseline and during follow-up between January 2020 and December 2020. Myocardial biopsy was performed as clinically indicated., Results: [68 Ga]Ga-FAPI-04 tracer uptake was increased in SSc-related MF with higher uptake in SSc patients with arrhythmias, elevated serum-NT-pro-BNP, and increased late gadolinium enhancement (LGE) in cMRI. Histologically, myocardial biopsies from cMRI- and [68 Ga]Ga-FAPI-04-positive regions confirmed the accumulation of FAP+ fibroblasts surrounded by collagen deposits. We observed similar but not equal spatial distributions of [68 Ga]Ga-FAPI-04 uptake and quantitative cMRI-based techniques. Using sequential [68 Ga]Ga-FAPI-04-PET-CTs, we observed dynamic changes of [68 Ga]Ga-FAPI-04 uptake associated with changes in the activity of SSc-related MF, while cMRI parameters remained stable after regression of molecular activity and rather indicated tissue damage., Conclusions: We present first in-human evidence that [68 Ga]Ga-FAPI-04 uptake visualizes fibroblast activation in SSc-related MF and may be a diagnostic option to monitor cardiac fibroblast activity in situ., (© 2022. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
224. Engrailed 1 coordinates cytoskeletal reorganization to induce myofibroblast differentiation.
- Author
-
Györfi AH, Matei AE, Fuchs M, Liang C, Rigau AR, Hong X, Zhu H, Luber M, Bergmann C, Dees C, Ludolph I, Horch RE, Distler O, Wang J, Bengsch B, Schett G, Kunz M, and Distler JHW
- Subjects
- Adult, Aged, Animals, Case-Control Studies, Cell Differentiation physiology, Cytoskeleton genetics, Female, Gene Expression Regulation, Homeodomain Proteins metabolism, Humans, Male, Mice, Knockout, Middle Aged, Myofibroblasts cytology, Myofibroblasts physiology, Skin pathology, Transforming Growth Factor beta metabolism, Young Adult, rho-Associated Kinases metabolism, Mice, Cytoskeleton metabolism, Homeodomain Proteins genetics, Myofibroblasts pathology, Scleroderma, Systemic pathology
- 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., Competing Interests: Disclosures: C. Bergmann reported personal fees from Boehringer Ingelheim and Pfizer during the conduct of the study. O. Distler reported personal fees from Abbvie, Acceleron Pharma, Amgen, AnaMar, Arxx Therapeutics, Baecon Discovery, Blade Therapeutics, Bayer, Böhringer Ingelheim, ChemomAb, Corbus Pharmacheuticals, CSL Behring, Galapagos NV, Glenmark Pharmaceuticals, GSK, Horizon Pharmaceuticals, Inventiva, Iqvia, Italfarmaco, Iqone, Kymera Therapeutics, Lupin Pharmaceuticals, Medac, Medscape, Mitsubishi Tanabe Pharma, MSD, Novartis, Pfizer, Roche, Roivant, Sanofi, Serodapharm, Topadur, Target Bioscience, and UCB during the conduct of the study; in addition, O. Distler had a patent to mir-29 for the treatment of systemic sclerosis issued (US8247389, EP2331143). J.H.W. Distler reported personal fees from Janssen, Anamar, ARXX, Bayer Pharma, Boehringer Ingelheim, Galapagos, GSK, Inventiva, Novartis, and UCB; grants from Anamar, ARXX, aTyr, Bayer Pharma, Boehringer Ingelheim, Cantargia, Celgene, CSL Behring, Galapagos, Inventiva, Kiniksa, and UCB outside the submitted work; and owns stock in 4D Science. No other disclosures were reported., (© 2021 Györfi et al.)
- Published
- 2021
- Full Text
- View/download PDF
225. X-linked inhibitor of apoptosis protein (XIAP) inhibition in systemic sclerosis (SSc).
- Author
-
Bergmann C, Hallenberger L, Chenguiti Fakhouri S, Merlevede B, Brandt A, Dees C, Zhu H, Zehender A, Zhou X, Schwab A, Chen CW, Györfi AH, Matei AE, Chakraborty D, Trinh-Minh T, Rauber S, Coras R, Bozec A, Kreuter A, Ziemer M, Schett G, and Distler JHW
- Subjects
- Animals, Bleomycin pharmacology, Disease Models, Animal, Fibroblasts metabolism, Fibrosis, Humans, Mice, Skin pathology, Transforming Growth Factor beta metabolism, X-Linked Inhibitor of Apoptosis Protein antagonists & inhibitors, Scleroderma, Systemic pathology, X-Linked Inhibitor of Apoptosis Protein metabolism, beta Catenin metabolism
- Abstract
Objective: X-linked inhibitor of apoptosis protein (XIAP) is a multifunctional protein with important functions in apoptosis, cellular differentiation and cytoskeletal organisation and is emerging as potential target for the treatment of various cancers. The aim of the current study was to investigate the role of XIAP in the pathogenesis of systemic sclerosis (SSc)., Methods: The expression of XIAP in human skin samples of patients with SSc and chronic graft versus host disease (cGvHD) and healthy individuals was analysed by quantitative PCR, immunofluorescence (IF) and western blot. XIAP was inactivated by siRNA-mediated knockdown and pharmacological inhibition. The effects of XIAP inactivation were analysed in cultured fibroblasts and in the fibrosis models bleomycin-induced and topoisomerase-I-(topoI)-induced fibrosis and in Wnt10b-transgenic mice., Results: The expression of XIAP, but not of other inhibitor of apoptosis protein family members, was increased in fibroblasts in SSc and sclerodermatous cGvHD. Transforming growth factor beta (TGF-β) induced the expression of XIAP in a SMAD3-dependent manner. Inactivation of XIAP reduced WNT-induced fibroblast activation and collagen release. Inhibition of XIAP also ameliorated fibrosis induced by bleomycin, topoI and overexpression of Wnt10b in well-tolerated doses. The profibrotic effects of XIAP were mediated via WNT/β-catenin signalling. Inactivation of XIAP reduces binding of β-catenin to TCF to in a TLE-dependent manner to block WNT/β-catenin-dependent transcription., Conclusions: Our data characterise XIAP as a novel link between two core pathways of fibrosis. XIAP is overexpressed in SSc and cGvHD in a TGF-β/SMAD3-dependent manner and in turn amplifies the profibrotic effects of WNT/β-catenin signalling on fibroblasts via transducin-like enhancer of split 3. Targeted inactivation of XIAP inhibits the aberrant activation of fibroblasts in murine models of SSc., Competing Interests: Competing interests: JHWD has consultancy relationships and/or has received research funding from Anamar, Active Biotech, Array Biopharma, ARXX, aTyr, BMS, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, Novartis, Sanofi-Aventis, RedX and UCB; personal fees from Actelion, Anamar, ARXX, Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, JB Therapeutics and UCB, outside the submitted work, and is stock owner of 4D Science., (© Author(s) (or their employer(s)) 2021. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
226. Targeting of canonical WNT signaling ameliorates experimental sclerodermatous chronic graft-versus-host disease.
- Author
-
Zhang Y, Shen L, Dreißigacker K, Zhu H, Trinh-Minh T, Meng X, Tran-Manh C, Dees C, Matei AE, Chen CW, Ditschkowski M, Krauss S, Winkler J, Wolff D, Ziemer M, Beilhack A, Karrer S, Herr W, Mackensen A, Schett G, Spriewald BM, and Distler JHW
- Subjects
- Animals, Anti-Bacterial Agents pharmacology, Female, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, Graft vs Host Disease etiology, Graft vs Host Disease metabolism, Graft vs Host Disease pathology, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Middle Aged, Scleroderma, Systemic etiology, Scleroderma, Systemic metabolism, Scleroderma, Systemic pathology, Graft vs Host Disease prevention & control, Hematopoietic Stem Cell Transplantation adverse effects, Pyrans pharmacology, Pyrvinium Compounds pharmacology, Scleroderma, Systemic prevention & control, Sulfones pharmacology, Triazoles pharmacology, Wnt Signaling Pathway drug effects
- Abstract
Chronic graft-versus-host disease (cGVHD) is a major life-threatening complication of allogeneic hematopoietic stem cell transplantation. The molecular mechanisms underlying cGVHD remain poorly understood, and targeted therapies for clinical use are not well established. Here, we examined the role of the canonical WNT pathway in sclerodermatous cGVHD (sclGVHD). WNT signaling was activated in human sclGVHD with increased nuclear accumulation of the transcription factor β-catenin and a WNT-biased gene expression signature in lesional skin. Treatment with the highly selective tankryase inhibitor G007-LK, the CK1α agonist pyrvinium, or the LRP6 inhibitor salinomycin abrogated the activation of WNT signaling and protected against experimental cGVHD, without a significant impact on graft-versus-leukemia effect (GVL). Treatment with G007-LK, pyrvinium, or salinomycin almost completely prevented the development of clinical and histological features in the B10.D2 (H-2d) → BALB/c (H-2d) and LP/J (H-2b) → C57BL/6 (H-2b) models of sclGVHD. Inhibition of canonical WNT signaling reduced the release of extracellular matrix from fibroblasts and reduced leukocyte influx, suggesting that WNT signaling stimulates fibrotic tissue remodeling by direct effects on fibroblasts and by indirect inflammation-dependent effects in sclGVHD. Our findings may have direct translational potential, because pyrvinium is in clinical use, and tankyrase inhibitors are in clinical trials for other indications., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
227. 68 Ga-FAPI-04 PET-CT for molecular assessment of fibroblast activation and risk evaluation in systemic sclerosis-associated interstitial lung disease: a single-centre, pilot study.
- Author
-
Bergmann C, Distler JHW, Treutlein C, Tascilar K, Müller AT, Atzinger A, Matei AE, Knitza J, Györfi AH, Lück A, Dees C, Soare A, Ramming A, Schönau V, Distler O, Prante O, Ritt P, Götz TI, Köhner M, Cordes M, Bäuerle T, Kuwert T, Schett G, and Schmidkonz C
- Abstract
Background: Interstitial lung disease (ILD) is the most common cause of death in systemic sclerosis. To date, the progression of systemic sclerosis-associated ILD is judged by the accrual of lung damage on CT and pulmonary function tests. However, diagnostic tools to assess disease activity are not available. Here, we tested the hypothesis that quantification of fibroblast activation by PET-CT using a
68 Ga-labelled selective inhibitor of prolyl endopeptidase FAP (68 Ga-FAPI-04) would correlate with ILD activity and disease progression in patients with systemic sclerosis-associated ILD., Methods: Between Sept 10, 2018, and April 8, 2020, 21 patients with systemic sclerosis-associated ILD confirmed by high-resolution CT (HRCT) within 12 months of inclusion and with onset of systemic sclerosis-associated ILD within 5 years or signs of progressive ILD and 21 controls without ILD were consecutively enrolled. All participants underwent68 Ga-FAPI-04 PET-CT imaging and standard-of-care procedures, including HRCT and pulmonary function tests at baseline. Patients with systemic sclerosis-associated ILD were followed for 6 months with HRCT and pulmonary function tests. We compared baseline68 Ga-FAPI-04 PET-CT uptake with standard diagnostic tools and predictors of ILD progression. The association of68 Ga-FAPI-04 uptake with changes in forced vital capacity was analysed using mixed-effects models. Follow-up68 Ga-FAPI-04 PET-CT scans were obtained in a subset of patients treated with nintedanib (follow-up between 6-10 months) to assess change over time., Findings:68 Ga-FAPI-04 accumulated in fibrotic areas of the lungs in patients with systemic sclerosis-associated ILD compared with controls, with a median standardised uptake value (SUV) mean over the whole lung of 0·80 (IQR 0·60-2·10) in the systemic sclerosis-ILD group and 0·50 (0·40-0·50) in the control group (p<0·0001) and a mean whole lung maximal SUV of 4·40 (range 3·05-5·20) in the systemic sclerosis-ILD group compared with 0·70 (0·65-0·70) in the control group (p<0·0001). Whole-lung FAPI metabolic active volume (wlFAPI-MAV) and whole-lung total lesion FAPI (wlTL-FAPI) were not measurable in control participants, because no68 Ga-FAPI-04 uptake above background level was observed. In the systemic sclerosis-ILD group the median wlFAPI-MAV was 254·00 cm3 (IQR 163·40-442·30), and the median wlTL-FAPI was 183·60 cm3 (98·04-960·70).68 Ga-FAPI-04 uptake was higher in patients with extensive disease, with previous ILD progression, or high EUSTAR activity scores than in those with with limited disease, previously stable ILD, or low EUSTAR activity scores. Increased68 Ga-FAPI-04 uptake at baseline was associated with progression of ILD independently of extent of involvement on HRCT scan and the forced vital capacity at baseline. In consecutive68 Ga-FAPI-04 PET-CTs, changes in68 Ga-FAPI-04 uptake was concordant with the observed response to the fibroblast-targeting antifibrotic drug nintedanib., Interpretation: Our study presents the first in-human evidence that fibroblast activation correlates with fibrotic activity and disease progression in the lungs of patients with systemic sclerosis-associated ILD and that68 Ga-FAPI-04 PET-CT might improve risk assessment of systemic sclerosis-associated ILD., Funding: German Research Foundation, Erlangen Anschubs-und Nachwuchsfinanzierung, Interdisziplinäres Zentrum für Klinische Forschung Erlangen, Bundesministerium für Bildung und Forschung, Deutsche Stiftung Systemische Sklerose, Wilhelm-Sander-Foundation, Else-Kröner-Fresenius-Foundation, European Research Council, Ernst-Jung-Foundation, and Clinician Scientist Program Erlangen., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
228. Cellular and molecular mechanisms in fibrosis.
- Author
-
Dees C, Chakraborty D, and Distler JHW
- Subjects
- Animals, DNA Methylation, Ephrins metabolism, Fibroblast Growth Factor 9 metabolism, Fibrosis genetics, Fibrosis pathology, Guanylate Cyclase metabolism, Histone Code, Humans, Idiopathic Pulmonary Fibrosis genetics, Idiopathic Pulmonary Fibrosis pathology, Janus Kinases metabolism, Myofibroblasts, Receptors, Cytoplasmic and Nuclear metabolism, STAT Transcription Factors metabolism, Scleroderma, Systemic genetics, Scleroderma, Systemic pathology, Serotonin metabolism, Fibrosis metabolism, Idiopathic Pulmonary Fibrosis metabolism, Scleroderma, Systemic metabolism, Signal Transduction, Skin pathology, Transforming Growth Factor beta metabolism
- Abstract
The activation of fibroblasts is required for physiological tissue remodelling such as wound healing. However, when the regulatory mechanisms are disrupted and fibroblasts remain persistently activated, the progressive deposition of extracellular matrix proteins leads to tissue fibrosis, which results in dysfunction or even loss of function of the affected organ. Although fibrosis has been recognized as a major cause of morbidity and mortality in modern societies, there are only few treatment options available that directly disrupt the release of extracellular matrix from fibroblasts. Intensive research in recent years, however, identified several pathways as core fibrotic mechanisms that are shared across different fibrotic diseases and organs. We discuss herein selection of those core pathways, especially downstream of the profibrotic TGF-β pathway, which are druggable and which may be transferable from bench to bedside., (© 2020 The Authors. Experimental Dermatology published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
229. Fibroblast growth factor receptor 3 activates a network of profibrotic signaling pathways to promote fibrosis in systemic sclerosis.
- Author
-
Chakraborty D, Zhu H, Jüngel A, Summa L, Li YN, Matei AE, Zhou X, Huang J, Trinh-Minh T, Chen CW, Lafyatis R, Dees C, Bergmann C, Soare A, Luo H, Ramming A, Schett G, Distler O, and Distler JHW
- Subjects
- Animals, Cells, Cultured, Fibroblasts pathology, Fibrosis, Mice, Receptor, Fibroblast Growth Factor, Type 3 genetics, Signal Transduction, Skin pathology, Transforming Growth Factor beta, Receptor, Fibroblast Growth Factor, Type 3 metabolism, Scleroderma, Systemic pathology
- Abstract
Aberrant activation of fibroblasts with progressive deposition of extracellular matrix is a key feature of systemic sclerosis (SSc), a prototypical idiopathic fibrotic disease. Here, we demonstrate that the profibrotic cytokine transforming growth factor β selectively up-regulates fibroblast growth factor receptor 3 (FGFR3) and its ligand FGF9 to promote fibroblast activation and tissue fibrosis, leading to a prominent FGFR3 signature in the SSc skin. Transcriptome profiling, in silico analysis and functional experiments revealed that FGFR3 induces multiple profibrotic pathways including endothelin, interleukin-4, and connective tissue growth factor signaling mediated by transcription factor CREB (cAMP response element-binding protein). Inhibition of FGFR3 signaling by fibroblast-specific knockout of FGFR3 or FGF9 or pharmacological inhibition of FGFR3 blocked fibroblast activation and attenuated experimental skin fibrosis in mice. These findings characterize FGFR3 as an upstream regulator of a network of profibrotic mediators in SSc and as a potential target for the treatment of fibrosis., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
230. Translational engagement of lysophosphatidic acid receptor 1 in skin fibrosis: from dermal fibroblasts of patients with scleroderma to tight skin 1 mouse.
- Author
-
Ledein L, Léger B, Dees C, Beyer C, Distler A, Vettori S, Boukaiba R, Bidouard JP, Schaefer M, Pernerstorfer J, Ruetten H, Jagerschmidt A, Janiak P, Distler JHW, Distler O, and Illiano S
- Subjects
- Animals, Disease Models, Animal, Fibroblasts pathology, Fibrosis, Humans, Mice, Skin pathology, Receptors, Lysophosphatidic Acid, Scleroderma, Systemic drug therapy, Scleroderma, Systemic pathology
- Abstract
Background and Purpose: Genetic deletion and pharmacological studies suggest a role for lysophosphatidic acid (LPA
1 ) receptor in fibrosis. We investigated the therapeutic potential in systemic sclerosis (SSc) of a new orally active selective LPA1 receptor antagonist using dermal fibroblasts from patients and an animal model of skin fibrosis., Experimental Approach: Dermal fibroblast and skin biopsies from systemic sclerosis patients were used. Myofibroblast differentiation, gene expression and cytokine secretion were measured following LPA and/or SAR100842 treatment. Pharmacolgical effect of SAR100842 was assessed in the tight skin 1 (Tsk1) mouse model., Key Results: SAR100842 is equipotent against various LPA isoforms. Dermal fibroblasts and skin biopsies from patients with systemic sclerosis expressed high levels of LPA1 receptor. The LPA functional response (Ca2+ ) in systemic sclerosis dermal fibroblasts was fully antagonized with SAR100842. LPA induced myofibroblast differentiation in systemic sclerosis dermal and idiopathic pulmonary fibrosis lung fibroblasts and the secretion of inflammatory markers and activated Wnt markers. Results from systemic sclerosis dermal fibroblasts mirror those obtained in a mouse Tsk1 model of skin fibrosis. Using a therapeutic protocol, SAR100842 consistently reversed dermal thickening, inhibited myofibroblast differentiation and reduced skin collagen content. Inflammatory and Wnt pathway markers were also inhibited by SAR100842 in the skin of Tsk1 mice., Conclusion and Implications: The effects of SAR100842 on LPA-induced inflammation and on mechanisms linked to fibrosis like myofibroblast differentiation and Wnt pathway activation indicate that LPA1 receptor activation plays a key role in skin fibrosis. Our results support the therapeutic potential of LPA1 receptor antagonists in systemic sclerosis., (© 2020 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)- Published
- 2020
- Full Text
- View/download PDF
231. PGC-1α regulates autophagy to promote fibroblast activation and tissue fibrosis.
- Author
-
Zhang Y, Shen L, Zhu H, Dreissigacker K, Distler D, Zhou X, Györfi AH, Bergmann C, Meng X, Dees C, Trinh-Minh T, Ludolph I, Horch R, Ramming A, Schett G, and Distler JHW
- Subjects
- Animals, Bleomycin pharmacology, Blotting, Western, Collagen biosynthesis, Disease Models, Animal, Fibrosis, Fluorescent Antibody Technique, Humans, Mice, Polymerase Chain Reaction, Receptor, Transforming Growth Factor-beta Type I metabolism, Signal Transduction drug effects, Transforming Growth Factor beta metabolism, Up-Regulation, Autophagy genetics, Fibroblasts physiology, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha physiology, Scleroderma, Systemic genetics, Scleroderma, Systemic pathology
- Abstract
Objectives: Coactivators are a heterogeneous family of transcriptional regulators that are essential for modulation of transcriptional outcomes and fine-tune numerous cellular processes. The aim of the present study was to evaluate the role of the coactivator peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) in the pathogenesis of systemic sclerosis (SSc)., Methods: Expression of PGC-1α was analysed by real-time PCR, western blot and immunofluorescence. Modulation of autophagy was analysed by reporter studies by expression of autophagy-related genes. The effects of PGC-1α knockdown on collagen production and myofibroblast differentiation were analysed in cultured human fibroblasts and in two mouse models with fibroblast-specific knockout of PGC-1α., Results: The expression of PGC-1α was induced in dermal fibroblasts of patients with SSc and experimental murine fibrosis. Transforming growth factor beta (TGFβ), hypoxia and epigenetic mechanisms regulate the expression of PGC-1α in fibroblasts. Knockdown of PGC-1α prevented the activation of autophagy by TGFβ and this translated into reduced fibroblast-to-myofibroblast differentiation and collagen release. Knockout of PGC-1α in fibroblasts prevented skin fibrosis induced by bleomycin and by overexpression of a constitutively active TGFβ receptor type I. Moreover, pharmacological inhibition of PGC-1α by SR18292 induced regression of pre-established, bleomycin-induced skin fibrosis., Conclusion: PGC-1α is upregulated in SSc and promotes autophagy to foster TGFβ-induced fibroblast activation. Targeting of PGC-1α prevents aberrant autophagy, inhibits fibroblast activation and tissue fibrosis and may over therapeutic potential., Competing Interests: Competing interests: JHWD has consultancy relationships with Actelion, Active Biotech, Anamar, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, JB Therapeutics, Medac, Pfizer, RuiYi and UCB. JHWD has received research funding from Anamar, Active Biotech, Array Biopharma, aTyr, BMS, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, Novartis, Sanofi-Aventis, RedX, UCB. JHWD is stock owner of 4D Science., (© Author(s) (or their employer(s)) 2020. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
232. TGF-β-induced epigenetic deregulation of SOCS3 facilitates STAT3 signaling to promote fibrosis.
- Author
-
Dees C, Pötter S, Zhang Y, Bergmann C, Zhou X, Luber M, Wohlfahrt T, Karouzakis E, Ramming A, Gelse K, Yoshimura A, Jaenisch R, Distler O, Schett G, and Distler JH
- Subjects
- Animals, DNA (Cytosine-5-)-Methyltransferase 1 biosynthesis, DNA (Cytosine-5-)-Methyltransferases biosynthesis, DNA Methyltransferase 3A, Female, Fibrosis, Gene Expression Regulation, Enzymologic, Humans, Male, Mice, Myofibroblasts pathology, Scleroderma, Systemic pathology, Epigenesis, Genetic, Myofibroblasts metabolism, STAT3 Transcription Factor metabolism, Scleroderma, Systemic metabolism, Signal Transduction, Suppressor of Cytokine Signaling 3 Protein metabolism, Transforming Growth Factor beta metabolism
- Abstract
Fibroblasts are key effector cells in tissue remodeling. They remain persistently activated in fibrotic diseases, resulting in progressive deposition of extracellular matrix. Although fibroblast activation may be initiated by external factors, prolonged activation can induce an "autonomous," self-maintaining profibrotic phenotype in fibroblasts. Accumulating evidence suggests that epigenetic alterations play a central role in establishing this persistently activated pathologic phenotype of fibroblasts. We demonstrated that in fibrotic skin of patients with systemic sclerosis (SSc), a prototypical idiopathic fibrotic disease, TGF-β induced the expression of DNA methyltransferase 3A (DNMT3A) and DNMT1 in fibroblasts in a SMAD-dependent manner to silence the expression of suppressor of cytokine signaling 3 (SOCS3) by promoter hypermethylation. Downregulation of SOCS3 facilitated activation of STAT3 to promote fibroblast-to-myofibroblast transition, collagen release, and fibrosis in vitro and in vivo. Reestablishment of the epigenetic control of STAT3 signaling by genetic or pharmacological inactivation of DNMT3A reversed the activated phenotype of SSc fibroblasts in tissue culture, inhibited TGF-β-dependent fibroblast activation, and ameliorated experimental fibrosis in murine models. These findings identify a pathway of epigenetic imprinting of fibroblasts in fibrotic disease with translational implications for the development of targeted therapies in fibrotic diseases.
- Published
- 2020
- Full Text
- View/download PDF
233. Dipeptidylpeptidase 4 as a Marker of Activated Fibroblasts and a Potential Target for the Treatment of Fibrosis in Systemic Sclerosis.
- Author
-
Soare A, Györfi HA, Matei AE, Dees C, Rauber S, Wohlfahrt T, Chen CW, Ludolph I, Horch RE, Bäuerle T, von Hörsten S, Mihai C, Distler O, Ramming A, Schett G, and Distler JHW
- Subjects
- Adult, Aged, Animals, Bleomycin toxicity, Cell Movement, Cell Proliferation, Collagen, Dipeptidyl Peptidase 4 genetics, Dipeptidyl-Peptidase IV Inhibitors pharmacology, Disease Models, Animal, Female, Fibrosis, Fluorescent Antibody Technique, Gene Knockdown Techniques, Graft vs Host Disease, Humans, Lung drug effects, Lung pathology, MAP Kinase Signaling System, Male, Mice, Mice, Knockout, Middle Aged, Myofibroblasts, Real-Time Polymerase Chain Reaction, Scleroderma, Systemic metabolism, Scleroderma, Systemic pathology, Sitagliptin Phosphate pharmacology, Skin drug effects, Skin pathology, Transforming Growth Factor beta, Vildagliptin pharmacology, Dipeptidyl Peptidase 4 metabolism, Fibroblasts metabolism, Scleroderma, Systemic genetics, Skin metabolism
- Abstract
Objective: Expression of dipeptidylpeptidase 4 (DPP-4) identifies a dermal fibroblast lineage involved in scarring during wound healing. The role of DDP-4 in tissue fibrosis is, however, unknown. The aim of the present study was to evaluate DPP-4 as a potential target for the treatment of fibrosis in patients with systemic sclerosis (SSc)., Methods: Expression of DPP-4 in skin biopsy samples and dermal fibroblasts was analyzed by real-time polymerase chain reaction, immunofluorescence, and Western blot analyses. The activity of DPP-4 was modulated by overexpression, knockdown, and pharmacologic inhibition of DPP4 using sitagliptin and vildagliptin. The effects of DPP4 inhibition were analyzed in human dermal fibroblasts and in different mouse models of SSc (each n = 6)., Results: The expression of DPP-4 and the number of DPP-4-positive fibroblasts were increased in the fibrotic skin of SSc patients, in a transforming growth factor β (TGFβ)-dependent manner. DPP-4-positive fibroblasts expressed higher levels of myofibroblast markers and collagen (each P < 0.001 versus healthy controls). Overexpression of DPP4 promoted fibroblast activation, whereas pharmacologic inhibition or genetic inactivation of DPP4 reduced the proliferation, migration, and expression of contractile proteins and release of collagen (each P < 0.001 versus control mice) by interfering with TGFβ-induced ERK signaling. DPP4-knockout mice were less sensitive to bleomycin-induced dermal and pulmonary fibrosis (P < 0.0001 versus wild-type controls). Treatment with DPP4 inhibitors promoted regression of fibrosis in mice that had received bleomycin challenge and mice with chronic graft-versus-host disease, and ameliorated fibrosis in TSK1 mice (each P < 0.001 versus untreated controls). These antifibrotic effects were associated with a reduction in inflammation., Conclusion: DPP-4 characterizes a population of activated fibroblasts and shows that DPP-4 regulates TGFβ-induced fibroblast activation in the fibrotic skin of SSc patients. Inhibition of DPP4 exerts potent antifibrotic effects when administered in well-tolerated doses. As DPP4 inhibitors are already in clinical use for diabetes, these results may have direct translational implications for the treatment of fibrosis in patients with SSc., (© 2019, American College of Rheumatology.)
