Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Eur J Endocrinol ; 189(4): 448-459, 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37796032

ABSTRACT

Fibroblast growth factor 23 (FGF23) is produced and secreted by osteocytes and is essential for maintaining phosphate homeostasis. One of the main regulators of FGF23, 1,25-dihydroxyvitamin D (1,25(OH)2D3), is primarily synthesized in the kidney from 25-hydroxyvitamin D (25(OH)D) by 1α-hydroxylase (encoded by CYP27B1). Hitherto, it is unclear whether osteocytes can convert 25(OH)D and thereby allow for 1,25(OH)2D3 to induce FGF23 production and secretion locally. Here, we differentiated MC3T3-E1 cells toward osteocyte-like cells expressing and secreting FGF23. Treatment with 10-6 M 25(OH)D resulted in conversion of 25(OH)D to 150 pmol/L 1,25(OH)2D3 and increased FGF23 expression and secretion, but the converted amount of 1,25(OH)2D3 was insufficient to trigger an FGF23 response, so the effect on FGF23 was most likely directly caused by 25(OH)D. Interestingly, combining phosphate with 25(OH)D resulted in a synergistic increase in FGF23 expression and secretion, likely due to activation of additional signaling pathways by phosphate. Blockage of the vitamin D receptor (VDR) only partially abolished the effects of 25(OH)D or 25(OH)D combined with phosphate on Fgf23, while completely inhibiting the upregulation of cytochrome P450 family 24 subfamily A member 1 (Cyp24a1), encoding for 24-hydroxylase. RNA sequencing and in silico analyses showed that this could potentially be mediated by the nuclear receptors Retinoic Acid Receptor ß (RARB) and Estrogen Receptor 2 (ESR2). Taken together, we demonstrate that osteocytes are able to convert 25(OH)D to 1,25(OH)2D3, but this is insufficient for FGF23 activation, implicating a direct effect of 25(OH)D in the regulation of FGF23, which occurs at least partially independent from its cognate VDR. Moreover, phosphate and 25(OH)D synergistically increase expression and secretion of FGF23, which warrants investigating consequences in patients receiving a combination of vitamin D analogues and phosphate supplements. These observations help us to further understand the complex relations between phosphate, vitamin D, and FGF23.


Subject(s)
Calcitriol , Osteocytes , Humans , Calcifediol , Calcitriol/pharmacology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Mixed Function Oxygenases , Osteocytes/metabolism , Phosphates , Receptors, Calcitriol/genetics , Vitamin D/pharmacology , Animals , Mice
2.
Front Endocrinol (Lausanne) ; 14: 1121727, 2023.
Article in English | MEDLINE | ID: mdl-37293482

ABSTRACT

The skeleton is an organ of dual functionality; on the one hand, it provides protection and structural competence. On the other hand, it participates extensively in coordinating homeostasis globally given that it is a mineral and hormonal reservoir. Bone is the only tissue in the body that goes through strategically consistent bouts of bone resorption to ensure its integrity and organismal survival in a temporally and spatially coordinated process, known as bone remodeling. Bone remodeling is directly enacted by three skeletal cell types, osteoclasts, osteoblasts, and osteocytes; these cells represent the acting force in a basic multicellular unit and ensure bone health maintenance. The osteocyte is an excellent mechanosensory cell and has been positioned as the choreographer of bone remodeling. It is, therefore, not surprising that a holistic grasp of the osteocyte entity in the bone is warranted. This review discusses osteocytogenesis and associated molecular and morphological changes and describes the osteocytic lacunocanalicular network (LCN) and its organization. We highlight new knowledge obtained from transcriptomic analyses of osteocytes and discuss the regulatory role of osteocytes in promoting osteoclastogenesis with an emphasis on the case of osteoclastogenesis in anosteocytic bones. We arrive at the conclusion that osteocytes exhibit several redundant means through which osteoclast formation can be initiated. However, whether osteocytes are true "orchestrators of bone remodeling" cannot be verified from the animal models used to study osteocyte biology in vivo. Results from studying osteocyte biology using current animal models should come with the caveat that these models are not osteocyte-specific, and conclusions from these studies should be interpreted cautiously.


Subject(s)
Bone Resorption , Osteogenesis , Animals , Osteoclasts/metabolism , Bone and Bones/metabolism , Bone Resorption/metabolism , Osteocytes/metabolism
3.
Biomed Pharmacother ; 162: 114403, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37003034

ABSTRACT

OBJECTIVES: In the field of orthopedics, osteonecrosis of the femoral head (ONFH) is a common and refractory condition sometimes known as "immortal cancer" due to its complicated etiology, difficult treatment, and high disability rate. This paper's main goal is to examine the most recent literature on the pro-apoptotic effects of traditional Chinese medicine TCM monomers or compounds on osteocytes and to provide a summary of the potential signal routes. METHODS: The last ten years' worth of literature on ONFH as well as the anti-ONFH effects of aqueous extracts and monomers from traditional Chinese medicine were compiled. CONCLUSIONS: When all the relevant signal pathways are considered, the key apoptotic routes include those mediated by the mitochondrial pathway, the MAPK signaling pathway, the PI3K/Akt signaling pathway, the Wnt/-catenin signaling pathway, the HIF-1 signaling network, etc. As a result, we anticipate that this study will shed light on the value of TCM and its constituent parts for treating ONFH by inducing apoptosis in osteocytes and offer some guidance for the future development of innovative medications as anti-ONFH medications in clinical settings.


Subject(s)
Femur Head Necrosis , Osteonecrosis , Humans , Osteocytes/metabolism , Femur Head , Phosphatidylinositol 3-Kinases/metabolism , Osteonecrosis/metabolism , Femur Head Necrosis/metabolism , Wnt Signaling Pathway , Apoptosis
4.
J Clin Invest ; 133(3)2023 02 01.
Article in English | MEDLINE | ID: mdl-36512405

ABSTRACT

Disuse osteoporosis is a metabolic bone disease resulting from skeletal unloading (e.g., during extended bed rest, limb immobilization, and spaceflight), and the slow and insufficient bone recovery during reambulation remains an unresolved medical challenge. Here, we demonstrated that loading-induced increase in bone architecture/strength was suppressed in skeletons previously exposed to unloading. This reduction in bone mechanosensitivity was directly associated with attenuated osteocytic Ca2+ oscillatory dynamics. The unloading-induced compromised osteocytic Ca2+ response to reloading resulted from the HIF-1α/PDK1 axis-mediated increase in glycolysis, and a subsequent reduction in ATP synthesis. HIF-1α also transcriptionally induced substantial glutaminase 2 expression and thereby glutamine addiction in osteocytes. Inhibition of glycolysis by blockade of PDK1 or glutamine supplementation restored the mechanosensitivity in those skeletons with previous unloading by fueling the tricarboxylic acid cycle and rescuing subsequent Ca2+ oscillations in osteocytes. Thus, we provide mechanistic insight into disuse-induced deterioration of bone mechanosensitivity and a promising therapeutic approach to accelerate bone recovery after long-duration disuse.


Subject(s)
Calcium , Glutamine , Calcium/metabolism , Glutamine/pharmacology , Glutamine/metabolism , Osteocytes/metabolism , Glucose/metabolism , Energy Metabolism
5.
Front Endocrinol (Lausanne) ; 13: 1005189, 2022.
Article in English | MEDLINE | ID: mdl-36246908

ABSTRACT

Since phosphorus is a component of hydroxyapatite, its prolonged deprivation affects bone mineralization. Fibroblast growth factor 23 (FGF23) is essential for maintaining phosphate homeostasis and is mainly produced by osteocytes. FGF23 increases the excretion of inorganic phosphate (Pi) and decreases the production of 1,25-dihydroxyvitamin D in the kidneys. Osteocytes are cells of osteoblastic lineage that have undergone terminal differentiation and become embedded in mineralized bone matrix. Osteocytes express FGF23 and other multiple genes responsible for hereditary hypophosphatemic rickets, which include phosphate-regulating gene homologous to endopeptidase on X chromosome (PHEX), dentin matrix protein 1 (DMP1), and family with sequence similarity 20, member C (FAM20C). Since inactivating mutations in PHEX, DMP1, and FAM20C boost the production of FGF23, these molecules might be considered as local negative regulators of FGF23. Mouse studies have suggested that enhanced FGF receptor (FGFR) signaling is involved in the overproduction of FGF23 in PHEX-deficient X-linked hypophosphatemic rickets (XLH) and DMP1-deficient autosomal recessive hypophosphatemic rickets type 1. Since FGFR is involved in the transduction of signals evoked by extracellular Pi, Pi sensing in osteocytes may be abnormal in these diseases. Serum levels of sclerostin, an inhibitor Wnt/ß-catenin signaling secreted by osteocytes, are increased in XLH patients, and mouse studies have suggested the potential of inhibiting sclerostin as a new therapeutic option for the disease. The elucidation of complex abnormalities in the osteocytes of FGF23-related hypophosphatemic diseases will provide a more detailed understanding of their pathogenesis and more effective treatments.


Subject(s)
Familial Hypophosphatemic Rickets , Rickets, Hypophosphatemic , Animals , Calcium-Binding Proteins/metabolism , Endopeptidases/metabolism , Extracellular Matrix Proteins/genetics , Familial Hypophosphatemic Rickets/genetics , Fibroblast Growth Factors/metabolism , Hydroxyapatites/metabolism , Mice , Osteocytes/metabolism , Phosphates , Phosphorus/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Rickets, Hypophosphatemic/metabolism , beta Catenin/metabolism
6.
Sci Rep ; 11(1): 12258, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112892

ABSTRACT

Glucocorticoids (GCs), such as prednisolone, are widely used to treat inflammatory diseases. Continuously long-term or high dose treatment with GCs is one of the most common causes of secondary osteoporosis and is associated with sarcopenia and increased risk of debilitating osteoporotic fragility fractures. Abaloparatide (ABL) is a potent parathyroid hormone-related peptide analog, which can increase bone mineral density (aBMD), improve trabecular microarchitecture, and increase bone strength. The present study aimed to investigate whether GC excess blunts the osteoanabolic effect of ABL. Sixty 12-13-week-old female RjOrl:SWISS mice were allocated to the following groups: Baseline, Control, ABL, GC, and GC + ABL. ABL was administered as subcutaneous injections (100 µg/kg), while GC was delivered by subcutaneous implantation of a 60-days slow-release prednisolone-pellet (10 mg). The study lasted four weeks. GC induced a substantial reduction in muscle mass, trabecular mineral apposition rate (MAR) and bone formation rate (BFR/BS), and endocortical MAR compared with Control, but did not alter the trabecular microarchitecture or bone strength. In mice not receiving GC, ABL increased aBMD, bone mineral content (BMC), cortical and trabecular microarchitecture, mineralizing surface (MS/BS), MAR, BFR/BS, and bone strength compared with Control. However, when administered concomitantly with GC, the osteoanabolic effect of ABL on BMC, cortical morphology, and cortical bone strength was blunted. In conclusion, at cortical bone sites, the osteoanabolic effect of ABL is generally blunted by short-term GC excess.


Subject(s)
Bone Density/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Femur , Glucocorticoids/administration & dosage , Parathyroid Hormone-Related Protein/pharmacology , Adipocytes/metabolism , Animals , Biomarkers , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/metabolism , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Immunohistochemistry , Mechanical Phenomena , Mice , Osteocytes/metabolism , Osteogenesis/drug effects , Osteoporosis/drug therapy , Osteoporosis/etiology , Osteoporosis/metabolism , X-Ray Microtomography
7.
Arch Biochem Biophys ; 685: 108333, 2020 05 30.
Article in English | MEDLINE | ID: mdl-32194044

ABSTRACT

This study summarizes the available evidence from systematic reviews on the in vitro effects of photobiomodulation on the proliferation and differentiation of human bone and stromal cells by appraising their methodological quality. Improvements for future studies are also highlighted, with particular emphasis on in vitro protocols and cell-related characteristics. Six reviews using explicit eligibility criteria and methods selected in order to minimize bias were included. There was no compelling evidence on the cellular mechanisms of action or treatment parameters of photobiomodulation; compliance with quality assessment was poor. A rigorous description of laser parameters (wavelength, power, beam spot size, power density, energy density, repetition rate, pulse duration or duty cycle, exposure duration, frequency of treatments, and total radiant energy), exposure conditions (methods to ensure a uniform irradiation and to avoid cross-irradiation, laser-cell culture surface distance, lid presence during irradiation) and cell-related characteristics (cell type or line, isolation and culture conditions, donor-related factors where applicable, tissue source, cell phenotype, cell density, number of cell passages in culture) should be included among eligibility criteria for study inclusion. These methodological improvements will maximize the contribution of in vitro studies on the effects of photobiomodulation on human bone and stromal cells to evidence-based translational research.


Subject(s)
Low-Level Light Therapy , Osteocytes/metabolism , Stromal Cells/metabolism , Animals , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Humans , Osteocytes/radiation effects , Stromal Cells/radiation effects , Systematic Reviews as Topic
8.
Aging Cell ; 19(2): e13095, 2020 02.
Article in English | MEDLINE | ID: mdl-31880094

ABSTRACT

To determine whether 1,25-dihydroxyvitamin D (1,25(OH)2 D) can exert an anti-osteoporosis role through anti-aging mechanisms, we analyzed the bone phenotype of mice with 1,25(OH)2 D deficiency due to deletion of the enzyme, 25-hydroxyvitamin D 1α-hydroxylase, while on a rescue diet. 1,25(OH)2 D deficiency accelerated age-related bone loss by activating the p16/p19 senescence signaling pathway, inhibiting osteoblastic bone formation, and stimulating osteoclastic bone resorption, osteocyte senescence, and senescence-associated secretory phenotype (SASP). Supplementation of exogenous 1,25(OH)2 D3 corrected the osteoporotic phenotype caused by 1,25(OH)2 D deficiency or natural aging by inhibiting the p16/p19 pathway. The proliferation, osteogenic differentiation, and ectopic bone formation of bone marrow mesenchymal stem cells derived from mice with genetically induced deficiency of the vitamin D receptor (VDR) were significantly reduced by mechanisms including increased oxidative stress, DNA damage, and cellular senescence. We also demonstrated that p16 deletion largely rescued the osteoporotic phenotype caused by 1,25(OH)2 D3 deficiency, whereas 1,25(OH)2 D3 could up-regulate the enzyme Ezh2 via VDR-mediated transcription thereby enriching H3K27me3 and repressing p16/p19 transcription. Finally, we demonstrated that treatment with 1,25(OH)2 D3 improved the osteogenic defects of human BM-MSCs caused by repeated passages by stimulating their proliferation and inhibiting their senescence via the VDR-Ezh2-p16 axis. The results of this study therefore indicate that 1,25(OH)2 D3 plays a role in preventing age-related osteoporosis by up-regulating Ezh2 via VDR-mediated transcription, increasing H3K27me3 and repressing p16 transcription, thus promoting the proliferation and osteogenesis of BM-MSCs and inhibiting their senescence, while also stimulating osteoblastic bone formation, and inhibiting osteocyte senescence, SASP, and osteoclastic bone resorption.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Mesenchymal Stem Cells/drug effects , Osteoporosis/drug therapy , Receptors, Calcitriol/metabolism , Vitamin D/analogs & derivatives , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Aging/genetics , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/physiopathology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Damage/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Histones/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Osteocytes/drug effects , Osteocytes/metabolism , Osteogenesis/genetics , Osteoporosis/enzymology , Osteoporosis/metabolism , Osteoporosis/physiopathology , Oxidative Stress/genetics , Receptors, Calcitriol/genetics , Vitamin D/pharmacology , Vitamin D/therapeutic use
9.
Pediatr Nephrol ; 35(10): 1843-1854, 2020 10.
Article in English | MEDLINE | ID: mdl-31392510

ABSTRACT

Hypophosphatemic rickets is caused by renal phosphate wasting that is most commonly due to X-linked dominant mutations in PHEX. PHEX mutations cause hypophosphatemia indirectly, through the increased expression of fibroblast growth factor 23 (FGF23) by osteocytes. FGF23 decreases renal phosphate reabsorption and thereby increases phosphate excretion. The lack of phosphate leads to a mineralization defect at the level of growth plates (rickets), bone tissue (osteomalacia), and teeth, where the defect facilitates the formation of abscesses. The bone tissue immediately adjacent to osteocytes often remains unmineralized ("periosteocytic lesions"), highlighting the osteocyte defect in this disorder. Common clinical features of XLH include deformities of the lower extremities, short stature, enthesopathies, dental abscesses, as well as skull abnormalities such as craniosynostosis and Chiari I malformation. For the past four decades, XLH has been treated by oral phosphate supplementation and calcitriol, which improves rickets and osteomalacia and the dental manifestations, but often does not resolve all aspects of the mineralization defects. A newer treatment approach using inactivating FGF23 antibodies leads to more stable control of serum inorganic phosphorus levels and seems to heal rickets more reliably. However, the long-term benefits of FGF23 antibody treatment remain to be elucidated.


Subject(s)
Familial Hypophosphatemic Rickets/pathology , Fibroblast Growth Factors/metabolism , Osteomalacia/pathology , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Phosphates/metabolism , Absorptiometry, Photon , Bone Development/drug effects , Bone Development/genetics , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Calcitriol/administration & dosage , Familial Hypophosphatemic Rickets/diagnosis , Familial Hypophosphatemic Rickets/drug therapy , Familial Hypophosphatemic Rickets/genetics , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/antagonists & inhibitors , Humans , Osteocytes/metabolism , Osteomalacia/diagnosis , Osteomalacia/drug therapy , Osteomalacia/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Paracrine Communication/genetics , Phosphates/administration & dosage , Phosphates/blood , Renal Reabsorption/drug effects , Renal Reabsorption/genetics , Tooth/growth & development , Tooth/pathology , Treatment Outcome
10.
Article in English | MEDLINE | ID: mdl-31505801

ABSTRACT

A positive association between metabolic syndrome (MetS) and osteoporosis has been demonstrated in previous animal studies. The mechanisms of MetS in orchestrating the bone remodelling process have traditionally focused on the interactions between mature osteoblasts and osteoclasts, while the role of osteocytes is unexplored. Our earlier studies demonstrated the bone-promoting effects of tocotrienol using a rat model of osteoporosis induced by MetS. This study aimed to investigate the expression of osteocyte-derived peptides in the bone of rats with MetS-induced osteoporosis treated with tocotrienol. Age-matched male Wistar rats (12-week-old; n = 42) were divided into seven experimental groups. Two groups served as the baseline and normal group, respectively. The other five groups were fed with a high-carbohydrate high-fat (HCHF) diet to induce MetS. The five groups of HCHF animals were treated with tocopherol-stripped corn oil (vehicle), annatto tocotrienol (60 and 100 mg/kg), and palm tocotrienol (60 and 100 mg/kg) starting from week 8. At the end of the study, the rats were sacrificed and their right tibias were harvested. Protein was extracted from the metaphyseal region of the proximal right tibia and levels of bone peptides, including osteoprotegerin (OPG), soluble receptor activator of nuclear factor-kappa B ligand (sRANKL), sclerostin (SOST), Dickkopf-related protein 1 (DKK-1), fibroblast growth factor-23 (FGF-23), and parathyroid hormone (PTH), were measured. The vehicle-treated animals displayed higher levels of sRANKL, SOST, DKK-1, FGF-23, and PTH as compared to the normal animals. Oral supplementation of annatto and palm tocotrienol (60 and 100 mg/kg) reduced the levels of sRANKL and FGF-23 in the HCHF animals. Only 100 mg/kg annatto and palm tocotrienol lowered SOST and DKK-1 levels in the HCHF animals. In conclusion, tocotrienol exerts potential skeletal-promoting benefit by modulating the levels of osteocytes-derived bone-related peptides.


Subject(s)
Metabolic Syndrome/metabolism , Osteoporosis/metabolism , Tibia/drug effects , Tocotrienols/pharmacology , Animals , Bixaceae , Bone Morphogenetic Proteins/metabolism , Carotenoids , Disease Models, Animal , Fibroblast Growth Factors/metabolism , Genetic Markers , Intercellular Signaling Peptides and Proteins/metabolism , Male , Metabolic Syndrome/complications , Osteocytes/drug effects , Osteocytes/metabolism , Osteoporosis/etiology , Osteoprotegerin/metabolism , Parathyroid Hormone/metabolism , Plant Extracts , RANK Ligand/metabolism , Rats, Wistar , Tibia/metabolism
11.
Toxicol Sci ; 170(1): 199-209, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31120128

ABSTRACT

Once absorbed in the body, natural uranium [U(VI)], a radionucleotide naturally present in the environment, is targeted to the skeleton which is the long-term storage organ. We and others have reported the U(VI) negative effects on osteoblasts (OB) and osteoclasts (OC), the main two cell types involved in bone remodeling. In the present work, we addressed the U(VI) effect on osteocytes (OST), the longest living bone cell type and the more numerous (> 90%). These cells, which are embedded in bone matrix and thus are the more prone to U(VI) long-term exposure, are now considered as the chief orchestrators of the bone remodeling process. Our results show that the cytotoxicity index of OST is close to 730 µM, which is about twice the one reported for OB and OC. However, despite this resistance potential, we observed that chronic U(VI) exposure as low as 5 µM led to a drastic decrease of the OST mineralization function. Gene expression analysis showed that this impairment could potentially be linked to an altered differentiation process of these cells. We also observed that U(VI) was able to trigger autophagy, a highly conserved survival mechanism. Extended X-ray absorption fine structure analysis at the U LIII edge of OST cells exposed to U(VI) unambiguously shows the formation of an uranyl phosphate phase in which the uranyl local structure is similar to the one present in Autunite. Thus, our results demonstrate for the first time that OST mineralization function can be affected by U(VI) exposure as low as 5 µM, suggesting that prolonged exposure could alter the central role of these cells in the bone environment.


Subject(s)
Autophagy/drug effects , Gene Expression/drug effects , Organometallic Compounds/toxicity , Osteocytes/drug effects , Uranium/toxicity , Animals , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Mice , Osteocytes/metabolism , Osteocytes/ultrastructure
12.
Folia Morphol (Warsz) ; 78(4): 789-797, 2019.
Article in English | MEDLINE | ID: mdl-30949994

ABSTRACT

BACKGROUND: Osteoporosis results with the imbalance between osteoblastic formation and osteoclastic resorption, resulting in susceptibility to bone fractures. Ovariectomy leads to osteoporosis by triggering alterations in bone formation and structure. Tamoxifen as an anti-oestrogen is used for adjuvant therapy especially in metastatic diseases and known to have a bone mass protective effect after ovariectomy. MATERIALS AND METHODS: An animal model of ovariectomy induced osteoporosis after tamoxifen citrate administration was studied via biochemical and immunohistochemical methods. Female Wistar albino rats (n = 45), selected according to their oestrous cycle, were divided into three groups; I - control, II - ovariectomy, III - ovariectomy + tamoxifen. Following ovariectomy, tamoxifen citrate (10 mg/kg) was given intraperitoneally daily for 8 weeks. At the end of the period, animals were sacrificed under anaesthesia, blood samples were taken to measure oestrogen, calcium, and alkaline phosphate. Tibia bone samples were fixed in formalin solution and decalcified with 5% ethylene-diamine tetra acetic acid. After the routine histological follow up, samples were embedded in paraffin and cut with a microtome for semi-thin sections. Primary antibodies osteonectin and osteopontin were applied to sections and examined under light microscope. RESULTS: As a consequence, when oestrogen and calcium data were compared there was a decrease in ovariectomy group with an increase in alkaline phosphatase. In ovariectomy + tamoxifen group, these values were close to the control group. Osteonectin was observed to promote bone formation by influencing collagen fibre formation, extracellular matrix development, osteoblast differentiation and the capacity to affect osteoclast activity. CONCLUSIONS: It has been suggested that osteopontin, the cytokine and cell binding protein, stimulates cellular signalling pathways, induces bone remodelling and acts in osteoporosis.


Subject(s)
Bone Remodeling , Osteogenesis , Ovariectomy , Tamoxifen/pharmacology , Animals , Bone Remodeling/drug effects , Female , Immunohistochemistry , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocytes/drug effects , Osteocytes/metabolism , Osteogenesis/drug effects , Osteonectin/metabolism , Rats
13.
FEBS Open Bio ; 9(6): 1082-1096, 2019 06.
Article in English | MEDLINE | ID: mdl-31006177

ABSTRACT

Oxidative stress and abnormal osteocyte apoptosis are often related to dysregulation of bone turnover and chronic bone loss, and so fruit and vegetables with high antioxidant potential may play an important role in the prevention and/or management of osteoporosis. Osteocytes are the main regulators of bone remodelling. For the first time, we demonstrate here that blueberry juice (BJ), obtained from Vaccinium myrtillus, rich in polyphenols, shows antioxidant and antiosteoclastogenic properties in MLO-Y4 osteocytes. We report that BJ prevents oxidative stress-induced apoptosis and reverses the increase in receptor activator of nuclear factor κB ligand and sclerostin expression, crucial factors for osteoclast activation and bone resorption. BJ is also able to prevent oxidative stress-induced cell cytotoxicity in bone marrow mesenchymal stromal cells (MSCs), which are considered to be an important tool for cell therapy in bone disorders. No significant difference in preventing these events was observed between BJ and blueberry dry extract containing equal amounts of total soluble polyphenols. We have also shown that blueberry acts as both an antioxidant and an activator of sirtuin type 1, a class III histone deacetylase involved in cell death regulation and considered a molecular target for blocking bone resorption without affecting osteoclast survival. Overall, these novel data obtained in osteocytes and MSCs may help us clarify the mechanisms by which blueberry counteracts oxidative stress-induced damage in bone remodelling and osteogenesis at the cellular and molecular level. Our findings are consistent with the reported beneficial effects of blueberry on bone tissue reported in animal studies, which suggest that blueberry may be a useful supplement for the prevention and/or management of osteoporosis and osteogenic process.


Subject(s)
Antioxidants/pharmacology , Blueberry Plants/chemistry , Fruit and Vegetable Juices , Mesenchymal Stem Cells/metabolism , Osteocytes/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Sirtuin 1/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Fruit/chemistry , Humans , Mice , RANK Ligand/metabolism , Reactive Oxygen Species/metabolism
14.
Ann Clin Lab Sci ; 49(1): 63-71, 2019 01.
Article in English | MEDLINE | ID: mdl-30814079

ABSTRACT

This study aimed to evaluate the use of Insulin-Transferrin-Selenium (ITS) medium in place of fetal bovine serum (FBS) to culture human amnion mesenchymal stem cells (hAMSCs). Cell morphology, ultrastructure, proliferation, migration and MSC related markers were assessed accordingly. The hAMSCs were induced to osteocyte, chondrocyte, adipocyte and keratinocyte by culturing in appropriate induction medium. hAMSCs mRNA expression was detected for the matrix metalloproteinases 2 (MMP2), keratinocyte growth factor (KGF), vascular endothelial growth factor (VEGF), insulin-like growth factor-I (IGF-I), Platelet-derived Growth Factor (PDGF), and transforming growth factor beta 1 (TGF-ß) by real-time quantitative RT-PCR. Our results showed that hAMSCs cultured in ITS medium exhibited similar proliferation rates, demonstrated a statistically significant increased migration and expressed similar levels of MSC markers(CD73+, CD90+, CD105+, CD45-, CD34-) compared with those cultured in FBS. Osteoblasts, chondrocytes, adipocytes and keratinocytes were differentiated. Results of transmission electron microscope (TEM) revealed that hAMSCs cultured in ITS medium underwent active metabolism. The mRNA expression of MMP2, VEGF, KGF, TGF-ß, IGF-I and PDGF upregulated in ITS medium. In conclusion, ITS medium has the potential to be used for the expansion of hAMSCs before clinical application.


Subject(s)
Adipocytes/cytology , Amnion/cytology , Chondrocytes/cytology , Culture Media/pharmacology , Keratinocytes/cytology , Mesenchymal Stem Cells/cytology , Osteocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Amnion/drug effects , Amnion/metabolism , Antioxidants/pharmacology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteocytes/drug effects , Osteocytes/metabolism , Selenium/pharmacology , Signal Transduction , Transferrin/pharmacology
15.
J Bone Miner Metab ; 37(5): 773-779, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30607618

ABSTRACT

Mutations in Serpinf1 gene which encodes pigment epithelium-derived factor (PEDF) lead to osteogenesis imperfecta type VI whose hallmark is defective matrix mineralization. We reported previously that PEDF reduced expression and synthesis of Sost/Sclerostin as well as other osteocytes genes encoding proteins that regulate matrix mineralization [1]. To determine whether PEDF had an effect on osteocyte gene expression in bone, we used bone explant cultures. First, osteocytes were isolated from surgical waste of bone fragments obtained from patients undergoing elective foot surgeries under approved IRB protocol by Penn State College of Medicine IRB committee. Primary osteocytes treated with PEDF reduced expression and synthesis of Sost/Sclerostin and matrix phosphoglycoprotein (MEPE) as well as dentin matrix protein (DMP-1). On the whole, PEDF reduced osteocyte protein synthesis by 50% and by 75% on mRNA levels. For bone explants, following collagenase digestion, bone fragments were incubated in alpha-MEM supplemented with 250 ng/ml of PEDF or BSA. After 7 days of incubation in a medium supplemented with PEDF, analysis of mRNA by PCR and protein by western blotting of encoded osteocyte proteins showed reduced Sclerostin synthesis by 39% and MEPE by 27% when compared to fragments incubated in medium supplemented with BSA. mRNA expression levels of osteocytes in bone fragments treated with PEDF were reduced by 50% for both SOST and MEPE when compared to BSA-treated bone fragments. Taken together, the data indicate that PEDF has an effect on osteocyte gene expression in bone and encourage further studies to examine effect of PEDF on bone formation indices in animal models and its effect on osteocyte gene expression in vivo following PEDF administration.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone and Bones/metabolism , Eye Proteins/pharmacology , Gene Expression Regulation/drug effects , Nerve Growth Factors/pharmacology , Osteocytes/metabolism , Serpins/pharmacology , Tissue Culture Techniques , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Genetic Markers/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Middle Aged , Osteocytes/drug effects , Phosphoproteins/genetics , Phosphoproteins/metabolism
16.
Int J Biol Sci ; 15(1): 58-68, 2019.
Article in English | MEDLINE | ID: mdl-30662347

ABSTRACT

Accumulating studies have shown that oxidative stress increases with aging, which is related to the pathophysiology of postmenopausal osteoporosis. Pyrroloquinoline quinone (PQQ) is a natural anti-oxidant with anti-oxidative and anti-aging effects. However, it is unclear whether PQQ has a protective role against estrogen deficiency-induced osteoporosis. Here, we evaluated the efficacy of PQQ on bone mineral density, bone microarchitecture, bone turnover and biomechanical strength in ovariectomy (OVX)-induced osteoporosis mouse model. Although dietary PQQ supplement did not affect serum E2 levels and uterine weight in OVX mice, it could prevent OVX-induced bone loss and improve bone strength by inhibiting oxidative stress, osteocyte senescence and senescence-associated secretory phenotype (SASP), subsequently promoting osteoblastic bone formation and inhibiting osteoclastic bone resorption, which was comparable to treatment with exogenous estrogen. The results from our study provide experimental evidence for the clinical use of PQQ to prevent estrogen deficiency-induced osteoporosis.


Subject(s)
Estrogens/deficiency , Osteoporosis/etiology , Osteoporosis/prevention & control , PQQ Cofactor/therapeutic use , Animals , Cellular Senescence/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Inbred C57BL , Osteocytes/drug effects , Osteocytes/metabolism , Osteoporosis/metabolism , Oxidative Stress/drug effects
17.
J Bone Miner Metab ; 37(2): 212-223, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29603070

ABSTRACT

A link between vascular calcification and bone anomalies has been suggested in chronic kidney disease (CKD) patients with low bone turnover disease. We investigated the vascular expression of osteocyte markers in relation to bone microarchitecture and mineralization defects in a model of low bone turnover CKD rats with vascular calcification. CKD with vascular calcification was induced by 5/6 nephrectomy followed by high calcium and phosphate diet, and vitamin D supplementation (Ca/P/VitD). CKD + Ca/P/VitD group (n = 12) was compared to CKD + normal diet (n = 12), control + normal diet (n = 8) and control + Ca/P/VitD supplementation (n = 8). At week 6, tibia, femurs and the thoracic aorta were analysed by Micro-Ct, histomorphometry and for expression of osteocyte markers. High Ca/P/VitD treatment induced vascular calcification only in CKD rats, suppressed serum parathyroid hormone levels and led to higher sclerostin, DKK1 and FGF23 serum levels. Expression of sclerostin, DKK1 and DMP1 but not FGF23 were increased in calcified vessels from CKD + Ca/P/VitD rats. Despite low parathyroid hormone levels, tibia bone cortical thickness was significantly lower in CKD + Ca/P/VitD rats as compared to control rats fed a normal diet, which is likely the result of radial growth impairment. Finally, Ca/P/VitD treatment in CKD rats induced a bone mineralization defect, which is likely explained by the high calcitriol dose. In conclusion, Ca/P/VitD supplementation in CKD rats induces expression of osteocyte markers in vessels and bone mineralisation anomalies. Further studies should evaluate the mechanisms of high dose calcitriol-induced bone mineralisation defects in CKD.


Subject(s)
Calcification, Physiologic/drug effects , Calcitriol/adverse effects , Calcium/adverse effects , Dietary Supplements/adverse effects , Osteocytes/pathology , Phosphates/adverse effects , Uremia/complications , Vascular Calcification/chemically induced , Animals , Bone Remodeling/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cortical Bone/drug effects , Cortical Bone/pathology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Male , Minerals/metabolism , Osteocytes/drug effects , Osteocytes/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Uremia/blood , Uremia/pathology , Uremia/physiopathology , Vascular Calcification/blood , Vascular Calcification/complications , Vascular Calcification/physiopathology , Wnt Signaling Pathway
18.
Br J Nutr ; 120(11): 1217-1229, 2018 12.
Article in English | MEDLINE | ID: mdl-30309398

ABSTRACT

Both genetic selection and increasing nutrient density for improving growth performance had inadvertently increased leg problems of meat ducks, which adversely affects animal welfare. We hypothesised that slowing weight gain with improving tibia quality probably enhanced tibial mechanical properties and alleviated leg deformities. Therefore, the present study aimed to evaluate the effect of graded Ca supplementation in a low-nutrient density (LND) diet on tibia composition and bone turnover in meat ducks. A total of 720 15-d-old male meat ducks were randomly assigned and fed a standard nutrient density positive control (PC) diet containing 0·9 % Ca, and four LND diets with 0·5, 0·7, 0·9 and 1·1 % Ca, respectively. Ducks fed the 0·5 % Ca LND diet and the PC diet had higher incidence of tibial dyschondroplasia (TD). When compared with the 0·5 % Ca LND diet, LND diets with ≥0·7 % Ca significantly improved tibia composition, microarchitecture and mechanical properties, and consequently decreased the incidence of TD. Furthermore, LND diets with ≥0·7 % Ca increased osteocyte-specific gene mRNA expression, blocked the expression of osteoblast differentiation marker genes including osteocalcin, collagenase-1 and alkaline phosphatase (ALP), and also decreased the expression of osteoclast differentiation genes, such as vacuolar-type H+-ATPase, cathepsin K and receptor activator of NF-κB. Meanwhile bone markers such as serum ALP, osteocalcin (both osteoblast markers) and tartrate-resistant acid phosphatase (an osteoclast marker) were significantly decreased in at least 0·7 % Ca treated groups. These findings indicated that LND diets with ≥0·7 % Ca decreased bone turnover, which subsequently increased tibia quality for 35-d-old meat ducks.


Subject(s)
Animal Feed , Bone Remodeling , Bone and Bones/drug effects , Calcium/metabolism , Dietary Supplements , Tibia/drug effects , Animal Nutrition Sciences , Animals , Body Weight , Bone Density/drug effects , Ducks , Gene Expression Regulation , Male , Meat , Osteocytes/metabolism , Tibia/physiopathology
19.
Bone ; 112: 10-18, 2018 07.
Article in English | MEDLINE | ID: mdl-29635039

ABSTRACT

Glucocorticoid treatment, a major cause of drug-induced osteoporosis and fractures, is widely used to treat inflammatory conditions and diseases. By contrast, mechanical loading increases bone mass and decreases fracture risk. With these relationships in mind, we investigated whether mechanical loading interacts with GC treatment in bone. Three-month-old female C57BL/6 mice were treated with high-dose prednisolone (15 mg/60 day pellets/mouse) or vehicle for two weeks. During the treatment, right tibiae were subjected to short periods of cyclic compressive loading three times weekly, while left tibiae were used as physiologically loaded controls. The bones were analyzed using peripheral quantitative computed tomography, histomorphometry, real-time PCR, three-point bending and Fourier transform infrared micro-spectroscopy. Loading alone increased trabecular volumetric bone mineral density (vBMD), cortical thickness, cortical area, osteoblast-associated gene expression, osteocyte- and osteoclast number, and bone strength. Prednisolone alone decreased cortical area and thickness and osteoblast-associated gene expression. Importantly, prednisolone treatment decreased the load-induced increase in trabecular vBMD by 57% (p < 0.001) and expression of osteoblast-associated genes, while completely abolishing the load-induced increase in cortical area, cortical thickness, number of osteocytes and osteoclasts, and bone strength. When combined, loading and prednisolone decreased the collagen content. In conclusion, high-dose prednisolone treatment strongly inhibits the loading-induced increase in trabecular BMD, and abolishes the loading-induced increase in cortical bone mass. This phenomenon could be due to prednisolone inhibition of osteoblast differentiation and function.


Subject(s)
Osteogenesis/drug effects , Prednisolone/pharmacology , Anabolic Agents/pharmacology , Animals , Cancellous Bone/drug effects , Cancellous Bone/physiology , Collagen/metabolism , Female , Gene Expression Regulation/drug effects , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/metabolism , Osteogenesis/genetics , Weight-Bearing/physiology
20.
Chem Biol Interact ; 261: 11-17, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27833010

ABSTRACT

Postmenopausal osteoporosis is characterized by a reduction in the number of sinusoidal and arterial capillaries in the bone marrow and reduced bone perfusion. Thus, osteogenesis and angiogenesis are coupled in the process of osteoporosis formation and fracture healing. Naringin is the main ingredient of the root Rhizoma Drynariae, a traditional Chinese medicine, and it has potential effects on promoting fracture healing. However, whether naringin stimulates angiogenesis in the process of bone healing is unclear. Here, we show that naringin promotes fracture healing through stimulating angiogenesis by regulating the VEGF/VEGFR-2 signaling pathway in osteoporotic rats.


Subject(s)
Flavanones/pharmacology , Fracture Healing/drug effects , Neovascularization, Physiologic/drug effects , Osteoporosis/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Bone Density/drug effects , Bony Callus/drug effects , Bony Callus/pathology , Female , Gene Expression Regulation/drug effects , Immunohistochemistry , Osteocytes/drug effects , Osteocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL