Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 19(3): 358-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23396210

ABSTRACT

Hepatic insulin resistance is a driving force in the pathogenesis of type 2 diabetes mellitus (T2DM) and is tightly coupled with excessive storage of fat and the ensuing inflammation within the liver. There is compelling evidence that activation of the transcription factor nuclear factor-κB (NF-κB) and downstream inflammatory signaling pathways systemically and in the liver are key events in the etiology of hepatic insulin resistance and ß-cell dysfunction, although the molecular mechanisms involved are incompletely understood. We here test the hypothesis that receptor activator of NF-κB ligand (RANKL), a prototypic activator of NF-κB, contributes to this process using both an epidemiological and experimental approach. In the prospective population-based Bruneck Study, a high serum concentration of soluble RANKL emerged as a significant (P<0.001) and independent risk predictor of T2DM manifestation. In close agreement, systemic or hepatic blockage of RANKL signaling in genetic and nutritional mouse models of T2DM resulted in a marked improvement of hepatic insulin sensitivity and amelioration or even normalization of plasma glucose concentrations and glucose tolerance. Overall, this study provides evidence for a role of RANKL signaling in the pathogenesis of T2DM. If so, translation to the clinic may be feasible given current pharmacological strategies to lower RANKL activity to treat osteoporosis.


Subject(s)
Diabetes Mellitus, Type 2/prevention & control , Insulin Resistance/physiology , Liver/metabolism , RANK Ligand/metabolism , Adult , Aged , Animals , Cell Line , Enzyme Activation , Female , HEK293 Cells , Humans , Male , Mice , Middle Aged , NF-kappa B/metabolism , Prospective Studies , RANK Ligand/antagonists & inhibitors
2.
Ann Rheum Dis ; 72(5): 761-8, 2013 May.
Article in English | MEDLINE | ID: mdl-22975756

ABSTRACT

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.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autophagy/immunology , Joints/pathology , Tumor Necrosis Factor-alpha/immunology , Adoptive Transfer , Animals , Apoptosis Regulatory Proteins/genetics , Autophagy-Related Protein 7 , Beclin-1 , Biomarkers , Bone Marrow Transplantation , Bone Resorption/immunology , Bone Resorption/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Osteoclasts/immunology , Osteoclasts/pathology , Osteoclasts/ultrastructure , Tumor Necrosis Factor-alpha/genetics
3.
Ann Rheum Dis ; 71(5): 785-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22402139

ABSTRACT

OBJECTIVES: Tissue fibrosis is a leading cause of death in patients with systemic sclerosis (SSc). Effective antifibrotic treatments are not available. Here, the authors investigated inhibition of hedgehog signalling by targeting Smoothened (Smo) as a novel antifibrotic approach. METHODS: The activation status of the hedgehog pathway was assessed by immunohistochemistry for Gli transcription factors and by quantification of hedgehog target genes. Hedgehog signalling was inhibited by the selective inhibitor LDE223 and by small interfering RNA against Smo in the models of bleomycin-induced dermal fibrosis and in tight-skin-1 mice. RESULTS: Hedgehog signalling is activated in SSc and in murine models of SSc. Inhibition of Smo either by LDE223 or by small interfering RNA prevented dermal thickening, myofibroblast differentiation and accumulation of collagen upon challenge with bleomycin. Targeting Smo also exerted potent antifibrotic effects in tight-skin-1 mice and did prevent progression of fibrosis and induced regression of pre-established fibrosis. CONCLUSIONS: Inhibition of hedgehog signalling exerted potent antifibrotic effects in preclinical models of SSc in both preventive and therapeutic settings. These findings might have direct translational implications because inhibitors of Smo are already available and yielded promising results in initial clinical trials.


Subject(s)
Fibrosis , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction , Skin Diseases/pathology , Skin/pathology , Animals , Biphenyl Compounds/pharmacology , Bleomycin/toxicity , Disease Models, Animal , Drug Therapy, Combination , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/prevention & control , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism , Skin Diseases/drug therapy , Skin Diseases/metabolism , Smoothened Receptor
4.
Nat Commun ; 3: 735, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22415826

ABSTRACT

The transforming growth factor-ß (TGF-ß) signalling pathway is a key mediator of fibroblast activation that drives the aberrant synthesis of extracellular matrix in fibrotic diseases. Here we demonstrate a novel link between transforming growth factor-ß and the canonical Wnt pathway. TGF-ß stimulates canonical Wnt signalling in a p38-dependent manner by decreasing the expression of the Wnt antagonist Dickkopf-1. Tissue samples from human fibrotic diseases show enhanced expression of Wnt proteins and decreased expression of Dickkopf-1. Activation of the canonical Wnt pathway stimulates fibroblasts in vitro and induces fibrosis in vivo. Transgenic overexpression of Dickkopf-1 ameliorates skin fibrosis induced by constitutively active TGF-ß receptor type I signalling and also prevents fibrosis in other TGF-ß-dependent animal models. These findings demonstrate that canonical Wnt signalling is necessary for TGF-ß-mediated fibrosis and highlight a key role for the interaction of both pathways in the pathogenesis of fibrotic diseases.


Subject(s)
Fibrosis/pathology , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Adult , Animals , Cells, Cultured , Extracellular Matrix/metabolism , Female , Fibrosis/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Male , Mice , Mice, Transgenic , Middle Aged , RNA Interference , RNA, Small Interfering , Receptors, Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Ann Rheum Dis ; 71(5): 761-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22328737

ABSTRACT

OBJECTIVES: Pathologic fibroblast activation drives fibrosis of the skin and internal organs in patients with systemic sclerosis (SSc). ß-catenin is an integral part of adherens junctions and a central component of canonical Wnt signaling. Here, the authors addressed the role of ß-catenin in fibroblasts for the development of SSc dermal fibrosis. METHODS: Nuclear accumulation of ß-catenin in fibroblasts was assessed by triple staining for ß-catenin, prolyl-4-hydroxylase-ß and 4',6-diamidino-2-phenylindole (DAPI). The expression of Wnt proteins in the skin was analysed by real-time PCR and immunohistochemistry. Mice with fibroblast-specific stabilisation or fibroblast-specific depletion were used to evaluate the role of ß-catenin in fibrosis. RESULTS: The auhors found significantly increased nuclear levels of ß-catenin in fibroblasts in SSc skin compared to fibroblasts in the skin of healthy individuals. The accumulation of ß-catenin resulted from increased expression of Wnt-1 and Wnt-10b in SSc. The authors further showed that the nuclear accumulation of ß-catenin has direct implications for the development of fibrosis: Mice with fibroblast-specific stabilisation of ß-catenin rapidly developed fibrosis within 2 weeks with dermal thickening, accumulation of collagen and differentiation of resting fibroblasts into myofibroblasts. By contrast, fibroblast-specific deletion of ß-catenin significantly reduced bleomycin-induced dermal fibrosis. CONCLUSIONS: The present study findings identify ß-catenin as a key player of fibroblast activation and tissue fibrosis in SSc. Although further translational studies are necessary to test the efficacy and tolerability of ß-catenin/Wnt inhibition in SSc, the present findings may have clinical implications, because selective inhibitors of ß-catenin/Wnt signaling have recently entered clinical trials.


Subject(s)
Fibroblasts/metabolism , Fibrosis/metabolism , Scleroderma, Systemic/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adult , Aged , Cell Nucleus/metabolism , Cell Nucleus/pathology , Female , Fibroblasts/pathology , Fibrosis/pathology , Humans , Male , Middle Aged , Scleroderma, Systemic/pathology , Skin/metabolism , Skin/pathology , Wnt Proteins/genetics , Young Adult
6.
Hepatology ; 53(4): 1259-69, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21480331

ABSTRACT

UNLABELLED: Chronic diseases of the biliary system are common and may cause fibrosis and eventually progression to liver cirrhosis. The aim was to define a new mouse model of a cholangiopathy leading to liver fibrosis in fra-1tg mice. Liver pathology of fra-1tg mice was analyzed in detail by histology and flow cytometry. Transcript levels of fibrosis-related genes and matrix metalloproteinase (MMP) activities were quantified and immunohistochemical analysis additionally applied. The role of the immune system in this model was analyzed by crossing fra-1tg mice with rag2(-/-) mice. Furthermore, expression of Fra-1 in corresponding human liver diseases was investigated on transcription level and histologically. Fra-1tg mice spontaneously develop biliary fibrosis preceded by ductular proliferation and infiltration of inflammatory cells. Fra-1 protein is present in cholangiocytes and inflammatory cells within the liver. These findings were replicated in human biopsies of patients with advanced liver fibrosis. The inflammatory infiltrate showed a strong increase in activated T cells and decreased natural killer (NK), natural killer T cells (NKT), and B cells in fra-1tg mice as compared to wildtype mice. Moreover, fra-1tg mice develop biliary fibrosis with a time-dependent increase in hepatic collagen content and increase in relative messenger RNA (mRNA) expression of profibrotic genes. Attenuation but not complete prevention of collagen accumulation in liver was observed in the fra-1tg × rag2(-/-) mice. However, transplantation of fra-1tg bone marrow cells into wildtype mice could not induce disease. CONCLUSION: Fra-1tg mice spontaneously develop a progressive biliary disease. These mice are an attractive model for the investigation of cholangiopathies and their interaction with the immune system.


Subject(s)
Cholangitis/chemically induced , Liver Cirrhosis/chemically induced , Proto-Oncogene Proteins c-fos/physiology , Animals , Chemokines/biosynthesis , Disease Models, Animal , Fibrosis , Humans , Liver/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Transgenic , Transcription Factor AP-1/physiology
7.
Ann Rheum Dis ; 69(12): 2152-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20858621

ABSTRACT

UNLABELLED: Introduction Inflammation is a major risk factor for systemic bone loss. Proinflammatory cytokines like tumour necrosis factor (TNF) affect bone homeostasis and induce bone loss. It was hypothesised that impaired bone formation is a key component in inflammatory bone loss and that Dkk-1, a Wnt antagonist, is a strong inhibitor of osteoblast-mediated bone formation. METHODS: TNF transgenic (hTNFtg) mice were treated with neutralising antibodies against TNF, Dkk-1 or a combination of both agents. Systemic bone architecture was analysed by bone histomorphometry. The expression of ß-catenin, osteoprotegerin and osteocalcin was analysed. In vitro, primary osteoblasts were stimulated with TNF and analysed for their metabolic activity and expression of Dkk-1 and sclerostin. Sclerostin expression and osteocyte death upon Dkk-1 blockade were analysed in vivo. RESULTS: Neutralisation of Dkk-1 completely protected hTNFtg mice from inflammatory bone loss by preventing TNF-mediated impaired osteoblast function and enhanced osteoclast activity. These findings were accompanied by enhanced skeletal expression of ß-catenin, osteocalcin and osteoprotegerin. In vitro, TNF rapidly increased Dkk-1 expression in primary osteoblasts and effectively blocked osteoblast differentiation. Moreover, blockade of Dkk-1 not only rescued impaired osteoblastogenesis but also neutralised TNF-mediated sclerostin expression in fully differentiated osteoblasts in vitro and in vivo. CONCLUSIONS: These findings indicate that low bone formation and expression of Dkk-1 trigger inflammatory bone loss. Dkk-1 blocks osteoblast differentiation, induces sclerostin expression and leads to osteocyte death. Inhibition of Dkk-1 may thus be considered as a potent strategy to protect bone from inflammatory damage.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Bone Morphogenetic Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/metabolism , Bone Resorption/prevention & control , Cell Differentiation/drug effects , Cells, Cultured , Genetic Markers , Glycoproteins , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Osteoblasts/drug effects , Osteoblasts/physiology , Osteocytes/metabolism , Osteogenesis/drug effects , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
8.
J Bone Miner Res ; 23(10): 1650-60, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18505368

ABSTRACT

The objective of this study was to investigate the function of inflammatory bone marrow infiltrates found in vicinity to joints affected by inflammatory arthritis. These bone marrow infiltrates are rich in B cells and emerge at the interphase between bone marrow and synovial inflammatory tissue, where cortical bone has been broken. We deleted an essential molecule of B-cell development, Brutons tyrosine kinase (Btk), in arthritic TNF-transgenic mice and studied its effect on bone marrow inflammation. Although antigen responses, immunoglobulin levels, and autoantibody production were diminished in Btk(-/-)hTNFtg mice, synovial inflammation developed normally. However, bone marrow infiltrates were significantly diminished in Btk(-/-)hTNFtg mice, which lead to impaired bone formation at endosteal sites underneath bone erosions and an increased invasion of synovial inflammatory cells into the bone marrow. Expression of bone morphogenic protein-7 was dramatically decreased in Btk(-/-)hTNFtg mice. These results do not only indicate that bone formation at endosteal regions next to bone marrow infiltrates is driven by B cells but also show that bone marrow aggregates in the vicinity of inflamed joint appear as an attempt to counter the invasion of inflammatory tissue into the bone marrow.


Subject(s)
Arthritis, Rheumatoid/pathology , B-Lymphocytes/pathology , Bone Development , Animals , Base Sequence , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Transgenic , Polymerase Chain Reaction
9.
J Biol Chem ; 283(11): 7082-93, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18156182

ABSTRACT

Here we report on the structure, expression, and function of a novel cartilage-specific gene coding for a 17-kDa small, highly charged, and secreted protein that we termed Ucma (unique cartilage matrix-associated protein). The protein is processed by a furin-like protease into an N-terminal peptide of 37 amino acids and a C-terminal fragment (Ucma-C) of 74 amino acids. Ucma is highly conserved between mouse, rat, human, dog, clawed frog, and zebrafish, but has no homology to other known proteins. Remarkable are 1-2 tyrosine sulfate residues/molecule and dense clusters of acidic and basic residues in the C-terminal part. In the developing mouse skeleton Ucma mRNA is expressed in resting chondrocytes in the distal and peripheral zones of epiphyseal and vertebral cartilage. Ucma is secreted into the extracellular matrix as an uncleaved precursor and shows the same restricted distribution pattern in cartilage as Ucma mRNA. In contrast, antibodies prepared against the processed C-terminal fragment located Ucma-C in the entire cartilage matrix, indicating that it either diffuses or is retained until chondrocytes reach hypertrophy. During differentiation of an MC615 chondrocyte subclone in vitro, Ucma expression parallels largely the expression of collagen II and decreases with maturation toward hypertrophic cells. Recombinant Ucma-C does not affect expression of chondrocyte-specific genes or proliferation of chondrocytes, but interferes with osteogenic differentiation of primary osteoblasts, mesenchymal stem cells, and MC3T3-E1 pre-osteoblasts. These findings suggest that Ucma may be involved in the negative control of osteogenic differentiation of osteochondrogenic precursor cells in peripheral zones of fetal cartilage and at the cartilage-bone interface.


Subject(s)
Cartilage/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation , Osteogenesis/physiology , Proteins/physiology , Amino Acid Sequence , Animals , Base Sequence , Bone and Bones/metabolism , Chondrocytes/metabolism , Extracellular Matrix Proteins , Humans , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Mesenchymal Stem Cells/cytology , Mice , Molecular Sequence Data , Osteoblasts/metabolism , Proteins/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...