Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Bone Res ; 6: 4, 2018.
Article in English | MEDLINE | ID: mdl-29507818

ABSTRACT

Skeletal health relies on architectural integrity and sufficient bone mass, which are maintained through a tightly regulated equilibrium of bone resorption by osteoclasts and bone formation by osteoblasts. Genetic studies have linked the gene coding for low-density lipoprotein receptor-related protein1 (Lrp1) to bone traits but whether these associations are based on a causal molecular relationship is unknown. Here, we show that Lrp1 in osteoblasts is a novel regulator of osteoclast activity and bone mass. Mice lacking Lrp1 specifically in the osteoblast lineage displayed normal osteoblast function but severe osteoporosis due to highly increased osteoclast numbers and bone resorption. Osteoblast Lrp1 limited receptor activator of NF-κB ligand (RANKL) expression in vivo and in vitro through attenuation of platelet-derived growth factor (PDGF-BB) signaling. In co-culture, Lrp1-deficient osteoblasts stimulated osteoclastogenesis in a PDGFRß-dependent manner and in vivo treatment with the PDGFR tyrosine kinase inhibitor imatinib mesylate limited RANKL production and led to complete remission of the osteoporotic phenotype. These results identify osteoblast Lrp1 as a key regulator of osteoblast-to-osteoclast communication and bone mass through a PDGF-RANKL signaling axis in osteoblasts and open perspectives to further explore the potential of PDGF signaling inhibitors in counteracting bone loss as well as to evaluate the importance of functional LRP1 gene variants in the control of bone mass in humans.

2.
Injury ; 48(4): 841-848, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28237183

ABSTRACT

Hypothalamic-pituitary disconnection (HPD) leads to low bone turnover followed by bone loss and reduced biomechanical properties in sheep. To investigate the role of peripheral hormones in this centrally induced systemic bone loss model, we planned a hormone replacement experiment. Therefore, estrogen (OHE), thyroxin (OHT) or a combination of both (OHTE) was substituted in ovariectomized HPD sheep, as both hormones are decreased in HPD sheep and are known to have a significant but yet not fully understood impact on bone metabolism. Bone turnover and structural parameters were analyzed in comparison to different control groups - untreated sheep (C), ovariectomized (O) and ovariectomized+HPD sheep (OH). We performed histomorphometric and HR-pQCT analyses nine months after the HPD procedure, as well as biomechanical testing of all ewes studied. In HPD sheep (OH) the low bone turnover led to a significant bone loss. Treatment with thyroxin alone (OHT) mainly increased bone resorption, leading to a further reduction in bone volume. In contrast, the treatment with estrogen alone (OHE) and the combined treatment with estrogen and thyroxin (OHTE) prevented HPD-induced bone loss completely. In conclusion, peripheral hormone substitution was able to prevent HPD-induced low-turnover osteoporosis in sheep. But only the treatment with estrogen alone or in combination with thyroxin was able to completely preserve bone mass and structure. These findings demonstrate the importance of peripheral hormones for a balanced bone remodeling and a physiological bone turnover.


Subject(s)
Bone Resorption/pathology , Bone and Bones/pathology , Estrogens/pharmacology , Osteoporosis/pathology , Pituitary Gland/pathology , Thyroxine/pharmacology , Animals , Biomechanical Phenomena , Bone Density , Disease Models, Animal , Female , Hormone Replacement Therapy , Ovariectomy , Sheep
3.
Bone ; 64: 222-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24769333

ABSTRACT

Tumor-induced osteomalacia (TIO) is a paraneoplastic syndrome characterized by renal phosphate wasting, hypophosphatemia and low calcitriol levels as well as clinical symptoms like diffuse bone and muscle pain, fatigue fractures or increased fracture risk. Conventional imaging methods, however, often fail to detect the small tumors. Lately, tumor localization clearly improved by somatostatin-receptor (SSTR) imaging, such as octreotide scintigraphy or octreotide SPECT/CT. However, recent studies revealed that still a large number of tumors remained undetected by octreotide imaging. Hence, studies focused on different SSTR imaging methods such as 68Ga DOTA-NOC, 68Ga DOTA-TOC and 68Ga DOTA-TATE PET/CT with promising first results. Studies comparing different SSTR imaging methods for tumor localization in TIO are rare and thus little is known about diagnostic alternatives once a particular method failed to detect a tumor in patients with TIO. Here, we report the data of 5 consecutive patients suffering from TIO, who underwent both 111Indium-octreotide scintigraphy (111In-OCT) SPECT/CT as well as 68Ga DOTA-TATE PET/CT for tumor detection. While 111In-OCT SPECT/CT allowed tumor detection in only 1 of 5 patients, 68Ga DOTA-TATE PET/CT was able to localize the tumor in all patients. Afterwards, anatomical imaging of the region of interest was performed with CT and MRI. Thus, successful surgical resection of the tumor was achieved in all patients. Serum phosphate levels returned to normal and all patients reported relief of symptoms within weeks. Moreover, an iliac crest biopsy was obtained from every patient and revealed marked osteomalacia in all cases. Follow-up DXA revealed an increase in BMD of up to 34.5% 1-year postoperative, indicating remineralization. No recurrence was observed. In conclusion our data indicates that 68Ga DOTA-TATE PET/CT is an effective and promising diagnostic tool in the diagnosis of TIO, even in patients in whom 111In-OCT prior failed to detect a tumor.


Subject(s)
Gallium Radioisotopes , Neoplasms/diagnostic imaging , Osteomalacia/etiology , Radiopharmaceuticals , Adult , Cohort Studies , Female , Fibroblast Growth Factor-23 , Humans , Male , Middle Aged , Multimodal Imaging , Neoplasms/complications , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
4.
Brain ; 137(Pt 3): 683-92, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24459106

ABSTRACT

Many neurodegenerative disorders present with sensory loss. In the group of hereditary sensory and autonomic neuropathies loss of nociception is one of the disease hallmarks. To determine underlying factors of sensory neurodegeneration we performed whole-exome sequencing in affected individuals with the disorder. In a family with sensory neuropathy with loss of pain perception and destruction of the pedal skeleton we report a missense mutation in a highly conserved amino acid residue of atlastin GTPase 3 (ATL3), an endoplasmic reticulum-shaping GTPase. The same mutation (p.Tyr192Cys) was identified in a second family with similar clinical outcome by screening a large cohort of 115 patients with hereditary sensory and autonomic neuropathies. Both families show an autosomal dominant pattern of inheritance and the mutation segregates with complete penetrance. ATL3 is a paralogue of ATL1, a membrane curvature-generating molecule that is involved in spastic paraplegia and hereditary sensory neuropathy. ATL3 proteins are enriched in three-way junctions, branch points of the endoplasmic reticulum that connect membranous tubules to a continuous network. Mutant ATL3 p.Tyr192Cys fails to localize to branch points, but instead disrupts the structure of the tubular endoplasmic reticulum, suggesting that the mutation exerts a dominant-negative effect. Identification of ATL3 as novel disease-associated gene exemplifies that long-term sensory neuronal maintenance critically depends on the structural organisation of the endoplasmic reticulum. It emphasizes that alterations in membrane shaping-proteins are one of the major emerging pathways in axonal degeneration and suggests that this group of molecules should be considered in neuroprotective strategies.


Subject(s)
Bone Diseases/genetics , Endoplasmic Reticulum/genetics , GTP Phosphohydrolases/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Adult , Age of Onset , Bone Diseases/etiology , Bone Diseases/physiopathology , Cohort Studies , Cough/genetics , Cough/pathology , Cough/physiopathology , Endoplasmic Reticulum/pathology , Exome/genetics , Female , Fractures, Bone/genetics , Fractures, Bone/pathology , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/pathology , Gastroesophageal Reflux/physiopathology , Genes, Dominant/genetics , Haplotypes/genetics , Hereditary Sensory and Autonomic Neuropathies/complications , Hereditary Sensory and Autonomic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Intracellular Space/genetics , Male , Mutation , Mutation, Missense/genetics , Pedigree , Phenotype , Young Adult
5.
Bone ; 62: 90-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24440515

ABSTRACT

A number of unexpected molecules were recently identified as products of osteoblasts, linking bone homeostasis to systemic energy metabolism. Here we identify the lipolytic enzyme hepatic lipase (HL, encoded by Lipc) as a novel cell-autonomous regulator of osteoblast function. In an unbiased genome-wide expression analysis, we find Lipc to be highly induced upon osteoblast differentiation, verified by quantitative Taqman analyses of primary osteoblasts in vitro and of bone samples in vivo. Functionally, loss of HL in vitro leads to increased expression and secretion of osteoprotegerin (OPG), while expression of some osteoblast differentiation makers is impaired. When challenging energy metabolism in a diet-induced obesity (DIO) study, lack of HL leads to a significant increase in bone formation markers and a decrease in bone resorption markers. Accordingly, in the DIO setting, we observe in Lipc(-/-) animals but not in wild-type controls a significant increase in lumbar vertebral trabecular bone mass and formation rate as well as in femoral trabecular bone mass and cortical thickness. Taken together, we demonstrate that HL expressed by osteoblasts has an impact on osteoblast OPG expression and that lack of HL leads to increased bone mass in DIO. These data provide a novel and completely unexpected molecular link in the complex interplay of osteoblasts and systemic energy metabolism.


