Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 566(7744): 344-349, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30700907

RESUMEN

Fibroblasts are polymorphic cells with pleiotropic roles in organ morphogenesis, tissue homeostasis and immune responses. In fibrotic diseases, fibroblasts synthesize abundant amounts of extracellular matrix, which induces scarring and organ failure. By contrast, a hallmark feature of fibroblasts in arthritis is degradation of the extracellular matrix because of the release of metalloproteinases and degrading enzymes, and subsequent tissue destruction. The mechanisms that drive these functionally opposing pro-fibrotic and pro-inflammatory phenotypes of fibroblasts remain unknown. Here we identify the transcription factor PU.1 as an essential regulator of the pro-fibrotic gene expression program. The interplay between transcriptional and post-transcriptional mechanisms that normally control the expression of PU.1 expression is perturbed in various fibrotic diseases, resulting in the upregulation of PU.1, induction of fibrosis-associated gene sets and a phenotypic switch in extracellular matrix-producing pro-fibrotic fibroblasts. By contrast, pharmacological and genetic inactivation of PU.1 disrupts the fibrotic network and enables reprogramming of fibrotic fibroblasts into resting fibroblasts, leading to regression of fibrosis in several organs.


Asunto(s)
Diferenciación Celular/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/genética , Fibrosis/patología , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Epigénesis Genética , Femenino , Humanos , Inflamación/genética , Inflamación/patología , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores
2.
Connect Tissue Res ; 61(6): 509-516, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31340682

RESUMEN

Background: The pathophysiology of idiopathic frozen shoulder (FS) remains poorly described. There is a lack of differentiation between idiopathic and secondary cause. The aim of this systematic review was to summarize the evidence regarding the pathophysiology of idiopathic FS on a molecular level and emphasize the clinical relevance. Methods: A database search of Medline, EMBASE and Cochrane Central Register of Controlled Trials from inception to April 2018 was performed. Participants who underwent previous injections or surgeries were excluded. A thorough selection and quality assessment process using the Cochrane Risk of Bias assessment tool and the Joanna Briggs Institute Critical Appraisal Checklist was conducted by two reviewers independently. Results: A total of 15 studies analyzing 333 study subjects were included. Twelve studies evaluated capsular tissue and three studies investigated blood samples. The tissue samples revealed increased expression of various inflammatory cytokines including interleukins, cyclooxygenase and tumor necrosis factor. Several types of acid-sensing ion channels (ASIC1 and ASIC3) were associated with disturbed neurogenesis and melatonin-regulated pain mechanism. The blood samples showed prevalence of specific interleukin and metalloproteinase genotypes. A decreased matrix metalloproteinase/tissue inhibitor of metalloproteinase ratio was found both in tissue and blood. Conclusion: The findings indicate an abnormal local neurogenesis with possible regulation through melatonin. The disturbance in remodeling of the extracellular matrix and in collagen translation, together with a persistent inflammation and an impaired healing, all interact in the process that leads to persistent fibrosis. There is global fibroplasia with localized anterior capsule contracture.


Asunto(s)
Biomarcadores/análisis , Bursitis/patología , Biomarcadores/sangre , Bursitis/sangre , Humanos , Cápsula Articular/patología , Garantía de la Calidad de Atención de Salud
3.
Ann Rheum Dis ; 78(12): 1686-1692, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31540936

RESUMEN

OBJECTIVES: Fibrosis is a complex pathophysiological process involving interplay between multiple cell types. Experimental modelling of fibrosis is essential for the understanding of its pathogenesis and for testing of putative antifibrotic drugs. However, most current models employ either phylogenetically distant species or rely on human cells cultured in an artificial environment. Here we evaluated the potential of vascularised in vitro human skin equivalents as a novel model of skin fibrosis and a platform for the evaluation of antifibrotic drugs. METHODS: Skin equivalents were assembled on a three-dimensional extracellular matrix by sequential seeding of endothelial cells, fibroblasts and keratinocytes. Fibrotic transformation on exposure to transforming growth factor-ß (TGFß) and response to treatment with nintedanib as an established antifibrotic agent were evaluated by quantitative polymerase chain reaction (qPCR), capillary Western immunoassay, immunostaining and histology. RESULTS: Skin equivalents perfused at a physiological pressure formed a mature, polarised epidermis, a stratified dermis and a functional vessel system. Exposure of these models to TGFß recapitulated key features of SSc skin with activation of TGFß pathways, fibroblast to myofibroblast transition, increased release of collagen and excessive deposition of extracellular matrix. Treatment with the antifibrotic agent nintedanib ameliorated this fibrotic transformation. CONCLUSION: Our data provide evidence that vascularised skin equivalents can replicate key features of fibrotic skin and may serve as a platform for evaluation of antifibrotic drugs in a pathophysiologically relevant human setting.


Asunto(s)
Indoles/uso terapéutico , Enfermedades de la Piel/tratamiento farmacológico , Piel/patología , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Humanos , Inhibidores de Proteínas Quinasas/uso terapéutico , Piel/irrigación sanguínea , Piel/efectos de los fármacos , Enfermedades de la Piel/patología
4.
Ann Rheum Dis ; 75(1): 303-10, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25384965

RESUMEN

OBJECTIVES: To investigate the role of microRNA-193b-3p (miR-193b) in the vascular pathophysiology of systemic sclerosis (SSc). METHODS: Expression of miR-193b in skin biopsies and fibroblasts from patients with SSc and normal healthy (NH) controls were determined by real-time PCR. Transfection with miR-193b precursor and inhibitor were used to confirm targets of miR-193b. Proliferative effects of urokinase-type plasminogen activator (uPA) were determined by water-soluble tetrazolium salt-1 assay and by analysis of proliferating cell nuclear antigen expression. Fluorescence activated cell sorting analysis was performed to investigate the effect of uPA on apoptosis. For inhibition of the uPA-cellular receptor for uPA (uPAR) pathway, uPAR neutralising antibodies and low molecular weight uPA were used. RESULTS: We found that miR-193b was downregulated in SSc fibroblasts and skin sections as compared with NH controls. The expression of miR-193b was not affected by major profibrotic cytokines and hypoxia. Induction of miR-193b in SSc fibroblasts suppressed, and accordingly, knockdown of miR-193b increased the levels of messenger RNA and protein for uPA. uPA was found to be upregulated in SSc as compared with NH controls in a transforming growth factor-ß dependent manner, and uPA was strongly expressed in vascular smooth muscle cells in SSc skin section. Interestingly, uPA induced cell proliferation and inhibited apoptosis of human pulmonary artery smooth muscle cells, and these effects were independent of uPAR signalling. CONCLUSIONS: In SSc, the downregulation of miR-193b induces the expression of uPA, which increases the number of vascular smooth muscle cells in an uPAR-independent manner and thereby contributes to the proliferative vasculopathy with intimal hyperplasia characteristic for SSc.


Asunto(s)
Regulación hacia Abajo/fisiología , MicroARNs/biosíntesis , Músculo Liso Vascular/patología , Esclerodermia Sistémica/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Apoptosis/fisiología , Estudios de Casos y Controles , Hipoxia de la Célula/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Citocinas/fisiología , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/genética , MicroARNs/fisiología , Esclerodermia Sistémica/metabolismo , Transducción de Señal/fisiología , Piel/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
5.
Ann Rheum Dis ; 73(10): 1898-904, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897768

RESUMEN

BACKGROUND: Identification of parameters for early diagnosis and treatment response would be beneficial for patients with early rheumatoid arthritis (ERA) to prevent ongoing joint damage. miRNAs have features of potential biomarkers, and an altered expression of miRNAs was shown in established rheumatoid arthritis (RA). OBJECTIVE: To analyse RA associated miRNAs in the sera of patients with ERA to find markers of early disease, clinical activity or predictors of disease outcome. METHODS: Total RNA was isolated from whole sera in ERA patients (prior to and after 3 and 12 months of therapy with disease modifying antirheumatic drugs), in patients with established RA and in healthy controls (HC) using phenol-chloroform extraction. Expression of miR-146a, miR-155, miR-223, miR-16, miR-203, miR-132 and miR-124a was analysed by TaqMan Real Time PCR. RESULTS: From all analysed miRNAs, levels of miR-146a, miR-155 and miR-16 were decreased in the sera of ERA patients in comparison with established RA. A change in circulating miR-16 in the first 3 months of therapy was associated with a decrease in DAS28 in long term follow-up in ERA (p=0.002). Levels of circulating miR-223 in treatment naïve ERA correlated with C reactive protein (p=0.008), DAS28 (p=0.031) and change in DAS28 after 3 months (p=0.003) and 12 months (p=0.011) of follow-up. However, neither miR-16 nor miR-223 could distinguish ERA from HC. CONCLUSIONS: Differential expression of circulating miR-146a, miR-155 and miR-16 in the sera of ERA patients may characterise an early stage of the disease. We suggest miR-223 as a marker of disease activity and miR-16 and miR-223 as possible predictors for disease outcome in ERA.


Asunto(s)
Artritis Reumatoide/diagnóstico , MicroARNs/sangre , Adulto , Anciano , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/genética , Biomarcadores/sangre , Biomarcadores/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Diagnóstico Precoz , Femenino , Fibroblastos/metabolismo , Estudios de Seguimiento , Expresión Génica , Humanos , Recuento de Leucocitos , Masculino , MicroARNs/biosíntesis , MicroARNs/genética , Persona de Mediana Edad , Pronóstico , Índice de Severidad de la Enfermedad , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
6.
J Transl Med ; 11: 187, 2013 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-23937772

RESUMEN

INTRODUCTION: Malignant pleural mesothelioma (MPM) is an incurable malignant disease, which results from chronic exposition to asbestos in at least 70% of the cases. Fibroblast activation protein (FAP) is predominantly expressed on the surface of reactive tumor-associated fibroblasts as well as on particular cancer types. Because of its expression on the cell surface, FAP is an attractive target for adoptive T cell therapy. T cells can be re-directed by retroviral transfer of chimeric antigen receptors (CAR) against tumor-associated antigens (TAA) and therefore represent a therapeutic strategy of adoptive immunotherapy. METHODS: To evaluate FAP expression immunohistochemistry was performed in tumor tissue from MPM patients. CD8+ human T cells were retrovirally transduced with an anti-FAP-F19-∆CD28/CD3ζ-CAR. T cell function was evaluated in vitro by cytokine release and cytotoxicity assays. In vivo function was tested with an intraperitoneal xenograft tumor model in immunodeficient mice. RESULTS: FAP was found to be expressed in all subtypes of MPM. Additionally, FAP expression was evaluated in healthy adult tissue samples and was only detected in specific areas in the pancreas, the placenta and very weakly for cervix and uterus. Expression of the anti-FAP-F19-∆CD28/CD3ζ-CAR in CD8+ T cells resulted in antigen-specific IFNγ release. Additionally, FAP-specific re-directed T cells lysed FAP positive mesothelioma cells and inflammatory fibroblasts in an antigen-specific manner in vitro. Furthermore, FAP-specific re-directed T cells inhibited the growth of FAP positive human tumor cells in the peritoneal cavity of mice and significantly prolonged survival of mice. CONCLUSION: FAP re-directed CD8+ T cells showed antigen-specific functionality in vitro and in vivo. Furthermore, FAP expression was verified in all MPM histotypes. Therefore, our data support performing a phase I clinical trial in which MPM patients are treated with adoptively transferred FAP-specific re-directed T cells.


Asunto(s)
Gelatinasas/metabolismo , Inmunoterapia , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Proteínas de la Membrana/metabolismo , Mesotelioma/inmunología , Mesotelioma/terapia , Neoplasias Pleurales/inmunología , Neoplasias Pleurales/terapia , Serina Endopeptidasas/metabolismo , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Línea Celular , Citotoxicidad Inmunológica , Endopeptidasas , Humanos , Inmunohistoquímica , Mesotelioma Maligno , Ratones , Peritoneo/patología , Proteínas Recombinantes/metabolismo , Células del Estroma/metabolismo , Linfocitos T/metabolismo , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Am J Pathol ; 180(5): 2144-55, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22449947

RESUMEN

A key feature of granulomatosis with polyangiitis (GPA; or Wegener's granulomatosis) is the granulomatous inflammation of the upper respiratory tract, which leads to the subsequent destruction of adjacent tissues. The aim of our work was to study the histopathological and cellular components of tissue destruction of human GPA tissue transplanted into immunodeficient mice. Biopsy specimens from patients with active GPA (n = 10) or sinusitis (controls, n = 6) were s.c. co-implanted with healthy allogeneic human nasal cartilage into immunodeficient pfp/rag2(-/-) mice. Transplants were examined for their destructive capability of the allografted human cartilage. In addition, nasal fibroblasts from patients with GPA (n = 8) and control healthy nasal fibroblasts (n = 5) were cultured, and cell proliferation and apoptosis were quantified. mRNA and protein levels of matrix metalloproteinases and cytokines were evaluated at baseline and after proinflammatory stimulation. GPA implants showed massive destruction of the co-implanted human cartilage, whereas cartilage destruction was only marginal in control samples. Destruction was mediated by human fibroblasts and could be inhibited by corticoid treatment. The up-regulated production of matrix metalloproteinases 1, 3, and 13 and cytokines IL-6 and IL-8 was found in vivo and in vitro. Although proliferation of isolated fibroblasts was comparable between GPA and controls, GPA samples showed a significant delay of apoptosis. The destruction of nasal cartilage in GPA is mainly mediated by fibroblasts that can be blocked by corticosteroids, and this tissue destruction is not dependent on the influx of leukocytes.


Asunto(s)
Fibroblastos/fisiología , Granulomatosis con Poliangitis/patología , Cartílagos Nasales/patología , Adulto , Anciano , Animales , Apoptosis/fisiología , Proliferación Celular , Células Cultivadas , Citocinas/biosíntesis , Dexametasona/farmacología , Dexametasona/uso terapéutico , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Granulomatosis con Poliangitis/complicaciones , Granulomatosis con Poliangitis/tratamiento farmacológico , Humanos , Tolerancia Inmunológica , Masculino , Metaloproteinasas de la Matriz/biosíntesis , Ratones , Ratones Noqueados , Persona de Mediana Edad , Cartílagos Nasales/trasplante , Mucosa Nasal/patología , Mucosa Nasal/trasplante , Deformidades Adquiridas Nasales/etiología , Deformidades Adquiridas Nasales/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Adulto Joven
8.
Arthritis Rheum ; 64(6): 1771-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22161761

RESUMEN

OBJECTIVE: To investigate the expression and effect of the microRNA-34 (miR-34) family on apoptosis in rheumatoid arthritis synovial fibroblasts (RASFs). METHODS: Expression of the miR-34 family in synovial fibroblasts with or without stimulation with Toll-like receptor (TLR) ligands, tumor necrosis factor α (TNFα), interleukin-1ß (IL-1ß), hypoxia, or 5-azacytidine was analyzed by real-time polymerase chain reaction (PCR). Promoter methylation was studied by combined bisulfite restriction analysis. The effects of overexpression and silencing of miR-34a and miR-34a* on apoptosis were analyzed by annexin V/propidium iodide staining. Production of X-linked inhibitor of apoptosis protein (XIAP) was assessed by real-time PCR and immunohistochemistry analysis. Reporter gene assay was used to study the signaling pathways of miR-34a*. RESULTS: Basal expression levels of miR-34a* were found to be reduced in synovial fibroblasts from RA patients compared to osteoarthritis patients, whereas levels of miR-34a, miR-34b/b*, and miR-34c/c* did not differ. Neither TNFα, IL-1ß, TLR ligands, nor hypoxia altered miR-34a* expression. However, we demonstrated that the promoter of miR-34a/34a* was methylated and showed that transcription of the miR-34a duplex was induced upon treatment with demethylating agents. Enforced expression of miR-34a* led to an increased rate of FasL- and TRAIL-mediated apoptosis in RASFs. Moreover, levels of miR-34a* were highly correlated with expression of XIAP, which was found to be up-regulated in RA synovial cells. Finally, we identified XIAP as a direct target of miR-34a*. CONCLUSION: Our data provide evidence of a methylation-specific down-regulation of proapoptotic miR-34a* in RASFs. Decreased expression of miR- 34a* results in up-regulation of its direct target XIAP, thereby contributing to resistance of RASFs to apoptosis.


Asunto(s)
Apoptosis/fisiología , Artritis Reumatoide/metabolismo , Regulación hacia Abajo , Fibroblastos/metabolismo , MicroARNs/metabolismo , Membrana Sinovial/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Azacitidina/farmacología , Células Cultivadas , Fibroblastos/patología , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Interleucina-1beta/farmacología , MicroARNs/genética , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
9.
Arthritis Rheum ; 63(7): 2067-77, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21437873

RESUMEN

OBJECTIVE: Systemic sclerosis (SSc) is an autoimmune disease marked by aberrant activation and apoptosis of endothelial cells (ECs) and decreased numbers of circulating angiogenic cells (CACs). The aim of this study was to analyze whether microparticles might link pathologic activation and apoptosis of ECs with reduced numbers of CACs. METHODS: Apoptosis was quantified by staining for annexin V and measurement of caspase 3 activity. The uptake of microparticles by CACs was determined by fluorescence-activated cell sorting and by fluorescence microscopy. Tritiated arachidonic acid and phosphatidylinositol 3,5-bisphosphate were used to demonstrate the transfer of arachidonic acid and highlight the role of the acid sphingomyelinase in microparticle-induced apoptosis of endothelial progenitor cells. RESULTS: Microparticles derived from activated or apoptotic ECs, the expression of which is strongly increased in the blood of patients with SSc, induce apoptosis in CACs in a dose-dependent manner. Microparticles, which are rich in arachidonic acid, are phagocytosed by CACs. Inhibition of phagocytosis prevents the induction of apoptosis in CACs by microparticles. Microparticles can transport arachidonic acid from ECs to CACs, and purified arachidonic acid mimics the proapoptotic effects of microparticles. Arachidonic acid activates the acid sphingomyelinase, and inhibition of acid sphingomyelinase prevents microparticle-induced apoptosis of CACs. Thus, phagocytosis of microparticles might stimulate the activity of acid sphingomyelinase and activate the apoptotic machinery. CONCLUSION: The induction of apoptosis in CACs by microparticles derived from ECs provides a novel link between aberrant activation or apoptosis of ECs, decreased numbers of CACs, and impaired formation of new vessels in SSc.


Asunto(s)
Apoptosis/fisiología , Micropartículas Derivadas de Células/metabolismo , Células Endoteliales/metabolismo , Esclerodermia Sistémica/metabolismo , Adulto , Anexina A5/metabolismo , Apoptosis/genética , Ácido Araquidónico/farmacología , Caspasa 3/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Esclerodermia Sistémica/patología
10.
Cells ; 11(3)2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35159255

RESUMEN

(1) Background: Three-dimensional (3D) collagen I-based skin models are commonly used in drug development and substance testing but have major drawbacks such as batch-to-batch variations and ethical concerns. Recently, synthetic nanofibrous scaffolds created by electrospinning have received increasing interest as potential alternatives due to their morphological similarities to native collagen fibrils in size and orientation. The overall objective of this proof-of-concept study was to demonstrate the suitability of two synthetic polymers in creating electrospun scaffolds for 3D skin cell models. (2) Methods: Electrospun nanofiber mats were produced with (i) poly(acrylonitrile-co-methyl acrylate) (P(AN-MA)) and (ii) a blend of pullulan (Pul), poly(vinyl alcohol) (PVA) and poly(acrylic acid) (PAA) (Pul/PVA/PAA) and characterized by scanning electron microscopy (SEM) and diffuse reflectance infrared Fourier transform (DRIFT) spectra. Primary skin fibroblasts and keratinocytes were seeded onto the nanofiber mats and analyzed for phenotypic characteristics (phalloidin staining), viability (Presto Blue HS assay), proliferation (Ki-67 staining), distribution (H/E staining), responsiveness to biological stimuli (qPCR), and formation of skin-like structures (H/E staining). (3) Results: P(AN-MA) mats were more loosely packed than the Pul/PVA/PAA mats, concomitant with larger fiber diameter (340 nm ± 120 nm vs. 250 nm ± 120 nm, p < 0.0001). After sterilization and exposure to cell culture media for 28 days, P(AN-MA) mats showed significant adsorption of fetal calf serum (FCS) from the media into the fibers (DRIFT spectra) and increased fiber diameter (590 nm ± 290 nm, p < 0.0001). Skin fibroblasts were viable over time on both nanofiber mats, but suitable cell infiltration only occurred in the P(AN-MA) nanofiber mats. On P(AN-MA) mats, fibroblasts showed their characteristic spindle-like shape, produced a dermis-like structure, and responded well to TGFß stimulation, with a significant increase in the mRNA expression of PAI1, COL1A1, and αSMA (all p < 0.05). Primary keratinocytes seeded on top of the dermis equivalent proliferated and formed a stratified epidermis-like structure. (4) Conclusion: P(AN-MA) and Pul/PVA/PAA are both biocompatible materials suitable for nanofiber mat production. P(AN-MA) mats hold greater potential as future 3D skin models due to enhanced cell compatibility (i.e., adsorption of FCS proteins), cell infiltration (i.e., increased pore size due to swelling behavior), and cell phenotype preservation. Thus, our proof-of-concept study shows an easy and robust process of producing electrospun scaffolds for 3D skin cell models made of P(AN-MA) nanofibers without the need for bioactive molecule attachments.


Asunto(s)
Acrilonitrilo , Nanofibras , Colágeno , Glucanos , Nanofibras/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
11.
Ann Rheum Dis ; 70(8): 1357-62, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21628308

RESUMEN

The development of novel treatments for rheumatoid arthritis (RA) requires the interplay between clinical observations and studies in animal models. Given the complex molecular pathogenesis and highly heterogeneous clinical picture of RA, there is an urgent need to dissect its multifactorial nature and to propose new strategies for preventive, early and curative treatments. Research on animal models has generated new knowledge on RA pathophysiology and aetiology and has provided highly successful paradigms for innovative drug development. Recent focus has shifted towards the discovery of novel biomarkers, with emphasis on presymptomatic and emerging stages of human RA, and towards addressing the pathophysiological mechanisms and subsequent efficacy of interventions that underlie different disease variants. Shifts in the current paradigms underlying RA pathogenesis have also led to increased demand for new (including humanised) animal models. There is therefore an urgent need to integrate the knowledge on human and animal models with the ultimate goal of creating a comprehensive 'pathogenesis map' that will guide alignment of existing and new animal models to the subset of disease they mimic. This requires full and standardised characterisation of all models at the genotypic, phenotypic and biomarker level, exploiting recent technological developments in 'omics' profiling and computational biology as well as state of the art bioimaging. Efficient integration and dissemination of information and resources as well as outreach to the public will be necessary to manage the plethora of data accumulated and to increase community awareness and support for innovative animal model research in rheumatology.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Animales , Antirreumáticos/uso terapéutico , Artritis Experimental/fisiopatología , Artritis Experimental/prevención & control , Artritis Reumatoide/fisiopatología , Artritis Reumatoide/prevención & control , Descubrimiento de Drogas/métodos , Terapias en Investigación/métodos
12.
Arthritis Rheum ; 62(6): 1733-43, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20201077

RESUMEN

OBJECTIVE: To investigate the role of microRNA (miRNA) as posttranscriptional regulators of profibrotic genes in systemic sclerosis (SSc). METHODS: MicroRNA, which target collagens, were identified by in silico analysis. Expression of miRNA-29 (miR-29) was determined by TaqMan real-time polymerase chain reaction analysis of skin biopsy and fibroblast samples from SSc patients and healthy controls as well as in the mouse model of bleomycin-induced skin fibrosis. Cells were transfected with precursor miRNA (pre-miRNA)/anti-miRNA of miR-29 using Lipofectamine. Collagen gene expression was also studied in luciferase reporter gene assays. For stimulation, recombinant transforming growth factor beta (TGFbeta), platelet-derived growth factor B (PDGF-B), or interleukin-4 (IL-4) was used. The effects of inhibiting PDGF-B and TGFbeta signaling on the levels of miR-29 were studied in vitro and in the bleomycin model. RESULTS: We found that miR-29a was strongly down-regulated in SSc fibroblasts and skin sections as compared with the healthy controls. Overexpression in SSc fibroblasts significantly decreased, and accordingly, knockdown in normal fibroblasts increased, the levels of messenger RNA and protein for type I and type III collagen. In the reporter gene assay, cotransfection with pre-miR-29a significantly decreased the relative luciferase activity, which suggests a direct regulation of collagen by miR-29a. TGFbeta, PDGF-B, or IL-4 reduced the levels of miR-29a in normal fibroblasts to those seen in SSc fibroblasts. Similar to human SSc, the expression of miR-29a was reduced in the bleomycin model of skin fibrosis. Inhibition of PDGF-B and TGFbeta pathways by treatment with imatinib restored the levels of miR-29a in vitro and in the bleomycin model in vivo. CONCLUSION: These data add the posttranscriptional regulation of collagens by miR-29a as a novel aspect to the fibrogenesis of SSc and suggest miR-29a as a potential therapeutic target.


Asunto(s)
Colágeno Tipo III/genética , Colágeno Tipo I/genética , Fibroblastos/metabolismo , MicroARNs/genética , Esclerodermia Sistémica/genética , Piel/metabolismo , Western Blotting , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Regulación hacia Abajo/genética , Fibroblastos/patología , Humanos , MicroARNs/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Piel/patología
13.
Arthritis Rheum ; 62(5): 1224-35, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20155839

RESUMEN

OBJECTIVE: Since fibroblasts in the synovium of patients with rheumatoid arthritis (RA) express the serine proteases fibroblast activation protein (FAP) and dipeptidylpeptidase 4 (DPP-4)/CD26, we undertook the current study to determine the functional role of both enzymes in the invasion of RA synovial fibroblasts (RASFs) into articular cartilage. METHODS: Expression of FAP and DPP-4/CD26 by RASFs was analyzed using fluorescence-activated cell sorting and immunocytochemistry. Serine protease activity was measured by cleavage of fluorogenic substrates and inhibited upon treatment with L-glutamyl L-boroproline. The induction and expression of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) in RASFs were detected using real-time polymerase chain reaction. Densitometric measurements of MMPs using immunoblotting confirmed our findings on the messenger RNA level. Stromal cell-derived factor 1 (SDF-1 [CXCL12]), MMP-1, and MMP-3 protein levels were measured using enzyme-linked immunosorbent assay. The impact of FAP and DPP-4/CD26 inhibition on the invasiveness of RASFs was analyzed in the SCID mouse coimplantation model of RA using immunohistochemistry. RESULTS: Inhibition of serine protease activity of FAP and DPP-4/CD26 in vitro led to increased levels of SDF-1 in concert with MMP-1 and MMP-3, which are downstream effectors of SDF-1 signaling. Using the SCID mouse coimplantation model, inhibition of enzymatic activity in vivo significantly promoted invasion of xenotransplanted RASFs into cotransplanted human cartilage. Zones of cartilage resorption were infiltrated by FAP-expressing RASFs and marked by a significantly higher accumulation of MMP-1 and MMP-3, when compared with controls. CONCLUSION: Our results indicate a central role for the serine protease activity of FAP and DPP-4/CD26 in protecting articular cartilage against invasion by synovial fibroblasts in RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Cartílago Articular/patología , Dipeptidil Peptidasa 4/metabolismo , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Cartílago Articular/metabolismo , Células Cultivadas , Quimiocina CXCL12/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV , Modelos Animales de Enfermedad , Endopeptidasas , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Gelatinasas/antagonistas & inhibidores , Humanos , Metaloproteinasas de la Matriz/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones SCID , Membrana Sinovial/enzimología , Membrana Sinovial/patología
14.
Adv Biol (Weinh) ; 5(4): e2000168, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33852183

RESUMEN

Systemic sclerosis (SSc) is a complex multisystem disease with the highest case-specific mortality among all autoimmune rheumatic diseases, yet without any available curative therapy. Therefore, the development of novel therapeutic antifibrotic strategies that effectively decrease skin and organ fibrosis is needed. Existing animal models are cost-intensive, laborious and do not recapitulate the full spectrum of the disease and thus commonly fail to predict human efficacy. Advanced in vitro models, which closely mimic critical aspects of the pathology, have emerged as valuable platforms to investigate novel pharmaceutical therapies for the treatment of SSc. This review focuses on recent advancements in the development of SSc in vitro models, sheds light onto biological (e.g., growth factors, cytokines, coculture systems), biochemical (e.g., hypoxia, reactive oxygen species) and biophysical (e.g., stiffness, topography, dimensionality) cues that have been utilized for the in vitro recapitulation of the SSc microenvironment, and highlights future perspectives for effective drug discovery and validation.


Asunto(s)
Enfermedades Autoinmunes , Esclerodermia Sistémica , Animales , Enfermedades Autoinmunes/patología , Descubrimiento de Drogas , Fibrosis , Humanos , Esclerodermia Sistémica/tratamiento farmacológico , Piel/patología
15.
Circulation ; 120(23): 2367-76, 2009 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-19933934

RESUMEN

BACKGROUND: Microvascular damage is one of the first pathological changes in systemic sclerosis. In this study, we investigated the role of Fos-related antigen-2 (Fra-2), a transcription factor of the activator protein-1 family, in the peripheral vasculopathy of systemic sclerosis and examined the underlying mechanisms. METHODS AND RESULTS: Expression of Fra-2 protein was significantly increased in skin biopsies of systemic sclerosis patients compared with healthy controls, especially in endothelial and vascular smooth muscle cells. Fra-2 transgenic mice developed a severe loss of small blood vessels in the skin that was paralleled by progressive skin fibrosis at 12 weeks of age. The reduction in capillary density was preceded by a significant increase in apoptosis in endothelial cells at week 9 as detected by immunohistochemistry. Similarly, suppression of Fra-2 by small interfering RNA prevented human microvascular endothelial cells from staurosporine-induced apoptosis and improved both the number of tubes and the cumulative tube lengths in the tube formation assay. In addition, cell migration in the scratch assay and vascular endothelial growth factor-dependent chemotaxis in a modified Boyden chamber assay were increased after transfection of human microvascular endothelial cells with Fra-2 small interfering RNA, whereas proliferation was not affected. CONCLUSIONS: Fra-2 is present in human systemic sclerosis and may contribute to the development of microvasculopathy by inducing endothelial cell apoptosis and by reducing endothelial cell migration and chemotaxis. Fra-2 transgenic mice are a promising preclinical model to study the mechanisms and therapeutic approaches of the peripheral vasculopathy in systemic sclerosis.


Asunto(s)
Antígeno 2 Relacionado con Fos/fisiología , Enfermedades Vasculares Periféricas/metabolismo , Enfermedades Vasculares Periféricas/patología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Animales , Apoptosis/fisiología , Inhibición de Migración Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Quimiotaxis de Leucocito/fisiología , Progresión de la Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Transgénicos
16.
Curr Opin Rheumatol ; 22(3): 284-92, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20305560

RESUMEN

PURPOSE OF REVIEW: Rheumatoid arthritis (RA) is a systemic, autoimmune disease resulting in the destruction of affected joints. Even though current therapies with biologics such as tumor necrosis factor-alpha blockers yield significant improvement for the patients, the disease is not curable yet. Therefore, we need novel strategies for better therapies. RECENT FINDINGS: The growing knowledge of epigenetics might give us new insights into the pathogenesis of autoimmune diseases. In the last year, several new findings about epigenetic modifications of gene expression were reported in different arthritides. These modifications describe changes in the expression of DNA that result from methylation, posttranslational modifications of the histone proteins, including acetylation/deacetylation, sumoylation, methylation and microRNAs. Most interestingly, these modifications seem to act in concert and are associated with the circadian metabolic rhythm of cells. SUMMARY: This review summarizes reports from the last year about epigenetic modifications of gene expression via acetylation/deacetylation, including sirtuins, sumoylation, methylation, microRNAs in all in rheumatoid arthritis and other arthritides, providing potential strategies for better therapies and encourages the development of specific epigenetic drugs.


Asunto(s)
Artritis Reumatoide/genética , Epigénesis Genética/genética , Procesamiento Proteico-Postraduccional/genética , Enfermedades Reumáticas/genética , Acetilación , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/fisiopatología , Metilación de ADN/fisiología , Histonas/genética , Histonas/metabolismo , Humanos , MicroARNs/genética , Enfermedades Reumáticas/tratamiento farmacológico , Enfermedades Reumáticas/fisiopatología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo
17.
Ann Rheum Dis ; 69(5): 898-902, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19497915

RESUMEN

OBJECTIVE: To evaluate the decrease of cartilage destruction by a novel orally active and specific matrix metalloproteinase 13 (MMP-13) inhibitor in three different animal models of rheumatoid arthritis (RA). MATERIALS AND METHODS: The SCID mouse co-implantation model of RA, the collagen-induced arthritis (CIA) model in mice and the antigen-induced arthritis model (AIA) in rabbits were used. RESULTS: In the SCID mouse co-implantation model, the MMP-13 inhibitor reduced cartilage destruction by 75%. In the CIA model of RA, the MMP-13 inhibitor resulted in a significant and dose-dependent decrease in clinical symptoms as well as of cartilage erosion by 38% (30 mg/kg), 28% (10 mg/kg) and 21% (3 mg/kg). No significant effects were seen in the AIA model. No toxic effects were seen in all three animal models. CONCLUSION: Although several MMPs in concert with other proteinases have a role in the process of cartilage destruction, there is a need for highly selective MMP inhibitors to reduce severe side effects that occur with non-specific inhibitors. Significant inhibition of MMP-13 reduced cartilage erosions in two of three tested animal models of RA. These results strongly support the development of this class of drugs to reduce or halt joint destruction in patients with RA.


Asunto(s)
Antirreumáticos/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Inhibidores de la Metaloproteinasa de la Matriz , Administración Oral , Animales , Antirreumáticos/administración & dosificación , Artritis Experimental/enzimología , Artritis Experimental/patología , Artritis Reumatoide/enzimología , Artritis Reumatoide/patología , Cartílago Articular/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/administración & dosificación , Femenino , Humanos , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones SCID , Conejos , Membrana Sinovial/enzimología
18.
J Autoimmun ; 35(1): 15-22, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20079608

RESUMEN

The aggressive phenotype of RA synovial fibroblasts (RASF) is characterised by the increased expression of matrix metalloproteinase (MMP)-1 as well as the small ubiquitin like modifier (SUMO)-1 and decreased expression of SUMO-specific protease SENP1. Since we showed an increased activity of acetyltransferases in this autoimmune disease, we wanted to analyze whether this affects the expression of MMP-1 and can be reversed by the reconstitution of SENP1. In RASF, the acetylation of histone H4 was significantly increased in the distal region of the MMP-1 promoter by 274 +/- 36% compared to OASF. Most interestingly, overexpression of SENP1 in RASF decreased acetylation specifically in this region by 51 +/- 0.5% and globally by 73 +/- 11%. Furthermore, the overexpression of SENP1 resulted in a downregulation of MMP-1 at both the mRNA (58 +/- 7%) and protein levels (28 +/- 6%), significantly reduced the invasiveness of RASF (from 34 +/- 9% to 2 +/- 2%) and led to an accumulation of histone deacetylase 4 (HDAC4) on the MMP-1 promoter (197 +/- 36%). Interestingly, SENP1 failed to modulate the expression of MMP-1 in the cells silenced for HDAC4. This is the first study linking the SUMOylation pathway and the production of MMP-1 to an epigenetic control mechanism mediated through histone acetylation which has a functional consequence for the invasiveness of RASF.


Asunto(s)
Artritis Reumatoide/genética , Endopeptidasas/metabolismo , Fibroblastos/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Invasividad Neoplásica/genética , Acetilación , Animales , Artritis Reumatoide/metabolismo , Línea Celular , Clonación Molecular , Cisteína Endopeptidasas , Perros , Endopeptidasas/genética , Endopeptidasas/inmunología , Epigénesis Genética , Fibroblastos/inmunología , Fibroblastos/patología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/inmunología , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Membrana Sinovial/patología , Transgenes/genética
19.
Sci Transl Med ; 12(563)2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32998972

RESUMEN

Aberrant activation of fibroblasts with progressive deposition of extracellular matrix is a key feature of systemic sclerosis (SSc), a prototypical idiopathic fibrotic disease. Here, we demonstrate that the profibrotic cytokine transforming growth factor ß selectively up-regulates fibroblast growth factor receptor 3 (FGFR3) and its ligand FGF9 to promote fibroblast activation and tissue fibrosis, leading to a prominent FGFR3 signature in the SSc skin. Transcriptome profiling, in silico analysis and functional experiments revealed that FGFR3 induces multiple profibrotic pathways including endothelin, interleukin-4, and connective tissue growth factor signaling mediated by transcription factor CREB (cAMP response element-binding protein). Inhibition of FGFR3 signaling by fibroblast-specific knockout of FGFR3 or FGF9 or pharmacological inhibition of FGFR3 blocked fibroblast activation and attenuated experimental skin fibrosis in mice. These findings characterize FGFR3 as an upstream regulator of a network of profibrotic mediators in SSc and as a potential target for the treatment of fibrosis.


Asunto(s)
Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Esclerodermia Sistémica , Animales , Células Cultivadas , Fibroblastos/patología , Fibrosis , Ratones , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Esclerodermia Sistémica/patología , Transducción de Señal , Piel/patología , Factor de Crecimiento Transformador beta
20.
J Cell Mol Med ; 13(10): 4185-91, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18774958

RESUMEN

Systemic sclerosis (SSc) is a systemic autoimmune disease that is characterized by microangiopathy with progressive loss of capillaries and tissue fibrosis. Imatinib exerts potent anti-fibrotic effects and is currently evaluated in clinical trials. The aim of the present study was to exclude that the anti-fibrotic effects of imatinib are complicated by inhibitory effects on endothelial cell functions, which might augment vascular disease in SSc. Endothelial cells and mice were treated with pharmacologically relevant concentrations of imatinib. The expression of markers of vascular activation was assessed with real-time PCR. Proliferation was analysed with the cell counting experiments and the MTT assay. Apoptosis was quantified with caspase 3 assays, annexin V in vitro and with TUNEL staining in vivo. Migration was studied with scratch and transwell assays. Tube forming was investigated with the matrigel assay. Imatinib did not alter the expression of markers of vascular activation. Imatinib did not increase the percentage of annexin V positive cells or the activity of caspase 3. No reduction in proliferation or metabolic activity of endothelial cells was observed. Imatinib did not affect migration of endothelial cells and did not reduce the formation of capillary tubes. Consistent with the in vitro data, no difference in the number of apoptotic endothelial cells was observed in vivo in mice treated with imatinib. Imatinib does not inhibit activation, viability, proliferation, migration or tube forming of endothelial cells in vitro and in vivo. Thus, treatment with imatinib might not augment further endothelial cell damage in SSc.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Piperazinas/farmacología , Pirimidinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Benzamidas , Biomarcadores/metabolismo , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Células Endoteliales/metabolismo , Fibrosis , Humanos , Mesilato de Imatinib , Ratones , Neovascularización Patológica/patología , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA