Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 104(12): 5073-8, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17360386

ABSTRACT

The small ubiquitin-like modifier (SUMO)-1 is an important posttranslational regulator of different signaling pathways and involved in the formation of promyelocytic leukemia (PML) protein nuclear bodies (NBs). Overexpression of SUMO-1 has been associated with alterations in apoptosis, but the underlying mechanisms and their relevance for human diseases are not clear. Here, we show that the increased expression of SUMO-1 in rheumatoid arthritis (RA) synovial fibroblasts (SFs) contributes to the resistance of these cells against Fas-induced apoptosis through increased SUMOylation of nuclear PML protein and increased recruitment of the transcriptional repressor DAXX to PML NBs. We also show that the nuclear SUMO-protease SENP1, which is found at lower levels in RA SFs, can revert the apoptosis-inhibiting effects of SUMO-1 by releasing DAXX from PML NBs. Our findings indicate that in RA SFs overexpression of SENP1 can alter the SUMO-1-mediated recruitment of DAXX to PML NBs, thus influencing the proapoptotic effects of DAXX. Accumulation of DAXX in PML NBs by SUMO-1 may, therefore, contribute to the pathogenesis of inflammatory disorders.


Subject(s)
Apoptosis/drug effects , Arthritis, Rheumatoid/pathology , Cell Nucleus/metabolism , Fas Ligand Protein/pharmacology , Fibroblasts/pathology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , SUMO-1 Protein/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Co-Repressor Proteins , Cysteine Endopeptidases , Endopeptidases/genetics , Endopeptidases/metabolism , Fibroblasts/drug effects , Gene Expression/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Intranuclear Inclusion Bodies/drug effects , Molecular Chaperones , Promyelocytic Leukemia Protein , Protein Processing, Post-Translational/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synovial Fluid/cytology , Synovial Fluid/drug effects
2.
Respir Res ; 6: 37, 2005 Apr 17.
Article in English | MEDLINE | ID: mdl-15833141

ABSTRACT

BACKGROUND: An altered susceptibility of lung fibroblasts to Fas-induced apoptosis has been implicated in the pathogenesis of pulmonary fibrosis; however, the underlying mechanism is not completely understood. Here, we studied the susceptibility of lung fibroblasts, obtained from patients with (f-fibs) and without pulmonary fibrosis (n-fibs), to FasL- (CD95L/APO-1) induced apoptosis in relation to the expression and the amounts of membrane-bound and soluble Fas. We also analysed the effects of tumor necrosis factor-beta on FasL-induced cell death. METHODS: Apoptosis was induced with recombinant human FasL, with and without prior stimulation of the fibroblasts with tumor necrosis factor-alpha and measured by a histone fragmentation assay and flow cytometry. The expression of Fas mRNA was determined by quantitative PCR. The expression of cell surface Fas was determined by flow cytometry, and that of soluble Fas (sFas) was determined by enzyme-linked immunosorbent assay. RESULTS: When compared to n-fibs, f-fibs were resistant to FasL-induced apoptosis, despite significantly higher levels of Fas mRNA. F-fibs showed lower expression of surface-bound Fas but higher levels of sFas. While TNF-alpha increased the susceptibility to FasL-induced apoptosis in n-fibs, it had no pro-apoptotic effect in f-fibs. CONCLUSIONS: The data suggest that lower expression of surface Fas, but higher levels of apoptosis-inhibiting sFas, contribute to the resistance of fibroblasts in lung fibrosis against apoptosis, to increased cellularity and also to increased formation and deposition of extracellular matrix.


Subject(s)
Apoptosis/immunology , Cell Membrane/immunology , Fibroblasts/immunology , Lung/immunology , Membrane Glycoproteins/immunology , Pulmonary Fibrosis/immunology , Tumor Necrosis Factors/immunology , fas Receptor/immunology , Cell Membrane/chemistry , Cells, Cultured , Fas Ligand Protein , Fibroblasts/pathology , Humans , Immunity, Innate/immunology , Lung/pathology , Pulmonary Fibrosis/pathology , Solubility , fas Receptor/chemistry
3.
Arthritis Rheum ; 50(9): 2794-802, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15457447

