Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Frank Echtermeyer is active.

Publication


Featured researches published by Frank Echtermeyer.


Nature Medicine | 2009

Syndecan-4 regulates ADAMTS-5 activation and cartilage breakdown in osteoarthritis

Frank Echtermeyer; Jessica Bertrand; Rita Dreier; Ingmar Meinecke; Katja Neugebauer; Martin Fuerst; Yun Jong Lee; Yeong Wook Song; Christine Herzog; Gregor Theilmeier; Thomas Pap

Aggrecan cleavage by a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 5 (ADAMTS-5) is crucial for the breakdown of cartilage matrix during osteoarthritis, a degenerative joint disease that leads to the progressive destruction of articular structures. The mechanisms of ADAMTS-5 activation and their links to the pathogenesis of osteoarthritis remain poorly understood, but syndecans have been shown to be involved in the activation of ADAMTS-4 (ref. 3). Here we show that syndecan-4 is specifically induced in type X collagen–producing chondrocytes both in human osteoarthritis and in murine models of the disease. The loss of syndecan-4 in genetically modified mice and intra-articular injections of syndecan-4–specific antibodies into wild-type mice protect from proteoglycan loss and thereby prevent osteoarthritic cartilage damage in a surgically induced model of osteoarthritis. The occurrence of less severe osteoarthritis-like cartilage destruction in both syndecan-4–deficient mice and syndecan-4–specific antibody–treated wild-type mice results from a marked decrease in ADAMTS-5 activity. Syndecan-4 controls the activation of ADAMTS-5 through direct interaction with the protease and through regulating mitogen-activated protein kinase (MAPK)-dependent synthesis of matrix metalloproteinase-3 (MMP-3). Our data suggest that strategies aimed at the inhibition of syndecan-4 will be of great value for the treatment of cartilage damage in osteoarthritis.


Arthritis & Rheumatism | 2009

Calcification of articular cartilage in human osteoarthritis

Martin Fuerst; Jessica Bertrand; L. Lammers; Rita Dreier; Frank Echtermeyer; Y. Nitschke; F. Rutsch; F. K. W. Schäfer; Oliver Niggemeyer; J. Steinhagen; Christoph H. Lohmann; Thomas Pap; Wolfgang Rüther

OBJECTIVE Hypertrophic chondrocyte differentiation is a key step in endochondral ossification that produces basic calcium phosphates (BCPs). Although chondrocyte hypertrophy has been associated with osteoarthritis (OA), chondrocalcinosis has been considered an irregular event and linked mainly to calcium pyrophosphate dihydrate (CPPD) deposition. The aim of this study was to determine the prevalence and composition of calcium crystals in human OA and analyze their relationship to disease severity and markers of chondrocyte hypertrophy. METHODS One hundred twenty patients with end-stage OA undergoing total knee replacement were prospectively evaluated. Cartilage calcification was studied by conventional x-ray radiography, digital-contact radiography (DCR), field-emission scanning electron microscopy (FE-SEM), and synovial fluid analysis. Cartilage calcification findings were correlated with scores of knee function as well as histologic changes and chondrocyte hypertrophy as analyzed in vitro. RESULTS DCR revealed mineralization in all cartilage specimens. Its extent correlated significantly with the Hospital for Special Surgery knee score but not with age. FE-SEM analysis showed that BCPs, rather than CPPD, were the prominent minerals. On histologic analysis, it was observed that mineralization correlated with the expression of type X collagen, a marker of chondrocyte hypertrophy. Moreover, there was a strong correlation between the extent of mineralization in vivo and the ability of chondrocytes to produce BCPs in vitro. The induction of hypertrophy in healthy human chondrocytes resulted in a prominent mineralization of the extracellular matrix. CONCLUSION These results indicate that mineralization of articular cartilage by BCP is an indissociable process of OA and does not characterize a specific subset of the disease, which has important consequences in the development of therapeutic strategies for patients with OA.


American Journal of Pathology | 2002

Absence of Decorin Adversely Influences Tubulointerstitial Fibrosis of the Obstructed Kidney by Enhanced Apoptosis and Increased Inflammatory Reaction

Liliana Schaefer; Katarina Macakova; Igor Raslik; Miroslava Micegova; Hermann Josef Gröne; Elke Schönherr; Horst Robenek; Frank Echtermeyer; Susanne Grässel; Peter Bruckner; Roland M. Schaefer; Renato V. Iozzo; Hans Kresse

Decorin, a small dermatan-sulfate proteoglycan, participates in extracellular matrix assembly and influences directly and indirectly cell behavior via interactions with signaling membrane receptors and transforming growth factor (TGF)-beta. We have therefore compared the development of tubulointerstitial kidney fibrosis in wild-type (WT) and decorin-/- mice in the model of unilateral ureteral obstruction. Without obstruction, kidneys from decorin-/- mice did not differ in any aspect from their WT counterparts. However, already 12 hours after obstruction decorin-/- animals showed lower levels of p27(KIP1) and soon thereafter a more pronounced up-regulation and activation of initiator and effector caspases followed by enhanced apoptosis of tubular epithelial cells. Later, a higher increase of TGF-beta1 became apparent. After 7 days, there was an up to 15-fold transient up-regulation of the related proteoglycan biglycan, which was mainly caused by the appearance of biglycan-expressing mononuclear cells. Other small proteoglycans showed no similar response. Because of enhanced degradation of type I collagen, end-stage kidneys from decorin-/- animals were more atrophic than WT kidneys. These data suggest that decorin exerts beneficial effects on tubulointerstitial fibrosis, primarily by influencing the expression of a key cyclin-dependent kinase inhibitor and by limiting the degree of apoptosis, mononuclear cell infiltration, tubular atrophy, and expression of TGF-beta1.


Annals of the Rheumatic Diseases | 2012

Early structural changes in cartilage and bone are required for the attachment and invasion of inflamed synovial tissue during destructive inflammatory arthritis

Adelheid Korb-Pap; Athanasios Stratis; Katja Mühlenberg; Birgit Niederreiter; Silvia Hayer; Frank Echtermeyer; Richard Stange; Jochen Zwerina; Thomas Pap; Hermann Pavenstädt; Georg Schett; Josef S Smolen; Kurt Redlich

Objective To elucidate the mechanisms involved in cartilage damage in an experimental model of rheumatoid arthritis (RA) by specifically addressing the time course of extracellular matrix degradation and the contribution of cell–matrix interactions for initiation and perpetuation of this process. Methods The human tumour necrosis factor (TNF) transgenic (hTNFtg) mouse model of RA was used to analyse the time course of pannus attachment to the cartilage and cartilage destruction, respectively, and crossed hTNFtg mice with interleukin (IL)-1−/− animals were used to investigate the role of IL-1 on these TNF-induced mechanisms in vivo. In addition, an in vitro attachment assay using synovial fibroblasts (SFs) from hTNFtg mice and freshly isolated articular cartilage was used to determine the role of proteoglycan loss in attachment of SFs and the role of the transmembrane heparan sulfate proteoglycan syndecan-4. Results In vivo analyses of hTNFtg mice showed that proteoglycan loss induced by IL-1 precedes and constitutes an important prerequisite for these processes as, in hTNFtg mice, IL-1 deficiency protected from the loss of cartilage proteoglycans and almost completely prevented the attachment and subsequent invasion of inflamed synovial tissue into cartilage. In vitro studies confirmed that loss of cartilage proteoglycans is required for attachment of SFs and that syndecan-4 is prominently involved in SF attachment and activation. Conclusions The results of this study suggest that the loss of cartilage proteoglycans is an early event in the course of destructive arthritis that facilitates the attachment of the inflamed synovial membrane and also initiates matrix degradation and inflammation through cell–matrix interactions.


The Scientific World Journal | 2003

Syndecan-1 as a Regulator of Chemokine Function

Martin Götte; Frank Echtermeyer

Chemokines are a family of chemotactic cytokines that play critical roles in leukocyte recruitment to sites of inflammation. Characterized by the presence of conserved aminoterminal cysteine residues, these lowmolecular weight proteins signal through G-protein–coupled receptors with seven transmembrane domains[1,2]. Apart from their function in leukocyte recruitment, chemokines and their receptors have been ascribed roles in angiogenesis and tumor growth, infections, Th1 and Th2 responses, as well as the maturation and development of several leukocyte subpopulations[1]. Considering their fundamental role in orchestrating the inflammatory response, it is no surprise that a dysregulation of chemokine function is observed in a variety of pathological conditions, such as pulmonary fibrosis, hypersensitivity responses, chronic inflammation, and cancer[1,2,3,4]. For this reason, it is important to know how chemokine action is regulated in molecular detail, in order to develop more efficient therapies for diseases characterized by chemokine malfunction. Two recent papers shed more light on the regulation of chemokine function, uncovering a crucial role for the cell surface heparan sulfate (HS) proteoglycan syndecan-1 in the generation of chemokine gradients[5,6]. Using mice deficient in the matrix-metalloprotease matrilysin in an experimental model of lung fibrosis, Li et al.[5] observed an impaired transepithelial migration of neutrophils, which resulted in protection against the lethal effect of bleomycin-induced lung injury. They found that matrilysin sheds a complex of syndecan-1 and the CXC chemokine KC from the mucosal surface, which serves as a chemotactic gradient for neutrophils. Marshall et al.[6] used an in vitro model of transendothelial migration to demonstrate that syndecan-1 forms a chemotactic IL-8 gradient at the endothelial cell surface. An increase of constitutive syndecan-1/IL-8 complex shedding in the presence of a neutralizing antibody to plasminogen activator inhibitor-1 (PAI-1) resulted in an inhibition of transendothelial neutrophil migration. The syndecans are a family of cell surface heparan sulfate proteoglycans (HSPG), which act as adhesion molecules, modulators of growth factor function, and coreceptors in processes as diverse as morphogenesis, tissue repair, host defense, tumor development, and energy metabolism[7,8]. Protease-mediated cleavage of the intact syndecan ectodomains (“shedding”) converts the cell-surface molecules into soluble effectors[9]. Increasing evidence suggests an important role for the syndecans in the


Journal of Immunology | 2009

Role of the Heparan Sulfate Proteoglycan Syndecan-1 (CD138) in Delayed-Type Hypersensitivity

Behzad Kharabi Masouleh; Gerdy B. ten Dam; Martin K. Wild; Ruth Seelige; Johan van der Vlag; Angelique L. Rops; Frank Echtermeyer; Dietmar Vestweber; Toin H. van Kuppevelt; Ludwig Kiesel; Martin Götte

The cell surface heparan sulfate proteoglycan syndecan-1 (CD138) modulates the activity of chemokines, cytokines, integrins, and other adhesion molecules which play important roles in the regulation of inflammation. We have previously shown that syndecan-1-deficient murine leukocytes display increased interactions with endothelial cells and increased diapedesis in vivo and in vitro. In this study, we demonstrate that syndecan-1 has an important function as a negative modulator in the murine contact allergy model of oxazolone-mediated delayed-type hypersensitivity (DTH). Following elicitation of the DTH response, syndecan-1-deficient mice showed an increase in leukocyte recruitment, resulting in an increased and prolonged edema formation. Expression of the cytokines TNF-α and IL-6 of the chemokines CCL5/RANTES and CCL-3/MIP-1α and of the adhesion molecule ICAM-1 were significantly increased in syndecan-1-deficient compared with wild-type mice. In wild-type mice, syndecan-1 mRNA and protein expression was reduced during the DTH response. The differentially increased adhesion of syndecan-1-deficient leukocytes to ICAM-1 was efficiently inhibited in vitro by CD18-blocking Abs, which emerges as one mechanistic explanation for the anti-inflammatory effects of syndecan-1. Collectively, our results show an important role of syndecan-1 in the contact DTH reaction, identifying syndecan-1 as a novel target in anti-inflammatory therapy.


Cardiovascular Research | 2011

Syndecan-4 signalling inhibits apoptosis and controls NFAT activity during myocardial damage and remodelling

Frank Echtermeyer; Thomas Harendza; Svenja Hubrich; Anika Lorenz; Christine Herzog; Martin Mueller; Martina Schmitz; Andrea Grund; Jan Larmann; Jörg Stypmann; Bernhard Schieffer; Ralf Lichtinghagen; Denise Hilfiker-Kleiner; Kai C. Wollert; Jörg Heineke; Gregor Theilmeier

AIMS Myocardial infarction (MI) results in acute impairment of left ventricular (LV) function through the initial development of cardiomyocyte death and subsequent progression of LV remodelling. The expression of syndecan-4 (Sdc4), a transmembrane proteoglycan, is up-regulated after MI, but its function in the heart remains unknown. Here, we characterize the effects of Sdc4 deficiency in murine myocardial ischaemia and permanent infarction. METHODS AND RESULTS Targeted deletion of Sdc4 (Sdc4(-/-)) leads to increased myocardial damage after ischaemic-reperfusion injury due to enhanced cardiomyocyte apoptosis associated with reduced activation of extracellular signal-regulated kinase in cardiomyocytes in vitro and in vivo. After ischaemic-reperfusion injury and permanent infarction, we observed an increase in cardiomyocyte area, nuclear translocation of nuclear factor of activated T cells (NFAT), and transcription of the NFAT target rcan1.4 in wild-type mice. NFAT pathway activation was enhanced in Sdc4(-/-) mice. In line with the in vivo data, NFAT activation and hypertrophy occurs in isolated cardiomyocytes with reduced Sdc4 expression during phenylephrine stimulation in vitro. Despite the initially increased myocardial damage, echocardiography revealed improved LV geometry and function in Sdc4(-/-) mice 7 days after MI. CONCLUSION Interception of the Sdc4 pathway enhances infarct expansion and hypertrophic remodelling during early infarct healing in ischaemic-reperfusion injury and permanent infarction mouse models and exerts net beneficial effects on LV function.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

The Proteoglycan Syndecan 4 Regulates Transient Receptor Potential Canonical 6 Channels via RhoA/Rho-associated Protein Kinase Signaling

Ying Liu; Frank Echtermeyer; Florian Thilo; Gregor Theilmeier; Antje Schmidt; Ralf Schülein; Boye L. Jensen; Christoph Loddenkemper; Vera Jankowski; Niels Marcussen; Maik Gollasch; William J. Arendshorst; Martin Tepel

Objective—Syndecan 4 (Sdc4) modulates signal transduction and regulates activity of protein channels. Sdc4 is essential for the regulation of cellular permeability. We hypothesized that Sdc4 may regulate transient receptor potential canonical 6 (TRPC6) channels, a determinant of glomerular permeability, in a RhoA/Rho-associated protein kinase-dependent manner. Methods and Results—Sdc4 knockout (Sdc4−/−) mice showed increased glomerular filtration rate and ameliorated albuminuria under baseline conditions and after bovine serum albumin overload (each P<0.05). Using reverse transcription–polymerase chain reaction and immunoblotting, Sdc4−/− mice showed reduced TRPC6 mRNA by 79% and TRPC6 protein by 82% (each P<0.05). Sdc4−/− mice showed an increased RhoA activity by 87% and increased phosphorylation of ezrin in glomeruli by 48% (each P<0.05). Sdc4 knockdown in cultured podocytes reduced TRPC6 gene expression and reduced the association of TRPC6 with plasma membrane and TRPC6-mediated calcium influx and currents. Sdc4 knockdown inactivated negative regulatory protein Rho GTPase activating protein by 33%, accompanied by a 41% increase in RhoA activity and increased phosphorylation of ezrin (P<0.05). Conversely, overexpression of Sdc4 reduced RhoA activity and increased TRPC6 protein and TRPC6-mediated calcium influx and currents. Conclusion—Our results establish a previously unknown function of Sdc4 for regulation of TRPC6 channels and support the role of Sdc4 for the regulation of glomerular permeability.


Cardiovascular Research | 2014

Thrombomodulin's lectin-like domain reduces myocardial damage by interfering with HMGB1-mediated TLR2 signalling

Christine Herzog; Anika Lorenz; Hans-Jörg Gillmann; Arpita Chowdhury; Jan Larmann; Thomas Harendza; Frank Echtermeyer; Martin Müller; Martina Schmitz; Jörg Stypmann; Daniela G. Seidler; Martin Damm; Sebastian Stehr; Thea Koch; Kai C. Wollert; Edward M. Conway; Gregor Theilmeier

AIMS Thrombomodulin (TM), via its lectin-like domain (LLD), exhibits anti-inflammatory properties partly by sequestering the pro-inflammatory cytokine, high-mobility group box 1 (HMGB1). Since myocardial damage after ischaemia and reperfusion is mediated by inflammation, we evaluated the cardioprotective effects of the LLD of TM. Using an in vivo mouse model of transient ischaemia and in vitro models of cardiomyocyte hypoxia, we assessed the ability of the LLD to suppress HMGB1-mediated activation of the receptors, receptor for advanced glycation endproducts (RAGEs) and Toll-like receptors (TLRs) 2 and 4. METHODS AND RESULTS Thirty-minute myocardial ischaemia was induced in isoflurane-anaesthetized mice followed by 24 h of reperfusion in wild-type (WT) mice, in mice lacking the LLD of TM (TM(LeD/LeD) mice), and in WT with systemic overexpression of the LLD of TM induced by hydrodynamic transfection. Infarct size, HMGB1 protein, and apoptotic cells were significantly increased in TM(LeD/LeD) mice when compared with WT. Neonatal rat cardiomyocytes transfected with TLR2-, TLR4-, and RAGE-siRNA were exposed to hypoxia (0.8% O2) and reoxygenation (21% O2). HMGB1 augmented hypoxia-induced apoptosis in TLR2- but not in RAGE- or TLR4-suppressed cells. Administration of HMGB1- and TLR2-blocking antibodies in TM(LeD/LeD) mice prior to myocardial ischaemia diminished apoptosis. Therapeutic systemic gene therapy using the LLD reduced the infarct size and HMGB1 protein levels 24 h after reperfusion. CONCLUSION The LLD of TM suppresses HMGB1-induced and TLR2-mediated myocardial reperfusion injury and apoptosis in vitro and in vivo.


Arthritis & Rheumatism | 2013

Syndecan 4 supports bone fracture repair, but not fetal skeletal development, in mice

Jessica Bertrand; Richard Stange; Heriburg Hidding; Frank Echtermeyer; G. Nalesso; Lars Godmann; Melanie Timmen; Peter Bruckner; Francesco Dell'Accio; Michael J. Raschke; Thomas Pap; Rita Dreier

OBJECTIVE Syndecan 4, a heparan sulfate proteoglycan, has been associated with osteoarthritis. The present study was undertaken to analyze the functional role of syndecan 4 in endochondral ossification of mouse embryos and in adult fracture repair, which, like osteoarthritis, involves an inflammatory component. METHODS Sdc4 promoter activity was analyzed in Sdc4(-/-) lacZ-knockin mice, using β-galactosidase staining. Endochondral ossification in embryos from embryonic day 16.5 was assessed by histologic and immunohistologic staining. Bone fracture repair was analyzed in femora of adult mice on days 7 and 14 postfracture. To evaluate Sdc2 and Sdc4 gene expression with and without tumor necrosis factor α (TNFα) and Wnt-3a stimulation, quantitative real-time polymerase chain reaction was performed. RESULTS In Sdc4(-/-) lacZ-knockin animals, syndecan 4 promoter activity was detectable at all stages of chondrocyte differentiation, and Sdc4 deficiency inhibited chondrocyte proliferation. Aggrecan turnover in the uncalcified cartilage of the epiphysis was decreased transiently in vivo, but this did not lead to a growth phenotype at birth. In contrast, among adult mice, fracture healing was markedly delayed in Sdc4(-/-) animals and was accompanied by increased callus formation. Blocking of inflammation via anti-TNFα treatment during fracture healing reduced these changes in Sdc4(-/-) mice to levels observed in wild-type controls. We analyzed the differences between the mild embryonic and the severe adult phenotype, and found a compensatory up-regulation of syndecan 2 in the developing cartilage of Sdc4(-/-) mice that was absent in adult tissue. Stimulation of chondrocytes with Wnt-3a in vitro led to increased expression of syndecan 2, while stimulation with TNFα resulted in up-regulation of syndecan 4 but decreased expression of syndecan 2. TNFα stimulation reduced syndecan 2 expression and increased syndecan 4 expression even in the presence of Wnt-3a, suggesting that inflammation has a strong effect on the regulation of syndecan expression. CONCLUSION Our results demonstrate that syndecan 4 is functionally involved in endochondral ossification and that its loss impairs fracture healing, due to inhibition of compensatory mechanisms under inflammatory conditions.

Collaboration


Dive into the Frank Echtermeyer's collaboration.

Top Co-Authors

Avatar

Thomas Pap

University of Münster

View shared research outputs
Top Co-Authors

Avatar

Jessica Bertrand

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rita Dreier

University of Münster

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard Stange

Humboldt University of Berlin

View shared research outputs
Top Co-Authors

Avatar

Gregor Theilmeier

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jan Larmann

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

G. Nalesso

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Gregor Theilmeier

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge