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










Publication year range
1.
Photodermatol Photoimmunol Photomed ; 34(5): 288-297, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29855075

ABSTRACT

BACKGROUND: Human and animal studies have shown that exposure to ultraviolet light can incite a chain of endocrine, immunologic, and neurohumoral reactions that might affect mood. This review focuses on the evidence from clinical trials and observational studies on the effect of ultraviolet light on mood, depressive disorders, and well-being. METHODS: A search was made in PubMed, Embase, Web of Science, Cochrane, Psychinfo, CINAHL, Academic Search Premier and Science Direct, and the references of key papers, for clinical trials and observational studies describing the effect of ultraviolet light applied to skin or eyes on mood, depressive disorders, and well-being. RESULTS: Of the seven studies eligible for this review, the effect of ultraviolet light on mood, depressive symptoms and seasonal affective disorders was positive in six of them. CONCLUSIONS: Of the seven studies, six demonstrated benefit of exposure to ultraviolet radiation and improvement in mood which supports a positive effect of ultraviolet light on mood. Because of the small number of the studies and their heterogeneity, more research is warranted to confirm and document this correlation.


Subject(s)
Affect , Depressive Disorder , Ultraviolet Rays , Animals , Clinical Trials as Topic , Humans
2.
Bone ; 50(5): 1107-14, 2012 May.
Article in English | MEDLINE | ID: mdl-22353552

ABSTRACT

There is little information on the distribution of osteocytes within the individual cortical osteon, but using direct 3-D imaging in a single subject, Hannah et al. found a gradient with a two-fold higher density of cells adjacent to the cement line compared to near the canal. Since a limiting factor for bone formation might be the availability of osteoblasts due to their recruitment as osteocytes, we studied distributions of osteonal osteocytes in frozen sections of the femoral neck cortex. Osteocytes were stained with an anti-sclerostin antibody and counter-stained with toluidine blue. Adjacent sections were stained for alkaline phosphatase (ALP). Each osteonal osteocyte was categorised as being sclerostin-positive (scl+) or negative (scl-). ImageJ was used to measure the perimeter and area of each osteon and canal, while special purpose routines were used to measure the minimum distances of each osteocyte from the cement line and the canal. Canal area was strongly correlated with osteon area. Osteocytes were most dense close to the cement line; and their areal density within the matrix declined up to three-fold between the cement line and the canal, depending on osteon diameter. Large and small osteons had similar densities of osteocytes close to the cement line, but fractured neck of femur cases had significantly lower densities of osteocytes close to the canal. Higher osteocyte density close to the canal was associated with ALP expression. It is concluded that entombment of osteocytes newly drawn from the osteoblast pool into the mineralising matrix is independent of preceding bone resorption depth. As osteonal infilling proceeds, osteocyte formation declines more rapidly than matrix formation, leading to a progressive reduction in osteocyte density. A shrinking supply of precursor osteoblasts due to previous osteocyte recruitment, apoptosis, or both could produce this effect. In a statistically significant contrast, sclerostin negative osteocytes adjacent to the canal had the expected effect of reducing canal size in controls but this was not seen in hip fracture. This demonstrated the failure of osteonal osteoblasts to sustain bone formation through a complete remodelling cycle in osteoporosis, perhaps due to insufficient osteoblasts remaining capable of mineralized matrix formation. The failure of osteocytic sclerostin suppression to associate with bone formation in these osteons might alternatively be explained by downstream interference with sclerostin's effect on wnt signalling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Femur Neck/pathology , Haversian System/pathology , Hip Fractures/pathology , Osteocytes/pathology , Adaptor Proteins, Signal Transducing , Aged , Cell Count , Cell Death , Female , Genetic Markers , Haversian System/metabolism , Hip Fractures/metabolism , Humans , Male , Microscopy, Polarization , Models, Biological , Organ Size , Osteocytes/metabolism
3.
J Bone Miner Res ; 26(12): 2804-11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21786318

ABSTRACT

Sclerosteosis is a rare bone sclerosing dysplasia, caused by loss-of-function mutations in the SOST gene, encoding sclerostin, a negative regulator of bone formation. The purpose of this study was to determine how the lack of sclerostin affects bone turnover in patients with sclerosteosis and to assess whether sclerostin synthesis is decreased in carriers of the SOST mutation and, if so, to what extent this would affect their phenotype and bone formation. We measured sclerostin, procollagen type 1 amino-terminal propeptide (P1NP), and cross-linked C-telopeptide (CTX) in serum of 19 patients with sclerosteosis, 26 heterozygous carriers of the C69T SOST mutation, and 77 healthy controls. Chips of compact bone discarded during routine surgery were also examined from 6 patients and 4 controls. Sclerostin was undetectable in serum of patients but was measurable in all carriers (mean 15.5 pg/mL; 95% confidence interval [CI] 13.7 to 17.2 pg/mL), in whom it was significantly lower than in healthy controls (mean 40.0 pg/mL; 95% CI 36.9 to 42.7 pg/mL; p < 0.001). P1NP levels were highest in patients (mean 153.7 ng/mL; 95% CI 100.5 to 206.9 ng/mL; p = 0.01 versus carriers, p = 0.002 versus controls), but carriers also had significantly higher P1NP levels (mean 58.3 ng/mL; 95% CI 47.0 to 69.6 ng/mL) than controls (mean 37.8 ng/mL; 95% CI 34.9 to 42.0 ng/mL; p = 0.006). In patients and carriers, P1NP levels declined with age, reaching a plateau after the age of 20 years. Serum sclerostin and P1NP were negatively correlated in carriers and age- and gender-matched controls (r = 0.40, p = 0.008). Mean CTX levels were well within the normal range and did not differ between patients and disease carriers after adjusting for age (p = 0.22). Our results provide in vivo evidence of increased bone formation caused by the absence or decreased synthesis of sclerostin in humans. They also suggest that inhibition of sclerostin can be titrated because the decreased sclerostin levels in disease carriers did not lead to any of the symptoms or complications of the disease but had a positive effect on bone mass. Further studies are needed to clarify the role of sclerostin on bone resorption.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone Remodeling/physiology , Heterozygote , Hyperostosis/physiopathology , Models, Biological , Syndactyly/physiopathology , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Biomarkers/metabolism , Bone Morphogenetic Proteins/blood , Calcium/metabolism , Case-Control Studies , Child , Collagen Type I/blood , Female , Genetic Markers , Humans , Hyperostosis/blood , Hyperostosis/pathology , Male , Middle Aged , Peptide Fragments/blood , Peptides/blood , Procollagen/blood , Syndactyly/blood , Syndactyly/pathology , Young Adult
4.
J Biol Chem ; 285(53): 41614-26, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-20952383

ABSTRACT

Sclerostin is expressed by osteocytes and has catabolic effects on bone. It has been shown to antagonize bone morphogenetic protein (BMP) and/or Wnt activity, although at present the underlying mechanisms are unclear. Consistent with previous findings, Sclerostin opposed direct Wnt3a-induced but not direct BMP7-induced responses when both ligand and antagonist were provided exogenously to cells. However, we found that when both proteins are expressed in the same cell, sclerostin can antagonize BMP signaling directly by inhibiting BMP7 secretion. Sclerostin interacts with both the BMP7 mature domain and pro-domain, leading to intracellular retention and proteasomal degradation of BMP7. Analysis of sclerostin knock-out mice revealed an inhibitory action of sclerostin on Wnt signaling in both osteoblasts and osteocytes in cortical and cancellous bones. BMP7 signaling was predominantly inhibited by sclerostin in osteocytes of the calcaneus and the cortical bone of the tibia. Our results suggest that sclerostin exerts its potent bone catabolic effects by antagonizing Wnt signaling in a paracrine and autocrine manner and antagonizing BMP signaling selectively in the osteocytes that synthesize simultaneously both sclerostin and BMP7 proteins.


Subject(s)
Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Proteins/chemistry , Genetic Markers/physiology , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alleles , Animals , Bone Morphogenetic Proteins/physiology , Female , Glycoproteins , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Signal Transduction , Surface Plasmon Resonance , Transcription Factors/metabolism
5.
Hum Mutat ; 31(7): E1526-43, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20583295

ABSTRACT

Sclerosteosis is a rare bone dysplasia characterized by greatly increased bone mass, especially of the long bones and the skull. Patients are tall, show facial asymmetry and often have syndactyly. Clinical complications are due to entrapment of cranial nerves. The disease is thought to be due to loss-of-function mutations in the SOST gene. The SOST gene product, sclerostin, is secreted by osteocytes and transported to the bone surface where it inhibits osteoblastic bone formation by antagonizing Wnt signaling. In a small Turkish family with sclerosteosis, we identified a missense mutation (c.499T>C; p.Cys167Arg) in exon 2 of the SOST gene. This type of mutation has not been previously reported and using different functional approaches, we show that it has a devastating effect on the biological function of sclerostin. The affected cysteine is the last cysteine residue of the cystine-knot motif and loss of this residue leads to retention of the mutant protein in the ER, possibly as a consequence of impaired folding. Together with a significant reduced ability to bind to LRP5 and inhibit Wnt signaling, the p.Cys167Arg mutation leads to a complete loss of function of sclerostin and thus to the characteristic sclerosteosis phenotype.


Subject(s)
Bone Morphogenetic Proteins/genetics , Genetic Markers/genetics , Genetic Predisposition to Disease , Hyperostosis/genetics , Mutation, Missense , Adaptor Proteins, Signal Transducing , Adult , Blotting, Western , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Line, Tumor , DNA Mutational Analysis , Family Health , Female , Humans , Hyperostosis/metabolism , Hyperostosis/pathology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Microscopy, Confocal , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Red Fluorescent Protein
6.
J Bone Miner Res ; 25(8): 1867-76, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20200987

ABSTRACT

Remodeling imbalance in the elderly femoral neck can result in thin cortices and porosity predisposing to hip fracture. Hip osteoarthritis protects against intracapsular hip fracture. By secreting sclerostin, osteocytes may inhibit Wnt signaling and reduce bone formation by osteoblasts. We hypothesised that differences in osteocytic sclerostin expression might account for differences in osteonal bone-formation activity between controls and subjects with hip fracture or hip osteoarthritis. Using specific antibody staining, we determined the osteocytic expression of sclerostin within osteons of the femoral neck cortex in bone removed from subjects undergoing surgery for hip osteoarthritis (hOA: 5 males, 5 females, 49 to 92 years of age) or hip fracture fixation (FNF: 5 males, 5 females, 73 to 87 years of age) and controls (C: 5 males, 6 females, 61 to 90 years of age). Sclerostin expression and distances of each osteocyte to the canal surface and cement line were assessed for all osteonal osteocytes in 636 unremodeled osteons chosen from fields ( approximately 0.5 mm in diameter) with at least one canal staining for alkaline phosphatase (ALP), a marker of bone formation. In adjacent sections, ALP staining was used to classify basic multicellular unit (BMUs) as quiescent or actively forming bone (ALP(+)). The areal densities of scl(-) and scl(+) osteocytes (number of cells per unit area) in the BMU were inversely correlated and were strong determinants of ALP status in the BMU. In controls and hip fracture patients only, sclerostin-negative osteocytes were closer to osteonal surfaces than positively stained cells. Osteon maturity (progress to closure) was strongly associated with the proportion of osteonal osteocytes expressing sclerostin, and sclerostin expression was the chief determinant of ALP status. hOA patients had 18% fewer osteocytes per unit bone area than controls, fewer osteocytes expressed sclerostin on average than in controls, but wide variation was seen between subjects. Thus, in most hOA patients, there was increased osteonal ALP staining and reduced sclerostin staining of osteocytes. In FNF patients, newly forming osteons were similar in this respect to hOA osteons, but with closure, there was a much sharper reduction in ALP staining that was only partly accounted for by the increased proportions of osteonal osteocytes staining positive for sclerostin. There was no evidence for a greater effect on ALP expression by osteocytes near the osteonal canal. In line with data from blocking antibody experiments, osteonal sclerostin appears to be a strong determinant of whether osteoblasts actively produce bone. In hOA, reduced sclerostin expression likely mediates increased osteoblastic activity in the intracapsular cortex. In FNF, full osteonal closure is postponed, with increased porosity, in part because the proportion of osteocytes expressing sclerostin increases sharply with osteonal maturation.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Femoral Neck Fractures/complications , Femoral Neck Fractures/metabolism , Osteoarthritis, Hip/complications , Osteoarthritis, Hip/metabolism , Osteogenesis , Adaptor Proteins, Signal Transducing , Aged , Alkaline Phosphatase/metabolism , Female , Femoral Neck Fractures/pathology , Femoral Neck Fractures/physiopathology , Genetic Markers , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Osteoarthritis, Hip/pathology , Osteoarthritis, Hip/physiopathology , Osteocytes/enzymology , Osteocytes/pathology , beta Catenin/metabolism
7.
J Bone Joint Surg Am ; 90 Suppl 1: 31-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18292354

ABSTRACT

Sclerosteosis and Van Buchem disease are rare, high-bone-mass disorders that have been linked to deficiency in the SOST gene, encoding sclerostin. Sclerostin belongs to the DAN family of glycoproteins, of which multiple family members have been shown to antagonize bone morphogenetic protein (BMP) and/or Wnt activity. Sclerostin is specifically expressed by osteocytes and inhibits BMP-induced osteoblast differentiation and ectopic bone formation. Sclerostin binds only weakly to BMPs and does not inhibit direct BMP-induced responses. Instead, sclerostin antagonizes canonical Wnt signaling by binding to Wnt coreceptors, low-density lipoprotein receptor-related protein 5 and 6. Several lipoprotein receptor-related protein-5 mutants that cause the high-bone-mass trait are defective in sclerostin binding. Thus, high bone mass in sclerosteosis and Van Buchem disease may result from increased Wnt signaling due to the absence of or insensitivity to sclerostin.


Subject(s)
Bone Diseases, Developmental/genetics , Bone Morphogenetic Proteins/physiology , Genetic Markers/genetics , Osteocytes/physiology , Osteogenesis/physiology , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing , Bone Density , Bone Morphogenetic Proteins/genetics , Genetic Markers/physiology , Humans , Osteogenesis/genetics , Signal Transduction
8.
Curr Opin Investig Drugs ; 8(4): 293-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17458178

ABSTRACT

The need to develop novel drugs that stimulate bone formation and thereby elevate bone mass (anabolics), as opposed to preventing bone loss (anti-resorptives), has opened new research areas for therapeutic intervention in the treatment of osteoporosis. One of these areas is the Wnt/beta-catenin (canonical) pathway that plays an important role in regulating osteoblast proliferation and differentiation. Alterations in this pathway have been associated with bone disorders characterized by either low or high bone mass. However, as the Wnt/beta-catenin pathway is a ubiquitous mechanism not just exclusively involved in bone formation, targeting Wnts may be a challenge (eg, targeting Wnt activity may induce cancer). Nevertheless, specific pharmacological targets to influence bone formation have been identified in this pathway; these include the Wnt-lipoprotein receptor-related protein 5/6-frizzled complex and Wnt antagonists such as sclerostin. Since sclerostin expression is highly restricted to osteocytes, this specific target may be ideal for anabolic drug therapy.


Subject(s)
Drug Therapy/methods , Osteoporosis/prevention & control , Signal Transduction/drug effects , Wnt Proteins/physiology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Humans , Models, Biological , Osteoblasts/cytology , Osteoblasts/drug effects
9.
J Cell Sci ; 120(Pt 6): 964-72, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17311849

ABSTRACT

Genetic studies in mice and humans have shown that the transforming growth factor-beta (TGF-beta) type-I receptor activin receptor-like kinase 1 (ALK1) and its co-receptor endoglin play an important role in vascular development and angiogenesis. Here, we demonstrate that ALK1 is a signalling receptor for bone morphogenetic protein-9 (BMP-9) in endothelial cells (ECs). BMP-9 bound with high affinity to ALK1 and endoglin, and weakly to the type-I receptor ALK2 and to the BMP type-II receptor (BMPR-II) and activin type-II receptor (ActR-II) in transfected COS cells. Binding of BMP-9 to ALK2 was greatly facilitated when BMPR-II or ActR-II were co-expressed. Whereas BMP-9 predominantly bound to ALK1 and BMPR-II in ECs, it bound to ALK2 and BMPR-II in myoblasts. In addition, we observed binding of BMP-9 to ALK1 and endoglin in glioblastoma cells. BMP-9 activated Smad1 and/or Smad5, and induced ID1 protein and endoglin mRNA expression in ECs. Furthermore, BMP-9 was found to inhibit basic fibroblast growth factor (bFGF)-stimulated proliferation and migration of bovine aortic ECs (BAECs) and to block vascular endothelial growth factor (VEGF)-induced angiogenesis. Taken together, these results suggest that BMP-9 is a physiological ALK1 ligand that plays an important role in the regulation of angiogenesis.


Subject(s)
Activin Receptors, Type II/physiology , Bone Morphogenetic Proteins/physiology , Cell Proliferation/drug effects , Endothelial Cells/physiology , Fibroblast Growth Factor 2/pharmacology , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Activin Receptors, Type I/physiology , Animals , COS Cells , Cattle , Cell Line, Tumor , Cell Movement , Cells, Cultured , Chlorocebus aethiops , Endothelial Cells/drug effects , Growth Differentiation Factor 2 , Growth Differentiation Factors , Humans , Hypoxanthine Phosphoribosyltransferase/metabolism , Mice , Protein Binding , Receptors, Cell Surface/metabolism , Signal Transduction
10.
Am J Pathol ; 170(1): 160-75, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200191

ABSTRACT

Prostate and mammary cancer bone metastases can be osteoblastic or osteolytic, but the mechanisms determining these features are unclear. Bone morphogenetic and Wnt proteins are osteoinductive molecules. Their activity is modulated by antagonists such as noggin and dickkopf-1. Differential expression analysis of bone morphogenetic and Wnt protein antagonists in human prostate and mammary cancer cell lines showed that osteolytic cell lines constitutively express in vitro noggin and dickkopf-1 and at least one of the osteolytic cytokines parathyroid hormone-related protein, colony-stimulating factor-1, and interleukin-8. In contrast, osteoinductive cell lines express neither noggin nor dickkopf-1 nor osteolytic cytokines in vitro. The noggin differential expression profile observed in vitro was confirmed in vivo in prostate cancer cell lines xenografted into bone and in clinical samples of bone metastasis. Forced noggin expression in an osteoinductive prostate cancer cell line abolished the osteoblast response induced in vivo by its intraosseous xenografts. Basal bone resorption and tumor growth kinetics were marginally affected. Lack of noggin and possibly dickkopf-1 expression by cancer cells may be a relevant mechanism contributing to the osteoblast response in bone metastases. Concomitant lack of osteolytic cytokines may be permissive of this effect. Noggin is a candidate drug for the adjuvant therapy of bone metastasis.


Subject(s)
Bone Neoplasms , Carrier Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Osteoblasts , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Proliferation , Cytokines/biosynthesis , Female , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Transplantation , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
11.
Dev Dyn ; 236(2): 606-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17195180

ABSTRACT

Spatial-temporal regulation of bone morphogenetic protein (BMP) and Wnt activity is essential for normal cardiovascular development, and altered activity of these growth factors causes maldevelopment of the cardiac outflow tract and great arteries. In the present study, we show that SOST, a Dan family member reported to antagonize BMP and Wnt activity, is expressed within the medial vessel wall of the great arteries containing smooth muscle cells. The ascending aorta, aortic arch, brachiocephalic artery, common carotids, and pulmonary trunk were all associated with SOST expressing smooth muscle cells, while the heart itself, including the valves, and more distal arteries, that is, pulmonary arteries, subclavian arteries, and descending aorta, were negative. SOST was expressed from embryonic day 15.5 up to the neonatal period. SOST expression, however, did not correspond with inhibition of Smad-dependent BMP activity or beta-catenin-dependent Wnt activity in the great arteries. Activity of both signaling pathways was already down-regulated before induction of SOST expression.


