Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Calcif Tissue Int ; 114(3): 255-266, 2024 03.
Article in English | MEDLINE | ID: mdl-38226986

ABSTRACT

X-linked hypophosphatemia (XLH) is the most common monogenetic cause of chronic hypophosphatemia, characterized by rickets and osteomalacia. Disease manifestations and treatment of XLH patients in the Netherlands are currently unknown. Characteristics of XLH patients participating in the Dutch observational registry for genetic hypophosphatemia and acquired renal phosphate wasting were analyzed. Eighty XLH patients, including 29 children, were included. Genetic testing, performed in 78.8% of patients, showed a PHEX mutation in 96.8%. Median (range) Z-score for height was - 2.5 (- 5.5; 1.0) in adults and - 1.4 (- 3.7; 1.0) in children. Many patients were overweight or obese: 64.3% of adults and 37.0% of children. All children received XLH-related medication e.g., active vitamin D, phosphate supplementation or burosumab, while 8 adults used no medication. Lower age at start of XLH-related treatment was associated with higher height at inclusion. Hearing loss was reported in 6.9% of children and 31.4% of adults. Knee deformities were observed in 75.0% of all patients and osteoarthritis in 51.0% of adult patients. Nephrocalcinosis was observed in 62.1% of children and 33.3% of adults. Earlier start of XLH-related treatment was associated with higher risk of nephrocalcinosis and detection at younger age. Hyperparathyroidism longer than six months was reported in 37.9% of children and 35.3% of adults. This nationwide study confirms the high prevalence of adiposity, hearing loss, bone deformities, osteoarthritis, nephrocalcinosis and hyperparathyroidism in Dutch XLH patients. Early start of XLH-related treatment appears to be beneficial for longitudinal growth but may increase development of nephrocalcinosis.


Subject(s)
Familial Hypophosphatemic Rickets , Hearing Loss , Hyperparathyroidism , Hypophosphatemia , Nephrocalcinosis , Osteoarthritis , Child , Adult , Humans , Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/diagnosis , Nephrocalcinosis/genetics , Nephrocalcinosis/complications , Fibroblast Growth Factors/genetics , Hypophosphatemia/epidemiology , Hypophosphatemia/genetics , Phosphates , Hyperparathyroidism/complications , Obesity/complications , Hearing Loss/complications , Hearing Loss/drug therapy
2.
Bone ; 165: 116564, 2022 12.
Article in English | MEDLINE | ID: mdl-36150657

ABSTRACT

Studies on mice have shown a relationship between dietary intake of advanced glycation end-products (dAGEs) and deterioration of musculoskeletal health, but human studies are absent. We investigated the relationship between dietary intake of carboxymethyllysine (dCML) - an AGE prototype - and risk of sarcopenia at baseline and after 5 years of follow-up and a single evaluation of physical frailty in participants from the population-based Rotterdam Study. Appendicular lean mass (ALM) was obtained using insight dual-energy X-ray absorptiometry and hand grip strength (HGS) using a hydraulic hand dynamometer. Subjects with both low ALM and weak HGS were classified as having sarcopenia. Frailty (yes/no) was defined by presence of ≥3 and pre-frailty by presence of 1 or 2 components namely, exhaustion, weakness, slowness, weight loss or low physical activity. dCML was calculated using a food frequency questionnaire and dAGE databases. Logistic regression analysis was used to evaluate the odds of physical frailty and prevalent sarcopenia at baseline and follow-up and incident sarcopenia. 2782 participants with an age 66.4 ± 9.9 years and dCML intake 3.3 ± 1.3 mg/day, had data on sarcopenia at both time points. Of whom 84 had sarcopenia at baseline and 73 developed sarcopenia at follow-up. We observed an association of one SD increase in dCML intake with prevalent sarcopenia at baseline [odds ratio, OR = 1.27 (1.01-1.59)] and no association of dCML with incident sarcopenia at 5-year follow-up [OR = 1.12 (0.86-1.44)]. For frailty we analyzed 3577 participants, of whom 1972 were pre-frail and 158 were frail. We observed no association of dCML with either pre-frailty [OR = 0.99 (0.91-1.07)] or frailty [OR = 1.01 (0.83-1.22)] when non-frail subjects were used as reference. Our results show an association of dAGEs with sarcopenia cross-sectionally but not longitudinally where inconclusive findings are observed possibly due to a very low incidence of sarcopenia. There was no association with frailty cross-sectionally.


Subject(s)
Frailty , Sarcopenia , Humans , Mice , Animals , Child, Preschool , Middle Aged , Aged , Frailty/epidemiology , Sarcopenia/epidemiology , Hand Strength , Glycation End Products, Advanced , Eating
3.
Bone ; 164: 116526, 2022 11.
Article in English | MEDLINE | ID: mdl-35995334

ABSTRACT

A major precursor of advanced glycation end-products (AGEs) - methylglyoxal (MG) - is a reactive carbonyl metabolite that originates from glycolytic pathways. MG formation and accumulation has been implicated in the pathogenesis of diabetes and age-related chronic musculoskeletal disorders. Human bone marrow-derived stromal cells (BMSCs) are multipotent cells that have the potential to differentiate into cells of mesenchymal origin including osteoblasts, but the role of MG on their differentiation is unclear. We therefore evaluated the effect of MG on proliferation and differentiation of BMSC-derived osteoblasts. Cells were treated with different concentrations of MG (600, 800 and 1000 µM). Cell viability was assessed using a Cell Counting Kit-8 assay. Alkaline phosphatase (ALP) activity and calcium deposition assays were performed to evaluate osteoblast differentiation and mineralization. Gene expression was measured using qRT-PCR, whereas AGE specific receptor (RAGE) and collagen 1 were examined by immunocytochemistry and Western blotting. RAGE knockdown was performed by transducing RAGE specific short hairpin RNAs (shRNAs) using lentivirus. During osteogenic differentiation, MG treatment resulted in reduction of cell viability (27.7 %), ALP activity (45.5 %) and mineralization (82.3 %) compared to untreated cells. MG significantly decreased expression of genes involved in osteogenic differentiation - RUNX2 (2.8 fold), ALPL (3.2 fold), MG detoxification through glyoxalase - GLO1 (3 fold) and collagen metabolism - COL1A1 (4.9 fold), COL1A2 (6.8 fold), LOX (5.4 fold) and PLOD1 (1.7 fold). MG significantly reduced expression of collagen 1 (53.3 %) and RAGE (43.1 %) at protein levels. Co-treatment with a MG scavenger - aminoguanidine - prevented all negative effects of MG. RAGE-specific knockdown during MG treatment did not reverse the effects on cell viability, osteogenic differentiation or collagen metabolism. In conclusion, MG treatment can negatively influence the collagen metabolism and differentiation of BMSCs-derived osteoblasts through a RAGE independent mechanism.


Subject(s)
Glycation End Products, Advanced , Osteogenesis , Alkaline Phosphatase/metabolism , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacology , Humans , Osteoblasts/metabolism , Pyruvaldehyde/metabolism , Pyruvaldehyde/pharmacology , Receptor for Advanced Glycation End Products/metabolism
4.
Endocr Connect ; 10(3): 273-282, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33543729

ABSTRACT

BACKGROUND: Sex differences in calcium and phosphate have been observed. We aimed to assess a relation with age. METHODS: We used the laboratory values of serum calcium, phosphate and albumin from three different samples ( 2005, 2010 and 2014 years) using the hospital information system of Erasmus MC, Rotterdam. The samples were divided into three age groups: 1-17, 18-44 and ≥45 years. Sex differences in calcium and phosphate were analyzed using ANCOVA, adjusting for age and serum albumin. Furthermore, sex by age interactions were determined and we analyzed differences between age groups stratified by sex. RESULTS: In all three samples there was a significant sex × age interaction for serum calcium and phosphate, whose levels were significantly higher in women compared to men above 45 years. No sex differences in the younger age groups were found. In men, serum calcium and phosphate levels were highest in the youngest age group compared to age groups of 18-44 and ≥45 years. In women, serum calcium levels were significantly higher in the age group 1-17 and the age group ≥45 years compared to the 18-44 years age group. In women, serum phosphate was different between the three different age groups with highest level in the group 1-17 years and lowest in the group 18-44 years. CONCLUSION: There are age- dependent sex differences in serum calcium and phosphate. Furthermore, we found differences in serum calcium and phosphate between different age groups. Underlying mechanisms for these age- and sex- differences are not yet fully elucidated.

5.
Mater Today Bio ; 7: 100060, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32577614

ABSTRACT

The holy grail of orthopedic implant design is to ward off both aseptic and septic loosening for long enough that the implant outlives the patient. Questing this holy grail is feasible only if orthopedic biomaterials possess a long list of functionalities that enable them to discharge the onerous task of permanently replacing the native bone tissue. Here, we present a rationally designed and additive manufacturing (AM) topologically ordered porous metallic biomaterial that is made from Ti-6Al-4V using selective laser melting and packs most (if not all) of the required functionalities into a single implant. In addition to presenting a fully interconnected porous structure and form-freedom that enables realization of patient-specific implants, the biomaterials developed here were biofunctionalized using plasma electrolytic oxidation to locally release both osteogenic (i.e. strontium) and antibacterial (i.e. silver ions) agents. The same single-step biofunctionalization process also incorporated hydroxyapatite into the surface of the implants. Our measurements verified the continued release of both types of active agents up to 28 days. Assessment of the antibacterial activity in vitro and in an ex vivo murine model demonstrated extraordinarily high levels of bactericidal effects against a highly virulent and multidrug-resistant Staphylococcus aureus strain (i.e. USA300) with total eradication of both planktonic and adherent bacteria. This strong antibacterial behavior was combined with a significantly enhanced osteogenic behavior, as evidenced by significantly higher levels of alkaline phosphatase (ALP) activity compared with non-biofunctionalized implants. Finally, we discovered synergistic antibacterial behavior between strontium and silver ions, meaning that 4-32 folds lower concentrations of silver ions were required to achieve growth inhibition and total killing of bacteria. The functionality-packed biomaterial presented here demonstrates a unique combination of functionalities that make it an advanced prototype of future orthopedic biomaterials where implants will outlive patients.

6.
Biofabrication ; 11(3): 035012, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30921774

ABSTRACT

Decellularized tissue matrices are promising substrates for tissue generation by stem cells to replace poorly regenerating tissues such as cartilage. However, the dense matrix of decellularized cartilage impedes colonisation by stem cells. Here, we show that digestion of elastin fibre bundles traversing auricular cartilage creates channels through which cells can migrate into the matrix. Human chondrocytes and bone marrow-derived mesenchymal stromal cells efficiently colonise elastin-treated scaffolds through these channels, restoring a glycosaminoglycan-rich matrix and improving mechanical properties while maintaining size and shape of the restored tissue. The scaffolds are also rapidly colonised by endogenous cartilage-forming cells in a subcutaneously implanted osteochondral biopsy model. Creating channels for cells in tissue matrices may be a broadly applicable strategy for recellularization and restoration of tissue function.


Subject(s)
Ear Cartilage/cytology , Pancreatic Elastase/metabolism , Adolescent , Aged , Animals , Cattle , Child , Chondrogenesis , Elastin/metabolism , Extracellular Matrix/chemistry , Female , Glycosaminoglycans/metabolism , Humans , Mice, Nude , Middle Aged , Tissue Scaffolds/chemistry
7.
J Cell Physiol ; 233(6): 4895-4906, 2018 06.
Article in English | MEDLINE | ID: mdl-29194609

ABSTRACT

Osteoporosis is a common skeletal disorder characterized by low bone mass leading to increased bone fragility and fracture susceptibility. Identification of factors influencing osteoblast differentiation and bone formation is very important. Previously, we identified parbendazole to be a novel compound that stimulates osteogenic differentiation of human mesenchymal stromal cells (hMSCs), using gene expression profiling and bioinformatic analyzes, including the Connectivity Map (CMap), as an in-silico approach. The aim for this paper is to identify additional compounds affecting osteoblast differentiation using the CMap. Gene expression profiling was performed on hMSCs differentiated to osteoblasts using Illumina microarrays. Our osteoblast gene signature, the top regulated genes 6 hr after induction by dexamethasone, was uploaded into CMap (www.broadinstitute.org/cmap/). Through this approach we identified compounds with gene signatures positively correlating (withaferin-A, calcium folinate, amylocaine) or negatively correlating (salbutamol, metaraminol, diprophylline) to our osteoblast gene signature. All positively correlating compounds stimulated osteogenic differentiation, as indicated by increased mineralization compared to control treated cells. One of three negatively correlating compounds, salbutamol, inhibited dexamethasone-induced osteoblastic differentiation, while the other two had no effect. Based on gene expression data of withaferin-A and salbutamol, we identified HMOX1 and STC1 as being strongly differentially expressed . shRNA knockdown of HMOX1 or STC1 in hMSCs inhibited osteoblast differentiation. These results confirm that the CMap is a powerful approach to identify positively compounds that stimulate osteogenesis of hMSCs, and through this approach we can identify genes that play an important role in osteoblast differentiation and could be targets for novel bone anabolic therapies.


Subject(s)
Bone Density Conservation Agents/pharmacology , Cell Differentiation/drug effects , Gene Regulatory Networks/drug effects , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Osteogenesis/drug effects , Bone Density/drug effects , Bone Density/genetics , Cell Differentiation/genetics , Computational Biology , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Glycoproteins/genetics , Glycoproteins/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Osteogenesis/genetics , Protein Interaction Maps , Signal Transduction/drug effects
8.
Bone ; 95: 108-114, 2017 02.
Article in English | MEDLINE | ID: mdl-27845263

ABSTRACT

INTRODUCTION: Peripheral Blood Mononuclear Cells (PBMCs) have been extensively used as a culture model to generate osteoclasts in vitro. The aim of this study was to assess the osteoclastogenic potential of PBMCs derived from post-menopausal women with longstanding osteoporosis and compare this with PBMCs from healthy controls. MATERIAL AND METHODS: We selected from the population-based Rotterdam Study 82 participants of which 43 were diagnosed with osteoporosis (T-score below -2.5 at the lumbar spine) and the presence of at least 1 fracture and 29 healthy controls (T-score above 1; no fracture). PBMCs were differentiated into osteoclasts, and both differentiation capacity and activity were measured. Total RNA was obtained to assess gene expression of osteoclast markers. Deoxypyridinoline (DPD) was measured in plasma as a marker for bone resorption, in vivo. RESULTS: Neither the number of osteoclasts nor cathepsin K (CTSK) and dendritic cell-specific transmembrane protein (TM7SF4) gene expression was significantly different between both groups. There was also no significant difference in resorption pit area and plasma DPD levels. Stratification by fracture type into a group with vertebral, non-vertebral and both vertebral and non-vertebral fractures showed no difference in osteoclast formation or osteoclastic bone resorption. However, plasma DPD, but not the RNA expression markers, was significantly lower in the group of subjects with vertebral fracture group and those with vertebral and non-vertebral fractures compared to the healthy controls. No differences in osteoclastogenesis, osteoclastic resorption and plasma DPD levels were detected also after exclusion of past or present users of bisphosphonates and glucocorticoids. Stratification into high and low DPD levels showed higher osteoclastogenesis and more osteoclastic bone resorption in the high DPD group compared to the low DPD levels within the group of osteoporotic subjects. CONCLUSION: This study showed no difference in PBMC osteoclastogenic capacity and activity between women with and without osteoporosis and at least one previous fracture, who were on average 29.5years after menopause, suggesting that there is no difference in circulating osteoclast precursors. Although we cannot exclude that circulating precursors may behave differently at the bone site, it is possible that long after menopause a more stable phase of bone turnover is reached compared to earlier after the start of menopause in which differences in circulating osteoclast precursors and osteoclastogenic potential are more prominent.


Subject(s)
Leukocytes, Mononuclear/metabolism , Osteoclasts/pathology , Osteogenesis , Osteoporosis/blood , Osteoporosis/pathology , Aged , Bone Resorption/pathology , Case-Control Studies , Cell Differentiation , Female , Humans , Osteoporosis/drug therapy , Osteoporotic Fractures/drug therapy , Osteoporotic Fractures/pathology
9.
Endocrinology ; 157(12): 4930-4942, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27911148

ABSTRACT

Estrogen deficiency after ovariectomy (OVX) results in increased adiposity and bone loss, which can be prevented by systemic 17-ß estradiol (E2) replacement. Studies in transgenic mice suggested that in addition to direct actions of estrogen in peripheral tissues, also estrogen signaling in the hypothalamus regulates fat distribution and bone metabolism. We hypothesized that the protective effect of systemic E2 on fat and bone metabolism in the OVX model is partly mediated through the ventromedial nucleus of the hypothalamus (VMH). To test this hypothesis, we determined the effect of systemic, central, and targeted VMH administration of E2 on fat and bone metabolism in OVX rats. Subcutaneous administration of E2 for 4 weeks decreased body weight, gonadal and perirenal fat, and bone formation rate in OVX rats. This effect was completely mimicked by intracerebroventricular injections of E2, once every 4 days for 4 weeks. Administration of E2 locally in the VMH by retromicrodialysis (3 h) acutely increased expression of the lipolytic gene hormone-sensitive lipase in gonadal and perirenal fat. Finally, chronic administration of E2 in the VMH for 8 weeks decreased perirenal fat but did not affect body weight, trabecular bone volume, or cortical thickness. In conclusion, we demonstrated that intracerebroventricular E2 replacement reduces body weight gain, ameliorates intraabdominal fat accumulation, and reduces bone formation in the OVX rats. E2 administration selectively in the VMH also reduced intraabdominal fat but did not affect bone metabolism.


Subject(s)
Adipose Tissue/drug effects , Estradiol/administration & dosage , Femur/drug effects , Lipid Metabolism/drug effects , Osteogenesis/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Adipose Tissue/metabolism , Animals , Body Weight/drug effects , Female , Femur/metabolism , Ovariectomy , Rats , Sterol Esterase/genetics , Sterol Esterase/metabolism
10.
Nature ; 537(7620): 427-431, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27556946

ABSTRACT

Mice deficient in the DNA excision-repair gene Ercc1 (Ercc1∆/-) show numerous accelerated ageing features that limit their lifespan to 4-6 months. They also exhibit a 'survival response', which suppresses growth and enhances cellular maintenance. Such a response resembles the anti-ageing response induced by dietary restriction (also known as caloric restriction). Here we report that a dietary restriction of 30% tripled the median and maximal remaining lifespans of these progeroid mice, strongly retarding numerous aspects of accelerated ageing. Mice undergoing dietary restriction retained 50% more neurons and maintained full motor function far beyond the lifespan of mice fed ad libitum. Other DNA-repair-deficient, progeroid Xpg-/- (also known as Ercc5-/-) mice, a model of Cockayne syndrome, responded similarly. The dietary restriction response in Ercc1∆/- mice closely resembled the effects of dietary restriction in wild-type animals. Notably, liver tissue from Ercc1∆/- mice fed ad libitum showed preferential extinction of the expression of long genes, a phenomenon we also observed in several tissues ageing normally. This is consistent with the accumulation of stochastic, transcription-blocking lesions that affect long genes more than short ones. Dietary restriction largely prevented this declining transcriptional output and reduced the number of γH2AX DNA damage foci, indicating that dietary restriction preserves genome function by alleviating DNA damage. Our findings establish the Ercc1∆/- mouse as a powerful model organism for health-sustaining interventions, reveal potential for reducing endogenous DNA damage, facilitate a better understanding of the molecular mechanism of dietary restriction and suggest a role for counterintuitive dietary-restriction-like therapy for human progeroid genome instability syndromes and possibly neurodegeneration in general.


Subject(s)
Aging/genetics , Caloric Restriction , DNA Repair/genetics , Diet, Reducing , Genomic Instability , Animals , Brain/physiology , DNA Damage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endonucleases/deficiency , Endonucleases/genetics , Female , Male , Mice , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/prevention & control , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcriptome
11.
Bone ; 57(2): 443-54, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24084385

ABSTRACT

We explored the role of transient receptor potential vanilloid 4 (TRPV4) in murine bone metabolism and association of TRPV4 gene variants with fractures in humans. Urinary and histomorphometrical analyses demonstrated reduced osteoclast activity and numbers in male Trpv4(-/-) mice, which was confirmed in bone marrow-derived osteoclast cultures. Osteoblasts and bone formation as shown by serum procollagen type 1 amino-terminal propeptide and histomorphometry, including osteoid surface, osteoblast and osteocyte numbers were not affected in vivo. Nevertheless, osteoblast differentiation was enhanced in Trpv4(-/-) bone marrow cultures. Cortical and trabecular bone mass was 20% increased in male Trpv4(-/-) mice, compared to sex-matched wild type (Trpv4(+/+)) mice. However, at the same time intracortical porosity was increased and bone matrix mineralization was reduced. Together, these lead to a maximum load, stiffness and work to failure of the femoral bone, which were not different compared to Trpv4(+/+) mice, while the bone material was less resistant to stress and less elastic. The differential impacts on these determinants of bone strength were likely responsible for the lack of any changes in whole bone strength in the Trpv4(-/-) mice. None of these skeletal parameters were affected in female Trpv4(-/-) mice. The T-allele of rs1861809 SNP in the TRPV4 locus was associated with a 30% increased risk (95% CI: 1.1-1.6; p=0.013) for non-vertebral fracture risk in men, but not in women, in the Rotterdam Study. Meta-analyses with the population-based LASA study confirmed the association with non-vertebral fractures in men. This was lost when the non-population-based studies Mr. OS and UFO were included. In conclusion, TRPV4 is a male-specific regulator of bone metabolism, a determinant of bone strength, and a potential risk predictor for fractures through regulation of bone matrix mineralization and intra-cortical porosity. This identifies TRPV4 as a unique sexually dimorphic therapeutic and/or diagnostic candidate for osteoporosis.


Subject(s)
Bone and Bones/pathology , Osteoporotic Fractures/epidemiology , Sex Characteristics , TRPV Cation Channels/deficiency , Animals , Bone and Bones/metabolism , Elastic Modulus , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Mice , Netherlands/epidemiology , Osteoblasts/pathology , Osteoclasts/pathology , Osteoporotic Fractures/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Risk Factors , Stress, Mechanical , TRPV Cation Channels/genetics
12.
J Cell Physiol ; 228(11): 2167-74, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23589129

ABSTRACT

In healthy bones, mineralization has to be tightly controlled to avoid pathological phenotypes. In this study, we investigated interactions between 1α,25(OH)2 D3 (1,25D3) and activin A in the regulation of osteoblast induced mineralization. In human osteoblast cultures, we demonstrated that besides stimulation of mineralization, 1,25D3 also induced activin A, a strong inhibitor of mineralization. Simultaneously, follistatin (FST), the natural antagonist of activin A, was down-regulated by1,25D3. This resulted in an increase in activin A activity during 1,25D3 treatment. We also showed that in 1,25D3-treated osteoblasts, mineralization can be further increased when activin A activity was abrogated by adding exogenous FST. This observation implies that, besides stimulation of mineralization, 1,25D3 also controls activin A-mediated inhibition of mineralization. Besides activin A, 1,25D3 also induces osteocalcin (BGLAP), another inhibitor of mineralization. Warfarin, which has been shown to inactivate osteocalcin, increased 1,25D3-induced mineralization. Interaction between these two systems became evident from the synergistic increase in BGLAP expression upon blocking activin activity in 1,25D3-treated cultures. In conclusion, we demonstrate that 1,25D3 stimulation of mineralization by human osteoblasts is suppressed by concomitant induction of inhibitors of mineralization. Mineralization induction by 1,25D3 may actually be controlled via interplay with activin A and osteocalcin. Finally, this complex regulation of mineralization substantiates the significance of tight control of mineralization to prevent excessive mineralization and consequently reduction in bone quality and strength.


Subject(s)
Activins/biosynthesis , Calcification, Physiologic/drug effects , Osteoblasts/metabolism , Vitamin D/analogs & derivatives , Cell Line , Follistatin/biosynthesis , Gene Expression Regulation/drug effects , Humans , Lectins, C-Type/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/genetics , Osteocalcin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Smad7 Protein/metabolism , Vitamin D/pharmacology , Warfarin/pharmacology
13.
Gene ; 512(2): 438-43, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-22967709

ABSTRACT

Both vitamin D receptor (VDR) and peroxisome proliferator-activated receptor γ (PPAR-γ) are ligand-activated nuclear transcription factors that are instrumental for bone health. While 1α,25-dihydroxyvitamin D3 (1,25D3), the ligand for VDR, is essential for the development and maintenance of healthy bone, PPAR-γ agonists cause detrimental skeletal effects. Recent studies have revealed evidence for a cross-talk between 1,25D3- and PPAR-α/-δ ligand-mediated signaling but there is a current lack of knowledge regarding cross-talk between signaling of 1,25D3 and the PPAR-γ ligand mediated signaling. In this study, we investigated the cross-talk between 1,25D3- and PPAR-γ agonist rosiglitazone-mediated signaling in human osteoblasts. 1,25D3 slightly but significantly induced expression of the primary PPAR-γ target gene ANGPTL4 but did not influence FABP4. 1,25D3 did not change rosiglitazone regulation of ANGPTL4 and FABP4. The other way around, rosiglitazone reduced CYP24A1 gene expression but this did not change CYP24A1 induction by 1,25D3. The findings regarding CYP24A1 gene expression are in line with the observation that 1,25D3 levels in medium were not affected by rosiglitazone. Furthermore, rosiglitazone significantly inhibited 1,25D3-induction of BGLAP while rosiglitazone alone did not change BGLAP. Additionally, 1,25D3 and rosiglitazone increase osteoblast alkaline phosphatase activity and synergistically stimulated extracellular matrix mineralization. In conclusion, these data provide evidence for a cross-talk between rosiglitazone- and 1,25D3-mediated signaling leading to an acceleration of extracellular matrix mineralization. The data suggest that the reduction of the mineralization inhibitor BGLAP and the increased differentiation status underlie the increased mineralization.


Subject(s)
Bone Density Conservation Agents/agonists , Calcification, Physiologic/drug effects , Calcitriol/agonists , Hypoglycemic Agents/agonists , Osteoblasts/metabolism , Thiazolidinediones/agonists , Alkaline Phosphatase/biosynthesis , Angiopoietin-Like Protein 4 , Angiopoietins/metabolism , Bone Density Conservation Agents/pharmacology , Calcitriol/pharmacology , Cell Differentiation/drug effects , Cell Line , Drug Synergism , Extracellular Matrix/metabolism , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hypoglycemic Agents/pharmacology , Osteoblasts/cytology , PPAR alpha/metabolism , PPAR gamma/metabolism , PPAR-beta/metabolism , Receptors, Calcitriol/metabolism , Rosiglitazone , Signal Transduction/drug effects , Steroid Hydroxylases/biosynthesis , Thiazolidinediones/pharmacology , Vitamin D3 24-Hydroxylase
14.
J Cell Physiol ; 227(9): 3258-66, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22105341

ABSTRACT

It is well established that 1α-25-dihydroxyvitamin D3 (1,25D3) regulates osteoblast function and stimulates mineralization by human osteoblasts. The aim of this study was to identify processes underlying the 1,25D3 effects on mineralization. We started with gene expression profiling analyses of differentiating human pre-osteoblast treated with 1,25D3. Bioinformatic analyses showed interferon-related and -regulated genes (ISG) to be overrepresented in the set of 1,25D3-regulated genes. 1,25D3 down-regulated ISGs predominantly during the pre-mineralization period. This pointed to an interaction between the vitamin D and IFN signaling cascades in the regulation of osteoblast function. Separately, 1,25D3 enhances while IFNß inhibits mineralization. Treatment of human osteoblasts with 1,25D3 and IFNß showed that 1,25D3 completely overrules the IFNß inhibition of mineralization. This was supported by analyses of extracellular matrix gene expression, showing a dominant effect of 1,25D3 over the inhibitory effect of IFNß. We identified processes shared by IFNß- and 1,25D3-mediated signaling by performing gene expression profiling during early osteoblast differentiation. Bioinformatic analyses revealed that genes being correlated or anti-correlated with interferon-induced protein with tetratricopeptide repeats 1 (IFIT1) were associated with osteoblast proliferation. In conclusion, the current study demonstrates a cross talk between 1,25D3 and IFNß in osteoblast differentiation and bone formation/mineralization. The interaction is complex and depends on the process but importantly, 1,25D3 stimulation of mineralization is dominant over the inhibitory effect of IFNß. These observations are of potential clinical relevance considering the impact of the immune system on bone metabolism in conditions such as rheumatoid arthritis.


Subject(s)
Extracellular Matrix/genetics , Gene Expression Regulation/drug effects , Interferon-beta/genetics , Osteoblasts/metabolism , Vitamin D/metabolism , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Calcium/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Extracellular Matrix/metabolism , Genes, Dominant , Humans , Interferon-beta/metabolism , Interferon-beta/pharmacology , Signal Transduction , Vitamin D/analogs & derivatives , Vitamin D/genetics , Vitamin D/pharmacology
15.
J Cell Physiol ; 227(6): 2668-76, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21898404

ABSTRACT

Osteoimmunology is an emerging field of research focused on the interaction of the immune system and bone. In this study we demonstrate that human osteoblasts are sensitive to the immune cytokine interferon (IFN)ß. Osteoblasts respond to IFNß as shown by the induction of several known IFN target genes such as interferon-induced (IFI) proteins (IFIT1, IFI44L), interferon-stimulated gene factor 3 (ISGF3) complex and the induction of IFNß itself. We demonstrated that IFNß has severe inhibitory effects on mineralization of osteoblast-derived extracellular matrix (ECM). Analysis of the timing of the IFNß effects revealed that committed osteoblasts in early stage of differentiation are most sensitive to IFNß inhibition of mineralization. A single IFNß treatment was as effective as multiple treatments. During the progress of differentiation osteoblasts become desensitized for IFNß. This pinpoints to a complex pattern of IFNß sensitivity in osteoblasts. Focusing on early osteoblasts, we showed that IFNß decreased gene expression of ECM-related genes, such as type I Collagen (COL1A1), fibronectin (FN1), fibullin (FBLN1), fibrillin (FBN2), and laminin (LAMA1). Additionally, ECM produced by IFNß-treated osteoblasts contained less collagen protein. IFNß stimulated gene expression of osteopontin (OPN), annexin2 (ANXA2), and hyaluronan synthase 1 (HAS1), which are important factors in the adhesion of hematopoietic stem cells (HSC) in the HSC niche. In conclusion, IFNß directly modifies human osteoblast function by inhibiting ECM synthesis eventually resulting in delayed bone formation and mineralization and induces a HSC niche supporting phenotype. These effects are highly dependent on timing of treatment in the early phase of osteoblast differentiation.


Subject(s)
Calcification, Physiologic , Cell Differentiation , Extracellular Matrix/metabolism , Interferon-beta/metabolism , Osteoblasts/metabolism , Calcification, Physiologic/genetics , Cell Differentiation/genetics , Cell Line , Extracellular Matrix/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Osteoblasts/immunology , Phenotype , RNA, Messenger/metabolism , Stem Cell Niche , Time Factors
16.
J Cell Physiol ; 225(2): 593-600, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20506116

ABSTRACT

1Alpha,25-dihydroxyitamin D(3) (1,25D3) deficiency leads to impaired bone mineralization. We used the human pre-osteoblastic cell line SV-HFO, which forms within 19 days of culture an extracellular matrix that starts to mineralize around day 12, to examine the mechanism by which 1,25D3 regulates osteoblasts and directly stimulates mineralization. Time phase studies showed that 1,25D3 treatment prior to the onset of mineralization, rather than during mineralization led to accelerated and enhanced mineralization. This is supported by the observation of unaltered stimulation by 1,25D3 even when osteoblasts were devitalized just prior to onset of mineralization and after 1,25D3 treatment. Gene Chip expression profiling identified the pre-mineralization and mineralization phase as two strongly distinctive transcriptional periods with only 0.6% overlap of genes regulated by 1,25D3. In neither phase 1,25D3 significantly altered expression of extracellular matrix genes. 1,25D3 significantly accelerated the production of mature matrix vesicles (MVs) in the pre-mineralization. Duration rather than timing determined the extent of the 1,25D3 effect. We propose the concept that besides indirect effects via intestinal calcium uptake 1,25D3 directly accelerates osteoblast-mediated mineralization via increased production of mature MVs in the period prior to mineralization. The accelerated deposition of mature MVs leads to an earlier onset and higher rate of mineralization. These effects are independent of changes in extracellular matrix protein composition. These data on 1,25D3, mineralization, and MV biology add new insights into the role of 1,25D3 in bone metabolism and emphasize the importance of MVs in bone and maintaining bone health and strength by optimal mineralization status.


Subject(s)
Bone Matrix/metabolism , Calcification, Physiologic/drug effects , Calcitriol/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Calcification, Physiologic/physiology , Calcium/metabolism , Cell Differentiation , Cell Line , DNA/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
J Endocrinol ; 188(1): 37-47, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16394173

ABSTRACT

Recent studies demonstrate widespread expression of ghrelin among tissues and have uncovered its pleiotropic nature. We have examined gene expression of ghrelin and its two receptor splice variants, growth hormone secretagogue receptors (GHS-R) 1a and 1b, in human bone biopsies and in the human pre-osteoblastic SV-HFO cell line during differentiation. Additionally, we examined proliferative effects of ghrelin and unacylated ghrelin (UAG) in differentiating and non-differentiating cells. We detected GHS-R1b mRNA in human bone and osteoblasts but not ghrelin's cognate receptor GHS-R1a, using two different real-time PCR assays and both total RNA and mRNA. In osteoblasts GHS-R1b mRNA expression remained low during the first 14 days of culture, but increased 300% in differentiating cells by day 21. Both human bone biopsies and osteoblasts expressed ghrelin mRNA, and osteoblasts were found to secrete ghrelin. Overall, ghrelin gene expression was greater in differentiating than non-differentiating osteoblasts, but was not increased during culture in either group. Ghrelin and UAG induced thymidine uptake dose-dependently, peaking at 1 and 10 nM respectively, at day 6 of culture in both non-differentiating and differentiating osteoblasts. The proliferative response to ghrelin and UAG declined with culture time and state of differentiation. The proliferative effects of ghrelin and UAG were suppressed by inhibitors of extracellular-signal-regulated kinase (ERK) and phosphoinositide-3 kinase, and both peptides rapidly induced ERK phosphorylation. Overall, our data suggest new roles for ghrelin and UAG in modulating human osteoblast proliferation via a novel signal transduction pathway.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/cytology , Peptide Hormones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Alkaline Phosphatase/metabolism , Analysis of Variance , Biomarkers/analysis , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , DNA/biosynthesis , Femur Head , Ghrelin , Humans , Osteoblasts/drug effects , Receptors, Ghrelin , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , Thymidine/analysis , Thymidine/metabolism
18.
J Endocrinol ; 180(3): 457-67, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15012600

ABSTRACT

Estrogens are essential for bone mass accrual but their role before sexual maturation has remained elusive. Using in situ hybridization and immunohistochemistry, we investigated the expression of both estrogen receptor (ER) alpha and beta mRNA and protein as well as several mRNAs coding for enzymes involved in sex steroid metabolism (aromatase, type I and II 17 beta-hydroxysteroid dehydrogenase (17 beta-HSD), steroid sulfatase (STS) and type I 5 alpha-reductase) on sections of tibial metaphyses before (1- and 4-week-old), during (7-week-old) and after (16-week-old) sexual maturation in female and male rats. ER alpha and ER beta mRNA and protein were detected in metaphyseal bone in lining cells, osteoblasts, osteoclasts and some osteocytes with no apparent differences in expression during development or between the sexes. In contrast, aromatase, type I and II 17 beta-HSD and type I 5 alpha-reductase mRNAs were first detected in osteoblasts, osteoclasts and occasionally in osteocytes from sexual maturation (7-week-old rat) and onwards. Only STS was present before sexual maturation. To study the significance of ER alpha and beta expression in bone before sexual maturation when circulating sex steroid levels are low, 26-day-old female and male rats underwent gonadectomy or 17 beta-estradiol (E(2)) supplementation (0.5 mg/21 days) during 3 weeks. Following gonadectomy, trabecular bone volume (TBV) was lower in males (P=0.03) and there was a trend towards reduction in females (P=0.057). E(2) supplementation increased tibial TBV compared with controls in both genders as assessed by Masson-Goldner staining. These data suggest that the presence of ERs in bone cells before sex maturation might be of significance for bone mass accrual. Furthermore, based on the mRNA expression of the crucial enzymes aromatase and type I 17 beta-HSD, we suggest that bone cells in the tibial metaphysis acquire the intrinsic capacity to metabolize sex steroids from sexual maturation onwards. This process may contribute to the beneficial effects of estrogen on bone mass accrual, possibly by intracrinology.


Subject(s)
Gonadal Steroid Hormones/metabolism , Growth Plate/metabolism , Receptors, Estrogen/analysis , Sexual Maturation/physiology , Tibia , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/analysis , Animals , Aromatase/analysis , Aromatase/genetics , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Growth Plate/drug effects , Hydroxysteroid Dehydrogenases/analysis , Hydroxysteroid Dehydrogenases/genetics , Immunohistochemistry/methods , In Situ Hybridization/methods , Male , Orchiectomy , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Estrogen/genetics , Steryl-Sulfatase/analysis , Steryl-Sulfatase/genetics
19.
Endocr Rev ; 24(6): 782-801, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14671005

ABSTRACT

The growth plate is the final target organ for longitudinal growth and results from chondrocyte proliferation and differentiation. During the first year of life, longitudinal growth rates are high, followed by a decade of modest longitudinal growth. The age at onset of puberty and the growth rate during the pubertal growth spurt (which occurs under the influence of estrogens and GH) contribute to sex difference in final height between boys and girls. At the end of puberty, growth plates fuse, thereby ceasing longitudinal growth. It has been recognized that receptors for many hormones such as estrogen, GH, and glucocorticoids are present in or on growth plate chondrocytes, suggesting that these hormones may influence processes in the growth plate directly. Moreover, many growth factors, i.e., IGF-I, Indian hedgehog, PTHrP, fibroblast growth factors, bone morphogenetic proteins, and vascular endothelial growth factor, are now considered as crucial regulators of chondrocyte proliferation and differentiation. In this review, we present an update on the present perception of growth plate function and the regulation of chondrocyte proliferation and differentiation by systemic and local regulators of which most are now related to human growth disorders.


Subject(s)
Growth Plate/physiology , Animals , Cell Division , Chondrocytes/cytology , Growth Plate/anatomy & histology , Growth Plate/growth & development , Growth Substances/physiology , Hormones/physiology , Humans , Parturition
20.
Horm Res ; 59(1): 35-41, 2003.
Article in English | MEDLINE | ID: mdl-12566733

ABSTRACT

AIM: To study the effect of irradiation on the longitudinal growth and the expression of parathyroid hormone-related peptide (PTHrP) and Indian hedgehog (IHh) in tibial growth plates of rats. METHODS: At 3 weeks of age, 30 male rats received a single fraction of irradiation (8 Gy) to their right hind limb, and small groups of animals were sacrificed 1, 2, 3, 5, 7, 10, 15, and 26 weeks after irradiation. Weight and length of both irradiated and nonirradiated tibiae were measured, and sections of the tibiae were stained with HE. PTHrP and IHh were visualized using immunohistochemical techniques. RESULTS: Radiation resulted in persistent growth delay of the irradiated tibiae, with a difference in length of more than 10% between the irradiated and the nonirradiated tibiae 15 weeks or more after irradiation. The growth plate architecture was disturbed, and the expression of both PTHrP and IHh was decreased in the irradiated tibiae. CONCLUSION: As PTHrP and IHh are key regulators of both the pace and the synchronization of the differentiation of growth plate chondrocytes, the reduced expression of PTHrP and IHh may contribute to the changes found after irradiation.


Subject(s)
Bone Development/radiation effects , Growth Plate/physiology , Growth Plate/radiation effects , Peptide Hormones/metabolism , Tibia/physiology , Tibia/radiation effects , Trans-Activators/metabolism , Animals , Growth Plate/metabolism , Growth Plate/pathology , Hedgehog Proteins , Male , Parathyroid Hormone-Related Protein , Rats , Rats, Wistar , Tibia/metabolism , Tibia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...