Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 215
1.
Int J Mol Sci ; 23(10)2022 May 16.
Article En | MEDLINE | ID: mdl-35628342

Systemic sclerosis (SSc) is characterised by progressive multiple organ fibrosis leading to morbidity and mortality. Lysyl oxidases play a vital role in the cross-linking of collagens and subsequent build-up of fibrosis in the extracellular matrix. As such, their inhibition provides a novel treatment paradigm for SSc. A novel small molecule pan-lysyl oxidase inhibitor, PXS-5505, currently in clinical development for myelofibrosis treatment was evaluated using in vivo rodent models resembling the fibrotic conditions in SSc. Both lysyl oxidase and lysyl oxidase-like 2 (LOXL2) expression were elevated in the skin and lung of SSc patients. The oral application of PXS-5505 inhibited lysyl oxidase activity in the skin and LOXL2 activity in the lung. PXS-5505 exhibited anti-fibrotic effects in the SSc skin mouse model, reducing dermal thickness and α-smooth muscle actin. Similarly, in the bleomycin-induced mouse lung model, PXS-5505 reduced pulmonary fibrosis toward normal levels, mediated by its ability to normalise collagen/elastin crosslink formation. PXS-5505 also reduced fibrotic extent in models of the ischaemia-reperfusion heart, the unilateral ureteral obstruction kidney, and the CCl4-induced fibrotic liver. PXS-5505 consistently demonstrates potent anti-fibrotic efficacy in multiple models of organ fibrosis relevant to the pathogenesis of SSc, suggesting that it may be efficacious as a novel approach for treating SSc.


Collagen , Enzyme Inhibitors , Protein-Lysine 6-Oxidase , Scleroderma, Systemic , Animals , Collagen/antagonists & inhibitors , Collagen/metabolism , Disease Models, Animal , Fibrosis , Humans , Mice , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Protein-Lysine 6-Oxidase/metabolism , Rodentia/metabolism , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/enzymology
2.
Biochem Biophys Res Commun ; 549: 34-39, 2021 04 16.
Article En | MEDLINE | ID: mdl-33662666

Systemic sclerosis (SSc) is an inflammatory fibrotic disease characterized by an excessive extracellular matrix deposition in the skin and internal organs. One fibrotic key event remains the fibroblast-to-myofibroblast differentiation that is controlled by a combination of mechanical and soluble factors, such as transforming growth factor-ß1 (TGF-ß1) and interleukin-1ß (IL-1ß). One important myofibroblast biomarker is human xylosyltransferase-I (XT-I), the initial enzyme in proteoglycan biosynthesis. Increased serum XT activity was quantified in SSc, but the underlying cellular mechanisms remain elusive. This study aims to determine the cellular basis of XT-I induction in SSc by using a myofibroblast cell culture model with SSc fibroblasts (SScF) and healthy control fibroblasts. We found that SScF exhibit a higher extracellular XT-I activity compared to control fibroblasts. This increased XT-I activity in SScF was demonstrated to be mediated by an enhanced autocrine TGF-ß signaling. Upon IL-1ß treatment, SScF showed an increased mRNA expression level of XT-I and TGF-ß receptor II (TGFBR2), while healthy control fibroblasts did not, pointing towards an involvement of IL-1ß in the cytokine-mediated XT-I induction. Performing microRNA (miRNA) inhibition experiments in the presence of TGF-ß1, we showed that the pro-fibrotic effect of IL-1ß may be mediated by a miRNA-21/TGF-ß receptor II axis, enhancing the autocrine TGF-ß signaling in SScF. Taken together, this study improves the mechanistic understanding of fibrotic XT-I induction in SSc by identifying a hitherto unknown IL-1ß-mediated miRNA-21/TGFBR2 regulation contributing to the enhanced XYLT1 expression and XT-I activity in SScF.


Cytokines/pharmacology , Fibroblasts/enzymology , Fibroblasts/pathology , Pentosyltransferases/biosynthesis , Scleroderma, Systemic/enzymology , Skin/pathology , Enzyme Induction/drug effects , Fibroblasts/drug effects , Humans , Interleukin-1beta/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Pentosyltransferases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Scleroderma, Systemic/genetics , Scleroderma, Systemic/pathology , Transforming Growth Factor beta1/pharmacology , UDP Xylose-Protein Xylosyltransferase
3.
Transl Res ; 231: 139-158, 2021 05.
Article En | MEDLINE | ID: mdl-33422651

Systemic sclerosis (SSc) is an idiopathic autoimmune disease with a heterogeneous clinical phenotype ranging from limited cutaneous involvement to rapidly progressive diffuse SSc. The most severe SSc clinical and pathologic manifestations result from an uncontrolled fibrotic process involving the skin and various internal organs. The molecular mechanisms responsible for the initiation and progression of the SSc fibrotic process have not been fully elucidated. Recently it has been suggested that tyrosine protein kinases play a role. The implicated kinases include receptor-activated tyrosine kinases and nonreceptor tyrosine kinases. The receptor kinases are activated following specific binding of growth factors (platelet-derived growth factor, fibroblast growth factor, or vascular endothelial growth factor). Other receptor kinases are the discoidin domain receptors activated by binding of various collagens, the ephrin receptors that are activated by ephrins and the angiopoetin-Tie-2s receptors. The nonreceptor tyrosine kinases c-Abl, Src, Janus, and STATs have also been shown to participate in SSc-associated tissue fibrosis. Currently, there are no effective disease-modifying therapies for SSc-associated tissue fibrosis. Therefore, extensive investigation has been conducted to examine whether tyrosine kinase inhibitors (TKIs) may exert antifibrotic effects. Here, we review the role of receptor and nonreceptor tyrosine kinases in the pathogenesis of the frequently progressive cutaneous and systemic fibrotic alterations in SSc, and the potential of TKIs as SSc disease-modifying antifibrotic therapeutic agents.


Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/pathology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Protein-Tyrosine Kinases/genetics , Scleroderma, Systemic/drug therapy
4.
Am J Physiol Lung Cell Mol Physiol ; 320(1): L29-L40, 2021 01 01.
Article En | MEDLINE | ID: mdl-33026236

Pulmonary fibrosis is one of the important causes of morbidity and mortality in fibroproliferative disorders such as systemic sclerosis (SSc) and idiopathic pulmonary fibrosis (IPF). Lysyl oxidase (LOX) is a copper-dependent amine oxidase whose primary function is the covalent crosslinking of collagens in the extracellular matrix (ECM). We investigated the role of LOX in the pathophysiology of SSc. LOX mRNA and protein levels were increased in lung fibroblasts of SSc patients compared with healthy controls and IPF patients. In vivo, bleomycin induced LOX mRNA expression in lung tissues, and LOX activity increased in the circulation of mice with pulmonary fibrosis, suggesting that circulating LOX parallels levels in lung tissues. Circulating levels of LOX were reduced upon amelioration of fibrosis with an antifibrotic peptide. LOX induced ECM production at the transcriptional level in lung fibroblasts, human lungs, and human skin maintained in organ culture. In vivo, LOX synergistically exacerbated fibrosis in bleomycin-treated mice. Further, LOX increased the production of interleukin (IL)-6, and the increase was mediated by LOX-induced c-Fos expression, the nuclear localization of c-Fos, and its engagement with the IL-6 promoter region. Our findings demonstrate that LOX expression and activity correlate with fibrosis in vitro, ex vivo, and in vivo. LOX induced ECM production via upregulation of IL-6 and nuclear localization of c-Fos. Thus, LOX has a direct pathogenic role in SSc-associated fibrosis that is independent of its crosslinking function. Our findings also suggest that measuring circulating LOX levels and activity can be used for monitoring response to antifibrotic therapy.


Extracellular Matrix/pathology , Lung/pathology , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Fibrosis/pathology , Scleroderma, Systemic/pathology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Case-Control Studies , Extracellular Matrix/enzymology , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Interleukin-6/metabolism , Lung/enzymology , Male , Mice , Mice, Inbred C57BL , Protein-Lysine 6-Oxidase/genetics , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/etiology , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/etiology
5.
Int J Rheum Dis ; 22(12): 2119-2124, 2019 Dec.
Article En | MEDLINE | ID: mdl-31637859

AIM: It is commonly assumed that a genetically determined polymorphism of xenobiotic biotransformation plays a particular role in the development of such disease entities in which chemical compounds and environmental pollutants are relevant etiologic factors. Systemic sclerosis (SSc, scleroderma) belongs to diseases of connective tissue, characterized by chronic inflammation developing on an autoimmune background. The current state of knowledge on the etiopathogenesis of autoimmune diseases indicates the existence of many factors affecting the development of the disease, including factors of the external environment. Considering all the above, a study on a role of genetic polymorphisms of glutathione S-transferase has been undertaken in which predisposition to SSc in a Polish population was assessed. METHODS: The study was carried out in 161 subjects: 61 patients with SSc and 100 healthy volunteers. A determination of the polymorphism of GSTM1 and GSTT1 was performed with a multiplex PCR (polymerase chain reaction). The GSTP1 polymorphism was determined by using the PCR restriction fragment length polymorphism. RESULTS: The risk of developing SSc was 3-fold higher for persons with the null GSTM1 and GSTT1 genotypes (odds ratio [OR] = 3.3; P = .0051). The risk for SSc was also demonstrated to be over 2.5-fold greater in the GSTP1 Ile/Val genotype individuals (OR = 2.62; P = .0037). Carriers of the GSTP1 Val variant allele had a greater than 2-fold increase in SSc risk (OR = 2.41; P = .0006). CONCLUSION: The genetic polymorphism of glutathione S-transferase may affect the risk of SSc in a Polish population.


Glutathione S-Transferase pi/genetics , Glutathione Transferase/genetics , Polymorphism, Genetic , Scleroderma, Systemic/genetics , Adolescent , Adult , Aged , Case-Control Studies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Phenotype , Poland/epidemiology , Risk Assessment , Risk Factors , Scleroderma, Systemic/diagnosis , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/epidemiology , Young Adult
6.
Rheumatology (Oxford) ; 58(9): 1547-1555, 2019 09 01.
Article En | MEDLINE | ID: mdl-30770717

OBJECTIVE: Lysyl oxidase (LOX) is an extracellular enzyme that cross-links collagen fibrils. LOX was found to be increased in serum of SSc patients and was suggested to be related to skin fibrosis, yet a vascular source of LOX has been demonstrated in idiopathic pulmonary arterial hypertension (iPAH). We aimed to validate elevated LOX serum levels in SSc and to study its correlation with clinical characteristics and investigate its main source at the tissue level. METHODS: A total of 86 established SSc patients were compared with 86 patients with very early diagnosis of systemic sclerosis (VEDOSS), 110 patients with primary RP (PRP) and 80 healthy controls. LOX serum levels were determined by ELISA. Five lung and 12 skin biopsies from SSc patients were stained for LOX and compared with controls. RESULTS: Serum levels of LOX in SSc were significantly higher than in VEDOSS, PRP and healthy controls (P < 0.001). LOX inversely correlated with the diffusing capacity of the lung for carbon monoxide diffusing capacity (DLCO) in diffuse SSc (r = -0.376, P = 0.02). Patients with moderate to severe estimated systolic PAH had higher LOX levels (P < 0.01). Lung biopsies demonstrated intense LOX staining in SSc patients with PAH that was predominantly located in the endothelium of the remodelled pulmonary vessels. CONCLUSION: Serum LOX levels are increased in established SSc and inversely correlate with the DLCO. LOX is elevated in patients with moderate to severe PAH and is located in the proliferating endothelium in lung arterioles, suggesting a possible role for LOX in SSc-associated PAH.


Hypertension, Pulmonary/etiology , Protein-Lysine 6-Oxidase/physiology , Scleroderma, Systemic/complications , Adult , Biopsy , Case-Control Studies , Female , Fibrosis , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Lung/enzymology , Lung/pathology , Male , Middle Aged , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Diffusing Capacity/physiology , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Skin/enzymology , Skin/pathology
7.
Rheumatology (Oxford) ; 58(4): 708-718, 2019 04 01.
Article En | MEDLINE | ID: mdl-30608617

OBJECTIVE: Ubiquitination of proteins leads to their degradation by the proteasome, and is regulated by ubiquitin ligases and substrate-specific ubiquitin-specific peptidases (USPs). The ubiquitination process also plays important roles in the regulation of cell metabolism and cell cycle. Here, we found that the expression of several USPs is increased in SSc tenosynovial and skin biopsies, and we demonstrated that USP inhibition decreases TGF-ß signalling in primary fibroblast cell lines. METHODS: High-density transcriptomic studies were performed using total RNA obtained from SSc tenosynovial samples. Confirmatory immunostaining experiments were performed on tenosynovial and skin samples. In vitro experiments were conducted in order to study the influence of USP modulation on responses to TGF-ß stimulation. RESULTS: Tenosynovial biopsies from SSc patients overexpressed known disease-associated gene pathways: fibrosis, cytokines and chemokines, and Wnt/TGF-ß signalling, but also several USPs. Immunohistochemistry experiments confirmed the detection of USPs in the same samples, and in SSc skin biopsies. Exposure of primary fibroblast cell lines to TGF-ß induced USP gene expression. The use of a pan-USP inhibitor decreased SMAD3 phosphorylation, and expression of COL1A1, COL3A1 and fibronectin gene expression in TGF-ß-stimulated fibroblasts. The effect of the USP inhibitor resulted in increased SMAD3 ubiquitination, and was blocked by a proteasome inhibitor, thereby confirming the specificity of its action. CONCLUSION: Overexpression of several USPs, including USP15, amplifies fibrotic responses induced by TGF-ß, and is a potential therapeutic target in SSc.


Fibroblasts/metabolism , Scleroderma, Systemic/enzymology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Ubiquitin-Specific Proteases/metabolism , Humans , Scleroderma, Systemic/drug therapy
8.
Clin Exp Rheumatol ; 36 Suppl 113(4): 28-35, 2018.
Article En | MEDLINE | ID: mdl-29465357

OBJECTIVES: To evaluate the effect of autologous stem cell transplantation (aSCT) on antibody (ab) reactivity towards linear epitopes of topoisomerase-I (topo-I/Scl70) in patients with systemic sclerosis (SSc) and to correlate antibody reactivities with clinical outcome after aSCT. METHODS: Fourteen anti-topo-I/Scl70-positive SSc-patients were analysed before and after non-myeloablative aSCT. Five patients showed ongoing good response (group 1), 9 had primarily responded but later relapsed or did not respond (group 2). Patients' sera were tested by ELISA against full length (fl) topo-I and 45 overlapping 25-mer peptides. Furthermore, for comparison sera from patients with anti-topo-I-negative SSc (n=12), other collagen disorders (n=6), and from 21 healthy controls (HC) were analysed. RESULTS: Anti-topo-I-positive SSc-sera showed significantly higher IgG-reactivity as compared to HC towards 34 of the 45 peptides. Especially peptide 39 (aa647-671) emerged as a immunodominant epitope being recognised predominantly by anti-topo-I-positive SSc-sera. Reactivity towards 17 of the 45 peptides decreased after aSCT in group 1- and 2-patients. Before aSCT, group 1-patients had lower antibody reactivity towards peptide 39 than group 2-patients. There was no change in peptide-specificity after aSCT. CONCLUSIONS: Reactivity towards topo-I-epitopes is heterogeneous in SSc, but peptide 39 (aa647-671) may be another immunodominant epitope besides the published epitope aa489-573. Antibody reactivity to this peptide 39 was higher in group 2- than in group 1-patients. Peptide recognition pattern did not change after aSCT.


Autoantibodies/immunology , DNA Topoisomerases, Type I/immunology , Immunodominant Epitopes , Immunoglobulin G/immunology , Scleroderma, Systemic/surgery , Stem Cell Transplantation , Adult , Antibody Specificity , Autoantibodies/blood , Biomarkers/blood , Female , Humans , Immunoglobulin G/blood , Male , Middle Aged , Scleroderma, Systemic/blood , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/immunology , Transplantation, Autologous , Treatment Outcome , Young Adult
9.
Ann Rheum Dis ; 77(5): 744-751, 2018 05.
Article En | MEDLINE | ID: mdl-29431122

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-ß (TGFß) 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 TGFß-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 TGFß/Smad target genes. Inhibition of PARP-1 enhanced the effect of TGFß 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 TGFß signalling in experimental skin fibrosis. The downregulation of PARP-1 in SSc fibroblasts may thus directly contribute to hyperactive TGFß signalling and to persistent fibroblast activation in SSc.


Fibroblasts/physiology , Fibrosis/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Scleroderma, Systemic/genetics , Skin Diseases/genetics , 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
10.
Am J Respir Cell Mol Biol ; 58(1): 28-39, 2018 01.
Article En | MEDLINE | ID: mdl-28800254

Pulmonary fibrosis is the leading cause of death in systemic sclerosis (SSc). Sirtuin1 (SIRT1) is a deacetylase with known antiinflammatory and antifibrotic activity in the liver, kidney, and skin. The role of SIRT1 in SSc-related pulmonary fibrosis is unknown. In the present work, we determined that the expression of SIRT1 in peripheral blood mononuclear cells of patients with SSc with pulmonary fibrosis is lower than that in patients with SSc without pulmonary fibrosis. In in vivo studies of bleomycin-induced lung fibrosis in mice, SIRT1 activation with resveratrol reduced collagen production when it was administered either prophylactically during the inflammatory stage or after the development of fibrosis. Furthermore, SIRT1 activation or overexpression inhibited tumor necrosis factor-α-induced inflammatory responses in vitro in human fetal lung fibroblasts, depletion of SIRT1 in fibroblasts enhanced inflammation, and these effects were related to changes in the acetylation of NF-κB. In addition, SIRT1 activation or exogenous overexpression inhibited collagen production in vitro, and these manipulations also inhibited fibrosis via inactivation of transforming growth factor-ß/mothers against decapentaplegic homolog and mammalian target of rapamycin signaling. Taken together, our results show that a loss of SIRT1 may participate in the pathogenesis of SSc-related pulmonary fibrosis, and that SIRT1 activation is an effective treatment for both the early (inflammatory) and late (fibrotic) stages of pulmonary fibrosis. Thus, SIRT1 may be a promising therapeutic target in the management of SSc-related pulmonary fibrosis.


Pulmonary Fibrosis , Scleroderma, Systemic , Sirtuin 1/metabolism , Animals , Cell Line , Enzyme Activation , Female , Humans , Male , Mice , NF-kappa B/metabolism , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/prevention & control , Scleroderma, Systemic/complications , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/prevention & control , Tumor Necrosis Factor-alpha/metabolism
11.
Int J Rheum Dis ; 21(11): 1915-1923, 2018 Nov.
Article En | MEDLINE | ID: mdl-27990763

AIM: To quantify circulating fibroblast activation protein (cFAP) and dipeptidyl peptidase 4 (cDPP4) protease activities in patients with rheumatoid arthritis (RA), systemic sclerosis (SSc), and a control group with mechanical back pain and to correlate plasma levels with disease characteristics. METHODS: Plasma was collected from patients with RA (n = 73), SSc (n = 37) and control subjects (n = 26). DPP4 and FAP were quantified using specific enzyme activity assays. RESULTS: Median cDPP4 was significantly lower in the RA group (P = 0.02), and SSc group (P = 0.002) compared with controls. There were no significant differences in median cFAP between the three groups. DPP4 and FAP demonstrated a negative correlation with inflammatory markers and duration of disease. There were no associations with disease subtypes in RA, including seropositive and erosive disease. Decreased cDPP4 was found in SSc patients with myositis. Plasma FAP was lower in RA patients receiving prednisone (P = 0.001) or leflunomide (P = 0.04), but higher with biologic agents (P = 0.01). RA patients receiving leflunomide also had decreased cDPP4 (P = 0.014). SSc patients receiving prednisone (P = 0.02) had lower cDPP4 but there was no association with cFAP. CONCLUSIONS: No association was found between cFAP and RA or SSc. Plasma DPP4 was decreased in RA and SSc when compared with controls. cDPP4 and cFAP correlated negatively with inflammatory markers and there were no significant correlations with disease characteristics in this RA cohort.


Arthritis, Rheumatoid/blood , Dipeptidyl Peptidase 4/blood , Gelatinases/blood , Membrane Proteins/blood , Scleroderma, Systemic/blood , Serine Endopeptidases/blood , Aged , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Biological Products/therapeutic use , Biomarkers/blood , Case-Control Studies , Endopeptidases , Female , Glucocorticoids/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Inflammation Mediators/blood , Male , Middle Aged , Scleroderma, Systemic/diagnosis , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/enzymology
12.
Ann Rheum Dis ; 77(1): 150-158, 2018 Jan.
Article En | MEDLINE | ID: mdl-29070530

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 (TGFß)-dependent manner. Inactivation of JMJD3 reversed the activated fibroblast phenotype in SSc fibroblasts and prevented the activation of healthy dermal fibroblasts by TGFß. 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.


Fibroblasts/enzymology , Jumonji Domain-Containing Histone Demethylases/metabolism , Scleroderma, Systemic/enzymology , Adult , Aged , Animals , Bleomycin , Case-Control Studies , Cells, Cultured , Enzyme Activation , Female , Fibrosis/chemically induced , Fibrosis/enzymology , Humans , Male , Mice , Middle Aged , Young Adult
13.
Nat Commun ; 8(1): 1060, 2017 10 20.
Article En | MEDLINE | ID: mdl-29057934

Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of skin and internal organs. Protein tyrosine phosphatases have received little attention in the study of SSc or fibrosis. Here, we show that the tyrosine phosphatase PTP4A1 is highly expressed in fibroblasts from patients with SSc. PTP4A1 and its close homolog PTP4A2 are critical promoters of TGFß signaling in primary dermal fibroblasts and of bleomycin-induced fibrosis in vivo. PTP4A1 promotes TGFß signaling in human fibroblasts through enhancement of ERK activity, which stimulates SMAD3 expression and nuclear translocation. Upstream from ERK, we show that PTP4A1 directly interacts with SRC and inhibits SRC basal activation independently of its phosphatase activity. Unexpectedly, PTP4A2 minimally interacts with SRC and does not promote the SRC-ERK-SMAD3 pathway. Thus, in addition to defining PTP4A1 as a molecule of interest for TGFß-dependent fibrosis, our study provides information regarding the functional specificity of different members of the PTP4A subclass of phosphatases.


Immediate-Early Proteins/metabolism , MAP Kinase Signaling System , Protein Tyrosine Phosphatases/metabolism , Scleroderma, Systemic/enzymology , Transforming Growth Factor beta/physiology , Animals , Cell Line , Dermis/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Smad3 Protein/metabolism
14.
Curr Rheumatol Rep ; 19(5): 28, 2017 May.
Article En | MEDLINE | ID: mdl-28397126

PURPOSE OF REVIEW: The pathogenesis of systemic sclerosis depends on a complex interplay between autoimmunity, vasculopathy, and fibrosis. Reversible phosphorylation on tyrosine residues, in response to growth factors and other stimuli, critically regulates each one of these three key pathogenic processes. Protein tyrosine kinases, the enzymes that catalyze addition of phosphate to tyrosine residues, are known players in systemic sclerosis, and tyrosine kinase inhibitors are undergoing clinical trials for treatment of this disease. Until recently, the role of tyrosine phosphatases-the enzymes that counteract the action of tyrosine kinases by removing phosphate from tyrosine residues-in systemic sclerosis has remained largely unknown. Here, we review the function of tyrosine phosphatases in pathways relevant to the pathogenesis of systemic sclerosis and their potential promise as therapeutic targets to halt progression of this debilitating rheumatic disease. RECENT FINDINGS: Protein tyrosine phosphatases are emerging as important regulators of a multitude of signaling pathways and undergoing validation as molecular targets for cancer and other common diseases. Recent advances in drug discovery are paving the ways to develop new classes of tyrosine phosphatase modulators to treat human diseases. Although so far only few reports have focused on tyrosine phosphatases in systemic sclerosis, these enzymes play a role in multiple pathways relevant to disease pathogenesis. Further studies in this field are warranted to explore the potential of tyrosine phosphatases as drug targets for systemic sclerosis.


Molecular Targeted Therapy/methods , Protein Tyrosine Phosphatases/physiology , Scleroderma, Systemic/enzymology , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/therapeutic use , Fibrosis , Growth Substances/physiology , Humans , Protein Tyrosine Phosphatases/antagonists & inhibitors , Receptors, Interleukin/immunology , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/immunology , Signal Transduction/immunology , Translational Research, Biomedical/methods
15.
Exp Dermatol ; 26(1): 73-81, 2017 01.
Article En | MEDLINE | ID: mdl-27576129

Although there is increasing evidence that oxidative stress is involved in collagen synthesis and myofibroblast activation, the NADPH oxidase (Nox) system is incompletely investigated in the context of human dermal fibroblasts (HDFs) and skin fibrosis. Using the pan-Nox inhibitor diphenyleneiodonium (DPI) as an initial tool, we show that gene expression of collagen type I, α-smooth muscle actin (α-SMA) and fibronectin 1 is suppressed in HDFs. Detailed expression analysis of all Nox isoforms and adaptors revealed expression of RNA and protein expression of Nox4, p22phox and Poldip2 but neither Nox1 nor Nox2. Nox4 could be immunolocalized to the endoplasmic reticulum. Importantly, TGF-ß1 had a dose- and time-dependent upregulating effect on NADH activity and Nox4 gene expression in HDFs. Genetic silencing of Nox4 as demonstrated by siRNA in HDFs as well as in murine fibroblasts established from Nox4 knockout mice confirmed that TGF-ß1 -mediated collagen type I gene, α-SMA and fibronectin 1 gene expressions were Nox4-dependent. This TGF-ß1 effect was mediated by Smad3 as shown by in silico promoter analysis, pharmacological inhibition and gene silencing of Smad3. The relevance of these findings is highlighted in the bleomycin-induced scleroderma mouse model. DPI treatment attenuated skin fibrosis and myofibroblast activation. Moreover, Nox4 knockdown by siRNA reduced skin collagen synthesis, α-SMA and fibronectin 1 expression in vivo. Finally, analyses of HDFs from patients with systemic sclerosis confirmed the expression of Nox4 and its adaptors, whereas Nox1 and Nox2 were not detectable. Our findings indicate that Nox4 targeting is a promising future treatment for fibrotic skin diseases.


Fibroblasts/enzymology , NADPH Oxidase 4/genetics , Scleroderma, Systemic/enzymology , Skin/enzymology , Skin/pathology , Actins/genetics , Adult , Animals , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Computer Simulation , Cytokines/genetics , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Fibronectins , Fibrosis , Gene Expression/drug effects , Gene Expression Profiling , Gene Silencing , Humans , Infant, Newborn , Isoenzymes/genetics , Male , Mice , Middle Aged , Multienzyme Complexes/metabolism , Myofibroblasts , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1/genetics , NADPH Oxidase 2/genetics , NADPH Oxidase 4/metabolism , Onium Compounds/pharmacology , Primary Cell Culture , RNA, Messenger/metabolism , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/pathology , Transforming Growth Factor beta/pharmacology , Young Adult
16.
Oncotarget ; 7(43): 69321-69336, 2016 Oct 25.
Article En | MEDLINE | ID: mdl-27732568

Constitutive fibroblast activation is responsible for organ fibrosis in fibrotic disorders including systemic sclerosis (SSc), but the underlying mechanisms are not fully understood, and effective therapies are lacking. We investigated the expression of the mitochondrial deacetylase sirtuin 3 (SIRT3) and its modulation by hexafluoro, a novel fluorinated synthetic honokiol analogue, in the context of fibrosis. We find that augmenting cellular SIRT3 by forced expression in normal lung and skin fibroblasts, or by hexafluoro treatment, blocked intracellular TGF-ß signaling and fibrotic responses, and mitigated the activated phenotype of SSc fibroblasts. Moreover, hexafluoro attenuated mitochondrial and cytosolic reactive oxygen species (ROS) accumulation in TGF-ß-treated fibroblasts. Remarkably, we found that the expression of SIRT3 was significantly reduced in SSc skin biopsies and explanted fibroblasts, and was suppressed by TGF-ß treatment in normal fibroblasts. Moreover, tissue levels of acetylated MnSOD, a sensitive marker of reduced SIRT3 activity, were dramatically enhanced in lesional skin and lung biopsies from SSc patients. Mice treated with hexafluoro showed substantial attenuation of bleomycin-induced fibrosis in the lung and skin. Our findings reveal a cell-autonomous function for SIRT3 in modulating fibrotic responses, and demonstrate the ability of a novel pharmacological SIRT3 agonist to attenuate fibrosis in vitro and in vivo. In light of the impaired expression and activity of SIRT3 associated with organ fibrosis in SSc, pharmacological approaches for augmenting SIRT3 might have therapeutic potential.


Lung/enzymology , Scleroderma, Systemic/enzymology , Sirtuin 3/metabolism , Skin/enzymology , Adult , Aged , Animals , Bleomycin , Cells, Cultured , Enzyme Activation/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibrosis/chemically induced , Fibrosis/prevention & control , Humans , Hydrocarbons, Fluorinated/pharmacology , Lung/pathology , Male , Mice, Inbred C57BL , Middle Aged , RNA Interference , Scleroderma, Systemic/genetics , Scleroderma, Systemic/pathology , Sirtuin 3/genetics , Skin/pathology
17.
Rheumatology (Oxford) ; 55(11): 2066-2073, 2016 11.
Article En | MEDLINE | ID: mdl-27411481

OBJECTIVE: SSc is a disease characterized by inflammation and fibrosis. Heme Oxygenase-1 (HO-1) is a haem-degrading enzyme that mediates resolution of inflammation and is induced upon mediators abundantly present in SSc. We aimed to assess whether HO-1 expression/function is disturbed in SSc patients and could therefore be contributing to the ongoing inflammation. METHODS: In total, 92 SSc patients and 48 healthy controls were included. By measuring total bilirubin in plasma in vivo, HO-activity was assessed. HO-1 expression levels were determined with western blot in monocytes before and after induction of HO-1 with cobalt protoporphyrin (CoPP) with or without CXCL4. Monocyte-derived dendritic cells (DCs) were stimulated with several Toll-like receptor (TLR) ligands with or without pre-stimulation with CoPP for 24 h. Cytokine levels were measured in the supernatants using the Luminex Bead Array. RESULTS: SSc patients have lower plasma levels of bilirubin, suggestive of an aberrant HO-1 function. We demonstrated low HO-1 expression in immune cells from SSc patients, whereas induction with CoPP was able to restore HO-1 levels in DCs from SSc patients, almost normalizing the increased TLR response observed in SSc. Co-exposure to CXCL4 completely abrogated CoPP-induced HO-1 expression, suggesting that the high CXCL4 levels present in SSc patients block the normal induction of HO-1 and its function. CONCLUSION: We demonstrate that HO activity in SSc patients is decreased and show its functional consequences. Since CXCL4 blocks the induction of HO-1 expression, neutralization of CXCL4 in SSc patients could have clinical benefits by diminishing overactivation of immune cells and other anti-inflammatory effects of HO-1.


Heme Oxygenase-1/deficiency , Platelet Factor 4/physiology , Scleroderma, Systemic/enzymology , Toll-Like Receptors/physiology , Adult , Bilirubin/metabolism , Carbon Monoxide/metabolism , Case-Control Studies , Cytokines/metabolism , Dendritic Cells/metabolism , Female , Fibroblasts/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Male
18.
Clin Sci (Lond) ; 130(8): 575-86, 2016 Apr.
Article En | MEDLINE | ID: mdl-26744410

Fibroblasts derived from the lungs of patients with idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc) produce low levels of prostaglandin (PG) E2, due to a limited capacity to up-regulate cyclooxygenase-2 (COX-2). This deficiency contributes functionally to the fibroproliferative state, however the mechanisms responsible are incompletely understood. In the present study, we examined whether the reduced level of COX-2 mRNA expression observed in fibrotic lung fibroblasts is regulated epigenetically. The DNA methylation inhibitor, 5-aza-2'-deoxycytidine (5AZA) restored COX-2 mRNA expression by fibrotic lung fibroblasts dose dependently. Functionally, this resulted in normalization of fibroblast phenotype in terms of PGE2 production, collagen mRNA expression and sensitivity to apoptosis. COX-2 methylation assessed by bisulfite sequencing and methylation microarrays was not different in fibrotic fibroblasts compared with controls. However, further analysis of the methylation array data identified a transcriptional regulator, chromosome 8 open reading frame 4 (thyroid cancer protein 1, TC-1) (c8orf4), which is hypermethylated and down-regulated in fibrotic fibroblasts compared with controls. siRNA knockdown of c8orf4 in control fibroblasts down-regulated COX-2 and PGE2 production generating a phenotype similar to that observed in fibrotic lung fibroblasts. Chromatin immunoprecipitation demonstrated that c8orf4 regulates COX-2 expression in lung fibroblasts through binding of the proximal promoter. We conclude that the decreased capacity of fibrotic lung fibroblasts to up-regulate COX-2 expression and COX-2-derived PGE2 synthesis is due to an indirect epigenetic mechanism involving hypermethylation of the transcriptional regulator, c8orf4.


Cyclooxygenase 2/genetics , DNA Methylation , Epigenesis, Genetic , Fibroblasts/enzymology , Lung/enzymology , Neoplasm Proteins/genetics , Pulmonary Fibrosis/genetics , Scleroderma, Systemic/genetics , Aged , Binding Sites , Case-Control Studies , Cell Proliferation , Cells, Cultured , Cyclooxygenase 2/metabolism , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , Genotype , Humans , Lung/drug effects , Lung/pathology , Male , Middle Aged , Neoplasm Proteins/metabolism , Phenotype , Promoter Regions, Genetic , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/pathology , Transcription, Genetic , Transfection
20.
Clin Exp Rheumatol ; 33(4 Suppl 91): S15-22, 2015.
Article En | MEDLINE | ID: mdl-26148346

OBJECTIVES: The pathogenesis of fibrosis in scleroderma (SSc) is unknown. TGF-ß and platelet-derived growth factor are important in the development of fibrosis and tyrosine kinases are involved in these pathways. The possible antifibrotic effects of various kinase inhibitors in SSc have been studied before. Spleen tyro-sine kinase (Syk) is a protein tyrosine kinase which activates intracellular signal transduction pathways; and has been claimed to be involved in the pathogenesis of systemic autoimmune diseases. Inhibition of Syk suppresses IgE- and IgG-associated FcR signal activation in various cell types; and suppresses experimental arthritis and skin and kidney disease in lupus-prone mice. We investigated the ability of a small drug, the Syk inhibitor, fostamatinib, to protect mice from bleomycin-induced SSc. METHODS: Four study groups of BALB/c mice were included into this study: control, bleomycin (administered subcutaneously to BALB/c mice for 21 days), bleomycin and fostamatinib (mice fed with chow containing a Syk inhibitor for 21 days), and fostamatinib alone groups. Skin and lung tissue specimens were obtained and evaluated histologically. RESULTS: Treatment with fostamatinib significantly reduced skin thickness and fibrosis. Mice treated with fostamatinib also displayed less fibrosis and inflammation in the lung tissue. Following fostamatinib treatment, Syk, phospho-Syk, and TGF-ß expression decreased in both skin and lung tissues. CONCLUSIONS: The Syk inhibitor fostamatinib prevented bleomycin-induced fibrosis and inflammation in the skin and in the lung. The anti-fibrotic effect of fostamatinib is linked to reduced Syk phosphorylation and TGF-ß expression. The Syk pathway appears as a potential molecular target for therapeutic intervention in SSc.


Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lung/drug effects , Oxazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pulmonary Fibrosis/prevention & control , Pyridines/pharmacology , Scleroderma, Systemic/prevention & control , Skin/drug effects , Aminopyridines , Animals , Bleomycin , Cytoprotection , Disease Models, Animal , Immunoglobulin E/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung/enzymology , Lung/immunology , Lung/pathology , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Mice, Inbred C57BL , Morpholines , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Pyrimidines , Receptors, Fc/metabolism , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Signal Transduction/drug effects , Skin/enzymology , Skin/immunology , Skin/pathology , Syk Kinase , Transforming Growth Factor beta/metabolism
...