Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Calcif Tissue Int ; 114(3): 255-266, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38226986

RESUMO

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.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Perda Auditiva , Hiperparatireoidismo , Hipofosfatemia , Nefrocalcinose , Osteoartrite , Criança , Adulto , Humanos , Raquitismo Hipofosfatêmico Familiar/complicações , Raquitismo Hipofosfatêmico Familiar/genética , Raquitismo Hipofosfatêmico Familiar/diagnóstico , Nefrocalcinose/genética , Nefrocalcinose/complicações , Fatores de Crescimento de Fibroblastos/genética , Hipofosfatemia/epidemiologia , Hipofosfatemia/genética , Fosfatos , Hiperparatireoidismo/complicações , Obesidade/complicações , Perda Auditiva/complicações , Perda Auditiva/tratamento farmacológico
2.
J Cell Physiol ; 233(6): 4895-4906, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29194609

RESUMO

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.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Diferenciação Celular/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/genética , Diferenciação Celular/genética , Biologia Computacional , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Osteoblastos/metabolismo , Osteogênese/genética , Mapas de Interação de Proteínas , Transdução de Sinais/efeitos dos fármacos
3.
J Cell Physiol ; 228(11): 2167-74, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23589129

RESUMO

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.


Assuntos
Ativinas/biossíntese , Calcificação Fisiológica/efeitos dos fármacos , Osteoblastos/metabolismo , Vitamina D/análogos & derivados , Linhagem Celular , Folistatina/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lectinas Tipo C/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteocalcina/genética , Osteocalcina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad7/metabolismo , Vitamina D/farmacologia , Varfarina/farmacologia
4.
J Cell Physiol ; 227(9): 3258-66, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22105341

RESUMO

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.


Assuntos
Matriz Extracelular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon beta/genética , Osteoblastos/metabolismo , Vitamina D/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/genética , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Matriz Extracelular/metabolismo , Genes Dominantes , Humanos , Interferon beta/metabolismo , Interferon beta/farmacologia , Transdução de Sinais , Vitamina D/análogos & derivados , Vitamina D/genética , Vitamina D/farmacologia
5.
J Cell Physiol ; 227(6): 2668-76, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21898404

RESUMO

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.


Assuntos
Calcificação Fisiológica , Diferenciação Celular , Matriz Extracelular/metabolismo , Interferon beta/metabolismo , Osteoblastos/metabolismo , Calcificação Fisiológica/genética , Diferenciação Celular/genética , Linhagem Celular , Matriz Extracelular/genética , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Osteoblastos/imunologia , Fenótipo , RNA Mensageiro/metabolismo , Nicho de Células-Tronco , Fatores de Tempo
6.
Bone ; 164: 116526, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35995334

RESUMO

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.


Assuntos
Produtos Finais de Glicação Avançada , Osteogênese , Fosfatase Alcalina/metabolismo , Cálcio/metabolismo , Diferenciação Celular , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Produtos Finais de Glicação Avançada/farmacologia , Humanos , Osteoblastos/metabolismo , Aldeído Pirúvico/metabolismo , Aldeído Pirúvico/farmacologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo
7.
Bone ; 165: 116564, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36150657

RESUMO

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.


Assuntos
Fragilidade , Sarcopenia , Humanos , Camundongos , Animais , Pré-Escolar , Pessoa de Meia-Idade , Idoso , Fragilidade/epidemiologia , Sarcopenia/epidemiologia , Força da Mão , Produtos Finais de Glicação Avançada , Ingestão de Alimentos
8.
Endocr Connect ; 10(3): 273-282, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33543729

RESUMO

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.

9.
J Cell Physiol ; 225(2): 593-600, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20506116

RESUMO

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.


Assuntos
Matriz Óssea/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Calcitriol/farmacologia , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Calcificação Fisiológica/fisiologia , Cálcio/metabolismo , Diferenciação Celular , Linhagem Celular , DNA/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
Mater Today Bio ; 7: 100060, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32577614

RESUMO

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.

11.
Biofabrication ; 11(3): 035012, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30921774

RESUMO

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.


Assuntos
Cartilagem da Orelha/citologia , Elastase Pancreática/metabolismo , Adolescente , Idoso , Animais , Bovinos , Criança , Condrogênese , Elastina/metabolismo , Matriz Extracelular/química , Feminino , Glicosaminoglicanos/metabolismo , Humanos , Camundongos Nus , Pessoa de Meia-Idade , Alicerces Teciduais/química
12.
Endocr Rev ; 24(6): 782-801, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14671005

RESUMO

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.


Assuntos
Lâmina de Crescimento/fisiologia , Animais , Divisão Celular , Condrócitos/citologia , Lâmina de Crescimento/anatomia & histologia , Lâmina de Crescimento/crescimento & desenvolvimento , Substâncias de Crescimento/fisiologia , Hormônios/fisiologia , Humanos , Parto
13.
Bone ; 95: 108-114, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27845263

RESUMO

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.


Assuntos
Leucócitos Mononucleares/metabolismo , Osteoclastos/patologia , Osteogênese , Osteoporose/sangue , Osteoporose/patologia , Idoso , Reabsorção Óssea/patologia , Estudos de Casos e Controles , Diferenciação Celular , Feminino , Humanos , Osteoporose/tratamento farmacológico , Fraturas por Osteoporose/tratamento farmacológico , Fraturas por Osteoporose/patologia
14.
J Endocrinol ; 188(1): 37-47, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16394173

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoblastos/citologia , Hormônios Peptídicos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Fosfatase Alcalina/metabolismo , Análise de Variância , Biomarcadores/análise , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Cabeça do Fêmur , Grelina , Humanos , Osteoblastos/efeitos dos fármacos , Receptores de Grelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estimulação Química , Timidina/análise , Timidina/metabolismo
15.
Endocrinology ; 157(12): 4930-4942, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27911148

RESUMO

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.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Estradiol/administração & dosagem , Fêmur/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Feminino , Fêmur/metabolismo , Ovariectomia , Ratos , Esterol Esterase/genética , Esterol Esterase/metabolismo
16.
Endocrinology ; 143(10): 4048-55, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12239116

RESUMO

To assess whether growth plate-specific production of sex steroids is possible, we have surveyed the presence of several key-enzymes involved in androgen and estrogen metabolism in the tibial growth plate of female and male rats during development. Using in situ hybridization, mRNAs of aromatase p450, type I and II 17beta-hydroxysteroid dehydrogenase (HSD), steroid sulfatase (STS), and 5alpha-reductase were detected in proliferating and hypertrophic chondrocytes of the growth plate. The former three were strongly up-regulated around sexual maturation (7 wk), whereas the latter two were expressed at a relatively constant level during development. These data were supported by measuring aromatase, type I 17beta-HSD, and STS enzyme activities in chondrocytes collected from tibial growth plates at 1 and 7 wk of age. Of the enzymes studied, there were minor differences between the sexes in aromatase and 5alpha-reductase expression only. In conclusion, our findings clearly indicate the presence of various enzymes involved in sex steroid metabolism in the tibial growth plate, especially in sexually maturing rats, a timepoint at which sex steroids have major effects on longitudinal growth. Our data suggest that intracrinology in the rat growth plate can occur and may be a major source of local sex steroid delivery.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Lâmina de Crescimento/metabolismo , Tíbia/metabolismo , 17-Hidroxiesteroide Desidrogenases/genética , 17-Hidroxiesteroide Desidrogenases/metabolismo , Envelhecimento/metabolismo , Animais , Aromatase/genética , Aromatase/metabolismo , Arilsulfatases/genética , Arilsulfatases/metabolismo , Feminino , Hibridização In Situ , Masculino , RNA Mensageiro , Ratos , Ratos Wistar , Esteril-Sulfatase
17.
Bone ; 30(3): 478-85, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11882461

RESUMO

In this study we examine the spatial and temporal expression of estrogen receptor (ER) alpha and beta mRNA and protein in the tibial growth plate of the rat after birth, as well as the hormonal regulation of their expression. Using in situ hybridization and immunohistochemistry, we demonstrated ER alpha and ER beta mRNA and protein in tibial growth plates from 1 to 40 weeks after birth. ER alpha and beta mRNA and protein were localized in late proliferating and early hypertrophic chondrocytes during early life (1 and 4 weeks of age), whereas the immunohistochemistry also showed staining for ER alpha and beta in the resting cells. A similar expression pattern was observed during sexual maturation (7 weeks of age) except that ER beta mRNA was also detected in early proliferating chondrocytes. After sexual maturation (from 12 up to 40 weeks of age) ER alpha and beta mRNA and protein expression was confined to late proliferating and early hypertrophic chondrocytes. Apart from a relatively higher ER alpha mRNA expression in males after sexual maturation, we did not detect differences in expression of ERs between genders. Expression of ER beta mRNA in epiphyseal plates was increased in growth-retarded hypophysectomized rats compared with controls. Administration of growth hormone (GH) did not reverse the increased ER expression to normal. These data suggest that ER alpha and beta are coexpressed in growth plates of the rat after birth and that the level of expression of ERs in these tissues is hormonally regulated. Furthermore, our data indicate that the absence of growth-plate closure in the rat cannot be explained by disappearance of ER alpha expression during sexual maturation per se.


Assuntos
Lâmina de Crescimento/metabolismo , Receptores de Estrogênio/biossíntese , Animais , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hormônio do Crescimento/farmacologia , Hormônio do Crescimento/fisiologia , Lâmina de Crescimento/efeitos dos fármacos , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar
18.
Bone ; 30(6): 891-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12052459

RESUMO

In this study, we investigate the expression of the androgen receptor (AR) in the tibial growth plate and metaphyseal bone of male and female rats at the mRNA and protein level. Using in situ hybridization and immunohistochemistry, AR mRNA and protein were demonstrated in proliferating and early hypertrophic chondrocytes in the growth plate of 1-, 4-, and 7-week-old male and female rats. Immunostaining for AR was observed both in the nucleus and the cytoplasm. After sexual maturation at 12 and 16 weeks of age, AR expression decreased in both genders and was confined to a small rim of prehypertrophic chondrocytes. In female rats of 40 weeks of age, this expression pattern was still visible. In most age groups there was a tendency toward an increased AR mRNA expression in male vs. female rats except in the 7-week-old animals. At the protein level, sexually maturing 7-week-old male rats demonstrated a higher staining intensity compared to their female counterparts. At this stage, AR staining in the males was mainly confined to the nucleus, whereas in females staining was predominantly found in the cytoplasm. In the tibial metaphysis, AR mRNA was detected in lining cells, osteoblasts, osteocytes, and osteoclasts at all stages of development. At the protein level, a similar expression pattern was observed, except for an absence of immunostaining in the lining cells. The staining was both nuclear and cytoplasmic. In most age groups, mRNA and protein signals were higher in males compared with females. We have demonstrated the presence of AR mRNA and protein in the tibial growth plate and the underlying metaphyseal bone during development of the rat. In male rats, the presence of higher messenger and protein staining intensities, as well as preferential nuclear staining during sexual maturation, suggests that direct actions of androgens in chondrocytes and in bone forming cells may be involved in establishing the gender differences in the skeleton.


Assuntos
Lâmina de Crescimento/metabolismo , Receptores Androgênicos/biossíntese , Caracteres Sexuais , Tíbia/metabolismo , Animais , Epífises/química , Epífises/metabolismo , Epífises/fisiologia , Feminino , Expressão Gênica/fisiologia , Lâmina de Crescimento/química , Lâmina de Crescimento/fisiologia , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Receptores Androgênicos/análise , Maturidade Sexual/fisiologia , Tíbia/química , Tíbia/fisiologia
19.
J Endocrinol ; 180(3): 457-67, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15012600

RESUMO

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.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Lâmina de Crescimento/metabolismo , Receptores de Estrogênio/análise , Maturidade Sexual/fisiologia , Tíbia , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/análise , Animais , Aromatase/análise , Aromatase/genética , Estradiol/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Lâmina de Crescimento/efeitos dos fármacos , Hidroxiesteroide Desidrogenases/análise , Hidroxiesteroide Desidrogenases/genética , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Masculino , Orquiectomia , RNA Mensageiro/análise , Ratos , Ratos Wistar , Receptores de Estrogênio/genética , Esteril-Sulfatase/análise , Esteril-Sulfatase/genética
20.
J Endocrinol ; 175(2): 277-88, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12429026

RESUMO

Recently, both estrogen receptor (ER) alpha and beta were detected in growth plate chondrocytes of rats before sexual maturation, implying a role for estrogen at this stage. In this study, therefore, we investigated the effects of ovariectomy (OVX) or estrogen supplementation on parameters of longitudinal growth in 26-day-old rats, which were sexually immature at the start of the experiment. OVX caused an increase in body weight gain, tibial length and growth plate width due to an increased proliferating zone. This increase correlated with an increase in cell number, with a decrease in cell diameter and with increased proliferating cell nuclear antigen (PCNA) immunostaining compared with sham. Interestingly, the increase in proliferation was not caused by an increase in insulin-like growth factor-I (IGF-I) mRNA expression in the growth plate as assessed by real-time PCR. In contrast to OVX, 17beta-estradiol (E(2)) supplementation (0.5 mg/21 days) of 26-day-old female rats caused a strong decrease in body weight gain, tibial length and growth plate width. The latter was explained by a reduction of the proliferating zone width, which correlated with a reduced number of PCNA-positive cells (not significant) and by a reduction of the hypertrophic zone width. In male rats supplemented with E(2), similar effects were observed compared with the females. ERalpha and beta immunostaining was found predominantly in late proliferating and early hypertrophic chondrocytes. OVX did not affect ER expression but E(2) supplementation strongly decreased immunostaining for both ERalpha and beta in both sexes. Besides E(2), desoxyestrone (DE), an activator of nongenomic estrogen-like signaling (ANGEL) and 2-methoxyestradiol (2-MeO-E(2)), a tissue-selective naturally occurring metabolite of E(2), were administered to female and male rats of the same age. Compared with E(2), these compounds had less pronounced, though significant, effects on some parameters of longitudinal growth in both sexes, especially on growth plate characteristics. In conclusion, E(2) may exert effects on longitudinal growth before and at the onset of sexual maturation, despite very low endogenous serum levels at these stages. There may be a role for nongenomic signaling in body weight gain, tibial length and growth plate width but genomic signaling prevails.


Assuntos
Estradiol/análogos & derivados , Estrogênios/fisiologia , Lâmina de Crescimento/fisiologia , Maturidade Sexual/fisiologia , 2-Metoxiestradiol , Animais , Peso Corporal/fisiologia , Estradiol/fisiologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Estrogênios/administração & dosagem , Feminino , Masculino , Ovariectomia , Reação em Cadeia da Polimerase , Antígeno Nuclear de Célula em Proliferação/fisiologia , Ratos , Ratos Wistar , Receptores de Estrogênio/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa