Network


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

Hotspot


Dive into the research topics where Sylvia Thiele is active.

Publication


Featured researches published by Sylvia Thiele.


Journal of Bone and Mineral Research | 2012

Selective Glucocorticoid Receptor Modulation Maintains Bone Mineral Density in Mice

Sylvia Thiele; Nicole Ziegler; Elena Tsourdi; Karolien De Bosscher; Jan Tuckermann; Lorenz C. Hofbauer; Martina Rauner

Glucocorticoids (GCs) are potent anti‐inflammatory drugs, but their use is limited by their adverse effects on the skeleton. Compound A (CpdA) is a novel GC receptor modulator with the potential for an improved risk/benefit profile. We tested the effects of CpdA on bone in a mouse model of GC‐induced bone loss. Bone loss was induced in FVB/N mice by implanting slow‐release pellets containing either vehicle, prednisolone (PRED) (3.5 mg), or CpdA (3.5 mg). After 4 weeks, mice were killed to examine the effects on the skeleton using quantitative computed tomography, bone histomorphometry, serum markers of bone turnover, and gene expression analysis. To assess the underlying mechanisms, in vitro studies were performed with human bone marrow stromal cells (BMSCs) and murine osteocyte‐like cells (MLO‐Y4 cells). PRED reduced the total and trabecular bone density in the femur by 9% and 24% and in the spine by 11% and 20%, respectively, whereas CpdA did not influence these parameters. Histomorphometry confirmed these results and further showed that the mineral apposition rate was decreased by PRED whereas the number of osteoclasts was increased. Decreased bone formation was paralleled by a decline in serum procollagen type 1 N‐terminal peptide (P1NP), reduced skeletal expression of osteoblast markers, and increased serum levels of the osteoblast inhibitor dickkopf‐1 (DKK‐1). In addition, serum CTX‐1 and the skeletal receptor activator of NF‐κB ligand (RANKL)/osteoprotegerin (OPG) ratio were increased by PRED. None of these effects were observed with CpdA. Consistent with the in vivo data, CpdA did not increase the RANKL/OPG ratio in MLO‐Y4 cells or the expression of DKK‐1 in bone tissue, BMSCs, and osteocytes. Finally, CpdA also failed to transactivate DKK‐1 expression in bone tissue, BMSCs, and osteocytes. This study underlines the bone‐sparing potential of CpdA and suggests that by preventing increases in the RANKL/OPG ratio or DKK‐1 in osteoblast lineage cells, GC‐induced bone loss may be ameliorated.


Journal of Cellular Biochemistry | 2011

Expression profile of WNT molecules in prostate cancer and its regulation by aminobisphosphonates.

Sylvia Thiele; Martina Rauner; Claudia Goettsch; Tilman D. Rachner; Peggy Benad; Susanne Fuessel; Kati Erdmann; Christine Hamann; Gustavo Baretton; Manfred P. Wirth; Franz Jakob; Lorenz C. Hofbauer

Skeletal metastases represent a frequent complication in patients with advanced prostate cancer (PCa) and often require bisphosphonate treatment to limit skeletal‐related events. Metastasized PCa cells disturb bone remodeling. Since the WNT signaling pathway regulates bone remodeling and has been implicated in tumor progression and osteomimicry, we analyzed the WNT profile of primary PCa tissues and PCa cell lines and assessed its regulation by bisphosphonates. Prostate tissue (n = 18) was obtained from patients with benign prostate hyperplasia (BPH) and PCa patients with different disease stages. Serum samples were collected from 62 patients. Skeletal metastases were present in 17 patients of whom 6 had been treated with zoledronic acid. The WNT profile and its regulation by bisphoshonates were analyzed in tissue RNA extracts and serum samples as well as in osteotropic (PC3) and non‐osteotropic (DU145, LNCaP) PCa cell lines. Several members of the WNT pathway, including WNT5A, FZD5, and DKK1 were highly up‐regulated in PCa tissue from patients with advanced PCa. Interestingly, osteotropic cells showed a distinct WNT profile compared to non‐osteotropic cells. While WNT5A, FZD5, and DKK1 were highly expressed in PC3 cells, WNT1 and SFRP1 mRNA levels were higher in DU145 cells. Moreover, zoledronic acid down‐regulated mRNA levels of WNT5A (−34%), FZD5 (−60%), and DKK1 (−46%) in PC3 cells. Interestingly, patients with skeletal metastases who received zoledronic acid had twofold higher DKK1 serum levels compared to bisphosphonate‐naive patients. The WNT signaling pathway is up‐regulated in advanced PCa, differentially expressed in osteotropic versus non‐osteotropic cells, and is regulated by zoledronic acid. J. Cell. Biochem. 112: 1593–1600, 2011.