Subject(s)
Arteries/metabolism , Bone Morphogenetic Proteins/metabolism , Cardiovascular System/embryology , Cardiovascular System/growth & development , Gene Expression Regulation, Developmental , Muscle, Smooth/metabolism , Signal Transduction/genetics , Adaptor Proteins, Signal Transducing , Animals , Cardiovascular System/metabolism , Genetic Markers , Glycoproteins , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Mice , Wnt Proteins/metabolism
12.
J Bone Miner Res ; 22(1): 19-28, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17032150

ABSTRACT

UNLABELLED: Sclerostin is an osteocyte-derived negative regulator of bone formation. It inhibits BMP-stimulated bone formation both in vitro and in vivo but has no direct effect on BMP signaling. Instead, sclerostin inhibits Wnt signaling that is required for BMP-stimulated osteoblastic differentiation. INTRODUCTION: Sclerostin is a member of the Dan family of glycoproteins of which many members have been reported to antagonize BMP activity. Sclerostin has been shown to inhibit BMP-stimulated bone formation, but its mechanism of action seems to be different from classical BMP antagonists. In this study, we investigated the mechanism by which sclerostin inhibits BMP-stimulated bone formation. MATERIALS AND METHODS: DNA electroporation of calf muscle of mice using expression plasmids for BMP and sclerostin was used to study the effect of sclerostin on BMP-induced bone formation in vivo. Transcriptional profiling using microarrays of osteoblastic cells treated with BMP in the absence or presence of sclerostin was used to find specific growth factor signaling pathways affected by sclerostin. The affected pathways were further studied using growth factor-specific reporter constructs. RESULTS: BMP-induced ectopic bone formation in calf muscle of mice was prevented by co-expression of sclerostin in vivo. Transcriptional profiling analysis of osteoblastic cultures indicated that sclerostin specifically affects BMP and Wnt signaling out of many other growth signaling pathways. Sclerostin, however, did not inhibit stimulation of direct BMP target genes. Furthermore, we did not obtain any evidence for sclerostin acting as a direct BMP antagonist using a BMP-specific reporter construct. In contrast, sclerostin shared many characteristics with the Wnt antagonist dickkopf-1 in antagonizing BMP-stimulated bone formation and BMP- and Wnt-induced Wnt reporter construct activation. CONCLUSIONS: Sclerostin inhibits BMP-stimulated bone formation but does not affect BMP signaling. Instead, it antagonizes Wnt signaling in osteoblastic cells. High bone mass in sclerosteosis and van Buchem disease may, therefore, result from increased Wnt signaling.


Subject(s)
Bone Development/physiology , Bone Morphogenetic Proteins/physiology , Genetic Markers/physiology , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Development/drug effects , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Cells, Cultured , Electroporation , Gene Expression Regulation , Genes, Reporter , Glycoproteins , Humans , Intercellular Signaling Peptides and Proteins , Mesoderm/cytology , Mesoderm/physiology , Mice , Muscle, Skeletal/physiology , Oligonucleotide Array Sequence Analysis , Recombinant Proteins/pharmacology , Signal Transduction , Transfection
13.
J Bone Miner Res ; 20(10): 1867-77, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16160745

ABSTRACT

UNLABELLED: We examined the role of Wnt/beta-catenin signaling in successive stages of osteoblast differentiation. It has been shown that Wnt signaling in mature osteoblasts needs to be downregulated to enable the formation of a mineralized matrix. Using RNA interference, we showed that this is, at least in part, accomplished by upregulation of the Wnt antagonists Dickkopf-1 and -2. INTRODUCTION: The role of Wnt signaling in the initiation of osteoblast differentiation has been well studied. However, the role during late-stage differentiation is less clear. We have examined the role of Wnt/beta-catenin signaling in successive stages of osteoblast differentiation. MATERIALS AND METHODS: We treated murine bone marrow and mesenchymal stem cell-like KS483 cells with either LiCl or Wnt3A during several stages of osteoblast differentiation. In addition, we generated stable KS483 cell lines silencing either the Wnt antagonist Dkk-1 or -2 RESULTS: Activation of Wnt signaling by LiCl inhibits the formation of a mineralized bone matrix in both cell types. Whereas undifferentiated KS483 cells respond to Wnt3A by inducing nuclear beta-catenin translocation, differentiated cells do not. This is at least in part accomplished by upregulated expression of Dkk-1 and -2 during osteoblast differentiation. Using RNA interference, we showed that Dkk-1 plays a crucial role in blunting the BMP-induced alkaline phosphatase (ALP) response and in the transition of an ALP+ osteoblast in a mineralizing cell. In contrast, Dkk-2 plays a role in osteoblast proliferation and the initiation of osteoblast differentiation. CONCLUSIONS: Our data suggest that Wnt signaling in maturing osteoblasts needs to be downregulated to enable the formation of a mineralized bone matrix. Furthermore, they suggest that Dkk-1 and Dkk-2 may have distinct functions in osteoblast differentiation.


Subject(s)
Cell Differentiation/drug effects , Intercellular Signaling Peptides and Proteins/biosynthesis , Osteoblasts/metabolism , Proteins/metabolism , Up-Regulation/drug effects , Wnt Proteins/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Antimanic Agents/pharmacology , Bone Marrow Cells/metabolism , Cell Line , Lithium Chloride/pharmacology , Male , Mesenchymal Stem Cells/metabolism , Mice , Signal Transduction/drug effects , Wnt Proteins/metabolism , Wnt3 Protein , Wnt3A Protein , beta Catenin/metabolism
14.
J Clin Endocrinol Metab ; 90(12): 6392-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16189254

ABSTRACT

BACKGROUND: Sclerosteosis is an autosomal recessive sclerosing bone disorder due to deficiency of sclerostin, a protein secreted by the osteocytes that inhibits bone formation. In the present study we assessed the effect of variable expression of the genetic defect on bone mineral density (BMD) in patients and carriers of the determinant gene. METHODS: We studied 25 individuals (seven patients and 18 phenotypically normal heterozygotes). BMD was measured by dual x-ray absorptiometry at the lumbar spine, total hip, and distal forearm, and lateral radiographs of the skull were obtained. RESULTS: Individuals with sclerosteosis had markedly increased BMD at all skeletal sites (Z-score ranges: lumbar spine, +7.73 to +14.43; total hip, +7.84 to +11.51; forearm, +4.44 to +9.53). In heterozygotes, BMD was above the mean value of healthy age-matched individuals at all skeletal sites and had a wide range of normal and clearly increased values. Skull radiographs showed the typical hyperostotic changes in affected individuals and mild or no changes in heterozygotes. CONCLUSIONS: Heterozygous carriers of sclerosteosis have BMD values consistently higher than the mean of healthy subjects without any of the bone complications encountered in homozygotes. This finding suggests that the production and/or activity of sclerostin can be titrated in vivo, leading to variable increases in bone mass without any unwanted skeletal effects, a hypothesis of obvious significance for the development of new therapeutics for osteoporosis.


Subject(s)
Bone Density , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/metabolism , Bone Morphogenetic Proteins/deficiency , Genes, Recessive , Heterozygote , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/genetics , Child , Child, Preschool , Genetic Markers , Humans , Lumbar Vertebrae/metabolism , Middle Aged , Radiography , Skull/diagnostic imaging
15.
FASEB J ; 19(13): 1842-4, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16123173

ABSTRACT

Osteocytes are the most abundant cells in bone and are ideally located to influence bone turnover through their syncytial relationship with surface bone cells. Osteocyte-derived signals have remained largely enigmatic, but it was recently reported that human osteocytes secrete sclerostin, an inhibitor of bone formation. Absent sclerostin protein results in the high bone mass clinical disorder sclerosteosis. Here we report that within adult iliac bone, newly embedded osteocytes were negative for sclerostin staining but became positive at or after primary mineralization. The majority of mature osteocytes in mineralized cortical and cancellous bone was positive for sclerostin with diffuse staining along dendrites in the osteocyte canaliculi. These findings provide for the first time in vivo evidence to support the concept that osteocytes secrete sclerostin after they become embedded in a mineralized matrix to limit further bone formation by osteoblasts. Sclerostin did not appear to influence the formation of osteocytes. We propose that sclerostin production by osteocytes may regulate the linear extent of formation and the induction or maintenance of a lining cell phenotype on bone surfaces. In doing so, sclerostin may act as a key inhibitory signal governing skeletal microarchitecture.


Subject(s)
Bone Diseases, Developmental/metabolism , Bone Morphogenetic Proteins/physiology , Bone and Bones/metabolism , Genetic Markers/physiology , Osteocytes/metabolism , Adaptor Proteins, Signal Transducing , Aged , Alkaline Phosphatase/metabolism , Antibodies, Monoclonal/chemistry , Biopsy , Bone Development , Bone Diseases, Developmental/pathology , Bone Morphogenetic Proteins/chemistry , Bone Remodeling , Bone Resorption , Cell Differentiation , Coloring Agents/pharmacology , Female , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Microscopy, Fluorescence , Middle Aged , Models, Biological , Osteoblasts/cytology , Osteocytes/cytology , Osteogenesis , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Tolonium Chloride/pharmacology
16.
Cytokine Growth Factor Rev ; 16(3): 319-27, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15869900

ABSTRACT

Sclerosteosis and Van Buchem disease are two closely related bone disorders characterized by progressive bone thickening due to increased bone formation. Sclerosteosis is associated with mutations in the SOST gene and Van Buchem disease with a 52 kb deletion downstream of the SOST gene that probably affects transcription of the gene. Expression of the gene product sclerostin in bone is restricted to osteocytes and it is a negative regulator of bone formation. It inhibits BMP-stimulated bone formation, but cannot antagonize all BMP responses. The exclusive bone phenotype of good quality of patients with sclerosteosis and Van Buchem disease and the specific localization of sclerostin make it an attractive target for the development of bone forming therapeutics.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Osteogenesis , Adaptor Proteins, Signal Transducing , Animals , Genetic Markers , Humans , Hyperostosis/genetics , Hyperostosis/physiopathology , Osteocytes/metabolism , Osteosclerosis/genetics , Osteosclerosis/physiopathology
17.
J Exp Med ; 199(6): 805-14, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15024046

ABSTRACT

Sclerosteosis, a skeletal disorder characterized by high bone mass due to increased osteoblast activity, is caused by loss of the SOST gene product, sclerostin. The localization in bone and the mechanism of action of sclerostin are not yet known, but it has been hypothesized that it may act as a bone morphogenetic protein (BMP) antagonist. We show here that SOST/sclerostin is expressed exclusively by osteocytes in mouse and human bone and inhibits the differentiation and mineralization of murine preosteoblastic cells (KS483). Although sclerostin shares some of the actions of the BMP antagonist noggin, we show here that it also has actions distinctly different from it. In contrast to noggin, sclerostin did not inhibit basal alkaline phosphatase (ALP) activity in KS483 cells, nor did it antagonize BMP-stimulated ALP activity in mouse C2C12 cells. In addition, sclerostin had no effect on BMP-stimulated Smad phosphorylation and direct transcriptional activation of MSX-2 and BMP response element reporter constructs in KS483 cells. Its unique localization and action on osteoblasts suggest that sclerostin may be the previously proposed osteocyte-derived factor that is transported to osteoblasts at the bone surface and inhibits bone formation.


Subject(s)
Bone Development/physiology , Bone Diseases, Developmental/metabolism , Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental , Osteoblasts/metabolism , Adaptor Proteins, Signal Transducing , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Cells, Cultured , DNA Primers , DNA, Complementary/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Genetic Markers/genetics , Genetic Markers/physiology , Glycoproteins , Homeodomain Proteins , Humans , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Luciferases , Mice , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins , Trans-Activators/metabolism , Transfection
18.
Haematologica ; 88(8): 845-52, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12935972

ABSTRACT

BACKGROUND AND OBJECTIVES: We previously found that human fetal lung is a rich source of mesenchymal stem cells (MSC). Here we characterize and analyze the frequency and function of MSC in other second-trimester fetal tissues. DESIGN AND METHODS: Single cell suspensions of fetal bone marrow (BM), liver, lung, and spleen were made and analyzed by flow cytometry for the expression of CD90, CD105, CD166, SH3, SH4, HLA-ABC, HLA-DR, CD34 and CD45. We assessed the frequency of MSC by limiting dilution assay. RESULTS: The frequency of MSC in BM was significantly higher than in liver, lung, and spleen (p<0.05). On primary non-expanded cells from fetal liver, lung and spleen the number of cells positive for mesenchymal markers was significantly higher within the CD34 positive population than within the CD34 negative population. The phenotype of the culture-expanded MSC was similar for all fetal tissues, i.e. CD90, CD105, CD166, SH3, SH4 and HLA-ABC positive and CD34, CD45 and HLA-DR negative. Culture-expanded cells from all tissues were able to differentiate along adipogenic and osteogenic pathways. However, adipogenic differentiation was less in MSC derived from spleen, and osteogenic differentiation was reduced in liver-derived MSC (p<0.05). INTERPRETATION AND CONCLUSIONS: Our results indicate that culture-expanded MSC derived from second-trimester fetal tissues, although phenotypically similar, exhibit heterogeneity in differentiating potential. We speculate that these differences may be relevant for the clinical application of MSC.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Immunophenotyping , Liver/cytology , Lung/cytology , Mesoderm/cytology , Multipotent Stem Cells/cytology , Spleen/cytology , Abortion, Legal , Adipocytes/chemistry , Adipocytes/classification , Adipocytes/cytology , Adipocytes/metabolism , Antigens, CD34/analysis , Antigens, CD34/immunology , Cells, Cultured , Female , Fetus/cytology , Granulocytes/chemistry , Granulocytes/cytology , Granulocytes/metabolism , Humans , Immunophenotyping/methods , Liver/embryology , Lung/embryology , Lymphocytes/chemistry , Lymphocytes/cytology , Lymphocytes/metabolism , Mesoderm/classification , Monocytes/chemistry , Monocytes/cytology , Monocytes/metabolism , Multipotent Stem Cells/classification , Osteocytes/chemistry , Osteocytes/classification , Osteocytes/cytology , Osteocytes/metabolism , Pregnancy , Pregnancy Trimester, Second , Spleen/embryology
19.
Exp Hematol ; 30(8): 870-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12160838

ABSTRACT

OBJECTIVE: Mesenchymal stem cells (MSC) have been implicated as playing an important role in hematopoietic stem cell engraftment. We identified and characterized a new population of MSC derived from human fetal lung. In cotransplantation experiments, we examined the homing of MSC as well as the effect on engraftment of human umbilical cord blood (UCB)-derived CD34(+) cells in NOD/SCID mice. MATERIALS AND METHODS: Culture-expanded fetal lung-derived CD34(+) cells were characterized by immune phenotyping and cultured under conditions promoting differentiation to osteoblasts or adipocytes. Irradiated (3.5 Gy) NOD/SCID mice (n = 51) were transplanted intravenously with 0.03 to 1.0 x 10(6) UCB CD34(+) cells in the presence or absence of 1 x 10(6) culture-expanded fetal lung-derived MSC, irradiated CD34(-) cells, B cells, or with cultured MSC only. RESULTS: Culture-expanded fetal lung CD34(+) cells were identified as MSC based on phenotype (CD105(+), SH3(+), SH4(+), CD160(+)) and their multilineage potential. Cotransplantation of low doses of UCB CD34(+) cells and MSC resulted in a three-fold to four-fold increase in bone marrow engraftment after 6 weeks, whereas no such effect was observed after cotransplantation of irradiated CD34(-) or B cells. Homing experiments indicated the presence of MSC in the lung, but not in the bone marrow, of NOD/SCID mice. CONCLUSIONS: We identified a population of MSC derived from human fetal lung. Upon cotransplantation, MSC, but not irradiated CD34(-) or B cells, promote engraftment of UCB CD34(+) cells in bone marrow, spleen, and blood by mechanisms that may not require homing of MSC to the bone marrow.


Subject(s)
Fetal Blood/cytology , Fetal Tissue Transplantation , Graft Survival , Hematopoietic Stem Cell Transplantation , Mesoderm/cytology , Stem Cell Transplantation , Adipocytes/cytology , Animals , B-Lymphocytes/radiation effects , Bone Marrow/pathology , Cell Differentiation , Cell Lineage , Cell Movement , Cytokines/metabolism , Hematopoietic Stem Cells/radiation effects , Humans , Lung/cytology , Lung/embryology , Mice , Mice, Inbred NOD , Mice, SCID , Organ Specificity , Osteoblasts/cytology , Radiation Chimera , Stem Cells/metabolism , Stem Cells/physiology , Transplantation, Heterologous
20.
Endocrinology ; 143(4): 1545-53, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11897714

ABSTRACT

During bone formation and fracture healing there is a cross-talk between endothelial cells and osteoblasts. We previously showed that vascular endothelial growth factor A (VEGF-A) might be an important factor in this cross-talk, as osteoblast-like cells produce this angiogenic factor in a differentiation-dependent manner. Moreover, exogenously added VEGF-A enhances osteoblast differentiation. In the present study we investigated, given the coupling between angiogenesis and bone formation, whether bone morphogenetic proteins (BMPs) stimulate osteoblastogenesis and angiogenesis through the production of VEGF-A. For this we used the murine preosteoblast-like cell line KS483, which forms mineralized nodules in vitro, and an angiogenesis assay comprising 17-d-old fetal mouse bone explants that have the ability to form tube-like structures in vitro. Treatment of KS483 cells with BMP-2, -4, and -6 enhanced nodule formation, osteocalcin mRNA expression, and subsequent mineralization after 18 d of culture. This was accompanied by a dose-dependent increase in VEGF-A protein levels throughout the culture period. BMP-induced osteoblast differentiation, however, was independent of VEGF-A, as blocking VEGF-A activity by a VEGF-A antibody or a VEGF receptor 2 tyrosine kinase inhibitor did not affect BMP-induced mineralization. To investigate whether BMPs stimulate angiogenesis through VEGF-A, BMPs were assayed for their angiogenic activity. Treatment of bone explants with BMPs enhanced angiogenesis. This was inhibited by soluble BMP receptor 1A or noggin. In the presence of a VEGF-A antibody, both unstimulated and BMP-stimulated angiogenesis were arrested. Conditioned media of KS483 cells treated with BMPs also induced a strong angiogenic response, which was blocked by antimouse VEGF-A but not by noggin. These effects were specific for BMPs, as TGF beta inhibited osteoblast differentiation and angiogenesis while stimulating VEGF-A production. These findings indicate that BMPs stimulate angiogenesis through the production of VEGF-A by osteoblasts. In conclusion, VEGF-A produced by osteoblasts in response to BMPs is not involved in osteoblast differentiation, but couples angiogenesis to bone formation.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Endothelial Growth Factors/physiology , Neovascularization, Physiologic/drug effects , Osteoblasts/drug effects , Animals , Blotting, Northern , Calcium/metabolism , Cell Differentiation/drug effects , Cell Line , Culture Media, Conditioned , Dactinomycin/pharmacology , Enzyme-Linked Immunosorbent Assay , Mice , Osteoblasts/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A
SELECTION OF CITATIONS
SEARCH DETAIL
...