- Published
- 2020
- Full Text
- View/download PDF
234. Acyltransferase skinny hedgehog regulates TGFβ-dependent fibroblast activation in SSc.
- Author
-
Liang R, Kagwiria R, Zehender A, Dees C, Bergmann C, Ramming A, Krasowska D, Michalska-Jakubus M, Kreuter A, Kraner ME, Schett G, and Distler JHW
- Subjects
- Acyltransferases biosynthesis, Adult, Aged, Animals, Blotting, Western, Cell Differentiation, Cells, Cultured, Disease Models, Animal, Female, Fibroblasts metabolism, Fibroblasts pathology, Humans, Male, Mice, Middle Aged, Scleroderma, Systemic metabolism, Scleroderma, Systemic pathology, Signal Transduction, Skin metabolism, Young Adult, Acyltransferases genetics, Gene Expression Regulation, RNA genetics, Scleroderma, Systemic genetics, Skin pathology, Transforming Growth Factor beta metabolism
- Abstract
Objectives: Systemic sclerosis (SSc) is characterised by aberrant hedgehog signalling in fibrotic tissues. The hedgehog acyltransferase (HHAT) skinny hedgehog catalyses the attachment of palmitate onto sonic hedgehog (SHH). Palmitoylation of SHH is required for multimerisation of SHH proteins, which is thought to promote long-range, endocrine hedgehog signalling. The aim of this study was to evaluate the role of HHAT in the pathogenesis of SSc., Methods: Expression of HHAT was analysed by real-time polymerase chain reaction(RT-PCR), immunofluorescence and histomorphometry. The effects of HHAT knockdown were analysed by reporter assays, target gene studies and quantification of collagen release and myofibroblast differentiation in cultured human fibroblasts and in two mouse models., Results: The expression of HHAT was upregulated in dermal fibroblasts of patients with SSc in a transforming growth factor-β (TGFβ)/SMAD-dependent manner. Knockdown of HHAT reduced TGFβ-induced hedgehog signalling as well as myofibroblast differentiation and collagen release in human dermal fibroblasts. Knockdown of HHAT in the skin of mice ameliorated bleomycin-induced and topoisomerase-induced skin fibrosis., Conclusion: HHAT is regulated in SSc in a TGFβ-dependent manner and in turn stimulates TGFβ-induced long-range hedgehog signalling to promote fibroblast activation and tissue fibrosis. Targeting of HHAT might be a novel approach to more selectively interfere with the profibrotic effects of long-range hedgehog signalling., Competing Interests: Competing interests: JHWD has consultancy relationships with Actelion, Active Biotech, Anamar, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, JB Therapeutics, Medac, Pfizer, RuiYi and UCB. JHWD has received research funding from Anamar, Active Biotech, Array Biopharma, aTyr, BMS, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, Novartis, Sanofi-Aventis, RedX, UCB. JHWD is stock owner of 4D Science., (© Author(s) (or their employer(s)) 2019. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2019
- Full Text
- View/download PDF
235. PU.1 controls fibroblast polarization and tissue fibrosis.
- Author
-
Wohlfahrt T, Rauber S, Uebe S, Luber M, Soare A, Ekici A, Weber S, Matei AE, Chen CW, Maier C, Karouzakis E, Kiener HP, Pachera E, Dees C, Beyer C, Daniel C, Gelse K, Kremer AE, Naschberger E, Stürzl M, Butter F, Sticherling M, Finotto S, Kreuter A, Kaplan MH, Jüngel A, Gay S, Nutt SL, Boykin DW, Poon GMK, Distler O, Schett G, Distler JHW, and Ramming A
- Subjects
- Animals, Base Sequence, Epigenesis, Genetic, Female, Humans, Inflammation genetics, Inflammation pathology, Male, Mice, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins antagonists & inhibitors, Trans-Activators antagonists & inhibitors, Cell Differentiation genetics, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis genetics, Fibrosis pathology, Proto-Oncogene Proteins metabolism, Trans-Activators metabolism
- Abstract
Fibroblasts are polymorphic cells with pleiotropic roles in organ morphogenesis, tissue homeostasis and immune responses. In fibrotic diseases, fibroblasts synthesize abundant amounts of extracellular matrix, which induces scarring and organ failure. By contrast, a hallmark feature of fibroblasts in arthritis is degradation of the extracellular matrix because of the release of metalloproteinases and degrading enzymes, and subsequent tissue destruction. The mechanisms that drive these functionally opposing pro-fibrotic and pro-inflammatory phenotypes of fibroblasts remain unknown. Here we identify the transcription factor PU.1 as an essential regulator of the pro-fibrotic gene expression program. The interplay between transcriptional and post-transcriptional mechanisms that normally control the expression of PU.1 expression is perturbed in various fibrotic diseases, resulting in the upregulation of PU.1, induction of fibrosis-associated gene sets and a phenotypic switch in extracellular matrix-producing pro-fibrotic fibroblasts. By contrast, pharmacological and genetic inactivation of PU.1 disrupts the fibrotic network and enables reprogramming of fibrotic fibroblasts into resting fibroblasts, leading to regression of fibrosis in several organs.
- Published
- 2019
- Full Text
- View/download PDF
236. The tyrosine phosphatase SHP2 controls TGFβ-induced STAT3 signaling to regulate fibroblast activation and fibrosis.
- Author
-
Zehender A, Huang J, Györfi AH, Matei AE, Trinh-Minh T, Xu X, Li YN, Chen CW, Lin J, Dees C, Beyer C, Gelse K, Zhang ZY, Bergmann C, Ramming A, Birchmeier W, Distler O, Schett G, and Distler JHW
- Subjects
- Adult, Aged, Animals, Cell Line, Down-Regulation drug effects, Female, Fibroblasts drug effects, Fibrosis, Humans, Janus Kinase 2 metabolism, Male, Mice, Knockout, Middle Aged, Organ Specificity, Protein Tyrosine Phosphatase, Non-Receptor Type 11 genetics, Quinolines pharmacology, RNA, Messenger genetics, RNA, Messenger metabolism, Scleroderma, Systemic metabolism, Scleroderma, Systemic pathology, Young Adult, Fibroblasts metabolism, Fibroblasts pathology, Protein Tyrosine Phosphatase, Non-Receptor Type 11 metabolism, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Transforming Growth Factor beta metabolism
- Abstract
Uncontrolled activation of TGFβ signaling is a common denominator of fibrotic tissue remodeling. Here we characterize the tyrosine phosphatase SHP2 as a molecular checkpoint for TGFβ-induced JAK2/STAT3 signaling and as a potential target for the treatment of fibrosis. TGFβ stimulates the phosphatase activity of SHP2, although this effect is in part counterbalanced by inhibitory effects on SHP2 expression. Stimulation with TGFβ promotes recruitment of SHP2 to JAK2 in fibroblasts with subsequent dephosphorylation of JAK2 at Y570 and activation of STAT3. The effects of SHP2 on STAT3 activation translate into major regulatory effects of SHP2 on fibroblast activation and tissue fibrosis. Genetic or pharmacologic inactivation of SHP2 promotes accumulation of JAK2 phosphorylated at Y570, reduces JAK2/STAT3 signaling, inhibits TGFβ-induced fibroblast activation and ameliorates dermal and pulmonary fibrosis. Given the availability of potent SHP2 inhibitors, SHP2 might thus be a potential target for the treatment of fibrosis.
- Published
- 2018
- Full Text
- View/download PDF
237. Poly(ADP-ribose) polymerase-1 regulates fibroblast activation in systemic sclerosis.
- Author
-
Zhang Y, Pötter S, Chen CW, Liang R, Gelse K, Ludolph I, Horch RE, Distler O, Schett G, Distler JHW, and Dees C
- Subjects
- Adult, Aged, Animals, DNA Methylation genetics, Disease Models, Animal, Down-Regulation genetics, Female, Fibrosis chemically induced, Fibrosis enzymology, Humans, Male, Mice, Middle Aged, Protein Serine-Threonine Kinases, Scleroderma, Systemic enzymology, Signal Transduction, Skin metabolism, Skin pathology, Skin Diseases enzymology, Smad3 Protein metabolism, Transforming Growth Factor beta metabolism, Young Adult, Fibroblasts physiology, Fibrosis genetics, Poly (ADP-Ribose) Polymerase-1 metabolism, Scleroderma, Systemic genetics, Skin Diseases genetics
- Abstract
Objectives: The enzyme poly(ADP-ribose) polymerase-1 (PARP-1) transfers negatively charged ADP-ribose units to target proteins. This modification can have pronounced regulatory effects on target proteins. Recent studies showed that PARP-1 can poly(ADP-ribosyl)ate (PARylate) Smad proteins. However, the role of PARP-1 in the pathogenesis of systemic sclerosis (SSc) has not been investigated., Methods: The expression of PARP-1 was determined by quantitative PCR and immunohistochemistry. DNA methylation was analysed by methylated DNA immunoprecipitation assays. Transforming growth factor-β (TGFβ) signalling was assessed using reporter assays, chromatin immunoprecipitation assays and target gene analysis. The effect of PARP-1 inactivation was investigated in bleomycin-induced and topoisomerase-induced fibrosis as well as in tight-skin-1 (Tsk-1) mice., Results: The expression of PARP-1 was decreased in patients with SSc, particularly in fibroblasts. The promoter of PARP-1 was hypermethylated in SSc fibroblasts and in TGFβ-stimulated normal fibroblasts. Inhibition of DNA methyltransferases (DNMTs) reduced the promoter methylation and reactivated the expression of PARP-1. Inactivation of PARP-1 promoted accumulation of phosphorylated Smad3, enhanced Smad-dependent transcription and upregulated the expression of TGFβ/Smad target genes. Inhibition of PARP-1 enhanced the effect of TGFβ on collagen release and myofibroblast differentiation in vitro and exacerbated experimental fibrosis in vivo. PARP-1 deficiency induced a more severe fibrotic response to bleomycin with increased dermal thickening, hydroxyproline content and myofibroblast counts. Inhibition of PARylation also exacerbated fibrosis in Tsk-1 mice and in mice with topoisomerase-induced fibrosis., Conclusion: PARP-1 negatively regulates canonical TGFβ signalling in experimental skin fibrosis. The downregulation of PARP-1 in SSc fibroblasts may thus directly contribute to hyperactive TGFβ signalling and to persistent fibroblast activation in SSc., Competing Interests: Competing interests: OD has consultancy relationships and/or has received research funding from Actelion, Pfizer, Ergonex, BMS, Sanofi-Aventis, United BioSource Corporation, medac, Biovitrium, Boehringer Ingelheim, Novartis, 4D Science and Active Biotech in the area of potential treatments of SSc; JHWD has consultancy relationships and/or has received research funding from Actelion, Pfizer, Ergonex, 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 and is stock owner of 4D Science GmbH., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)
- Published
- 2018
- Full Text
- View/download PDF
238. Protein kinases G are essential downstream mediators of the antifibrotic effects of sGC stimulators.
- Author
-
Matei AE, Beyer C, Györfi AH, Soare A, Chen CW, Dees C, Bergmann C, Ramming A, Friebe A, Hofmann F, Distler O, Schett G, and Distler JHW
- Subjects
- Adult, Aged, Animals, Bleomycin pharmacology, Blotting, Western, Carbazoles pharmacology, Cell Culture Techniques, Female, Fibroblasts drug effects, Fibrosis metabolism, Fluorescent Antibody Technique, Humans, Male, Mice, Mice, Knockout, Middle Aged, Pyrazoles pharmacology, Pyridines pharmacology, Real-Time Polymerase Chain Reaction, Scleroderma, Systemic pathology, Signal Transduction drug effects, Transforming Growth Factor beta metabolism, Cyclic GMP-Dependent Protein Kinases metabolism, Fibroblasts metabolism, Scleroderma, Systemic metabolism, Soluble Guanylyl Cyclase metabolism
- Abstract
Objectives: Stimulators of soluble guanylate cyclase (sGC) are currently investigated in clinical trials for the treatment of fibrosis in systemic sclerosis (SSc). In this study, we aim to investigate the role of protein kinases G (PKG) as downstream mediators of sGC-cyclic guanosine monophosphate (cGMP) in SSc., Methods: Mice with combined knockout of PKG1 and 2 were challenged with bleomycin and treated with the sGC stimulator BAY 41-2272. Fibroblasts were treated with BAY 41-2272 and with the PKG inhibitor KT 5823., Results: PKG1 and 2 are upregulated in SSc in a transforming growth factor-β1 (TGFβ1)-dependent manner, as an attempt to compensate for the decreased signalling through the sGC-cGMP-PKG pathway. Inhibition or knockout of PKG1 and 2 abrogates the inhibitory effects of sGC stimulation on fibroblast activation in a SMAD-independent, but extracellular signal-regulated kinase (ERK)-dependent manner. In vivo, sGC stimulation fails to prevent bleomycin-induced fibrosis in PKG1 and 2 knockout mice., Conclusions: Our data provide evidence that PKGs are essential mediators of the antifibrotic effects of sGC stimulators through interfering with non-canonical TGFβ signalling. TGFβ1 promotes its profibrotic effects through inhibition of sGC-cGMP-PKG signalling, sGC stimulation exerts its antifibrotic effects by inhibition of TGFβ1-induced ERK phosphorylation., Competing Interests: Competing interests: OD has consulted for, or has received research funding from, 4D Science, Actelion, Active Biotech, Bayer-Schering, Biogen, Biovitrium, BMS, Boehringer, EpiPharm, Ergonex, GSK, Inventiva, Medac, Novartis, Pfizer, Roche/Genentech, Sanofi/Genzyme, Serodapharm, Sinoxa and United BioSource Corporation; JHWD has consultancy relationships and/or has received research funding from Actelion, BMS, Celgene, Bayer Pharma, Boehringer Ingelheim, JB Therapeutics, Sanofi-Aventis, Novartis, UCB, GSK, Array Biopharma, Galapagos, Inventiva and Active Biotech in the area of potential treatments of SSc and is stock owner of 4D Science., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)
- Published
- 2018
- Full Text
- View/download PDF
239. The histone demethylase Jumonji domain-containing protein 3 (JMJD3) regulates fibroblast activation in systemic sclerosis.
- Author
-
Bergmann C, Brandt A, Merlevede B, Hallenberger L, Dees C, Wohlfahrt T, Pötter S, Zhang Y, Chen CW, Mallano T, Liang R, Kagwiria R, Kreuter A, Pantelaki I, Bozec A, Abraham D, Rieker R, Ramming A, Distler O, Schett G, and Distler JHW
- Subjects
- Adult, Aged, Animals, Bleomycin, Case-Control Studies, Cells, Cultured, Enzyme Activation, Female, Fibrosis chemically induced, Fibrosis enzymology, Humans, Male, Mice, Middle Aged, Young Adult, Fibroblasts enzymology, Jumonji Domain-Containing Histone Demethylases metabolism, Scleroderma, Systemic enzymology
- Abstract
Objectives: Systemic sclerosis (SSc) fibroblasts remain activated even in the absence of exogenous stimuli. Epigenetic alterations are thought to play a role for this endogenous activation. Trimethylation of histone H3 on lysine 27 (H3K27me3) is regulated by Jumonji domain-containing protein 3 (JMJD3) and ubiquitously transcribed tetratricopeptide repeat on chromosome X (UTX) in a therapeutically targetable manner. The aim of this study was to explore H3K27me3 demethylases as potential targets for the treatment of fibrosis., Methods: JMJD3 was inactivated by small interfering RNA-mediated knockdown and by pharmacological inhibition with GSKJ4. The effects of targeted inactivation of JMJD3 were analysed in cultured fibroblasts and in the murine models of bleomycin-induced and topoisomerase-I (topoI)-induced fibrosis. H3K27me3 at the FRA2 promoter was analysed by ChIP., Results: The expression of JMJD3, but not of UTX, was increased in fibroblasts in SSc skin and in experimental fibrosis in a transforming growth factor beta (TGFβ)-dependent manner. Inactivation of JMJD3 reversed the activated fibroblast phenotype in SSc fibroblasts and prevented the activation of healthy dermal fibroblasts by TGFβ. Pharmacological inhibition of JMJD3 ameliorated bleomycin-induced and topoI-induced fibrosis in well-tolerated doses. JMJD3 regulated fibroblast activation in a FRA2-dependent manner: Inactivation of JMJD3 reduced the expression of FRA2 by inducing accumulation of H3K27me3 at the FRA2 promoter. Moreover, the antifibrotic effects of JMJD3 inhibition were reduced on knockdown of FRA2 ., Conclusion: We present first evidence for a deregulation of JMJD3 in SSc. JMJD3 modulates fibroblast activation by regulating the levels of H3K27me3 at the promoter of FRA2 . Targeted inhibition of JMJD3 limits the aberrant activation of SSc fibroblasts and exerts antifibrotic effects in two murine models., Competing Interests: Competing interests: O.D. has consulted for, or has received research funding from, 4D Science, Actelion, Active Biotech, Bayer-Schering, Biogen, Biovitrium, BMS, Boehringer, EpiPharm, Ergonex, GSK, Inventiva, Medac, Novartis, Pfizer, Roche/Genentech, Sanofi/Genzyme, Serodapharm, Sinoxa and United BioSource Corporation; JHWD has consultancy relationships and/or has received research funding from Actelion, BMS, Celgene, Bayer Pharma, Boehringer Ingelheim, JB Therapeutics, Sanofi-Aventis, Novartis, UCB, GSK, Array Biopharma, Galapagos, Inventiva and Active Biotech in the area of potential treatments of SSc and is stock owner of 4D Science., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)
- Published
- 2018
- Full Text
- View/download PDF
240. Nintedanib inhibits macrophage activation and ameliorates vascular and fibrotic manifestations in the Fra2 mouse model of systemic sclerosis.
- Author
-
Huang J, Maier C, Zhang Y, Soare A, Dees C, Beyer C, Harre U, Chen CW, Distler O, Schett G, Wollin L, and Distler JHW
- Subjects
- Animals, Cell Proliferation drug effects, Disease Models, Animal, Fibrosis, Fos-Related Antigen-2, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary etiology, Macrophage Activation drug effects, Mice, Mice, Transgenic, Muscle, Smooth, Vascular cytology, Protein-Tyrosine Kinases antagonists & inhibitors, Pulmonary Artery drug effects, Scleroderma, Systemic complications, Scleroderma, Systemic pathology, Vascular Endothelial Growth Factor A blood, Enzyme Inhibitors pharmacology, Indoles pharmacology, Scleroderma, Systemic drug therapy
- Abstract
Background: Nintedanib is an inhibitor targeting platelet-derived growth factor receptor, fibroblast growth factor receptor and vascular endothelial growth factor receptor tyrosine kinases that has recently been approved for the treatment of idiopathic pulmonary fibrosis. The aim of this study was to analyse the effects of nintedanib in the fos-related antigen-2 (Fra2) mouse model of systemic sclerosis (SSc)., Methods: The effects of nintedanib on pulmonary arterial hypertension with proliferation of pulmonary vascular smooth muscle cells (PVSMCs) and luminal occlusion, on microvascular disease with apoptosis of microvascular endothelial cells (MVECs) and on fibroblast activation with myofibroblast differentiation and accumulation of extracellular matrix were analysed. We also studied the effects of nintedanib on the levels of key mediators involved in the pathogenesis of SSc and on macrophage polarisation., Results: Nintedanib inhibited proliferation of PVSMCs and prevented thickening of the vessel walls and luminal occlusion of pulmonary arteries. Treatment with nintedanib also inhibited apoptosis of MVECs and blunted the capillary rarefaction in Fra2-transgenic mice. These effects were associated with a normalisation of the serum levels of vascular endothelial growth factor in Fra2 mice on treatment with nintedanib. Nintedanib also effectively blocked myofibroblast differentiation and reduced pulmonary, dermal and myocardial fibrosis in Fra2-transgenic mice. The antifibrotic effects of nintedanib were associated with impaired M2 polarisation of monocytes and reduced numbers of M2 macrophages., Conclusion: Nintedanib targets core features of SSc in Fra2-transgenic mice and ameliorates histological features of pulmonary arterial hypertension, destructive microangiopathy and pulmonary and dermal fibrosis. These data might have direct implications for the ongoing phase III clinical trial with nintedanib in SSc-associated interstitial lung disease., Competing Interests: Competing interests: OD has consultancy relationships and/or has received research funding from Actelion, Pfizer, Ergonex, BMS, Sanofi-Aventis, United BioSource Corporation, Roche/Genentech, Medac, Biovitrium, Boehringer Ingelheim, Novartis, 4D Science, Active Biotech, Bayer, Sinoxa, Serodapharm, EpiPharm, GSK, Pharmacyclics and Biogen. LW is an employee of Boehringer-Ingelheim. JHWD has consultancy relationships with Actelion, Active Biotech, Anamar, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, JB Therapeutics, Medac, Pfizer, RuiYi and UCB. JHWD has received research funding from Anamar, Active Biotech, Array Biopharma, BMS, Bayer Pharma, Boehringer Ingelheim, Celgene, GSK, Novartis, Sanofi-Aventis and UCB. JHWD is stock owner of 4D Science., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2017. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)
- Published
- 2017
- Full Text
- View/download PDF
241. Oxidant sensor in the cGMP-binding pocket of PKGIα regulates nitroxyl-mediated kinase activity.
- Author
-
Donzelli S, Goetz M, Schmidt K, Wolters M, Stathopoulou K, Diering S, Prysyazhna O, Polat V, Scotcher J, Dees C, Subramanian H, Butt E, Kamynina A, Schobesberger S, King SB, Nikolaev VO, de Wit C, Leichert LI, Feil R, Eaton P, and Cuello F
- Subjects
- Animals, Catalytic Domain, Cells, Cultured, Cyclic GMP-Dependent Protein Kinase Type I genetics, Cysteine genetics, Fluorescence Resonance Energy Transfer, HEK293 Cells, Humans, Male, Mass Spectrometry, Mice, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Oxidation-Reduction, Cyclic GMP metabolism, Cyclic GMP-Dependent Protein Kinase Type I chemistry, Cyclic GMP-Dependent Protein Kinase Type I metabolism, Disulfides metabolism, Mutagenesis, Site-Directed, Nitrogen Oxides pharmacology
- Abstract
Despite the mechanisms for endogenous nitroxyl (HNO) production and action being incompletely understood, pharmacological donors show broad therapeutic promise and are in clinical trials. Mass spectrometry and site-directed mutagenesis showed that chemically distinct HNO donors 1-nitrosocyclohexyl acetate or Angeli's salt induced disulfides within cGMP-dependent protein kinase I-alpha (PKGIα), an interdisulfide between Cys42 of the two identical subunits of the kinase and a previously unobserved intradisulfide between Cys117 and Cys195 in the high affinity cGMP-binding site. Kinase activity was monitored in cells transfected with wildtype (WT), Cys42Ser or Cys117/195Ser PKGIα that cannot form the inter- or intradisulfide, respectively. HNO enhanced WT kinase activity, an effect significantly attenuated in inter- or intradisulfide-deficient PKGIα. To investigate whether the intradisulfide modulates cGMP binding, real-time imaging was performed in vascular smooth muscle cells expressing a FRET-biosensor comprising the cGMP-binding sites of PKGIα. HNO induced FRET changes similar to those elicited by an increase of cGMP, suggesting that intradisulfide formation is associated with activation of PKGIα. Intradisulfide formation in PKGIα correlated with enhanced HNO-mediated vasorelaxation in mesenteric arteries in vitro and arteriolar dilation in vivo in mice. HNO induces intradisulfide formation in PKGIα, inducing the same effect as cGMP binding, namely kinase activation and thus vasorelaxation.
- Published
- 2017
- Full Text
- View/download PDF
242. JAK1-dependent transphosphorylation of JAK2 limits the antifibrotic effects of selective JAK2 inhibitors on long-term treatment.
- Author
-
Zhang Y, Liang R, Chen CW, Mallano T, Dees C, Distler A, Reich A, Bergmann C, Ramming A, Gelse K, Mielenz D, Distler O, Schett G, and Distler JHW
- Subjects
- Adult, Animals, Antibiotics, Antineoplastic toxicity, Benzoquinones pharmacology, Bleomycin toxicity, Blotting, Western, Disease Models, Animal, Fibroblasts metabolism, Fibrosis, HSP90 Heat-Shock Proteins antagonists & inhibitors, Humans, Immunohistochemistry, Janus Kinase 1 antagonists & inhibitors, Janus Kinase 1 metabolism, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 metabolism, Lactams, Macrocyclic pharmacology, Lung pathology, Male, Mice, Middle Aged, Nitriles, Phosphorylation drug effects, Pulmonary Fibrosis chemically induced, Real-Time Polymerase Chain Reaction, Transforming Growth Factor beta pharmacology, Fibroblasts drug effects, Janus Kinase 1 drug effects, Janus Kinase 2 drug effects, Lung drug effects, Protein Kinase Inhibitors pharmacology, Pulmonary Fibrosis metabolism, Pyrazoles pharmacology, Pyrimidines pharmacology, Scleroderma, Systemic, Sulfonamides pharmacology
- Abstract
Objectives: Janus kinase 2 (JAK2) has recently been described as a novel downstream mediator of the pro-fibrotic effects of transforming growth factor-β. Although JAK2 inhibitors are in clinical use for myelodysplastic syndromes, patients often rapidly develop resistance. Tumour cells can escape the therapeutic effects of selective JAK2 inhibitors by mutation-independent transactivation of JAK2 by JAK1. Here, we used selective JAK2 inhibition as a model to test the hypothesis that chronic treatment may provoke resistance by facilitating non-physiological signalling pathways in fibroblasts., Methods: The antifibrotic effects of long-term treatment with selective JAK2 inhibitors and reactivation of JAK2 signalling by JAK1-dependent transphosphorylation was analysed in cultured fibroblasts and experimental dermal and pulmonary fibrosis. Combined JAK1/JAK2 inhibition and co-treatment with an HSP90 inhibitor were evaluated as strategies to overcome resistance., Results: The antifibrotic effects of selective JAK2 inhibitors on fibroblasts decreased with prolonged treatment as JAK2 signalling was reactivated by JAK1-dependent transphosphorylation of JAK2. This reactivation could be prevented by HSP90 inhibition, which destabilised JAK2 protein, or with combined JAK1/JAK2 inhibitors. Treatment with combined JAK1/JAK2 inhibitors or with JAK2 inhibitors in combination with HSP90 inhibitors was more effective than monotherapy with JAK2 inhibitors in bleomycin-induced pulmonary fibrosis and in adTBR-induced dermal fibrosis., Conclusion: Fibroblasts can develop resistance to chronic treatment with JAK2 inhibitors by induction of non-physiological JAK1-dependent transactivation of JAK2 and that inhibition of this compensatory signalling pathway, for example, by co-inhibition of JAK1 or HSP90 is important to maintain the antifibrotic effects of JAK2 inhibition with long-term treatment., Competing Interests: Competing interests: None declared., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2017. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)
- Published
- 2017
- Full Text
- View/download PDF
243. The transcription factor GLI2 as a downstream mediator of transforming growth factor-β-induced fibroblast activation in SSc.
- Author
-
Liang R, Šumová B, Cordazzo C, Mallano T, Zhang Y, Wohlfahrt T, Dees C, Ramming A, Krasowska D, Michalska-Jakubus M, Distler O, Schett G, Šenolt L, and Distler JH
- Subjects
- Adult, Aged, Anilides pharmacology, Animals, Cells, Cultured, Collagen Type I genetics, Connective Tissue Growth Factor genetics, Female, Fibroblasts drug effects, Fibroblasts metabolism, Fibrosis, Gene Knockout Techniques, Humans, Kruppel-Like Transcription Factors antagonists & inhibitors, Kruppel-Like Transcription Factors genetics, Male, Mice, Mice, Knockout, Mice, Transgenic, Middle Aged, Plasminogen Activator Inhibitor 1 genetics, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Pteridines pharmacology, Pulmonary Fibrosis chemically induced, Pyridines pharmacology, Pyrimidines pharmacology, RNA, Messenger metabolism, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta antagonists & inhibitors, Receptors, Transforming Growth Factor beta genetics, Recombinant Proteins pharmacology, Signal Transduction drug effects, Skin drug effects, Smad3 Protein metabolism, Smoothened Receptor antagonists & inhibitors, Transforming Growth Factor beta pharmacology, Young Adult, Zinc Finger Protein Gli2, Hedgehog Proteins metabolism, Kruppel-Like Transcription Factors metabolism, Pulmonary Fibrosis metabolism, Scleroderma, Systemic genetics, Scleroderma, Systemic metabolism, Skin pathology, Transforming Growth Factor beta metabolism
- Abstract
Objectives: Hedgehog signalling plays a critical role during the pathogenesis of fibrosis in systemic sclerosis (SSc). Besides canonical hedgehog signalling with smoothened (SMO)-dependent activation of GLI transcription factors, GLI can be activated independently of classical hedgehog ligands and receptors (so-called non-canonical pathways). Here, we aimed to evaluate the role of non-canonical hedgehog signalling in SSc and to test the efficacy of direct GLI inhibitors that target simultaneously canonical and non-canonical hedgehog pathways., Methods: The GLI inhibitor GANT-61 was used to inhibit canonical as well as non-canonical hedgehog signalling, while the SMO inhibitor vismodegib was used to selectively target canonical hedgehog signalling. Furthermore, GLI2 was selectively depleted in fibroblasts using the Cre-LoxP system. The effects of pharmacological or genetic of GLI2 on transforming growth factor-β (TGF-β) signalling were analysed in cultured fibroblasts, in bleomycin-induced pulmonary fibrosis and in mice with overexpression of a constitutively active TGF-β receptor I., Results: TGF-β upregulated GLI2 in a Smad3-dependent manner and induced nuclear accumulation and DNA binding of GLI2. Fibroblast-specific knockout of GLI2 protected mice from TBR
act -induced fibrosis. Combined targeting of canonical and non-canonical hedgehog signalling with direct GLI inhibitors exerted more potent antifibrotic effects than selective targeting of canonical hedgehog signalling with SMO inhibitors in experimental dermal and pulmonary fibrosis., Conclusions: Our data demonstrate that hedgehog pathways and TGF-β signalling both converge to GLI2 and that GLI2 integrates those signalling to promote tissue fibrosis. These findings may have translational implications as non-selective inhibitors of GLI2 are in clinical use and selective molecules are currently in development., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)- Published
- 2017
- Full Text
- View/download PDF
244. Cannula-associated Ocular Injuries during Cataract Surgery: A Preventable Intraoperative Complication?
- Author
-
Ting DSJ, Dees C, and Ellerton C
- Subjects
- Aged, Aged, 80 and over, Eye Injuries, Penetrating diagnosis, Eye Injuries, Penetrating prevention & control, Female, Humans, Male, Retrospective Studies, Visual Acuity, Cannula adverse effects, Cataract Extraction adverse effects, Eye Injuries, Penetrating etiology, Intraoperative Complications
- Abstract
Although rare, inadvertently dislodged cannula can occur during cataract surgery. We report two cases of cannula-associated ocular injury during stromal hydration of the main corneal incision despite the use of Luer-lock syringes. Case 1 suffered from an initially occult intraocular injury which led to a delayed presentation of vitreous prolapsing into the anterior chamber, presumed posterior capsular rupture, vitreous hemorrhage, and multiple retinal tears, which required a three-port pars plana vitrectomy and cryotherapy. Case 2 sustained an iris laceration, anterior capsular tear, and postoperative raised intraocular pressure with no late sequelae. The former case highlights the need for close monitoring postoperatively despite the absence of initial apparent evidence of intraocular injury. Herein, we propose a systematic approach in reducing the risk of inadvertent cannula-associated ocular injury., Competing Interests: There are no conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
245. Composition of TWIST1 dimers regulates fibroblast activation and tissue fibrosis.
- Author
-
Palumbo-Zerr K, Soare A, Zerr P, Liebl A, Mancuso R, Tomcik M, Sumova B, Dees C, Chen CW, Wohlfahrt T, Mallano T, Distler A, Ramming A, Gelse K, Mihai C, Distler O, Schett G, and Distler JH
- Subjects
- Animals, Case-Control Studies, Female, Fibroblasts drug effects, Gene Expression Regulation drug effects, Gene Expression Regulation physiology, Gene Knockdown Techniques, Humans, Male, Mice, Knockout, Nuclear Proteins biosynthesis, Nuclear Proteins deficiency, Nuclear Proteins genetics, Nuclear Proteins metabolism, Protein Multimerization physiology, RNA, Messenger genetics, RNA, Small Interfering genetics, Scleroderma, Systemic pathology, Signal Transduction physiology, Skin pathology, Transforming Growth Factor beta pharmacology, Twist-Related Protein 1 biosynthesis, Twist-Related Protein 1 deficiency, Twist-Related Protein 1 genetics, Twist-Related Protein 1 metabolism, Fibroblasts metabolism, Nuclear Proteins physiology, Scleroderma, Systemic metabolism, Twist-Related Protein 1 physiology
- Abstract
Objectives: TWIST1 is a member of the class B of basic helix-loop-helix transcription factors that regulates cell lineage determination and differentiation and has been implicated in epithelial-to-mesenchymal transition. Here, we aimed to investigate the role of TWIST1 for the activation of resident fibroblasts in systemic sclerosis (SSc)., Methods: The expression of Twist1 in fibroblasts was modulated by forced overexpression or siRNA-mediated knockdown. Interaction of Twist1, E12 and inhibitor Of differentiation (Id) was analysed by co-immunoprecipitation. The role of Twist1 in vivo was evaluated using inducible, conditional knockout mice with either ubiquitous or fibroblast-specific depletion of Twist1. Mice were either challenged with bleomycin or overexpressing a constitutively active transforming growth factor (TGF)β receptor I., Result: The expression of TWIST1 was increased in fibroblasts in fibrotic human and murine skin in a TGFβ/SMAD3-dependent manner. TWIST1 in turn enhanced TGFβ-induced fibroblast activation in a p38-dependent manner. The stimulatory effects of TWIST1 on resident fibroblasts were mediated by TWIST1 homodimers. TGFβ promotes the formation of TWIST1 homodimers by upregulation of TWIST1 and by induction of inhibitor of DNA-binding proteins, which have high affinity for E12/E47 and compete against TWIST1 for E12/E47 binding. Mice with selective depletion of Twist1 in fibroblasts are protected from experimental skin fibrosis in different murine models to a comparable degree as mice with ubiquitous depletion of Twist1., Conclusions: Our data identify TWIST1 as a central pro-fibrotic factor in SSc, which facilitates fibroblast activation by amplifying TGFβ signalling. Targeting of TWIST1 may thus be a novel approach to normalise aberrant TGFβ signalling in SSc., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2017
- Full Text
- View/download PDF
246. Inhibition of Notch1 promotes hedgehog signalling in a HES1-dependent manner in chondrocytes and exacerbates experimental osteoarthritis.
- Author
-
Lin NY, Distler A, Beyer C, Philipi-Schöbinger A, Breda S, Dees C, Stock M, Tomcik M, Niemeier A, Dell'Accio F, Gelse K, Mattson MP, Schett G, and Distler JH
- Subjects
- Animals, Cartilage, Articular metabolism, Mice, Mice, Transgenic, Osteophyte metabolism, Real-Time Polymerase Chain Reaction, Severity of Illness Index, Signal Transduction, Arthritis, Experimental metabolism, Carrier Proteins metabolism, Chondrocytes metabolism, Membrane Glycoproteins metabolism, Osteoarthritis metabolism, Receptor, Notch1 metabolism, Transcription Factor HES-1 metabolism
- Abstract
Objectives: Notch ligands and receptors have recently been shown to be differentially expressed in osteoarthritis (OA). We aim to further elucidate the functional role of Notch signalling in OA using Notch1 antisense transgenic (Notch1 AS) mice., Methods: Notch and hedgehog signalling were analysed by real-time PCR and immunohistochemistry. Notch-1 AS mice were employed as a model of impaired Notch signalling in vivo. Experimental OA was induced by destabilisation of the medial meniscus (DMM). The extent of cartilage destruction and osteophyte formation was analysed by safranin-O staining with subsequent assessment of the Osteoarthritis Research Society International (OARSI) and Mankin scores and µCT scanning. Collagen X staining was used as a marker of chondrocyte hypertrophy. The role of hairy/enhancer of split 1 (Hes-1) was investigated with knockdown and overexpression experiments., Results: Notch signalling was activated in human and murine OA with increased expression of Jagged1, Notch-1, accumulation of the Notch intracellular domain 1 and increased transcription of Hes-1. Notch1 AS mice showed exacerbated OA with increases in OARSI scores, osteophyte formation, increased subchondral bone plate density, collagen X and osteocalcin expression and elevated levels of Epas1 and ADAM-TS5 mRNA. Inhibition of the Notch pathway induced activation of hedgehog signalling with induction of Gli-1 and Gli-2 and increased transcription of hedgehog target genes. The regulatory effects of Notch signalling on Gli-expression were mimicked by Hes-1., Conclusions: Inhibition of Notch signalling activates hedgehog signalling, enhances chondrocyte hypertrophy and exacerbates experimental OA including osteophyte formation. These data suggest that the activation of the Notch pathway may limit aberrant hedgehog signalling in OA., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2016
- Full Text
- View/download PDF
247. Type 2 innate lymphoid cell counts are increased in patients with systemic sclerosis and correlate with the extent of fibrosis.
- Author
-
Wohlfahrt T, Usherenko S, Englbrecht M, Dees C, Weber S, Beyer C, Gelse K, Distler O, Schett G, Distler JH, and Ramming A
- Subjects
- Adult, Aged, Case-Control Studies, Female, Fibrosis, Flow Cytometry, Humans, Immunohistochemistry, Lymphocyte Count, Lymphocytes cytology, Male, Middle Aged, Pulmonary Fibrosis blood, Pulmonary Fibrosis etiology, Scleroderma, Systemic blood, Scleroderma, Systemic complications, Scleroderma, Systemic pathology, Severity of Illness Index, Skin cytology, Skin pathology, Immunity, Innate immunology, Lymphocytes immunology, Pulmonary Fibrosis immunology, Scleroderma, Systemic immunology, Skin immunology
- Abstract
Objective: Type 2 innate lymphoid cells (ILC2s), a recently identified population of lymphoid cells lacking lineage-specific receptors, promote type 2 immunity and tissue remodelling. However, the contributive role of ILC2s in the pathogenesis of systemic sclerosis (SSc) is unknown. We aimed to evaluate the levels and correlations with fibrotic manifestations in SSc., Methods: 69 patients with SSc and 47 healthy controls were included. Blood samples and skin sections were analysed by flow cytometry and immunohistochemically by staining two complementary panels of markers., Results: Dermal and circulating ILC2s were significantly elevated in patients with SSc compared with controls. Dermal, but not circulating ILC2s were activated. Stratification of the SSc population in patients with limited cutaneous SSc (lcSSc) and diffuse cutaneous SSc (dcSSc) demonstrated increased levels of ILC2s in both subgroups with significantly higher frequencies in dcSSc compared with lcSSc. Moreover, dermal and circulating ILC2 counts correlated closely with the modified Rodnan skin score and with the presence of pulmonary fibrosis., Conclusions: ILC2 counts are elevated in patients with SSc and correlate with the extent of skin fibrosis and the presence of interstitial lung disease providing compelling evidence for profibrotic effect of ILC2s in SSc., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
248. Tribbles homologue 3 stimulates canonical TGF-β signalling to regulate fibroblast activation and tissue fibrosis.
- Author
-
Tomcik M, Palumbo-Zerr K, Zerr P, Sumova B, Avouac J, Dees C, Distler A, Becvar R, Distler O, Schett G, Senolt L, and Distler JH
- Subjects
- Adult, Aged, Animals, Antibiotics, Antineoplastic toxicity, Bleomycin toxicity, Case-Control Studies, Cell Cycle Proteins metabolism, Cells, Cultured, Collagen metabolism, Dermis cytology, Disease Models, Animal, Female, Fibrosis chemically induced, Fibrosis genetics, Gene Knock-In Techniques, Gene Knockdown Techniques, Humans, Immunohistochemistry, Male, Mice, Middle Aged, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Real-Time Polymerase Chain Reaction, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta, Repressor Proteins metabolism, Scleroderma, Systemic metabolism, Signal Transduction genetics, Skin Diseases chemically induced, Skin Diseases genetics, Young Adult, Cell Cycle Proteins genetics, Fibroblasts metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Repressor Proteins genetics, Scleroderma, Systemic genetics, Smad3 Protein metabolism, Transforming Growth Factor beta metabolism
- Abstract
Objectives: Tribbles homologue 3 (TRB3) is a pseudokinase that modifies the activation of various intracellular signalling pathways to control fundamental processes extending from mitosis and cell activation to apoptosis and modulation of gene expression. Here, we aimed to analyse the role of TRB3 in fibroblast activation in systemic sclerosis (SSc)., Methods: The expression of TRB3 was quantified by quantitative PCR, western blot and immunohistochemistry. The role of TRB3 was analysed in cultured fibroblasts and in experimental fibrosis using small interfering RNA (siRNA)-mediated knockdown and overexpression of TRB3., Results: TRB3 expression was increased in fibroblasts of patients with SSc and in murine models of SSc in a transforming growth factor-β (TGF-β)/Smad-dependent manner. Overexpression of TRB3 stimulated canonical TGF-β signalling and induced an activated phenotype in resting fibroblasts. In contrast, knockdown of TRB3 reduced the profibrotic effects of TGF-β and decreased the collagen synthesis. Moreover, siRNA-mediated knockdown of TRB3 exerted potent antifibrotic effects and ameliorated bleomycin as well as constitutively active TGF-β receptor I-induced fibrosis with reduced dermal thickening, decreased hydroxyproline content and impaired myofibroblast differentiation., Conclusions: The present study characterises TRB3 as a novel profibrotic mediator in SSc. TGF-β induces TRB3, which in turn activates canonical TGF-β/Smad signalling and stimulates the release of collagen, thereby inducing a positive feedback loop that may contribute to aberrant TGF-β signalling in SSc., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
249. Activating transcription factor 3 regulates canonical TGFβ signalling in systemic sclerosis.
- Author
-
Mallano T, Palumbo-Zerr K, Zerr P, Ramming A, Zeller B, Beyer C, Dees C, Huang J, Hai T, Distler O, Schett G, and Distler JH
- Subjects
- Adult, Aged, Animals, Blotting, Western, Case-Control Studies, Dermis cytology, Female, Fibrosis genetics, Fluorescent Antibody Technique, Gene Expression Profiling, Gene Expression Regulation, Humans, Immunohistochemistry, Male, Mice, Mice, Knockout, Middle Aged, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins c-jun metabolism, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta, Reverse Transcriptase Polymerase Chain Reaction, Scleroderma, Systemic metabolism, Signal Transduction genetics, Transcription Factor AP-1 metabolism, Young Adult, Activating Transcription Factor 3 genetics, Fibroblasts metabolism, Scleroderma, Systemic genetics, Smad3 Protein metabolism, Transforming Growth Factor beta metabolism
- Abstract
Background: Activating transcription factor 3 (ATF3), a member of the ATF/cAMP-responsive element binding (CREB) family of transcription factors, regulates cellular response to stress including oxidative stress. The aim of this study was to analyse the role of ATF3 in fibroblast activation in systemic sclerosis (SSc)., Methods: ATF3 was analysed by reverse transcription quantitative PCR, western blot and immunohistochemistry. ATF3 knockout fibroblasts and mice were used to study the functional role of ATF3. Knockdown experiments, reporter assays and coimmunoprecipitation were performed to study the effects of ATF3 on Smad and activation protein 1 (AP-1) signalling. The role of c-Jun was analysed by costaining, specific inactivation and coimmunoprecipitation., Results: Transforming growth factor-β (TGFβ) upregulates the expression of ATF3 in SSc fibroblasts. ATF3-deficient fibroblasts were less sensitive to TGFβ, whereas ectopic expression of ATF3 enhanced the profibrotic effects of TGFβ. Mechanistically, ATF3 interacts with Smad3 directly on stimulation with TGFβ and regulates Smad activity in a c-Jun-dependent manner. Knockout of ATF3 protected mice from bleomycin-induced fibrosis and fibrosis induced by overexpression of a constitutively active TGFβ receptor I. Reporter assays and analyses of the expression of Smad target genes demonstrated that binding of ATF3 regulates the transcriptional activity of Smad3., Conclusions: We demonstrate for the first time a key role for ATF3 in fibrosis. Knockout of the ATF3 gene reduced the stimulatory effect of TGFβ on fibroblasts by interfering with canonical Smad signalling and protected the mice from experimental fibrosis in two different models. ATF3 might thus be a candidate for molecular targeted therapies for SSc., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
250. From pathogenesis to therapy--Perspective on treatment strategies in fibrotic diseases.
- Author
-
Ramming A, Dees C, and Distler JH
- Subjects
- Animals, Cytokines metabolism, Endothelium drug effects, Endothelium metabolism, Endothelium pathology, Extracellular Matrix drug effects, Extracellular Matrix metabolism, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis metabolism, Humans, Inflammation drug therapy, Inflammation metabolism, Inflammation pathology, Skin drug effects, Skin metabolism, Skin pathology, Fibrosis drug therapy, Fibrosis pathology
- Abstract
Although fibrosis is becoming increasingly recognized as a major cause of morbidity and mortality in modern societies, there are very few treatment strategies available that specifically target the pathogenesis of fibrosis. Early in disease, inflammation and vascular changes and an increase in reactive oxygen species play pivotal roles. After inflammation has subsided, fibrosis and scarring are predominant in later phases. Fibrosis is driven by a complex, not-yet fully understood interplay between inflammatory cells on one hand and endothelium and fibroblasts on the other hand. The latter are regarded as the key players due to their extensive synthesis of extracellular matrix components which results in skin and organ fibrosis. Various cytokines orchestrate altered functions of the mentioned cell types. There are promising targets with therapeutic potential that have been extensively characterized in recent years connected with the hope to translate these preclinical results into clinical practice., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.