Endocrinology | 2011

Dissociation of Osteogenic and Immunological Effects by the Selective Glucocorticoid Receptor Agonist, Compound A, in Human Bone Marrow Stromal Cells

Martina Rauner; Claudia Goettsch; Nicola Stein; Sylvia Thiele; Martin Bornhaeuser; Karolien De Bosscher; Guy Haegeman; Jan Tuckermann; Lorenz C. Hofbauer

Glucocorticoids (GCs) regulate various physiological processes, including bone remodeling. Whereas physiological amounts of GCs are required for proper human osteoblast differentiation, prolonged exposure to GCs leads to substantial bone loss in vivo predominantly by inhibiting osteoblast functions. Compound A (CpdA) is a novel GC receptor modulator with the potential of an improved benefit/risk profile. Here we tested the osteoimmunological effects of CpdA on primary human osteoblasts and their paracrine interactions with osteoclasts. To assess the antiinflammatory potential of CpdA in human bone marrow stromal cell (BMSC)-derived osteoblasts, cells were stimulated with lipopolysaccharide and cytokine expression was determined. Similar to dexamethasone (DEX), CpdA profoundly suppressed lipopolysaccharide-induced TNF-α (-63%), IL-1β (-38%), and IL-6 (-36%) (P < 0.05) mRNA levels. Of note, CpdA failed to induce osteogenic differentiation of BMSCs, whereas DEX and budesonide enhanced matrix mineralization an d increased runt-related transcription factor 2 and alkaline phosphatase mRNA levels up to 5-fold in a dose-dependent manner. Interestingly, each substance promoted cell proliferation by 7-10% and suppressed apoptosis by 25-30% at low concentrations and early differentiation stages, whereas high concentrations (1 μm) suppressed proliferation and stimulated apoptosis in mature osteoblasts. Finally, CpdA did not increase the receptor activator of nuclear factor-κB ligand to osteoprotegerin mRNA ratio as compared with DEX and did not stimulate the formation of osteoclasts in coculture with BMSCs. In summary, CpdA displays dissociated osteogenic and immunological effects in human BMSCs that are distinct from those of conventional GCs. Whether the specific osteoimmunological profile of CpdA translates into a relevant in vivo effect needs to be further explored.


Endocrinology | 2013

Effects of the Selective Glucocorticoid Receptor Modulator Compound A on Bone Metabolism and Inflammation in Male Mice With Collagen-Induced Arthritis

Martina Rauner; Sylvia Thiele; Kathrin Sinningen; Maria Winzer; Juliane Salbach-Hirsch; Katrin Peschke; Guy Haegeman; Jan Tuckermann; Lorenz C. Hofbauer

Glucocorticoids (GCs) are potent drugs to treat rheumatoid arthritis but exert adverse skeletal effects. Compound A (CpdA) is a selective GC receptor modulator with an improved risk/benefit profile in mouse models of inflammation and bone loss. Here we tested whether CpdA also exerts bone-sparing effects under proinflammatory circumstances using the collagen-induced arthritis model, a murine model of rheumatoid arthritis. CpdA decreased disease activity, paw swelling, and the paw temperature by 43%, 12%, and 7%, respectively, but was less potent than dexamethasone (DEX), which reduced these parameters by 72%, 22%, and 10%, respectively. Moreover, T cells isolated from CpdA- and DEX-treated animals were less active based on proliferation rates after challenge with type II collagen and produced smaller amounts of interferon-γ and TNF as compared with T cells from PBS-treated mice. Histological assessment of the joints confirmed the weaker potency of CpdA as compared with DEX in preventing infiltration of inflammatory cells, induction of osteoclastogenesis, and destruction of articular cartilage. Due to the lack of GC-susceptible arthritis models, we were not able to fully address the bone-sparing potential of CpdA in inflammatory conditions. Nevertheless, the bone formation marker procollagen type 1 N-terminal peptide, a surrogate marker for GC-mediated suppression of bone formation, was significantly decreased by DEX in arthritic mice but not by CpdA. Our data indicate that CpdA moderately suppresses inflammation, whereas the concurrent effects on bone remain unknown. In light of its narrow therapeutic range, CpdA may be more useful as a molecular tool for dissecting GC actions rather than a therapeutic agent.


Journal of Cellular Biochemistry | 2014

Sulfated Glycosaminoglycans Support Osteoblast Functions and Concurrently Suppress Osteoclasts

Juliane Salbach-Hirsch; Nicole Ziegler; Sylvia Thiele; Stephanie Moeller; Matthias Schnabelrauch; Vera Hintze; Dieter Scharnweber; Martina Rauner; Lorenz C. Hofbauer

In order to improve bone regeneration, development and evaluation of new adaptive biomaterials is warranted. Glycosaminoglycans (GAGs) such as hyaluronan (HA) and chondroitin sulfate (CS) are major extracellular matrix (ECM) components of bone, and display osteogenic properties that are potentially useful for biomaterial applications. Using native and synthetic sulfate‐modified GAGs, we manufactured artificial collagen/GAG ECM (aECMs) coatings, and evaluated how the presence of GAGs and their degree of sulfation affects the differentiation of murine mesenchymal stem cells to osteoblasts (OB) cultivated on these aECMs. GAG sulfation regulated osteogenesis at all key steps of OB development. Adhesion, but not migration, was diminished by 50% (P < 0.001). Proliferation and metabolic activity were slightly (P < 0.05) and cell death events strongly (P < 0.001) down‐regulated due to a switch from proliferative to matrix synthesis state. When exposed to sulfated GAGs, OB marker genes, such as alkaline phosphatase, osteoprotegerin (OPG), and osteocalcin increased by up to 28‐fold (P < 0.05) and calcium deposition up to 4‐fold (P < 0.05). Furthermore, GAG treatment of OBs suppressed their ability to support osteoclast (OC) differentiation and resorption. In conclusion, GAG sulfation controls bone cell homeostasis by concurrently promoting osteogenesis and suppressing their paracrine support of OC functions, thus displaying a favorable profile on bone remodeling. Whether these cellular properties translate into improved bone regeneration needs to be validated in vivo. J. Cell. Biochem. 115: 1101–1111, 2014.


International Archives of Allergy and Immunology | 2013

Advances in osteoimmunology: pathophysiologic concepts and treatment opportunities.

Martina Rauner; Wolfgang Sipos; Sylvia Thiele; Peter Pietschmann

Osteoimmunology is an emerging research area that deals with the mutual interactions between bone and the immune system. Osteoclasts have long been the center of attention in osteoimmunological research due to their hematopoietic origin and strong activation through cytokines. However, also the osteoclast’s opponent – the osteoblast – has recently sought the spotlight, and novel functions of its descendant – the osteocyte – have been unraveled. A considerable number of investigations carried out over the past decade have identified critical proteins with osteoimmune functions including the pro-osteoclastic cytokine receptor activator of NF-ĸB ligand and inhibitors of the pro-osteoblastic Wnt signaling pathway. These discoveries have also led to the development of targeted therapies to counteract not only inflammation-induced bone loss but also postmenopausal osteoporosis and osteoporosis associated with aging.


Journal of Bone and Mineral Research | 2016

Milk Fat Globule‐Epidermal Growth Factor 8 (MFG‐E8) is a Novel Anti‐inflammatory Factor in Rheumatoid Arthritis in Mice and Humans

Elise Albus; Kathrin Sinningen; Maria Winzer; Sylvia Thiele; Ulrike Baschant; Anke Hannemann; J. Fantana; Anne-Kathrin Tausche; Henri Wallaschofski; Matthias Nauck; Henry Völzke; Sylvia Grossklaus; Triantafyllos Chavakis; Mark C. Udey; Lorenz C. Hofbauer; Martina Rauner

Milk fat globule-epidermal growth factor 8 (MFG-E8) is an anti-inflammatory glycoprotein that mediates the clearance of apoptotic cells and is implicated in the pathogenesis of autoimmune and inflammatory diseases. Because MFG-E8 also controls bone metabolism, we investigated its role in rheumatoid arthritis (RA), focusing on inflammation and joint destruction. The regulation of MFG-E8 by inflammation was assessed in vitro using osteoblasts, in arthritic mice and in patients with RA. K/BxN serum transfer arthritis (STA) was applied to MFG-E8 knock-out mice to assess its role in the pathogenesis of arthritis. Stimulation of osteoblasts with lipopolysaccharide (LPS) and tumor necrosis factor (TNF)-α downregulated the expression of MFG-E8 by 30% to 35%. MFG-E8-deficient osteoblasts responded to LPS with a stronger production of pro-inflammatory cytokines. In vivo, MFG-E8 mRNA levels were 52% lower in the paws of collagen-induced arthritic (CIA) mice and 24% to 42% lower in the serum of arthritic mice using two different arthritis models (CIA and STA). Similarly, patients with RA (n = 93) had lower serum concentrations of MFG-E8 (-17%) compared with healthy controls (n = 140). In a subgroup of patients who had a moderate to high disease activity (n = 21), serum concentrations of MFG-E8 rose after complete or partial remission had been achieved (+67%). Finally, MFG-E8-deficient mice subjected to STA exhibited a stronger disease burden, an increased number of neutrophils in the joints, and a more extensive local and systemic bone loss. This was accompanied by an increased activation of osteoclasts and a suppression of osteoblast function in MFG-E8-deficient mice. Thus, MFG-E8 is a protective factor in the pathogenesis of RA and subsequent bone loss. Whether MFG-E8 qualifies as a novel biomarker or therapeutic target for the treatment of RA is worth addressing in further studies.


bonekey Reports | 2014

Instructions for producing a mouse model of glucocorticoid-induced osteoporosis

Sylvia Thiele; Ulrike Baschant; Alexander Rauch; Martina Rauner

Glucocorticoids are effective drugs used for the treatment of inflammatory diseases such as rheumatoid arthritis or asthma. Furthermore, they regulate various physiological processes, including bone remodeling. However, long-term high- and even low-dose glucocorticoid use is associated with a compromised bone quality and an increased fracture risk. At the cellular level, glucocorticoids suppress bone formation and stimulate bone resorption, which leads to loss of bone mass. To investigate the underlying mechanisms and new therapeutic strategies, the in vivo model for glucocorticoid-induced bone loss is widely used. This protocol outlines the common procedure that is currently used for the induction of bone loss in mice using glucocorticoids. It further provides useful hints and highlights possible pitfalls to take into account before starting an experiment.


Bone | 2015

Loss of milk fat globule-epidermal growth factor 8 (MFG-E8) in mice leads to low bone mass and accelerates ovariectomy-associated bone loss by increasing osteoclastogenesis.

Kathrin Sinningen; Elise Albus; Sylvia Thiele; Sylvia Grossklaus; Thomas Kurth; Mark C. Udey; Triantafyllos Chavakis; Lorenz C. Hofbauer; Martina Rauner

Milk fat globule-epidermal growth factor 8 (MFG-E8) is a glycoprotein that controls the engulfment of apoptotic cells and exerts inflammation-modulatory effects. Recently, it has been implicated in osteoclastogenesis and the pathogenesis of inflammatory periodontal bone loss, but its role in physiological bone homeostasis is still not well defined. Here, we evaluated the influence of MFG-E8 on osteoblasts and osteoclasts and its impact on bone remodeling in healthy and ovariectomized mice as a model for post-menopausal osteoporosis. Total and trabecular bone mineral densities at the lumbar spine in 6-week-old MFG-E8 KO mice were reduced by 11% (p < 0.05) and 17% (p < 0.01), respectively, as compared to wild-type (WT) mice. Accordingly, serum levels of the bone formation marker P1NP were decreased by 37% (p < 0.01) in MFG-E8 KO mice as were the ex vivo mineralization capacity and expression of osteoblast genes (Runx2, alkaline phosphatase, osteocalcin) in MFG-E8 KO osteoblasts. In contrast, serum bone resorption markers CTX1 and TRAP5b were increased by 30% and 60% (p < 0.05), respectively, in MFG-E8 KO mice. Furthermore, bone marrow macrophages from MFG-E8-KO mice differentiated more effectively into osteoclasts, as compared to WT cells. MFG-E8-deficient osteoclasts displayed increased bone resorption ex vivo, which could be reversed by the presence of recombinant MFG-E8. To determine the significance of the enhanced osteoclastogenesis in MFG-E8 KO mice, we performed an ovariectomy, which is associated with bone loss due to increased osteoclast activity. Indeed, MFG-E8 KO mice lost 12% more trabecular bone density than WT mice after ovariectomy. Together, these data indicate that MFG-E8 controls steady-state and pathological bone turnover and may therefore represent a new target gene in the treatment of bone diseases.


Journal of Bone and Mineral Research | 2018

Postnatal Skeletal Deletion of Dickkopf-1 Increases Bone Formation and Bone Volume in Male and Female Mice, Despite Increased Sclerostin Expression: POSTNATAL SKELETAL DKK1 DELETION INCREASES BONE FORMATION

Juliane Colditz; Sylvia Thiele; Ulrike Baschant; Christof Niehrs; Lynda F. Bonewald; Lorenz C. Hofbauer; Martina Rauner

The Wnt antagonist Dickkopf‐1 (Dkk1) is a negative regulator of osteoblast function and bone mass. However, because of the lack of appropriate models, many aspects of its role in the regulation of postnatal bone turnover and its cellular source have remained unknown. In this study, we deleted Dkk1 postnatally and in different cell types using various Cre‐drivers (Rosa26‐ERT2‐Cre, Osx‐cre, Dmp1‐Cre) and assessed to which extent cells of the osteoblastic lineage contribute to the effects of Dkk1 on bone turnover and homeostasis. Female and male mice were examined at 12 weeks of age. Mice with a global or cell type–specific deletion of Dkk1 showed a two‐ to threefold higher bone volume compared with their Cre‐negative littermates. The mineral apposition rate and the bone formation rate were increased two‐ to fourfold in all three mouse lines, despite a significant increase in systemic and skeletal levels of sclerostin. Dkk1 deletion further reduced the number of osteoclasts about twofold, which was accompanied by a strong decrease in the receptor activator of nuclear factor‐κB ligand/osteoprotegerin mRNA ratio in femoral bone. Despite similar increases in bone mass, the deletion of Dkk1 in osterix‐expressing cells reduced circulating Dkk1 significantly (males, –79%; females, –77%), whereas they were not changed in Dkk1fl/fl;Dmp1‐Cre mice. However, both lines showed significantly reduced Dkk1 mRNA levels in bone. In summary, we show that lack of Dkk1 in cells of the osteoblastic lineage leads to high bone mass with increased bone formation, despite increased levels of sclerostin. Moreover, the majority of systemic Dkk1 appears to originate from osteoprogenitors but not from mature osteoblasts or osteocytes. Nevertheless, the amount of Dkk1 produced locally by more mature osteogenic cells is sufficient to modulate bone mass. Thus, this study highlights the importance of local Wnt signaling on postnatal bone homeostasis.

Collaboration


Dive into the Sylvia Thiele's collaboration.

Top Co-Authors

Avatar

Martina Rauner

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Lorenz C. Hofbauer

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ulrike Baschant

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Kathrin Sinningen

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Claudia Goettsch

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Elena Tsourdi

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Juliane Salbach-Hirsch

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Sylvia Grossklaus

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Triantafyllos Chavakis

Dresden University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge