Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Bone ; 155: 116267, 2022 02.
Article in English | MEDLINE | ID: mdl-34856421

ABSTRACT

Leucine-rich repeat containing G-protein-coupled receptor 6 (LGR6) is a marker of osteoprogenitor cells and is dynamically expressed during in vitro osteodifferentation of mouse and human mesenchymal stem cells (MSCs). While the Lgr6 genomic locus has been associated with osteoporosis in human cohorts, the precise molecular function of LGR6 in osteogenesis and maintenance of bone mass are not yet known. In this study, we performed in vitro Lgr6 knockdown and overexpression experiments in murine osteoblastic cells and find decreased Lgr6 levels results in reduced osteoblast proliferation, differentiation, and mineralization. Consistent with these data, overexpression of Lgr6 in these cells leads to significantly increased proliferation and osteodifferentiation. To determine whether these findings are recapitulated in vivo, we performed microCT and ex vivo osteodifferentiation analyses using our newly generated CRISPR-Cas9 mediated Lgr6 mouse knockout allele (Lgr6-KO). We find that ex vivo osteodifferentiation of Lgr6-KO primary MSCs is significantly reduced, and 8 week-old Lgr6-KO mice have less trabecular bone mass as compared to Lgr6 wildtype controls, indicating that Lgr6 is necessary for normal osteogenesis and bone mass. Towards mechanism, we analyzed in vitro signaling in the context of two LGR6 ligands, RSPO2 and MaR1. We find that RSPO2 stimulates LGR6-mediated WNT/ß-catenin signaling whereas MaR1 stimulates LGR6-mediated cAMP activity, suggesting two ligand-dependent functions for LGR6 receptor signaling during osteogenesis. Collectively, this study reveals that Lgr6 is necessary for wildtype levels of proliferation and differentiation of osteoblasts, and achieving normal bone mass.


Subject(s)
Osteogenesis , Wnt Signaling Pathway , Animals , Cell Differentiation/genetics , Ligands , Mice , Osteoblasts , Receptors, G-Protein-Coupled/genetics
2.
Bone ; 129: 115055, 2019 12.
Article in English | MEDLINE | ID: mdl-31476437

ABSTRACT

CONTEXT: The mechanism behind hypophosphatemia in the setting of neurofibromatosis type 1 (NF1) is not known. We describe a possible role of fibroblast growth factor-23 (FGF23) in the pathophysiology of hypophosphatemia in a patient with NF1. CASE DESCRIPTION: A 34-year woman with NF1 presented with severe hypophosphatemia, osteomalacia, and elevated plasma FGF23. The patient had considerable improvement on replacement of oral phosphate. Two Ga68 DOTANOC PET-CT scans over a period of 2 years failed to detect any localized uptake. Immuno-staining for FGF23 was absent in the neural-derived tumour cells of the neurofibromas in the proband. CONCLUSION: The patient with NF1 had elevated circulating FGF23. Tumour cells in the neurofibroma tissues did not stain for FGF23 on IHC. It is unlikely for neurofibromas to contribute to high circulating FGF23 levels in the proband.


Subject(s)
Fibroblast Growth Factors/metabolism , Hypophosphatemia/complications , Hypophosphatemia/metabolism , Neurofibromatosis 1/complications , Neurofibromatosis 1/metabolism , Osteomalacia/complications , Osteomalacia/metabolism , Adult , Female , Fibroblast Growth Factor-23 , Humans , Hypophosphatemia/diagnostic imaging , Hypophosphatemia/pathology , Neurofibromatosis 1/diagnostic imaging , Neurofibromatosis 1/pathology , Osteomalacia/diagnostic imaging , Osteomalacia/pathology
3.
JBMR Plus ; 3(2): e10075, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30828690

ABSTRACT

Bone marrow-derived mesenchymal stem cells are an important source of osteoblasts critical for both bone homeostasis and repair. The ability to isolate, or specifically target, mesenchymal stem cells committed to the osteogenic lineage is necessary for orthopedic translational therapy efforts; however the precise molecular signature of these cells remains elusive. Previously, we identified a population of osteoprogenitor cells expressing the Wnt signaling agonist Lgr6, which contributes to the development and regeneration of the mouse digit tip bone. In our present study we build upon this data and investigate the expression of Lgr6 more broadly in the skeleton. We find that Lgr6, and closely related Lgr4, are expressed in mouse primary calvarial cells, bone marrow cells, and bone marrow-derived mesenchymal stem cells. In addition, our data demonstrates that Lgr4 expression is modestly increased throughout the differentiation and mineralization of mesenchymal stem cells. In contrast, we find Lgr6 expression to be strikingly increased upon osteogenic induction and subsequently decreased upon differentiation and mineralization. These findings provide evidence for Lgr6 as a novel marker of osteoprogenitor cells in bone marrow, which could prove useful for isolation of this population toward future research and clinical applications.

4.
Mol Ther Nucleic Acids ; 14: 536-549, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30769134

ABSTRACT

Post-menopausal condition augments the biological aging process, characterized by multiple metabolic disorders in which bone loss is the most prevalent outcome and usually coupled with sarcopenia. Coexistence of such associated pathogenesis have much worse health outcomes, compared to individuals with osteoporosis only. Pre- and post-natal bone development demands calcium from mother to fetus during pregnancy and lactation leading to a significant maternal skeletal loss. It follows an anabolic phase around weaning during which there is a notable recovery of the maternal skeleton. Here, we have studied the therapeutic effect of microRNA-672-5p identified during weaning when it is predominantly expressed, in ovariectomized mice for both osteopenia and sarcopenia. miR-672-5p induced osteoblast differentiation and mineralization. These actions were mediated through inhibition of Smurf1 with enhanced Runx2 transcriptional activation. In vivo, miR-672-5p significantly increased osteoblastogenesis and mineralization, thus reversing bone loss caused by ovariectomy. It also improved bone-mineral density, load-bearing capacity, and bone quality. Sarcopenia was also alleviated by miR-672-5p, as we observed increased cross-sectional area and Feret's diameter of muscle fibers. We hypothesize that elevated miR-672-5p expression has therapeutic efficacy in estrogen-deficiency-induced osteopenia along with sarcopenia.

5.
Bone ; 117: 98-115, 2018 12.
Article in English | MEDLINE | ID: mdl-30218791

ABSTRACT

Parathyroid hormone (PTH; amino acid 1-34, known as teriparatide) has reported promoting differentiation and glucose uptake in osteoblasts. However, how PTH regulates glucose metabolism to facilitate osteoblast differentiation is not understood. Here, we report that PTH promotes glucose dependent miR-451a expression which stimulates osteoblast differentiation. In addition to glucose uptake, PTH suppresses AMPK phosphorylation via PI3K-mTOR-AKT axis thereby preventing phosphorylation and inactivation of octamer-binding transcription factor 1 (OCT-1) which has been reported to act on the promoter region of miR-451a. Modulation of AMPK activity controls miR-451a levels in differentiating osteoblasts. Moreover, pharmacological inhibition of PI3K-mTOR-AKT axis suppressed miR-451a via increased AMPK activity. We report that this glucose regulated miRNA is an anabolic target and transfection of miR-451a mimic induces osteoblast differentiation and mineralization in vitro. These actions were mediated through the suppression of Odd-skipped related 1 (Osr1) and activation of Runx2 transcription. When injected in vivo, the miR-451a mimic significantly increased osteoblastogenesis, mineralization, reversed ovariectomy induced bone loss and improved bone strength. Together, these findings suggest that enhanced osteoblast differentiation associated with bone formation in case of PTH therapy is also a consequence of elevated miR-451a levels via glucose regulation. Consequently, this miRNA has the potential to be a therapeutic target for conditions of bone loss.


Subject(s)
Cell Differentiation/drug effects , Gene Expression Regulation , Glucose/metabolism , MicroRNAs/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Adenylate Kinase/metabolism , Animals , Bone Resorption/pathology , Cell Differentiation/genetics , Enzyme Activation/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Mice, Inbred BALB C , MicroRNAs/metabolism , Octamer Transcription Factor-1/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Ovariectomy , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
6.
Medchemcomm ; 9(7): 1213-1225, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-30109010

ABSTRACT

ß-Carbolines have been assessed for osteoclastogenesis. However, their effect on osteoblasts during estrogen deficiency is still unclear. Here, a series of novel piperazine and tetrazole tag ß-carbolines have been synthesized and examined for osteoblast differentiation in vitro. In vitro data suggest that compound 8g is the most promising osteoblast differentiating agent that was evaluated for in vivo studies. Compound 8g promoted osteoblast mineralization, stimulated Runx2, BMP-2 and OCN expression levels, increased BrdU incorporation and inhibited generation of free radicals as well as nitric oxide. Since a piperazine group is involved in bone repair activity and ß-carboline in IκB kinase (IKK) inhibition, compound 8g inhibited tumor necrosis factor α (TNFα) directed IκBα phosphorylation, preventing nuclear translocation of NF-κB thereby alleviating osteoblast apoptosis. In vivo studies show that compound 8g was able to restore estrogen deficiency-induced bone loss in ovariectomized rats without any toxicity, thus signifying its potential in bone-protection chemotherapy under postmenopausal conditions.

7.
PLoS One ; 12(11): e0188963, 2017.
Article in English | MEDLINE | ID: mdl-29190819

ABSTRACT

During development, a multi-potent group of cells known as the cranial neural crest (CNC) migrate to form craniofacial structures. Proper migration of these cells requires proteolysis of cell adhesion molecules, such as cadherins. In Xenopus laevis, preventing extracellular cleavage of cadherin-11 impairs CNC migration. However, overexpression of the soluble cleavage product (EC1-3) is capable of rescuing this phenotype. The mechanism by which EC1-3 promotes CNC migration has not been investigated until now. Here we show that EC1-3 stimulates phosphorylation of Akt, a target of PI3K, in X.laevis CNC. Through immunoprecipitation experiments, we determined that EC1-3 interacts with all ErbB receptors, PDGFRα, and FGFR1. Of these receptors, only ErbB2 was able to produce an increase in Akt phosphorylation upon treatment with a recombinant EC1-3. This increase was abrogated by mubritinib, an inhibitor of ErbB2. We were able to recapitulate this decrease in Akt phosphorylation in vivo by knocking down ErbB2 in CNC cells. Knockdown of the receptor also significantly reduced CNC migration in vivo. We confirmed the importance of ErbB2 and ErbB receptor signaling in CNC migration using mubritinib and canertinib, respectively. Mubritinib and the PI3K inhibitor LY294002 significantly decreased cell migration while canertinib nearly prevented it altogether. These data show that ErbB2 and Akt are important for CNC migration and implicate other ErbB receptors and Akt-independent signaling pathways. Our findings provide the first example of a functional interaction between the extracellular domain of a type II classical cadherin and growth factor receptors.


Subject(s)
Brain/cytology , Cadherins/metabolism , Cell Movement , Neural Crest/cytology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Animals , Brain/metabolism , Cadherins/chemistry , HEK293 Cells , Humans , Phosphorylation , Protein Binding , Xenopus laevis/embryology
8.
Elife ; 62017 08 22.
Article in English | MEDLINE | ID: mdl-28829038

ABSTRACT

Adam13/33 is a cell surface metalloprotease critical for cranial neural crest (CNC) cell migration. It can cleave multiple substrates including itself, fibronectin, ephrinB, cadherin-11, pcdh8 and pcdh8l (this work). Cleavage of cadherin-11 produces an extracellular fragment that promotes CNC migration. In addition, the adam13 cytoplasmic domain is cleaved by gamma secretase, translocates into the nucleus and regulates multiple genes. Here, we show that adam13 interacts with the arid3a/dril1/Bright transcription factor. This interaction promotes a proteolytic cleavage of arid3a and its translocation to the nucleus where it regulates another transcription factor: tfap2α. Tfap2α in turn activates multiple genes including the protocadherin pcdh8l (PCNS). The proteolytic activity of adam13 is critical for the release of arid3a from the plasma membrane while the cytoplasmic domain appears critical for the cleavage of arid3a. In addition to this transcriptional control of pcdh8l, adam13 cleaves pcdh8l generating an extracellular fragment that also regulates cell migration.


Subject(s)
ADAM Proteins/metabolism , Cadherins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Neural Crest/physiology , Transcription Factor AP-2/metabolism , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Animals , Protocadherins , Transcription, Genetic , Xenopus laevis
9.
J Pharm Pharmacol ; 69(10): 1381-1397, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28664619

ABSTRACT

OBJECTIVES: This study was undertaken to investigate the effects of a heartwood ethanolic extract (HEE) made from the Dalbergia sissoo on facture healing and in the prevention of pathological bone loss resulting from estrogen deficiency in ovariectomized (Ovx) rats. METHODS: Heartwood ethanolic extract (250, 500 and 1000 mg/kg per day) was administered orally immediately next day after drill-hole injury and continued for 2 weeks. Ovx rats received HEE at same doses for 12 weeks and compared with 17-ß estradiol (E2; 100 µg/kg for 5 days/week subcutaneously) group. Confocal imaging for fracture healing, micro-architecture of long bones, biomechanical strength, formation of mineralized nodule by bone marrow osteoprogenitor cells, bone turnover markers and gene expression were studied. One-way ANOVA was used to test significance. KEY FINDINGS: Heartwood ethanolic extract treatment promoted fracture healing, formation of new bone at the drill-hole site and stimulated osteogenic genes at callus region. HEE administration to the Ovx rats exhibited better micro-architectural parameters at various anatomical positions, better bone biomechanical strength and more osteoprogenitor cells in the bone marrow compared with Ovx + vehicle group. HEE exhibited no uterine estrogenicity. CONCLUSIONS: Oral administration of HEE was found to promote fracture healing and exhibited osteoprotective effect by possibly stimulation of osteoblast function.