Subject(s)
Bone Remodeling , Lipase/metabolism , Obesity/enzymology , Obesity/pathology , Osteoblasts/enzymology , Osteoblasts/pathology , Animals , Biomarkers/blood , Biomarkers/urine , Cell Differentiation , Cells, Cultured , Diet, High-Fat , Feeding Behavior , Femur/diagnostic imaging , Femur/pathology , Lipase/deficiency , Lumbar Vertebrae/pathology , Male , Mice, Inbred C57BL , Organ Size , Osteoprotegerin/metabolism , X-Ray Microtomography
6.
J Bone Miner Res ; 29(4): 982-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24108692

ABSTRACT

Osteopetrosis is an inherited disorder of impaired bone resorption, with the most commonly affected genes being CLCN7 and TCIRG1, encoding the Cl(-) /H(+) exchanger CLC-7 and the a3 subunit of the vacuolar H(+) -ATPase, respectively. We and others have previously shown that the disease is frequently accompanied by osteomalacia, and that this additional pathology is also found in Tcirg1-deficient oc/oc mice. The remaining question was whether osteoid enrichment is specifically associated with TCIRG1 inactivation, or whether CLCN7 mutations would also cause skeletal mineralization defects. Here we describe a complete osteologic assessment of one family carrying a novel mutation in CLCN7 (D145G), which impairs the activation and relaxation kinetics of the CLC-7 ion transporter. The two siblings carrying the mutation in the homozygous state displayed high bone mass, increased serum levels of bone formation markers, but no impairment of calcium homeostasis when compared to the other family members. Most importantly, however, undecalcified processing of an iliac crest biopsy from one of the affected children clearly demonstrated a pathological increase of trabecular bone mass, but no signs of osteomalacia. Given the potential relevance of these findings we additionally performed undecalcified histology of iliac crest biopsies from seven additional cases with osteopetrosis caused by a mutation in TNFRSF11A (n=1), CLCN7 (n=3), or TCIRG1 (n=3). Here we observed that all cases with TCIRG1-dependent osteopetrosis displayed severe osteoid accumulation and decreased calcium content within the mineralized matrix. In contrast, there was no detectable bone mineralization defect in the cases with TNFRSF11A-dependent or CLCN7-dependent osteopetrosis. Taken together, our analysis demonstrates that CLCN7 and TCIRG1 mutations differentially affect bone matrix mineralization, and that there is a need to modify the current classification of osteopetrosis.


Subject(s)
Calcification, Physiologic , Chloride Channels/genetics , Mutation , Osteopetrosis/genetics , Vacuolar Proton-Translocating ATPases/genetics , Calcium/metabolism , Child , Child, Preschool , Female , Genes, Recessive , Homeostasis , Humans , Infant , Male , Pedigree
7.
Clin Oral Implants Res ; 25(11): 1239-1244, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24102843

ABSTRACT

PURPOSE: It is supposed that the demographic change will lead to an increase in patients with impaired alveolar bone conditions. Large animal models are of particular interest in this context as they are suitable for developing and testing new dental implants. Recently, we demonstrated that surgical hypothalamo-pituitary disconnection (HPD) causes a pronounced low-turnover situation leading to cortical and trabecular bone loss in sheep. In this study, we aimed to investigate the influence of the HPD procedure on the alveolar bone. METHODS: Ten adult Merino ewes were randomly assigned to two groups: Control and HPD. After 6 months, we analysed the cortical and trabecular bone of all mandibles by histomorphometry and high-resolution peripheral quantitative computed tomography (HR-pQCT). RESULTS: HPD ewes showed a significant decrease in cortical thickness by ~20%, a significant increase in cortical porosity by ~20% and a significant decrease in bone volume by ~30% in comparison with Control ewes. CONCLUSION: Our results underline the importance of central regulatory mechanisms of bone turnover. However, further studies are needed to understand these central regulatory elements of bone turnover in detail and to judge the value of the HPD sheep for dental research.


Subject(s)
Alveolar Bone Loss/etiology , Hypothalamic Diseases/complications , Hypothalamo-Hypophyseal System/physiopathology , Mandibular Diseases/etiology , Alveolar Bone Loss/diagnostic imaging , Alveolar Bone Loss/pathology , Animals , Bone Density/physiology , Bone Remodeling/physiology , Disease Models, Animal , Female , Mandibular Diseases/diagnostic imaging , Mandibular Diseases/pathology , Ovariectomy/methods , Random Allocation , Sheep , Time Factors , Tomography, X-Ray Computed/methods , X-Ray Microtomography/methods
8.
J Bone Miner Res ; 28(10): 2070-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23553711

ABSTRACT

Chemokines play crucial roles in the recruitment of specific hematopoietic cell types, and some of them have been suggested to be involved in the regulation of bone remodeling. Because we have previously observed that chemokine (C-C motif) ligand 2 (Ccl2) and Ccl5 are direct target genes of noncanonical Wnt signaling in osteoblasts, we analyzed the skeletal phenotypes of Ccl2-deficient and Ccl5-deficient mice. In line with previous studies, Ccl2-deficient mice display a moderate reduction of osteoclastogenesis at the age of 6 months. In contrast, 6-month-old Ccl5-deficient mice display osteopenia associated with decreased bone formation and increased osteoclastogenesis. Moreover, unlike in wild-type and Ccl2-deficient mice, large areas of their trabecular and endocortical bone surfaces are not covered by osteoblasts or bone-lining cells, and this is associated with a severe reduction of endosteal bone formation. Although this phenotype diminishes with age, it is important that we could further identify a reduced number of osteal macrophages in 6-month-old Ccl5-deficient mice, because this cell type has previously been reported to promote endosteal bone formation. Because Ccl5-deficient mice also display increased osteoclastogenesis, we finally addressed the question of whether osteal macrophages could differentiate into osteoclasts and/or secrete inhibitors of osteoclastogenesis. For that purpose we isolated these cells by CD11b affinity purification from calvarial cultures and characterized them ex vivo. Here we found that they are unable to differentiate into osteoblasts or osteoclasts, but that their conditioned medium mediates an antiosteoclastogenic effect, possibly caused by interleukin-18 (IL-18), an inhibitor of osteoclastogenesis expressed by osteal macrophages. Taken together, our data provide in vivo evidence supporting the previously suggested role of Ccl5 in bone remodeling. Moreover, to the best of our knowledge, Ccl5-deficient mice represent the first model with a spontaneous partial deficiency of osteal macrophages, a recently identified cell type, whose impact on bone remodeling is just beginning to be understood.


Subject(s)
Chemokine CCL5/deficiency , Osteoclasts/metabolism , Osteogenesis , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Remodeling , Cell Differentiation , Cell Separation , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Culture Media, Conditioned/pharmacology , Hematopoiesis , Interleukin-18/pharmacology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Spine/metabolism , Spine/pathology , Stem Cells/metabolism , Tibia/metabolism , Tibia/pathology
9.
J Bone Miner Res ; 28(2): 236-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22991192

ABSTRACT

The primary role of apolipoprotein E (apoE) is to mediate the cellular uptake of lipoproteins. However, a new role for apoE as a regulator of bone metabolism in mice has recently been established. In contrast to mice, the human APOE gene is characterized by three common isoforms APOE ε2, ε3, and ε4 that result in different metabolic properties of the apoE isoforms, but it remains controversial whether the APOE polymorphism influences bone traits in humans. To clarify this, we investigated bone phenotypes of apoE knock-in (k.i.) mice, which express one human isoform each (apoE2 k.i., apoE3 k.i., apoE4 k.i.) in place of the mouse apoE. Analysis of 12-week-old female k.i. mice revealed increased levels of biochemical bone formation and resorption markers in apoE2 k.i. animals as compared to apoE3 k.i. and apoE4 k.i., with a reduced osteoprotegerin (OPG)/receptor activator of NF-κB ligand (RANKL) ratio in apoE2 k.i., indicating increased turnover with prevailing resorption in apoE2 k.i. Accordingly, histomorphometric and micro-computed tomography (µCT) analyses demonstrated significantly lower trabecular bone mass in apoE2 than in apoE3 and apoE4 k.i. animals, which was reflected by a significant reduction of lumbar vertebrae maximum force resistance. Unlike trabecular bone, femoral cortical thickness, and stability was not differentially affected by the apoE isoforms. To extend these observations to the human situation, plasma from middle-aged healthy men homozygous for ε2/ε2, ε3/ε3, and ε4/ε4 (n = 21, n = 80, n = 55, respectively) was analyzed with regard to bone turnover markers. In analogy to apoE2 k.i. mice, a lower OPG/RANKL ratio was observed in the serum of ε2/ε2 carriers as compared to ε3/ε3 and ε4/ε4 individuals (p = 0.02 for ε2/ε2 versus ε4/ε4). In conclusion, the current data strongly underline the general importance of apoE as a regulator of bone metabolism and identifies the APOE ε2 allele as a potential genetic risk factor for low trabecular bone mass and vertebral fractures in humans.


Subject(s)
Apolipoproteins E/metabolism , Bone Remodeling/physiology , Bone and Bones/anatomy & histology , Bone and Bones/physiology , Animals , Apolipoprotein E2/blood , Apolipoprotein E2/genetics , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoproteins E/genetics , Biomarkers/metabolism , Biomechanical Phenomena , Bone Density/physiology , Female , Femur/physiology , Gene Knock-In Techniques , Homozygote , Humans , Lumbar Vertebrae/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Organ Size , Osteogenesis , Osteoprotegerin/blood , Osteoprotegerin/metabolism , Protein Isoforms , RANK Ligand/blood , RANK Ligand/metabolism
10.
J Cell Biol ; 192(6): 1057-72, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21402791

ABSTRACT

Although Wnt signaling in osteoblasts is of critical importance for the regulation of bone remodeling, it is not yet known which specific Wnt receptors of the Frizzled family are functionally relevant in this process. In this paper, we show that Fzd9 is induced upon osteoblast differentiation and that Fzd9(-/-) mice display low bone mass caused by impaired bone formation. Our analysis of Fzd9(-/-) primary osteoblasts demonstrated defects in matrix mineralization in spite of normal expression of established differentiation markers. In contrast, we observed a reduced expression of chemokines and interferon-regulated genes in Fzd9(-/-) osteoblasts. We also identified the ubiquitin-like modifier Isg15 as one potential downstream mediator of Fzd9 in these cells. Importantly, our molecular analysis further revealed that canonical Wnt signaling is not impaired in the absence of Fzd9, thus explaining the absence of a bone resorption phenotype. Collectively, our results reveal a previously unknown function of Fzd9 in osteoblasts, a finding that may have therapeutic implications for bone loss disorders.


Subject(s)
Receptors, Neurotransmitter/metabolism , Wnt Proteins/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Chemokines/metabolism , Frizzled Receptors , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Receptors, Neurotransmitter/genetics , Signal Transduction/physiology , Tissue Distribution , Wnt Proteins/genetics
11.
Osteoporos Int ; 22(10): 2667-75, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21267545

ABSTRACT

UNLABELLED: Histomorphometry and quantitative backscattered electron microscopy of iliac crest biopsies from patients with adult hypophosphatasia not only confirmed the expected enrichment of non-mineralized osteoid, but also demonstrated an altered trabecular microarchitecture, an increased number of osteoblasts, and an impaired calcium distribution within the mineralized bone matrix. INTRODUCTION: Adult hypophosphatasia is an inherited disorder of bone metabolism caused by inactivating mutations of the ALPL gene, encoding tissue non-specific alkaline phosphatase. While it is commonly accepted that the increased fracture risk of the patients is the consequence of osteomalacia, there are only few studies describing a complete histomorphometric analysis of bone biopsies from affected individuals. Therefore, we analyzed iliac crest biopsies from eight patients and set them in direct comparison to biopsies from healthy donors or from individuals with other types of osteomalacia. METHODS: Histomorphometric analysis was performed on non-decalcified sections stained either after von Kossa/van Gieson or with toluidine blue. Bone mineral density distribution was quantified by backscattered electron microscopy. RESULTS: Besides the well-documented enrichment of non-mineralized bone matrix in individuals suffering from adult hypophosphatasia, our histomorphometric analysis revealed alterations of the trabecular microarchitecture and an increased number of osteoblasts compared to healthy controls or to individuals with other types of osteomalacia. Moreover, the analysis of the mineralized bone matrix revealed significantly decreased calcium content in patients with adult hypophosphatasia. CONCLUSIONS: Taken together, our data show that adult hypophosphatasia does not solely result in an enrichment of osteoid, but also in a considerable degradation of bone quality, which might contribute to the increased fracture risk of the affected individuals.


Subject(s)
Bone Matrix/pathology , Calcification, Physiologic , Hypophosphatasia/pathology , Ilium/pathology , Osteomalacia/pathology , Adult , Aged , Bone Density , Case-Control Studies , Humans , Male , Microscopy, Electron , Middle Aged , Osteoblasts/metabolism , Young Adult
12.
Bone ; 48(2): 339-46, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20817130

ABSTRACT

Liver X receptors (LXRs) are nuclear receptors that play a crucial role in the transcriptional control of lipid metabolism. Pharmacological LXR activation is an attractive concept for the treatment of atherosclerosis. Genetic LXR deficiency in mice has been shown to have an effect on bone turnover and structure and LXR activation is known to influence the osteogenic differentiation of bone marrow stromal cells. Therefore, therapeutic pharmacological LXR activation may have relevant effects on bone. Here, using two synthetic LXR ligands, T0901317 and GW3965, we investigated the effect of LXR activation on murine osteoblasts and the influence of long-term LXR activation on bone in vivo in mice. Short term (48 h) in vitro treatment of primary murine osteoblasts with T0901317 resulted in a dose-dependent decrease of osteocalcin and alkaline phosphatase mRNA and protein. In vivo, a 6-day treatment of C57BL/6J mice with T0901317 led to a 40% reduction of serum osteocalcin concentrations. Long-term (12-week) oral administration of T0901317 or GW3965 influenced the expression of established LXR target genes in liver and intestine, but did not alter trabecular and cortical bone structure or bone turnover as determined by total skeleton radiography, histomorphometric analysis of lumbar vertebral trabecular bone, micro CT analysis of femur cortical bone and biochemical determination of bone formation and resorption markers. We conclude that short-term pharmacological LXR activation has the potential to profoundly influence osteoblast function, but that long-term LXR activation in vivo has no adverse effects on the murine skeleton.


Subject(s)
Orphan Nuclear Receptors/metabolism , Osteoblasts/metabolism , Animals , Biomechanical Phenomena , Cells, Cultured , Creatinine/urine , Enzyme-Linked Immunosorbent Assay , Intestinal Mucosa/metabolism , Liver/metabolism , Liver X Receptors , Mice , Mice, Inbred C57BL , Osteocalcin/blood , Polymerase Chain Reaction
13.
J Bone Miner Res ; 26(4): 704-17, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20939024

ABSTRACT

Since the hematopoetic system is located within the bone marrow, it is not surprising that recent evidence has demonstrated the existence of molecular interactions between bone and immune cells. While interleukin 1 (IL-1) and IL-18, two cytokines of the IL-1 family, have been shown to regulate differentiation and activity of bone cells, the role of IL-33, another IL-1 family member, has not been addressed yet. Since we observed that the expression of IL-33 increases during osteoblast differentiation, we analyzed its possible influence on bone formation and observed that IL-33 did not affect matrix mineralization but enhanced the expression of Tnfsf11, the gene encoding RANKL. This finding led us to analyze the skeletal phenotype of Il1rl1-deficient mice, which lack the IL-33 receptor ST2. Unexpectedly, these mice displayed normal bone formation but increased bone resorption, thereby resulting in low trabecular bone mass. Since this finding suggested a negative influence of IL-33 on osteoclastogenesis, we next analyzed osteoclast differentiation from bone marrow precursor cells and observed that IL-33 completely abolished the generation of TRACP(+) multinucleated osteoclasts, even in the presence of RANKL and macrophage colony-stimulating factor (M-CSF). Although our molecular studies revealed that IL-33 treatment of bone marrow cells caused a shift toward other hematopoetic lineages, we further observed a direct negative influence of IL-33 on the osteoclastogenic differentiation of RAW264.7 macrophages, where IL-33 repressed the expression of Nfatc1, which encodes one of the key transciption factors of osteoclast differentiation. Taken together, these findings have uncovered a previously unknown function of IL-33 as an inhibitor of bone resorption.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Interleukins/physiology , Osteoblasts/metabolism , Osteoclasts/cytology , Acid Phosphatase/metabolism , Animals , Basophils/cytology , Basophils/metabolism , Bone Marrow Cells/drug effects , Bone Resorption/metabolism , Bone and Bones/pathology , Calcitriol/pharmacology , Cell Count , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , Eosinophils/cytology , Eosinophils/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/pharmacology , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , RANK Ligand/genetics , RANK Ligand/pharmacology , Receptors, Interleukin/genetics , Stromal Cells/cytology , Stromal Cells/physiology , Tartrate-Resistant Acid Phosphatase
14.
J Trauma ; 69(6): 1473-80, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21150526

ABSTRACT

BACKGROUND: Long-term administration of glucocorticoids may lead to bone loss and osteoporosis as reported in previous experimental and clinical studies. On the other hand, several in vitro studies have demonstrated that dexamethasone treatment induces proliferation and differentiation of human and murine osteoblast precursors. Thereby, a positive interaction of dexamethasone with the osteoinductive bone morphogenetic proteins (BMPs) is reported in vitro, but in vivo studies are still missing. Thus, the aim of this study was to determine whether short-term application of dexamethasone may improve BMP-7-induced bone formation in vivo. METHODS: Ectopic bone formation was induced in control and dexamethasone-treated mice by application of BMP-7 into the hamstring muscles. After 20 days of treatment, each ectopic bone nodule was analyzed by contact radiography, microcomputed tomography, and histomorphometry. Furthermore, mice were subjected to histomorphometric analyses of their lumbar vertebrae and proximal tibiae to assess the systemic effect of short-term dexamethasone treatment on bone metabolism. RESULTS: Dexamethasone application significantly increased the bone volume and osteoblast number of the ectopic bone nodules compared with untreated controls. Histomorphometric analyses of the lumbar vertebrae and proximal tibiae revealed no significant differences between the control and dexamethasone-treated mice. CONCLUSIONS: This study demonstrates that BMP-7-induced ectopic bone formation is significantly enhanced by systemic short-term application of dexamethasone. These in vivo data confirm the results of previous in vitro studies and could be of interest for further studies with the intention to improve BMP-induced bone formation by short-term application of dexamethasone.


Subject(s)
Bone Morphogenetic Protein 7/physiology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Osteoblasts/drug effects , Osteogenesis/drug effects , Animals , Female , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Thigh/diagnostic imaging , X-Ray Microtomography
15.
Bone ; 47(4): 736-45, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20633710

ABSTRACT

The long prevailing view that obesity is generally associated with beneficial effects on the skeleton has recently been challenged. Apolipoprotein E (apoE) is known to influence both adipose tissue and bone. The goal of the current study was to examine the impact of apoE on the development of fat mass and bone mass in mice under conditions of diet-induced obesity (DIO). Four week-old male C57BL/6 (WT) and apoE-deficient (apoE(-/-)) mice received a control or a diabetogenic high-fat diet (HFD) for 16 weeks. The control-fed apoE(-/-) animals displayed less total fat mass and higher lumbar trabecular bone volume (BV/TV) than WT controls. When stressed with HFD to induce obesity, apoE(-/-) mice had a lower body weight, lower serum glucose, insulin and leptin levels and accumulated less white adipose tissue mass at all sites including bone marrow. While WT animals showed no significant change in BV/TV and bone formation rate (BFR), apoE deficiency led to a decrease of BV/TV and BFR when stressed with HFD. Bone resorption parameters were not affected by HFD in either genotype. Taken together, under normal dietary conditions, apoE-deficient mice acquire less fat mass and more bone mass than WT littermates. When stressed with HFD to develop DIO, the difference of total body fat mass becomes larger and the difference of bone mass smaller between the genotypes. We conclude that apoE is involved in an inverse regulation of bone mass and fat mass in growing mice and that this effect is modulated by diet-induced obesity.


Subject(s)
Adipose Tissue/pathology , Apolipoproteins E/metabolism , Diet , Obesity/metabolism , Obesity/pathology , Spine/pathology , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Apolipoproteins E/deficiency , Blood Glucose/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Remodeling/drug effects , Cell Proliferation/drug effects , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Epididymis/drug effects , Epididymis/metabolism , Epididymis/pathology , Insulin/metabolism , Leptin/blood , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/physiopathology , Organ Size/drug effects , Spine/metabolism , Spine/physiopathology , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/metabolism , Subcutaneous Tissue/pathology , Viscera/drug effects , Viscera/metabolism , Viscera/pathology , Weight Gain/drug effects
16.
J Bone Miner Res ; 25(8): 1724-35, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20200993

ABSTRACT

Midkine (Mdk) and pleiotrophin (Ptn) comprise a family of heparin-binding growth factors known primarily for their effects on neuronal cells. Since transgenic mice overexpressing Ptn have been reported to display increased bone density, we have previously analyzed Ptn-deficient mice but failed to detect any abnormality of skeletal development and remodeling. Together with the finding that Mdk expression increases in the course of primary osteoblast differentiation, we reasoned that Mdk, rather than Ptn, could play a physiologic role in bone formation. Here, we show that Mdk-deficient mice display an increased trabecular bone volume at 12 and 18 months of age, accompanied by cortical porosity. Histomorphometric quantification demonstrated an increased bone-formation rate compared with wild-type littermates, whereas bone resorption was differentially affected in trabecular and cortical bone of Mdk-deficient mice. To understand the effect of Mdk on bone formation at the molecular level, we performed a genome-wide expression analysis of primary osteoblasts and identified Ank and Enpp1 as Mdk-induced genes whose decreased expression in Mdk-deficient osteoblasts may explain, at least in part, the observed skeletal phenotype. Finally, we performed ovariectomy and observed bone loss only in wild-type but not in Mdk-deficient animals. Taken together, our data demonstrate that Mdk deficiency, at least in mice, results in an increased trabecular bone formation, thereby raising the possibility that Mdk-specific antagonists might prove beneficial in osteoporosis therapy.


Subject(s)
Cytokines/deficiency , Osteogenesis/physiology , Aging/metabolism , Analysis of Variance , Animals , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Cytokines/metabolism , Female , Gene Expression Regulation , Genotype , Mice , Midkine , Organ Size , Osteoblasts/metabolism , Osteoblasts/pathology , Ovariectomy , Phenotype
17.
J Orthop Res ; 28(6): 785-91, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20063306

ABSTRACT

Nonsteroidal antiinflammatory drugs (NSAIDs) are known to potentially impair the fracture healing process. The aim of the present study was to determine if the impairment of bone healing by systemic NSAID application is, at least in part, due to an interaction of NSAIDs with the bone anabolic BMP-7 pathway. Therefore, we first analyzed fracture healing in control and diclofenac-treated mice, where we not only found a significant impairment of fracture healing due to diclofenac treatment as assessed by biomechanical testing and microCT imaging, but also found high coexpression of bone morphogenetic protein-7 (BMP-7) and cyclooxygenase-2 (COX-2) within the fracture callus of both groups. To experimentally address the possible interaction between BMP-7 and COX-2, we then induced ectopic bone formation in control (n = 10) and diclofenac-treated mice (n = 10) by application of BMP-7 (recombinant human OP-1, rhOP-1) into the hamstring muscles. After 20 days of treatment, each ectopic bone nodule was analyzed by contact-radiography, microCT, histology, and histomorphometry. Diclofenac application decreased the trabecular number and bone mass in the ectopic bone nodules significantly due to reduced osteoblast number and activity. These data demonstrate that the bone anabolic effect of BMP-7 and fracture healing is impaired by diclofenac application, and suggest that the potential negative impact of NSAIDs on fracture healing is, at least in part, due to interference with BMP-7 signaling.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Bone Morphogenetic Protein 7/pharmacology , Diclofenac/toxicity , Fracture Healing/drug effects , Osteogenesis/drug effects , Animals , Cyclooxygenase 2/physiology , Female , Humans , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology
18.
Nat Med ; 15(6): 674-81, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19448635

ABSTRACT

Activation of osteoclasts and their acidification-dependent resorption of bone is thought to maintain proper serum calcium levels. Here we show that osteoclast dysfunction alone does not generally affect calcium homeostasis. Indeed, mice deficient in Src, encoding a tyrosine kinase critical for osteoclast activity, show signs of osteopetrosis, but without hypocalcemia or defects in bone mineralization. Mice deficient in Cckbr, encoding a gastrin receptor that affects acid secretion by parietal cells, have the expected defects in gastric acidification but also secondary hyperparathyroidism and osteoporosis and modest hypocalcemia. These results suggest that alterations in calcium homeostasis can be driven by defects in gastric acidification, especially given that calcium gluconate supplementation fully rescues the phenotype of the Cckbr-mutant mice. Finally, mice deficient in Tcirg1, encoding a subunit of the vacuolar proton pump specifically expressed in both osteoclasts and parietal cells, show hypocalcemia and osteopetrorickets. Although neither Src- nor Cckbr-deficient mice have this latter phenotype, the combined deficiency of both genes results in osteopetrorickets. Thus, we find that osteopetrosis and osteopetrorickets are distinct phenotypes, depending on the site or sites of defective acidification.


Subject(s)
Acids , Bone Density/physiology , Calcium/metabolism , Gastric Mucosa/metabolism , Homeostasis , Amino Acid Sequence , Animals , Base Sequence , Bone Diseases, Developmental/etiology , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/metabolism , Bone Diseases, Developmental/pathology , Calcium/pharmacology , Homeostasis/drug effects , Hydrogen-Ion Concentration , Hypocalcemia/complications , Hypocalcemia/genetics , Hypocalcemia/metabolism , Mice , Mice, Transgenic , Phenotype , Vacuolar Proton-Translocating ATPases/metabolism
19.
J Bone Miner Res ; 24(1): 62-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18767930

ABSTRACT

Although Paget's disease of bone (PDB) is the second most common metabolic bone disease, to our knowledge, there is only one quantitative analysis on the histological and especially on the histomorphometric level. Therefore, the aim of this study was to analyze, on the basis of the Hamburg Bone Register, PBD in terms of incidence, skeletal distribution, malignant transformation, and histological and histomorphometric characteristics. Bone biopsies and patient files of 754 cases with histologically proven PDB were reviewed in a retrospective study. Quantitative static histomorphometry was performed on a representative subgroup of 247 biopsies derived from patients with manifestation of PDB at the iliac crest and compared with an age- and sex-matched control group. The peak incidence of PDB was between 70 and 80 yr of age. The majority of monostotic skeletal manifestation was localized at the os ilium, followed by the spine and femur. Histomorphometric results showed a high bone turnover with a significant increase in bone resorption and bone formation indices leading to an increased bone volume. Paget sarcoma was diagnosed in 6 of 754 patients, indicating a malignant transformation in 0.8% of the affected patients. Taken together, our study characterizes PDB in Germany on the basis of one of the largest cohorts of patients with histologically proven PDB. Moreover, for the first time, a quantitative histomorphometric approach was taken for >200 cases, where we could show local high bone mass lesions as a result of an increase of both osteoclast and osteoblast indices.


Subject(s)
Osteitis Deformans/diagnosis , Osteitis Deformans/pathology , Sarcoma/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Bone and Bones/pathology , Child , Cohort Studies , Female , Humans , Male , Middle Aged , Retrospective Studies , Sarcoma/pathology
20.
Eur J Trauma Emerg Surg ; 34(6): 527-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-26816275

ABSTRACT

Osteoporosis is a disease that leads to fragility fractures due to the loss of bone mass and bone microstructure. This review presents an update on the fundamental pathophysiological and pathomorphological mechanisms of bone loss. Pathomorphological characteristics such as perforations and microcallus formations are explained. The physiological relevance of the remodeling process and its control by local paracrine, systemic endocrine, and central neural signaling pathways are discussed. Hormones, such as estrogen, follicle stimulating hormone, and leptin, transcription factors, such as Runx2 and osterix, and the wnt signaling pathway are discussed in terms of their roles in bone cell differentiation and function. On the basis of current knowledge, osteoporosis can be diagnosed and treated and fractures can be prevented. However, it is likely that new and even more effective diagnostic and therapeutic strategies will emerge as our understanding of the remodeling process that controls osteoblast and osteoclast function increases.

SELECTION OF CITATIONS
SEARCH DETAIL
...