ABSTRACT

OBJECTIVE: To study the specific contribution of MAP kinase activator c-Raf-1 and one of its downstream transcription factors, c-Myc, to the growth and invasive behavior of rheumatoid arthritis synovial fibroblasts (RASFs). METHODS: RASFs were transduced with retroviral constructs expressing dominant-negative mutants of c-Raf-1 or c-Myc (DN c-Raf-1 or DN c-Myc, respectively) or with the mock vector. The expression of wild-type and mutant proteins was confirmed by Western blotting. Growth curves of RASFs were recorded, and apoptosis was measured by flow cytometry. Invasiveness of RASFs was assessed in the SCID mouse model of RA. Immunohistochemistry was used to study the effects of DN c-Raf-1 on phosphorylated c-Jun and matrix metalloproteinase 1 (MMP-1) in RASFs implanted into SCID mice. The phosphorylation of ERK and JNK in DN c-Raf-1- and mock-transduced RASFs was determined in vitro by Western blotting. The levels of MMPs in these cells were measured by quantitative polymerase chain reaction (PCR). RESULTS: Neither DN c-Raf-1 alone nor DN c-Myc alone significantly altered proliferation or apoptosis of RASFs, but both mutants together rapidly induced apoptosis. Inhibition of c-Raf-1 or c-Myc significantly reduced the invasiveness of RASFs in the SCID mouse model. DN c-Raf-1 decreased the phosphorylation of ERK and JNK in vitro and reduced the in vivo expression of phosphorylated c-Jun as well as the expression of disease-relevant MMPs. As determined by quantitative PCR, the inhibition was most pronounced for MMP-1 and MMP-3. CONCLUSION: The data demonstrate that Ras- and c-Myc-dependent signaling events cooperate to regulate the growth and invasiveness of RASFs. Targeting of both c-Raf-1 and c-Myc may constitute an interesting therapeutic approach in RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Fibroblasts/immunology , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins c-raf/immunology , Synovial Fluid/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Arthritis, Rheumatoid/genetics , Female , Gene Transfer Techniques , Humans , Matrix Metalloproteinase 1/metabolism , Mice , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism , Models, Animal , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-raf/genetics , Signal Transduction/immunology , Synovial Fluid/cytology
5.
Arthritis Res Ther ; 5(5): R253-61, 2003.
Article in English | MEDLINE | ID: mdl-12932288

ABSTRACT

The aim of the present study was to investigate the expression of Fas in periarticular tenocytes of patients with osteoarthritis (OA) and to study their susceptibility to Fas ligand-mediated apoptosis. Tendon samples were obtained from the quadriceps femoris muscle of patients with knee OA and used for histological evaluation, for immunohistochemical detection of Fas, and to establish tenocyte cultures. The expression of Fas mRNA was determined by quantitative PCR. Levels of soluble Fas and soluble tumour necrosis factor (TNF) receptor I were measured using ELISA. Apoptosis was induced with recombinant human Fas ligand and measured by a histone fragmentation assay and flow cytometry. The effects of TNF-alpha were studied by stimulation with TNF-alpha alone or 24 hours before the induction of apoptosis. Tendon samples from non-OA patients were used as controls. Histological evaluation revealed degenerative changes in the tendons of all OA patients but not in the controls. Fas was detected by immunohistochemistry in all specimens, but quantitative PCR revealed significantly higher levels of Fas mRNA in OA tenocytes. In contrast, lower levels of soluble Fas were found in OA tenocytes by ELISA. OA tenocytes were significantly more susceptible to Fas ligand induced apoptosis than were control cells. TNF-alpha reduced the Fas ligand induced apoptosis in OA tenocytes but had no effects on control tenocytes. These data suggest that knee OA is associated with higher susceptibility of periarticular tenocytes to Fas ligand induced apoptosis because of higher expression of Fas but lower levels of apoptosis-inhibiting soluble Fas. These changes may contribute to decreased cellularity in degenerative tendons and promote their rupturing. The antiapoptotic effects of TNF-alpha in OA tenocytes most likely reflect regenerative attempts and must be taken into account when anti-TNF strategies are considered for OA.


Subject(s)
Apoptosis/physiology , Knee Joint/pathology , Membrane Glycoproteins/physiology , Osteoarthritis, Knee/pathology , Tendons/pathology , Tumor Necrosis Factor-alpha/physiology , Arthroplasty, Replacement, Knee/methods , Cell Death/physiology , Fas Ligand Protein , Femur/chemistry , Femur/pathology , Humans , Knee Joint/chemistry , Knee Joint/surgery , Muscle, Skeletal/chemistry , Muscle, Skeletal/pathology , Muscle, Skeletal/surgery , RNA, Messenger/biosynthesis , Solubility , Tendons/chemistry , Tendons/surgery , fas Receptor/biosynthesis , fas Receptor/metabolism
6.
Curr Opin Rheumatol ; 15(3): 274-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12707581

ABSTRACT

Apoptosis is a key mechanism that regulates tissue composition and homeostasis. Alterations in the apoptosis of synovial cells have been described in residential synoviocytes as well as inflammatory cells and associated with the pathogenesis of rheumatoid arthritis. These changes constitute hallmarks of synovial cell activation and contribute to both chronic inflammation and hyperplasia. Rheumatoid arthritis synovial fibroblasts are affected most prominently, and their resistance to apoptosis has been linked closely to the progressive destruction of articular cartilage. Although a detailed understanding of mechanisms that prevent synovial fibroblasts from programmed cell death is lacking, several antiapoptotic molecules have been identified. Among them, downstream modulators of Fas-signaling, such as sentrin-1/small ubiquitin-like modifier (SUMO)-1 and Fas-associated death domain-like interleukin (IL)-1beta-converting enzyme-inhibitory protein (FLIP), as well as transcriptional regulators such as NFkappaB, Stat3, and p53, have been suggested to regulate apoptosis most prominently. Current efforts are aimed at elucidating the specific role of these molecules in regulating the apoptosis of rheumatoid fibroblasts and at identifying molecular targets to interfere with altered apoptosis.


Subject(s)
Antirheumatic Agents/administration & dosage , Apoptosis/drug effects , Apoptosis/physiology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/physiopathology , Intracellular Signaling Peptides and Proteins , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Male , Primary Prevention/methods , Prognosis , Proteins , Risk Assessment , Sensitivity and Specificity , Severity of Illness Index , Synovial Membrane/cytology , Synovial Membrane/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...