Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Ann Rheum Dis ; 76(1): 244-251, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27113414

RESUMEN

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.


Asunto(s)
Fibroblastos/metabolismo , Proteínas Nucleares/fisiología , Esclerodermia Sistémica/metabolismo , Proteína 1 Relacionada con Twist/fisiología , Animales , Estudios de Casos y Controles , Femenino , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones Noqueados , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Multimerización de Proteína/fisiología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Esclerodermia Sistémica/patología , Transducción de Señal/fisiología , Piel/patología , Factor de Crecimiento Transformador beta/farmacología , Proteína 1 Relacionada con Twist/biosíntesis , Proteína 1 Relacionada con Twist/deficiencia , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
2.
Ann Rheum Dis ; 76(8): 1467-1475, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28478401

RESUMEN

OBJECTIVES: Janus kinase 2 (JAK2) has recently been described as a novel downstream mediator of the pro-fibrotic effects of transforming growth factor-ß. Although JAK2 inhibitors are in clinical use for myelodysplastic syndromes, patients often rapidly develop resistance. Tumour cells can escape the therapeutic effects of selective JAK2 inhibitors by mutation-independent transactivation of JAK2 by JAK1. Here, we used selective JAK2 inhibition as a model to test the hypothesis that chronic treatment may provoke resistance by facilitating non-physiological signalling pathways in fibroblasts. METHODS: The antifibrotic effects of long-term treatment with selective JAK2 inhibitors and reactivation of JAK2 signalling by JAK1-dependent transphosphorylation was analysed in cultured fibroblasts and experimental dermal and pulmonary fibrosis. Combined JAK1/JAK2 inhibition and co-treatment with an HSP90 inhibitor were evaluated as strategies to overcome resistance. RESULTS: The antifibrotic effects of selective JAK2 inhibitors on fibroblasts decreased with prolonged treatment as JAK2 signalling was reactivated by JAK1-dependent transphosphorylation of JAK2. This reactivation could be prevented by HSP90 inhibition, which destabilised JAK2 protein, or with combined JAK1/JAK2 inhibitors. Treatment with combined JAK1/JAK2 inhibitors or with JAK2 inhibitors in combination with HSP90 inhibitors was more effective than monotherapy with JAK2 inhibitors in bleomycin-induced pulmonary fibrosis and in adTBR-induced dermal fibrosis. CONCLUSION: Fibroblasts can develop resistance to chronic treatment with JAK2 inhibitors by induction of non-physiological JAK1-dependent transactivation of JAK2 and that inhibition of this compensatory signalling pathway, for example, by co-inhibition of JAK1 or HSP90 is important to maintain the antifibrotic effects of JAK2 inhibition with long-term treatment.


Asunto(s)
Fibroblastos/efectos de los fármacos , Janus Quinasa 1/efectos de los fármacos , Janus Quinasa 2/efectos de los fármacos , Pulmón/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Fibrosis Pulmonar/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Esclerodermia Sistémica , Sulfonamidas/farmacología , Adulto , Animales , Antibióticos Antineoplásicos/toxicidad , Benzoquinonas/farmacología , Bleomicina/toxicidad , Western Blotting , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Inmunohistoquímica , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/metabolismo , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Lactamas Macrocíclicas/farmacología , Pulmón/patología , Masculino , Ratones , Persona de Mediana Edad , Nitrilos , Fosforilación/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Crecimiento Transformador beta/farmacología
3.
Ann Rheum Dis ; 75(6): 1203-10, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26113650

RESUMEN

OBJECTIVES: Autophagy has recently been shown to regulate osteoclast activity and osteoclast differentiation. Here, we aim to investigate the impact of autophagy inhibition as a potential therapeutic approach for the treatment of osteoporosis in preclinical models. METHODS: Systemic bone loss was induced in mice by glucocorticoids and by ovariectomy (OVX). Autophagy was targeted by conditional inactivation of autophagy-related gene 7 (Atg7) and by treatment with chloroquine (CQ). Bone density was evaluated by microCT. The role of autophagy on osteoclastogenesis was analysed by osteoclastogenesis and bone resorption assays. The quantification of receptor activator of nuclear factor κ B ligand and osteoprotegerin proteins in cocultures was performed using ELISA whereas that of osteoclast and osteoblast differentiation markers was by qPCR. RESULTS: Selective deletion of Atg7 in monocytes from Atg7(fl/fl)_x_LysM-Cre mice mitigated glucocorticoid-induced and OVX-induced osteoclast differentiation and bone loss compared with Atg7(fl/fl) littermates. Pharmacological inhibition of autophagy by treatment with CQ suppressed glucocorticoid-induced osteoclastogenesis and protected mice from bone loss. Similarly, inactivation of autophagy shielded mice from OVX-induced bone loss. Inhibition of autophagy led to decreased osteoclast differentiation with lower expression of osteoclast markers such as NFATc1, tartrate-resistant acid phosphatase, OSCAR and cathepsin K and attenuated bone resorption in vitro. In contrast, osteoblast differentiation was not affected by inhibition of autophagy. CONCLUSIONS: Pharmacological or genetic inactivation of autophagy ameliorated glucocorticoid-induced and OVX-induced bone loss by inhibiting osteoclastogenesis. These findings may have direct translational implications for the treatment of osteoporosis, since inhibitors of autophagy such as CQ are already in clinical use.


Asunto(s)
Autofagia/efectos de los fármacos , Osteoporosis/prevención & control , Animales , Proteína 7 Relacionada con la Autofagia/genética , Células Cultivadas , Cloroquina/farmacología , Cloroquina/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Glucocorticoides , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia Molecular Dirigida , Monocitos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Osteogénesis/efectos de los fármacos , Osteoporosis/inducido químicamente , Osteoporosis/etiología , Osteoporosis/patología , Ovariectomía
4.
Ann Rheum Dis ; 75(3): 609-16, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25603829

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/genética , Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Represoras/genética , Esclerodermia Sistémica/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Anciano , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Estudios de Casos y Controles , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Colágeno/metabolismo , Dermis/citología , Modelos Animales de Enfermedad , Femenino , Fibrosis/inducido químicamente , Fibrosis/genética , Técnicas de Sustitución del Gen , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Masculino , Ratones , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta , Proteínas Represoras/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal/genética , Enfermedades de la Piel/inducido químicamente , Enfermedades de la Piel/genética , Adulto Joven
5.
Ann Rheum Dis ; 75(5): 883-90, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25858641

RESUMEN

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.


Asunto(s)
Fibroblastos/efectos de los fármacos , Indoles/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Esclerodermia Sistémica/tratamiento farmacológico , Animales , Bleomicina , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Fibroblastos/patología , Fibroblastos/fisiología , Fibrosis , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Indoles/administración & dosificación , Indoles/farmacología , Masculino , Ratones Endogámicos BALB C , Ratones Mutantes , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Esclerodermia Sistémica/patología , Piel/efectos de los fármacos , Piel/patología
6.
Ann Rheum Dis ; 75(11): 2037-2044, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26851274

RESUMEN

OBJECTIVES: Notch ligands and receptors have recently been shown to be differentially expressed in osteoarthritis (OA). We aim to further elucidate the functional role of Notch signalling in OA using Notch1 antisense transgenic (Notch1 AS) mice. METHODS: Notch and hedgehog signalling were analysed by real-time PCR and immunohistochemistry. Notch-1 AS mice were employed as a model of impaired Notch signalling in vivo. Experimental OA was induced by destabilisation of the medial meniscus (DMM). The extent of cartilage destruction and osteophyte formation was analysed by safranin-O staining with subsequent assessment of the Osteoarthritis Research Society International (OARSI) and Mankin scores and µCT scanning. Collagen X staining was used as a marker of chondrocyte hypertrophy. The role of hairy/enhancer of split 1 (Hes-1) was investigated with knockdown and overexpression experiments. RESULTS: Notch signalling was activated in human and murine OA with increased expression of Jagged1, Notch-1, accumulation of the Notch intracellular domain 1 and increased transcription of Hes-1. Notch1 AS mice showed exacerbated OA with increases in OARSI scores, osteophyte formation, increased subchondral bone plate density, collagen X and osteocalcin expression and elevated levels of Epas1 and ADAM-TS5 mRNA. Inhibition of the Notch pathway induced activation of hedgehog signalling with induction of Gli-1 and Gli-2 and increased transcription of hedgehog target genes. The regulatory effects of Notch signalling on Gli-expression were mimicked by Hes-1. CONCLUSIONS: Inhibition of Notch signalling activates hedgehog signalling, enhances chondrocyte hypertrophy and exacerbates experimental OA including osteophyte formation. These data suggest that the activation of the Notch pathway may limit aberrant hedgehog signalling in OA.


Asunto(s)
Artritis Experimental/metabolismo , Proteínas Portadoras/metabolismo , Condrocitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Osteoartritis/metabolismo , Receptor Notch1/metabolismo , Factor de Transcripción HES-1/metabolismo , Animales , Cartílago Articular/metabolismo , Ratones , Ratones Transgénicos , Osteofito/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Índice de Severidad de la Enfermedad , Transducción de Señal
7.
Ann Rheum Dis ; 74(8): 1621-5, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25817717

RESUMEN

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.


Asunto(s)
Guanilato Ciclasa/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Piel/patología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibrosis , Ratones , Ratones Endogámicos , Inhibidores de Fosfodiesterasa 5/farmacología , Pirazoles/administración & dosificación , Pirazoles/uso terapéutico , Pirimidinas/administración & dosificación , Pirimidinas/uso terapéutico , Esclerodermia Sistémica/tratamiento farmacológico , Citrato de Sildenafil/farmacología
8.
Ann Rheum Dis ; 74(5): 936-43, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-24431397

RESUMEN

OBJECTIVES: Casein kinase II (CK2) is a constitutively active serine/threonine protein kinase that plays a key role in cellular transformation and tumorigenesis. The purpose of the study was to characterise whether CK2 contributes to the pathologic activation of fibroblasts in patients with SSc and to evaluate the antifibrotic potential of CK2 inhibition. METHODS: Activation of CK2, JAK2 and STAT3 in human skin and in experimental fibrosis was analysed by immunohistochemistry. CK2 signalling was inhibited by the selective CK2 inhibitor 4, 5, 6, 7-Tetrabromobenzotriazole (TBB). The mouse models of bleomycin-induced and TGFß receptor I (TBR)-induced dermal fibrosis were used to evaluate the antifibrotic potential of specific CK2 inhibition in vivo. RESULT: Increased expression of CK2 was detected in skin fibroblasts of SSc patients. Inhibition of CK2 by TBB abrogated the TGFß-induced activation of JAK2/STAT3 signalling and prevented the stimulatory effects of TGFß on collagen release and myofibroblasts differentiation in cultured fibroblasts. Inhibition of CK2 prevented bleomycin-induced and TBR-induced skin fibrosis with decreased dermal thickening, lower myofibroblast counts and reduced accumulation of collagen. Treatment with TBB also induced regression of pre-established fibrosis. The antifibrotic effects of TBB were accompanied by reduced activation of JAK2/STAT3 signalling in vivo. CONCLUSIONS: We provide evidence that CK2 is activated in SSc and contributes to fibroblast activation by regulating JAK2/STAT3 signalling. Inhibition of CK2 reduced the pro-fibrotic effects of TGFß and inhibited experimental fibrosis. Targeting of CK2 may thus be a novel therapeutic approach for SSc and other fibrotic diseases.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Fibroblastos/metabolismo , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT3/metabolismo , Esclerodermia Sistémica/metabolismo , Piel/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Anciano , Animales , Quinasa de la Caseína II/antagonistas & inhibidores , Modelos Animales de Enfermedad , Femenino , Fibroblastos/efectos de los fármacos , Fibrosis , Humanos , Janus Quinasa 2/efectos de los fármacos , Masculino , Ratones , Persona de Mediana Edad , Factor de Transcripción STAT3/efectos de los fármacos , Esclerodermia Sistémica/patología , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos , Piel/patología , Factor de Crecimiento Transformador beta/efectos de los fármacos , Triazoles/farmacología , Adulto Joven
9.
Ann Rheum Dis ; 74(7): 1408-16, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24567525

RESUMEN

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.


Asunto(s)
Fibroblastos/patología , Guanilato Ciclasa/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/fisiopatología , Transducción de Señal/fisiología , Piel/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Estudios de Casos y Controles , Células Cultivadas , Colágeno/metabolismo , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis/metabolismo , Fibrosis/prevención & control , Guanilato Ciclasa/deficiencia , Humanos , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Pirazoles/farmacología , Piridinas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal/efectos de los fármacos , Piel/metabolismo , Proteínas Smad/metabolismo , Guanilil Ciclasa Soluble , Factor de Crecimiento Transformador beta/metabolismo
10.
Ann Rheum Dis ; 74(9): 1748-55, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24709861

RESUMEN

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.


Asunto(s)
Fibroblastos/metabolismo , Proteínas S100/metabolismo , Esclerodermia Sistémica/metabolismo , Piel/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteína de Unión al Calcio S100A4 , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Adulto Joven
11.
Ann Rheum Dis ; 74(3): e20, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24448349

RESUMEN

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.


Asunto(s)
Fibroblastos/metabolismo , Receptores de Calcitriol/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal/fisiología , Piel/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Anciano , Animales , Bleomicina/toxicidad , Modelos Animales de Enfermedad , Ergocalciferoles/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , ARN Interferente Pequeño/metabolismo , Receptores de Calcitriol/agonistas , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos , Piel/patología , Proteínas Smad/efectos de los fármacos , Factor de Crecimiento Transformador beta/efectos de los fármacos , Adulto Joven
12.
Ann Rheum Dis ; 74(6): 1317-24, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24618263

RESUMEN

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.


Asunto(s)
Fibroblastos/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/genética , Esclerodermia Difusa/metabolismo , Enfermedades de la Piel/metabolismo , Piel/patología , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibrosis , Humanos , Hidrocarburos Fluorados/farmacología , Receptores X del Hígado , Macrófagos/efectos de los fármacos , Ratones , Ratones Noqueados , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/metabolismo , Piel/efectos de los fármacos , Enfermedades de la Piel/inducido químicamente , Sulfonamidas/farmacología
13.
Ann Rheum Dis ; 74(3): e18, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24515954

RESUMEN

BACKGROUND: Osteoarthritis is the most common form of arthritis and a major socioeconomic burden. Our study is the first to explore the association between serum microRNA levels and the development of severe osteoarthritis of the knee and hip joint in the general population. METHODS: We followed 816 Caucasian individuals from 1995 to 2010 and assessed joint arthroplasty as a definitive outcome of severe osteoarthritis of the knee and hip. After a microarray screen, we validated 12 microRNAs by real-time PCR in the entire cohort at baseline. RESULTS: In Cox regression analysis, three microRNAs were associated with severe knee and hip osteoarthritis. let-7e was a negative predictor for total joint arthroplasty with an adjusted HR of 0.75 (95% CI 0.58 to 0.96; p=0.021) when normalised to U6, and 0.76 (95% CI 0.6 to 0.97; p=0.026) after normalisation to the Ct average. miRNA-454 was inversely correlated with severe knee or hip osteoarthritis with an adjusted HR of 0.77 (95% CI 0.61 to 0.97; p=0.028) when normalised to U6. This correlation was lost when data were normalised to Ct average (p=0.118). Finally, miRNA-885-5p showed a trend towards a positive relationship with arthroplasty when normalised to U6 (HR 1.24; 95% CI 0.95 to 1.62; p=0.107) or to Ct average (HR 1.30; 95% CI 0.99 to 1.70; p=0.056). CONCLUSIONS: Our study is the first to identify differentially expressed circulating microRNAs in osteoarthritis patients necessitating arthroplasty in a large, population-based cohort. Among these microRNAs, let-7e emerged as potential predictor for severe knee or hip osteoarthritis.


Asunto(s)
MicroARNs/sangre , Osteoartritis de la Cadera/genética , Osteoartritis de la Rodilla/genética , Anciano , Artroplastia de Reemplazo de Cadera , Artroplastia de Reemplazo de Rodilla , Estudios de Cohortes , Femenino , Humanos , Estudios Longitudinales , Masculino , MicroARNs/genética , Persona de Mediana Edad , Osteoartritis de la Cadera/sangre , Osteoartritis de la Cadera/cirugía , Osteoartritis de la Rodilla/sangre , Osteoartritis de la Rodilla/cirugía , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Índice de Severidad de la Enfermedad
14.
Ann Rheum Dis ; 73(10): 1880-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918036

RESUMEN

OBJECTIVES: High levels of vascular endothelial growth factor (VEGF), a key angiogenic factor, are present in patients with systemic sclerosis (SSc), but its role in the pathogenesis of fibrosis and its contribution to the disturbed angiogenesis of SSc remains hypothetical. METHODS: Mono (+/-) and double (+/+) VEGF transgenic (tg) mice and their wildtype (wt) controls were analysed. The bleomycin model was applied to VEGF tg mice to evaluate effects of VEGF under proinflammatory conditions. Additionally, tight skin (TSK) 1/VEGF+/+ mice were generated to mimic later non-inflammatory stages of SSc. RESULTS: VEGF+/+, but not VEGF+/- tg mice, spontaneously developed significant skin fibrosis, indicating profibrotic effect of VEGF in a gene-dosing manner. In the proinflammatory bleomycin model, the profibrotic effect became more pronounced with induction of skin fibrosis in VEGF+/- tg mice and even more enhanced fibrosis in VEGF+/+ tg mice. Analysis in TSK1/VEGF+/+ mice showed similar profibrotic effects of VEGF also under non-inflammatory in vivo conditions. In vitro analysis revealed that VEGF is able to directly induce collagen synthesis in dermal fibroblasts. Additionally, there was an inverse gene-dosing effect on the efficacy of angiogenesis in that a higher number of microvessels was observed in VEGF+/- tg mice than in VEGF+/+ tg mice. CONCLUSIONS: These data provide the first evidence for VEGF as a novel molecular link between fibrosis and vasculopathy in the pathogenesis of SSc. They suggest that high levels of VEGF potently induce fibrosis in inflammatory and non-inflammatory stages, and also contribute to the relatively insufficient angiogenesis characteristic for SSc.


Asunto(s)
Neovascularización Patológica/fisiopatología , Esclerodermia Sistémica/patología , Piel/patología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Bleomicina , Células Cultivadas , Colágeno/biosíntesis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis/inducido químicamente , Fibrosis/patología , Fibrosis/fisiopatología , Masculino , Ratones Transgénicos , Esclerodermia Sistémica/inducido químicamente , Esclerodermia Sistémica/fisiopatología , Piel/irrigación sanguínea , Piel/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
15.
Ann Rheum Dis ; 73(6): 1232-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23698475

RESUMEN

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.


Asunto(s)
Metilación de ADN/genética , Regulación hacia Abajo/genética , Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Leucocitos Mononucleares/metabolismo , Proteínas de la Membrana/genética , Esclerodermia Sistémica/genética , Vía de Señalización Wnt/genética , Adulto , Anciano , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Estudios de Casos y Controles , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , Decitabina , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Vía de Señalización Wnt/efectos de los fármacos , Adulto Joven
16.
Ann Rheum Dis ; 73(3): 624-7, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24257024

RESUMEN

OBJECTIVES: Canonical as well as non-canonical Wnt signalling pathways have emerged as core pathways of fibrosis. Their profibrotic effects are mediated via distinct intracellular cascades independently of each other. Thus, inhibition of both pathways may have additive antifibrotic effects. Here, we knocked down evenness interrupted (EVI) to simultaneously target for the first time canonical and non-canonical Wnt signalling in experimental fibrosis. METHODS: The antifibrotic effects of siRNA-mediated knockdown of EVI were evaluated in the mouse models of bleomycin-induced skin fibrosis and in fibrosis induced by adenoviral overexpression of a constitutively active TGF-ß receptor I (AdTBRI). RESULTS: Knockdown of EVI decreased the release of canonical and non-canonical Wnt ligands by fibroblasts and reduced the activation of canonical and non-canonical Wnt cascades in experimental fibrosis with decreased accumulation of ß-catenin and phosphorylated JNK and cJun. Inactivation of EVI exerted potent antifibrotic effects and reduced dermal thickening, myofibroblast differentiation and accumulation of collagen in the mouse models of bleomycin-induced and AdTBR-induced fibrosis. CONCLUSIONS: Inhibition of Wnt secretion by knockdown of EVI inhibits canonical and non-canonical Wnt signalling and effectively reduces experimental fibrosis in different preclinical models. Inhibition of Wnt secretion may thus be an interesting approach for the treatment of fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Receptores Acoplados a Proteínas G/genética , Esclerodermia Sistémica/prevención & control , Piel/patología , Vía de Señalización Wnt/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , MAP Quinasa Quinasa 4/metabolismo , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/fisiología , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
17.
Ann Rheum Dis ; 73(6): 1264-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24445254

RESUMEN

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.


Asunto(s)
Fibrosis/tratamiento farmacológico , Proteínas Hedgehog/antagonistas & inhibidores , Receptores Notch/antagonistas & inhibidores , Esclerodermia Sistémica/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos , Proteínas Wnt/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Bleomicina , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Pirimidinonas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Alcaloides de Veratrum/farmacología , Vía de Señalización Wnt/efectos de los fármacos
18.
Blood ; 120(14): 2909-17, 2012 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-22915638

RESUMEN

Chronic graft-versus-host disease (cGVHD) is a prognosis limiting complication of allogeneic stem cell transplantation. The molecular mechanisms underlying cGVHD are incompletely understood, and targeted therapies are not yet established for clinical use. Here we examined the role of the hedgehog pathway in sclerodermatous cGVHD. Hedgehog signaling was activated in human and murine cGVHD with increased expression of sonic hedgehog and accumulation of the transcription factors Gli-1 and Gli-2. Treatment with LDE223, a highly selective small-molecule antagonist of the hedgehog coreceptor Smoothened (Smo), abrogated the activation of hedgehog signaling and protected against experimental cGVHD. Preventive therapy with LDE223 almost completely impeded the development of clinical and histologic features of sclerodermatous cGVHD. Treatment with LDE223 was also effective, when initiated after the onset of clinical manifestations of cGVHD. Hedgehog signaling stimulated the release of collagen from cultured fibroblasts but did not affect leukocyte influx in murine cGVHD, suggesting direct, leukocyte-independent stimulatory effects on fibroblasts as the pathomechanism of hedgehog signaling in cGVHD. Considering the high morbidity of cGVHD, the current lack of efficient molecular therapies for clinical use, and the availability of well-tolerated inhibitors of Smo, targeting hedgehog signaling might be a novel strategy for clinical trials in cGVHD.


Asunto(s)
Compuestos de Bifenilo/uso terapéutico , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/prevención & control , Proteínas Hedgehog/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Esclerodermia Sistémica/prevención & control , Animales , Trasplante de Médula Ósea , Enfermedad Crónica , Colágeno/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/patología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/metabolismo , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Receptor Smoothened
19.
Ann Rheum Dis ; 72(5): 761-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22975756

RESUMEN

OBJECTIVES: Autophagy is a homeostatic process to recycle dispensable and damaged cell organelles. Dysregulation of autophagic pathways has recently been implicated in the pathogenesis of various diseases. Here, we investigated the role of autophagy during joint destruction in arthritis. METHODS: Autophagy in osteoclasts was analysed in vitro and ex vivo by transmission electron microscopy, Western blotting and immunohistochemistry for Beclin1 and Atg7. Small molecule inhibitors, LysMCre-mediated knockout of Atg7 and lentiviral overexpression of Beclin1 were used to modulate autophagy in vitro and in vivo. Osteoclast differentiation markers were quantified by real-time PCR. The extent of bone and cartilage destruction was analysed in human tumour necrosis factor α transgenic (hTNFα tg) mice after adoptive transfer with myeloid specific Atg7-deficient bone marrow. RESULTS: Autophagy was activated in osteoclasts of human rheumatoid arthritis (RA) showing increased expression of Beclin1 and Atg7. TNFα potently induced the expression of autophagy-related genes and activated autophagy in vitro and in vivo. Activation of autophagy by overexpression of Beclin1-induced osteoclastogenesis and enhanced the resorptive capacity of cultured osteoclasts, whereas pharmacologic or genetic inactivation of autophagy prevented osteoclast differentiation. Arthritic hTNFα tg mice transplanted with Atg7(fl/fl)×LysMCre(+) bone marrow cells (BMC) showed reduced numbers of osteoclasts and were protected from TNFα-induced bone erosion, proteoglycan loss and chondrocyte death. CONCLUSIONS: These findings demonstrate that autophagy is activated in RA in a TNFα-dependent manner and regulates osteoclast differentiation and bone resorption. We thus provide evidence for a central role of autophagy in joint destruction in RA.


Asunto(s)
Artritis Experimental/inmunología , Artritis Experimental/patología , Autofagia/inmunología , Articulaciones/patología , Factor de Necrosis Tumoral alfa/inmunología , Traslado Adoptivo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteína 7 Relacionada con la Autofagia , Beclina-1 , Biomarcadores , Trasplante de Médula Ósea , Resorción Ósea/inmunología , Resorción Ósea/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/genética , Osteoclastos/inmunología , Osteoclastos/patología , Osteoclastos/ultraestructura , Factor de Necrosis Tumoral alfa/genética
20.
Ann Rheum Dis ; 72(4): 614-20, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22915621

RESUMEN

OBJECTIVES: Epigenetic modifications such as DNA methylation and histone acetylation have been implicated in the pathogenesis of systemic sclerosis. However, histone methylation has not been investigated so far. We therefore aimed to evaluate the role of the trimethylation of histone H3 on lysine 27 (H3K27me3) on fibroblast activation and fibrosis. METHODS: H3K27me3 was inhibited by 3-deazaneplanocin A (DZNep) in cultured fibroblasts and in two murine models of dermal fibrosis. Fibrosis was analysed by assessment of the dermal thickening, determination of the hydroxyproline content and by quantification of the numbers of myofibroblasts. The expression of fos-related antigen 2 (fra-2) was assessed by real-time PCR, western blot and immunohistochemistry and modulated by siRNA. RESULTS: Inhibition of H3K27me3 stimulated the release of collagen in cultured fibroblasts in a time and dose-dependent manner. Treatment with DZNep exacerbated fibrosis induced by bleomycin or by overexpression of a constitutively active transforming growth factor ß receptor type I. Moreover, treatment with DZNep alone was sufficient to induce fibrosis. Inhibition of H3K27me3 induced the expression of the profibrotic transcription factor fra-2 in vitro and in vivo. Knockdown of fra-2 completely prevented the profibrotic effects of DZNep. CONCLUSIONS: These data demonstrate a novel role of H3 Lys27 histone methylation in fibrosis. In contrast to other epigenetic modifications such as DNA methylation and histone acetylation, H3 Lys27 histone methylation acts as a negative regulator of fibroblast activation in vitro and in vivo by repressing the expression of fra-2.


Asunto(s)
Fibroblastos/enzimología , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Esclerodermia Difusa/metabolismo , Esclerodermia Difusa/patología , Adulto , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Células Cultivadas , Colágeno/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/fisiología , Dermis/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/fisiología , Fibroblastos/patología , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Fibrosis/patología , Antígeno 2 Relacionado con Fos/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos DBA , Persona de Mediana Edad , Esclerodermia Difusa/inducido químicamente , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA