Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nature ; 629(8010): 184-192, 2024 May.
Article in English | MEDLINE | ID: mdl-38600378

ABSTRACT

Glucocorticoids represent the mainstay of therapy for a broad spectrum of immune-mediated inflammatory diseases. However, the molecular mechanisms underlying their anti-inflammatory mode of action have remained incompletely understood1. Here we show that the anti-inflammatory properties of glucocorticoids involve reprogramming of the mitochondrial metabolism of macrophages, resulting in increased and sustained production of the anti-inflammatory metabolite itaconate and consequent inhibition of the inflammatory response. The glucocorticoid receptor interacts with parts of the pyruvate dehydrogenase complex whereby glucocorticoids provoke an increase in activity and enable an accelerated and paradoxical flux of the tricarboxylic acid (TCA) cycle in otherwise pro-inflammatory macrophages. This glucocorticoid-mediated rewiring of mitochondrial metabolism potentiates TCA-cycle-dependent production of itaconate throughout the inflammatory response, thereby interfering with the production of pro-inflammatory cytokines. By contrast, artificial blocking of the TCA cycle or genetic deficiency in aconitate decarboxylase 1, the rate-limiting enzyme of itaconate synthesis, interferes with the anti-inflammatory effects of glucocorticoids and, accordingly, abrogates their beneficial effects during a diverse range of preclinical models of immune-mediated inflammatory diseases. Our findings provide important insights into the anti-inflammatory properties of glucocorticoids and have substantial implications for the design of new classes of anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents , Glucocorticoids , Inflammation , Macrophages , Mitochondria , Succinates , Animals , Female , Humans , Male , Mice , Anti-Inflammatory Agents/pharmacology , Carboxy-Lyases/metabolism , Carboxy-Lyases/antagonists & inhibitors , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Cytokines/immunology , Cytokines/metabolism , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Inflammation/drug therapy , Inflammation/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Receptors, Glucocorticoid/metabolism , Succinates/metabolism , Enzyme Activation/drug effects
2.
Sci Rep ; 10(1): 8428, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32439961

ABSTRACT

Bone turnover, which is determined by osteoclast-mediated bone resorption and osteoblast-mediated bone formation, represents a highly energy consuming process. The metabolic requirements of osteoblast differentiation and mineralization, both essential for regular bone formation, however, remain incompletely understood. Here we identify the nuclear receptor peroxisome proliferator-activated receptor (PPAR) δ as key regulator of osteoblast metabolism. Induction of PPARδ was essential for the metabolic adaption and increased rate in mitochondrial respiration necessary for the differentiation and mineralization of osteoblasts. Osteoblast-specific deletion of PPARδ in mice, in turn, resulted in an altered energy homeostasis of osteoblasts, impaired mineralization and reduced bone mass. These data show that PPARδ acts as key regulator of osteoblast metabolism and highlight the relevance of cellular metabolic rewiring during osteoblast-mediated bone formation and bone-turnover.


Subject(s)
Bone Remodeling/physiology , Osteoblasts/metabolism , Osteogenesis/physiology , PPAR delta/genetics , PPAR delta/metabolism , Animals , Bone Density/physiology , Cell Differentiation , Cells, Cultured , Energy Metabolism/genetics , Energy Metabolism/physiology , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Mitochondria/metabolism , Osteoblasts/cytology , Osteoclasts/metabolism , Oxidative Phosphorylation
3.
J Bone Miner Res ; 33(11): 2035-2047, 2018 11.
Article in English | MEDLINE | ID: mdl-29949664

ABSTRACT

NR4A1 (Nur77 or NGFI-B), an orphan member of the nuclear receptor superfamily, has been identified as a key regulator of the differentiation and function of myeloid, lymphoid, and mesenchymal cells. The detailed role of NR4A1 in bone biology is incompletely understood. Here, we report a role for NR4A1 as novel factor controlling the migration and recruitment of osteoclast precursors during bone remodeling. Myeloid-specific but not osteoblast-specific deletion of NR4A1 resulted in osteopenia due to an increase in the number of bone-lining osteoclasts. Although NR4A1-deficient osteoclast precursors displayed a regular differentiation into mature osteoclasts, they showed a hyper-motile phenotype that was largely dependent on increased osteopontin expression, suggesting that expression of NR4A1 negatively controlled osteopontin-mediated recruitment of osteoclast precursors to the trabecular bone. Pharmacological activation of NR4A1, in turn, inhibited osteopontin expression and osteopontin-dependent migration of osteoclast precursors resulted in reduced abundance of bone-resorbing osteoclasts in vivo as well as in an ameliorated bone loss after ovariectomy in mice. This study identifies NR4A1 as a crucial player in the regulation of osteoclast biology and bone remodeling and highlights this nuclear receptor as a promising target for therapeutic intervention during the treatment of osteoporosis. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.


Subject(s)
Bone Remodeling , Cell Movement , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Animals , Bone Resorption/pathology , Cancellous Bone/metabolism , Cell Count , Cell Differentiation , Cell Fusion , Gene Deletion , Homeostasis , Mice, Inbred C57BL , Myeloid Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Osteoblasts/metabolism , Osteopontin/metabolism , Ovariectomy , Repressor Proteins/metabolism
4.
Ann Rheum Dis ; 76(1): 244-251, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27113414

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)ß receptor I. RESULT: The expression of TWIST1 was increased in fibroblasts in fibrotic human and murine skin in a TGFß/SMAD3-dependent manner. TWIST1 in turn enhanced TGFß-induced fibroblast activation in a p38-dependent manner. The stimulatory effects of TWIST1 on resident fibroblasts were mediated by TWIST1 homodimers. TGFß 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 TGFß signalling. Targeting of TWIST1 may thus be a novel approach to normalise aberrant TGFß signalling in SSc.


Subject(s)
Fibroblasts/metabolism , Nuclear Proteins/physiology , Scleroderma, Systemic/metabolism , Twist-Related Protein 1/physiology , 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
5.
Ann Rheum Dis ; 75(3): 586-92, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25589515

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-ß (TGFß) upregulates the expression of ATF3 in SSc fibroblasts. ATF3-deficient fibroblasts were less sensitive to TGFß, whereas ectopic expression of ATF3 enhanced the profibrotic effects of TGFß. Mechanistically, ATF3 interacts with Smad3 directly on stimulation with TGFß 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 TGFß 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 TGFß 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.


Subject(s)
Activating Transcription Factor 3/genetics , Fibroblasts/metabolism , Scleroderma, Systemic/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , 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
6.
Ann Rheum Dis ; 75(3): 609-16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25603829

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-ß (TGF-ß)/Smad-dependent manner. Overexpression of TRB3 stimulated canonical TGF-ß signalling and induced an activated phenotype in resting fibroblasts. In contrast, knockdown of TRB3 reduced the profibrotic effects of TGF-ß and decreased the collagen synthesis. Moreover, siRNA-mediated knockdown of TRB3 exerted potent antifibrotic effects and ameliorated bleomycin as well as constitutively active TGF-ß 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-ß induces TRB3, which in turn activates canonical TGF-ß/Smad signalling and stimulates the release of collagen, thereby inducing a positive feedback loop that may contribute to aberrant TGF-ß signalling in SSc.


Subject(s)
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 , 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
7.
Ann Rheum Dis ; 75(1): 226-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25180292

ABSTRACT

BACKGROUND: Sirt1 is a member of the sirtuin family of proteins. Sirt1 is a class III histone deacetylase with important regulatory roles in transcription, cellular differentiation, proliferation and metabolism. As aberrant epigenetic modifications have been linked to the pathogenesis of systemic sclerosis (SSc), we aimed to investigate the role of Sirt1 in fibroblast activation. METHODS: Sirt1 expression was analysed by real-time PCR, western blot and immunohistochemistry. Sirt1 signalling was modulated with the Sirt1 agonist resveratrol and by fibroblast-specific knockout. The role of Sirt1 was evaluated in bleomycin-induced skin fibrosis and in mice overexpressing a constitutively active transforming growth fac-tor-ß (TGF-ß) receptor I (TBRIact). RESULTS: The expression of Sirt1 was decreased in patients with SSc and in experimental fibrosis in a TGF-ß-dependent manner. Activation of Sirt1 potentiated the profibrotic effects of TGF-ß with increased Smad reporter activity, elevated transcription of TGF-ß target genes and enhanced release of collagen. In contrast, knockdown of Sirt1 inhibited TGF-ß/SMAD signalling and reduced release of collagen in fibroblasts. Consistently, mice with fibroblast-specific knockdown of Sirt1 were less susceptible to bleomycin- or TBRIact-induced fibrosis. CONCLUSIONS: We identified Sirt1 as a crucial regulator of TGF-ß/Smad signalling in SSc. Although Sirt1 is downregulated, this decrease is not sufficient to counterbalance the excessive activation of TGF-ß signalling in SSc. However, augmentation of this endogenous regulatory mechanism, for example, by knockdown of Sirt1, can effectively inhibit TGF-ß signalling and exerts potent antifibrotic effects. Sirt1 may thus be a key regulator of fibroblast activation in SSc.


Subject(s)
Fibroblasts/metabolism , Scleroderma, Systemic/metabolism , Sirtuin 1/physiology , Skin/pathology , Transforming Growth Factor beta/physiology , Adult , Aged , Animals , Bleomycin , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Down-Regulation/physiology , Female , Fibrosis , Humans , Male , Mice, Knockout , Middle Aged , Scleroderma, Systemic/pathology , Signal Transduction/physiology , Skin/metabolism
8.
Ann Rheum Dis ; 75(5): 883-90, 2016 May.
Article in English | MEDLINE | ID: mdl-25858641

ABSTRACT

BACKGROUND: Nintedanib is a tyrosine kinase inhibitor that has recently been shown to slow disease progression in idiopathic pulmonary fibrosis in two replicate phase III clinical trials. The aim of this study was to analyse the antifibrotic effects of nintedanib in preclinical models of systemic sclerosis (SSc) and to provide a scientific background for clinical trials in SSc. METHODS: The effects of nintedanib on migration, proliferation, myofibroblast differentiation and release of extracellular matrix of dermal fibroblasts were analysed by microtitre tetrazolium and scratch assays, stress fibre staining, qPCR and SirCol assays. The antifibrotic effects of nintedanib were evaluated in bleomycin-induced skin fibrosis, in a murine sclerodermatous chronic graft-versus-host disease model and in tight-skin-1 mice. RESULTS: Nintedanib dose-dependently reduced platelet-derived growth factor-induced and transforming growth factor-ß-induced proliferation and migration as well as myofibroblast differentiation and collagen release of dermal fibroblasts from patients with and healthy individuals. Nintedanib also inhibited the endogenous activation of SSc fibroblasts. Nintedanib prevented bleomycin-induced skin fibrosis in a dose-dependent manner and was also effective in the treatment of established fibrosis. Moreover, treatment with nintedanib ameliorated fibrosis in the chronic graft-versus-host disease model and in tight-skin-1 mice in well-tolerated doses. CONCLUSIONS: We demonstrate that nintedanib effectively inhibits the endogenous as well as cytokine-induced activation of SSc fibroblasts and exerts potent antifibrotic effects in different complementary mouse models of SSc. These data have direct translational implications for clinical trials with nintedanib in SSc.


Subject(s)
Fibroblasts/drug effects , Indoles/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Scleroderma, Systemic/drug therapy , Animals , Bleomycin , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Fibroblasts/pathology , Fibroblasts/physiology , Fibrosis , Graft vs Host Disease/prevention & control , Humans , Indoles/administration & dosage , Indoles/pharmacology , Male , Mice, Inbred BALB C , Mice, Mutant Strains , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Scleroderma, Systemic/pathology , Skin/drug effects , Skin/pathology
9.
Rheumatology (Oxford) ; 54(12): 2273-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26231346

ABSTRACT

OBJECTIVES: IL-35 is a member of the IL-12 family consisting of p35/IL-12a and EBI3/IL-27b subunits. IL-35 exerts immunomodulatory activities in experimental and human autoimmune inflammatory conditions. Our aim was to assess IL-35 expression in the skin and circulation of SSc patients and to characterize its potential association with SSc-related features. METHODS: Expression of IL-35 in skin and dermal fibroblasts was quantified by quantitative PCR, immunohistochemistry and immunofluorescence. Serum levels of IL-35 (by ELISA), CRP (by turbidimetry), ANA (by immunofluorescence) and autoantibodies of the ENA complex (by immunoblot) were measured in 40 SSc patients. Serum IL-35 was determined in 40 age- and sex-matched healthy controls. RESULTS: IL-35 expression was increased in SSc skin and dermal fibroblasts in a TGF-ß-dependent manner. IL-35 induced an activated phenotype in resting fibroblasts and enhanced the release of collagen. IL-35 serum levels were increased in patients with SSc compared with healthy controls [median 83.9 (interquartile range 45.1-146.1) vs 36.2 (interquartile range 17.2-49.4) pg/ml, P < 0.0001]. Serum IL-35 was negatively correlated with disease duration (r = -0.4339, P = 0.0052). In line with this finding, serum IL-35 was increased in patients with an early SSc pattern on capillaroscopy assessment compared with those with active and late SSc patterns. CONCLUSION: The present study demonstrates overexpression of IL-35 in SSc skin, dermal fibroblasts and serum. TGF-ß induces IL-35, which in turn activates resting fibroblasts and enhances the release of collagen, thereby contributing to aberrant TGF-ß signalling in SSc. Increased serum IL-35 is associated with early, inflammatory stages of SSc.


Subject(s)
Interleukins/biosynthesis , Scleroderma, Systemic/immunology , Adult , Aged , Case-Control Studies , Cells, Cultured , Collagen/biosynthesis , Female , Fibroblasts/immunology , Humans , Interleukins/blood , Interleukins/genetics , Male , Middle Aged , RNA, Messenger/genetics , Skin/immunology , Transforming Growth Factor beta/immunology , Up-Regulation/immunology
10.
Ann Rheum Dis ; 74(8): 1621-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25817717

ABSTRACT

OBJECTIVES: Stimulators of the soluble guanylate cyclase (sGC) have recently been shown to inhibit transforming growth factor-ß signalling. Here, we aimed to demonstrate that riociguat, the drug candidate for clinical trials in systemic sclerosis (SSc), is effective in experimental fibrosis and to compare its efficacy to that of phosphodiesterase V inhibitors that also increase the intracellular levels of cyclic guanosine monophosphate. METHODS: The antifibrotic effects of riociguat and sildenafil were compared in the tight-skin 1 model, in bleomycin-induced fibrosis and in a model of sclerodermatous chronic graft-versus-host-disease (cGvHD). Doses of 0.1-3 mg/kg twice a day for riociguat and of 3-10 mg/kg twice a day for sildenafil were used. RESULT: Riociguat dose-dependently reduced skin thickening, myofibroblast differentiation and accumulation of collagen with potent antifibrotic effects at 1 and 3 mg/kg. Riociguat also ameliorated fibrosis of the gastrointestinal tract in the cGvHD model. The antifibrotic effects were associated with reduced phosphorylation of extracellular signal-regulated kinases. Sildenafil at doses of 3 and 10 mg/kg exerted mild antifibrotic effects that were significantly less pronounced compared with 1 and 3 mg/kg riociguat. CONCLUSIONS: These data demonstrated potent antifibrotic effects of riociguat on experimental skin and organ fibrosis. These findings suggest a role for riociguat for the treatment of fibrotic diseases, especially for the treatment of SSc. A phase II study with riociguat in patients with SSc is currently starting.


Subject(s)
Guanylate Cyclase/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Skin/pathology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrosis , Mice , Mice, Inbred Strains , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Scleroderma, Systemic/drug therapy , Sildenafil Citrate/pharmacology
11.
Nat Med ; 21(2): 150-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25581517

ABSTRACT

Mesenchymal responses are an essential aspect of tissue repair. Failure to terminate this repair process correctly, however, results in fibrosis and organ dysfunction. Therapies that block fibrosis and restore tissue homeostasis are not yet available for clinical use. Here we characterize the nuclear receptor NR4A1 as an endogenous inhibitor of transforming growth factor-ß (TGF-ß) signaling and as a potential target for anti-fibrotic therapies. NR4A1 recruits a repressor complex comprising SP1, SIN3A, CoREST, LSD1, and HDAC1 to TGF-ß target genes, thereby limiting pro-fibrotic TGF-ß effects. Even though temporary upregulation of TGF-ß in physiologic wound healing induces NR4A1 expression and thereby creates a negative feedback loop, the persistent activation of TGF-ß signaling in fibrotic diseases uses AKT- and HDAC-dependent mechanisms to inhibit NR4A1 expression and activation. Small-molecule NR4A1 agonists can overcome this lack of active NR4A1 and inhibit experimentally-induced skin, lung, liver, and kidney fibrosis in mice. Our data demonstrate a regulatory role of NR4A1 in TGF-ß signaling and fibrosis, providing the first proof of concept for targeting NR4A1 in fibrotic diseases.


Subject(s)
Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Liver Cirrhosis, Alcoholic/metabolism , Liver/metabolism , Lung/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Scleroderma, Systemic/metabolism , Skin/metabolism , Transforming Growth Factor beta/metabolism , Adolescent , Adult , Aged , Animals , Case-Control Studies , Cells, Cultured , Co-Repressor Proteins/metabolism , Female , Fibrosis , Histone Deacetylase 1/metabolism , Histone Demethylases/metabolism , Humans , Idiopathic Pulmonary Fibrosis/pathology , Liver/pathology , Liver Cirrhosis, Alcoholic/pathology , Lung/pathology , Male , Mice , Mice, Knockout , Middle Aged , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Repressor Proteins/metabolism , Scleroderma, Systemic/pathology , Signal Transduction , Sin3 Histone Deacetylase and Corepressor Complex , Skin/cytology , Skin/pathology , Sp1 Transcription Factor/metabolism , Wound Healing , Young Adult
12.
Arthritis Rheumatol ; 67(2): 508-16, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25371068

ABSTRACT

OBJECTIVE: Cardiomyopathy has emerged as a leading cause of death in patients with systemic sclerosis (SSc). However, the pathogenesis of SSc-related cardiomyopathy is poorly understood, and new therapies as well as platforms for testing are needed. The aim of this study was to characterize the histopathologic features of cardiomyopathy in patients with SSc and in common mouse models of SSc. METHODS: The histopathologic features of myocardial tissue specimens obtained at autopsy from 5 subjects with SSc and 5 control subjects matched for sex, age, and cardiovascular risk factors were evaluated and compared with those of myocardial tissue specimens obtained from 3 common mouse models of SSc with systemic manifestations: Fra-2-transgenic mice, mice with sclerodermatous chronic graft-versus-host disease (GVHD), and TSK-1 mice. RESULTS: Myocardial tissue from autopsy subjects with SSc and no clinically manifest cardiac involvement showed endothelial cell apoptosis with reduced capillary density, perivascular inflammation, myofibroblast differentiation, and accumulation of collagen. Only selected features of SSc-related cardiomyopathy were observed in the mice with chronic GVHD and TSK-1 mice. However, the myocardial tissue of Fra-2-transgenic mice mimicked all features of SSc-related cardiomyopathy and also demonstrated comparable vascular, inflammatory, and fibrotic manifestations. Of note, the expression of Fra-2 was also increased in the myocardium of autopsy subjects with SSc. CONCLUSION: We demonstrate that all typical manifestations of SSc-related cardiomyopathy are mimicked in Fra-2-transgenic mice. Moreover, overexpression of Fra-2 in the myocardium of autopsy subjects with SSc may suggest similar underlying pathogenic mechanisms. Thus, Fra-2-transgenic mice might be a suitable preclinical model with which to study the mechanisms of and therapeutic approaches to myocardial involvement in SSc.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/pathology , Disease Models, Animal , Myocardium/pathology , Scleroderma, Systemic/complications , Scleroderma, Systemic/pathology , Adult , Animals , Apoptosis , Autopsy , Cardiomyopathies/epidemiology , Cardiovascular Diseases/epidemiology , Case-Control Studies , Chronic Disease , Female , Fos-Related Antigen-2/genetics , Fos-Related Antigen-2/metabolism , Graft vs Host Disease/complications , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Middle Aged , Myocardium/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Risk Factors
13.
Ann Rheum Dis ; 74(9): 1748-55, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24709861

ABSTRACT

OBJECTIVES: S100A4 is a calcium binding protein with regulatory functions in cell homeostasis, proliferation and differentiation that has been shown to promote cancer progression and metastasis. In the present study, we evaluated the role of S100A4 in fibroblast activation in systemic sclerosis (SSc). METHODS: The expression of S100A4 was analysed in human samples, murine models of SSc and in cultured fibroblasts by real-time PCR, immunohistochemistry and western blot. The functional role of S100A4 was evaluated using siRNA, overexpression, recombinant protein and S100A4 knockout (S100A4(-/-)) mice. Transforming growth factor ß (TGF-ß) signalling was assessed by reporter assays, staining for phosphorylated Smad2/3 and analyses of target genes. RESULTS: The expression of S100A4 was increased in SSc skin and in experimental fibrosis in a TGF-ß/Smad-dependent manner. Overexpression of S100A4 or stimulation with recombinant S100A4 induced an activated phenotype in resting normal fibroblasts. In contrast, knockdown of S100A4 reduced the pro-fibrotic effects of TGF-ß and decreased the release of collagen. S100A4(-/-) mice were protected from bleomycin-induced skin fibrosis with reduced dermal thickening, decreased hydroxyproline content and lower myofibroblast counts. Deficiency of S100A4 also ameliorated fibrosis in the tight-skin-1 (Tsk-1) mouse model. CONCLUSIONS: We characterised S100A4 as a downstream mediator of the stimulatory effects of TGF-ß on fibroblasts in SSc. TGF-ß induces the expression of S100A4 to stimulate the release of collagen in SSc fibroblasts and induce fibrosis. Since S100A4 is essentially required for the pro-fibrotic effects of TGF-ß and neutralising antibodies against S100A4 are currently evaluated, S100A4 might be a candidate for novel antifibrotic therapies.


Subject(s)
Fibroblasts/metabolism , S100 Proteins/metabolism , Scleroderma, Systemic/metabolism , Skin/metabolism , Transforming Growth Factor beta/metabolism , Adult , Aged , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , S100 Calcium-Binding Protein A4 , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Young Adult
14.
Ann Rheum Dis ; 74(7): 1408-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24567525

ABSTRACT

OBJECTIVES: We have previously described the antifibrotic role of the soluble guanylate cyclase (sGC). The mode of action, however, remained elusive. In the present study, we describe a novel link between sGC signalling and transforming growth factor ß (TGFß) signalling that mediates the antifibrotic effects of the sGC. METHODS: Human fibroblasts and murine sGC knockout fibroblasts were treated with the sGC stimulator BAY 41-2272 or the stable cyclic guanosine monophosphate (cGMP) analogue 8-Bromo-cGMP and stimulated with TGFß. sGC knockout fibroblasts were isolated from sGCI(fl/fl) mice, and recombination was induced by Cre-adenovirus. In vivo, we studied the antifibrotic effects of BAY 41-2272 in mice overexpressing a constitutively active TGF-ß1 receptor. RESULTS: sGC stimulation inhibited TGFß-dependent fibroblast activation and collagen release. sGC knockout fibroblasts confirmed that the sGC is essential for the antifibrotic effects of BAY 41-2272. Furthermore, 8-Bromo-cGMP reduced TGFß-dependent collagen release. While nuclear p-SMAD2 and 3 levels, SMAD reporter activity and transcription of classical TGFß target genes remained unchanged, sGC stimulation blocked the phosphorylation of ERK. In vivo, sGC stimulation inhibited TGFß-driven dermal fibrosis but did not change p-SMAD2 and 3 levels and TGFß target gene expression, confirming that non-canonical TGFß pathways mediate the antifibrotic sGC activity. CONCLUSIONS: We elucidated the antifibrotic mode of action of the sGC that increases cGMP levels, blocks non-canonical TGFß signalling and inhibits experimental fibrosis. Since sGC stimulators have shown excellent efficacy and tolerability in phase 3 clinical trials for pulmonary arterial hypertension, they may be further developed for the simultaneous treatment of fibrosis and vascular disease in systemic sclerosis.


Subject(s)
Fibroblasts/pathology , Guanylate Cyclase/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Signal Transduction/physiology , Skin/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Case-Control Studies , Cells, Cultured , Collagen/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/prevention & control , Guanylate Cyclase/deficiency , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/metabolism , Scleroderma, Systemic/metabolism , Signal Transduction/drug effects , Skin/metabolism , Smad Proteins/metabolism , Soluble Guanylyl Cyclase , Transforming Growth Factor beta/metabolism
15.
Ann Rheum Dis ; 74(6): 1317-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24618263

ABSTRACT

OBJECTIVES: To investigate the role of liver X receptors (LXRs) in experimental skin fibrosis and evaluate their potential as novel antifibrotic targets. METHODS: We studied the role of LXRs in bleomycin-induced skin fibrosis, in the model of sclerodermatous graft-versus-host disease (sclGvHD) and in tight skin-1 (Tsk-1) mice, reflecting different subtypes of fibrotic disease. We examined both LXR isoforms using LXRα-, LXRß- and LXR-α/ß-double-knockout mice. Finally, we investigated the effects of LXRs on fibroblasts and macrophages to establish the antifibrotic mode of action of LXRs. RESULTS: LXR activation by the agonist T0901317 had antifibrotic effects in bleomycin-induced skin fibrosis, in the sclGvHD model and in Tsk-1 mice. The antifibrotic activity of LXRs was particularly prominent in the inflammation-driven bleomycin and sclGvHD models. LXRα-, LXRß- and LXRα/ß-double-knockout mice showed a similar response to bleomycin as wildtype animals. Low levels of the LXR target gene ABCA-1 in the skin of bleomycin-challenged and control mice suggested a low baseline activation of the antifibrotic LXR signalling, which, however, could be specifically activated by T0901317. Fibroblasts were not the direct target cells of LXRs agonists, but LXR activation inhibited fibrosis by interfering with infiltration of macrophages and their release of the pro-fibrotic interleukin-6. CONCLUSIONS: We identified LXRs as novel targets for antifibrotic therapies, a yet unknown aspect of these nuclear receptors. Our data suggest that LXR activation might be particularly effective in patients with inflammatory disease subtypes. Activation of LXRs interfered with the release of interleukin-6 from macrophages and, thus, inhibited fibroblast activation and collagen release.


Subject(s)
Fibroblasts/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , Orphan Nuclear Receptors/genetics , Scleroderma, Diffuse/metabolism , Skin Diseases/metabolism , Skin/pathology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Disease Models, Animal , Fibroblasts/drug effects , Fibrosis , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors , Macrophages/drug effects , Mice , Mice, Knockout , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , Skin/drug effects , Skin Diseases/chemically induced , Sulfonamides/pharmacology
16.
Ann Rheum Dis ; 74(3): e20, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24448349

ABSTRACT

BACKGROUND: Vitamin D receptor (VDR) is a member of the nuclear receptor superfamily. Its ligand, 1,25-(OH)2D, is a metabolically active hormone derived from vitamin D3. The levels of vitamin D3 are decreased in patients with systemic sclerosis (SSc). Here, we aimed to analyse the role of VDR signalling in fibrosis. METHODS: VDR expression was analysed in SSc skin, experimental fibrosis and human fibroblasts. VDR signalling was modulated by siRNA and with the selective agonist paricalcitol. The effects of VDR on Smad signalling were analysed by reporter assays, target gene analyses and coimmunoprecipitation. The effects of paricalcitol were evaluated in the models of bleomycin-induced fibrosis and fibrosis induced by overexpression of a constitutively active transforming growth factor-ß (TGF-ß) receptor I (TBRI(CA)). RESULTS: VDR expression was decreased in fibroblasts of SSc patients and murine models of SSc in a TGF-ß-dependent manner. Knockdown of VDR enhanced the sensitivity of fibroblasts towards TGF-ß. In contrast, activation of VDR by paricalcitol reduced the stimulatory effects of TGF-ß on fibroblasts and inhibited collagen release and myofibroblast differentiation. Paricalcitol stimulated the formation of complexes between VDR and phosphorylated Smad3 in fibroblasts to inhibit Smad-dependent transcription. Preventive and therapeutic treatment with paricalcitol exerted potent antifibrotic effects and ameliorated bleomycin- as well as TBRI(CA)-induced fibrosis. CONCLUSIONS: We characterise VDR as a negative regulator of TGF-ß/Smad signalling. Impaired VDR signalling with reduced expression of VDR and decreased levels of its ligand may thus contribute to hyperactive TGF-ß signalling and aberrant fibroblast activation in SSc.


Subject(s)
Fibroblasts/metabolism , Receptors, Calcitriol/metabolism , Scleroderma, Systemic/metabolism , Signal Transduction/physiology , Skin/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adult , Aged , Animals , Bleomycin/toxicity , Disease Models, Animal , Ergocalciferols/pharmacology , Female , Fibroblasts/drug effects , Fibrosis/chemically induced , Fibrosis/metabolism , Humans , Male , Mice , Middle Aged , RNA, Small Interfering/metabolism , Receptors, Calcitriol/agonists , Signal Transduction/drug effects , Skin/drug effects , Skin/pathology , Smad Proteins/drug effects , Transforming Growth Factor beta/drug effects , Young Adult
17.
Arthritis Rheumatol ; 66(11): 3140-50, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25155144

ABSTRACT

OBJECTIVE: Nuclear receptors regulate cell growth, differentiation, and homeostasis. Selective nuclear receptors promote fibroblast activation, which leads to tissue fibrosis, the hallmark of systemic sclerosis (SSc). This study was undertaken to investigate the effects of constitutive androstane receptor (CAR)/NR1I3, an orphan nuclear receptor, on fibroblast activation and experimental dermal fibrosis. METHODS: CAR expression was quantified by quantitative polymerase chain reaction, Western blotting, immunohistochemistry, and immunofluorescence. CAR expression was modulated by small molecules, small interfering RNA, forced overexpression, and site-directed mutagenesis. The effects of CAR activation were analyzed in cultured fibroblasts, in bleomycin-induced dermal fibrosis, and in mice overexpressing a constitutively active transforming growth factor ß (TGFß) receptor type I (TßRI-CA). RESULTS: Up-regulation of CAR was detected in the skin and in dermal fibroblasts in SSc patients. Stimulation of healthy fibroblasts with TGFß induced the expression of CAR messenger RNA and protein in a Smad-dependent manner. Pharmacologic activation or overexpression of CAR in healthy fibroblasts significantly increased the stimulatory effects of TGFß on collagen synthesis and myofibroblast differentiation, and amplified the stimulatory effects of TGFß on COL1A2 transcription activity. Treatment with CAR agonist increased the activation of canonical TGFß signaling in murine models of SSc and exacerbated bleomycin-induced and TßRI-CA-induced fibrosis with increased dermal thickening, myofibroblast counts, and collagen accumulation. CONCLUSION: Our findings indicate that CAR is up-regulated in SSc and regulates TGFß signaling. Activation of CAR increases the profibrotic effects of TGFß in cultured fibroblasts and in different preclinical models of SSc. Thus, inactivation of CAR might be a novel approach to target aberrant TGFß signaling in SSc and in other fibrotic diseases.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Scleroderma, Systemic/metabolism , Skin/metabolism , Skin/pathology , Transforming Growth Factor beta/metabolism , Adult , Aged , Animals , Bleomycin/adverse effects , Cells, Cultured , Collagen Type I/metabolism , Constitutive Androstane Receptor , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/chemically induced , Fibrosis/metabolism , Fibrosis/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Receptors, Transforming Growth Factor beta/metabolism , Scleroderma, Systemic/pathology , Signal Transduction/physiology , Up-Regulation/physiology
18.
Ann Rheum Dis ; 73(6): 1264-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24445254

ABSTRACT

OBJECTIVES: The morphogen pathways Hedgehog, Wnt and Notch are attractive targets for antifibrotic therapies in systemic sclerosis. Interference with stem cell regeneration, however, may complicate the use of morphogen pathway inhibitors. We therefore tested the hypothesis that combination therapies with low doses of Hedgehog, Wnt and Notch inhibitors maybe safe and effective for the treatment of fibrosis. METHODS: Skin fibrosis was induced by bleomycin and by overexpression of a constitutively active TGF-ß receptor type I. Adverse events were assessed by clinical monitoring, pathological evaluation and quantification of Lgr5-positive intestinal stem cells. RESULTS: Inhibition of Hedgehog, Wnt and Notch signalling dose-dependently ameliorated bleomycin-induced and active TGF-ß receptor type I-induced fibrosis. Combination therapies with low doses of Hedgehog/Wnt inhibitors or Hedgehog/Notch inhibitors demonstrated additive antifibrotic effects in preventive as well as in therapeutic regimes. Combination therapies were well tolerated. In contrast with high dose monotherapies, combination therapies did not reduce the number of Lgr5 positive intestinal stem cells. CONCLUSIONS: Combined inhibition of morphogen pathways exerts additive antifibrotic effects. Combination therapies are well tolerated and, in contrast to high dose monotherapies, may not impair stem cell renewal. Combined targeting of morphogen pathways may thus help to overcome dose-limiting toxicity of Hedgehog, Wnt and Notch signalling.


Subject(s)
Fibrosis/drug therapy , Hedgehog Proteins/antagonists & inhibitors , Receptors, Notch/antagonists & inhibitors , Scleroderma, Systemic/drug therapy , Signal Transduction/drug effects , Skin/drug effects , Wnt Proteins/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Bleomycin , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Pyrimidinones/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Veratrum Alkaloids/pharmacology , Wnt Signaling Pathway/drug effects
19.
Ann Rheum Dis ; 73(6): 1232-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23698475

ABSTRACT

OBJECTIVES: Activated Wnt signalling with decreased expression of endogenous inhibitors has recently been characterised as a central pathomechanism in systemic sclerosis (SSc). Aberrant epigenetic modifications also contribute to the persistent activation of SSc fibroblasts. We investigated whether increased Wnt signalling and epigenetic changes in SSc are causally linked via promoter hypermethylation-induced silencing of Wnt antagonists. METHODS: The methylation status of endogenous Wnt antagonists in leucocytes and fibroblasts was evaluated by methylation-specific PCR. 5-aza-2'-deoxycytidine was used to inhibit DNA methyltransferases (Dnmts) in cultured fibroblasts and in the mouse model of bleomycin-induced skin fibrosis. Activation of Wnt signalling was assessed by analysing Axin2 mRNA levels and by staining for ß-catenin. RESULTS: The promoters of DKK1 and SFRP1 were hypermethylated in fibroblasts and peripheral blood mononuclear cells of patients with SSc. Promoter hypermethylation resulted in impaired transcription and decreased expression of DKK1 and SFRP1 in SSc. Treatment of SSc fibroblasts or bleomycin-challenged mice with 5-aza prevented promoter methylation-induced silencing and increased the expression of both genes to normal levels. Reactivation of DKK1 and SFRP1 transcription by 5-aza inhibited canonical Wnt signalling in vitro and in vivo and effectively ameliorated experimental fibrosis. CONCLUSIONS: We demonstrate that hypermethylation of the promoters of DKK1 and SFRP1 contributes to aberrant Wnt signalling in SSc and that Dnmt inhibition effectively reduces Wnt signalling. These data provide a novel link between epigenetic alterations and increased Wnt signalling in SSc and also have translational implications because Dnmt inhibitors are already approved for clinical use.


Subject(s)
DNA Methylation/genetics , Down-Regulation/genetics , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Leukocytes, Mononuclear/metabolism , Membrane Proteins/genetics , Scleroderma, Systemic/genetics , Wnt Signaling Pathway/genetics , Adult , Aged , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Case-Control Studies , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Decitabine , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Female , Fibroblasts/drug effects , Humans , Male , Mice , Middle Aged , Promoter Regions, Genetic/genetics , Wnt Signaling Pathway/drug effects , Young Adult
20.
Ann Rheum Dis ; 73(6): 1215-22, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23661493

ABSTRACT

OBJECTIVES: Targeted therapies for systemic sclerosis (SSc) and other fibrotic diseases are not yet available. We evaluated the efficacy of heat shock protein 90 (Hsp90) inhibition as a novel approach to inhibition of aberrant transforming growth factor (TGF)-ß signalling and for the treatment of fibrosis in preclinical models of SSc. METHODS: Expression of Hsp90 was quantified by quantitative PCR, western blot and immunohistochemistry. The effects of Hsp90 inhibition were analysed in cultured fibroblasts, in bleomycin-induced dermal fibrosis, in tight-skin (Tsk-1) mice and in mice overexpressing a constitutively active TGF-ß receptor I (TßRI). RESULTS: Expression of Hsp90ß was increased in SSc skin and in murine models of SSc in a TGF-ß-dependent manner. Inhibition of Hsp90 by 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG) inhibited canonical TGF-ß signalling and completely prevented the stimulatory effects of TGF-ß on collagen synthesis and myofibroblast differentiation. Treatment with 17-DMAG decreased the activation of canonical TGF-ß signalling in murine models of SSc and exerted potent antifibrotic effects in bleomycin-induced dermal fibrosis, in Tsk-1 mice and in mice overexpressing a constitutively active TßRI. Dermal thickness, number of myofibroblasts and hydroxyproline content were all significantly reduced on treatment with 17-DMAG. No toxic effects were observed with 17-DMAG at antifibrotic doses. CONCLUSIONS: Hsp90 is upregulated in SSc and is critical for TGF-ß signalling. Pharmacological inhibition of Hsp90 effectively blocks the profibrotic effects of TGF-ß in cultured fibroblasts and in different preclinical models of SSc. These results have translational implications, as several Hsp90 inhibitors are in clinical trials for other indications.


Subject(s)
Fibroblasts/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Scleroderma, Systemic/metabolism , Transforming Growth Factor beta/metabolism , Adult , Aged , Animals , Benzoquinones/pharmacology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/drug effects , Fibrosis , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Male , Mice , Mice, Transgenic , Middle Aged , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Scleroderma, Systemic/prevention & control , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism , Transforming Growth Factor beta/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...