Subject(s)
Dalbergia , Fracture Healing/drug effects , Osteoporosis/drug therapy , Ovariectomy/adverse effects , Plant Extracts/therapeutic use , Animals , Bone Density/drug effects , Bone Density/physiology , Female , Fracture Healing/physiology , Osteoporosis/etiology , Osteoporosis/pathology , Ovariectomy/trends , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley
10.
Br J Nutr ; 117(5): 645-661, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28367764

ABSTRACT

Balance between adipocyte and osteoblast differentiation is the key link of disease progression in obesity and osteoporosis. We have previously reported that formononetin (FNT), an isoflavone extracted from Butea monosperma, stimulates osteoblast formation and protects against postmenopausal bone loss. The inverse relationship between osteoblasts and adipocytes prompted us to analyse the effect of FNT on adipogenesis and in vivo bone loss, triggered by high-fat diet (HFD)-induced obesity. The anti-obesity effect and mechanism of action of FNT was determined in 3T3-L1 cells and HFD-induced obese male mice. Our findings show that FNT suppresses the adipogenic differentiation of 3T3-L1 fibroblasts, through down-regulation of key adipogenic markers such as PPARγ, CCAAT/enhancer-binding protein alpha (C/EBPα) and sterol regulatory element-binding protein (SREBP) and inhibits intracellular TAG accumulation. Increased intracellular reactive oxygen species levels and AMP-activated protein kinase (AMPK) activation accompanied by stabilisation of ß-catenin were attributed to the anti-adipogenic action of FNT. In vivo, 12 weeks of FNT treatment inhibited the development of obesity in mice by attenuating HFD-induced body weight gain and visceral fat accumulation. The anti-obesity effect of FNT results from increased energy expenditure. FNT also protects against HFD-induced dyslipidaemia and rescues deterioration of trabecular bone volume by increasing bone formation and decreasing bone resorbtion caused by HFD. FNT's rescuing action against obesity-induced osteoporosis commenced at the level of progenitors, as bone marrow progenitor cells, obtained from the HFD mice group supplemented with FNT, showed increased osteogenic and decreased adipogenic potentials. Our findings suggest that FNT inhibits adipogenesis through AMPK/ß-catenin signal transduction pathways and protects against HFD-induced obesity and bone loss.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipogenesis/drug effects , Isoflavones/pharmacology , Obesity/prevention & control , Osteoporosis/prevention & control , beta Catenin/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Osteoporosis/etiology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Uncoupling Protein 1/genetics , Up-Regulation/drug effects
11.
Bioorg Med Chem Lett ; 27(8): 1765-1775, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28274632

ABSTRACT

The chemical modifications of the hydroxyl group of dalbergin have been described via the introduction of cyclic amine, ester and amide groups. Among the twenty-three prepared novel analogues of dalbergin, compound 4d (EC50 2.3µM) showed significantly increased proliferation as assessed by alkaline phosphatase activity and mineralization in calvarial osteoblast cells in vitro. Compound 4d, at a dose of 1.0mg/kg body weight exhibited the significant osteoprotective effect. It showed a significant increase in osteogenic gene expression RunX2 (∼4fold), ALP (∼5fold), OCN (∼4fold) and COL1 (∼4fold) as compared to control group at the same dose in vivo assay.


Subject(s)
Coumarins/chemistry , Coumarins/therapeutic use , Osteoblasts/drug effects , Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Animals , Bone Morphogenetic Protein 2/metabolism , Calcification, Physiologic/drug effects , Cells, Cultured , Coumarins/chemical synthesis , Drug Design , Female , Gene Expression Regulation/drug effects , Humans , Mice, Inbred BALB C , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoporosis, Postmenopausal/genetics , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/pathology , Skull/cytology , Skull/drug effects , Skull/metabolism , Skull/pathology
12.
Biomed Pharmacother ; 88: 804-813, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28160755

ABSTRACT

OBJECTIVE: In this study, we have evaluated the skeletal effects of butanolic fraction (BF) from Passiflora foetida in an estrogen deficient mice bone loss model. STUDY DESIGN: Skeletal effect of BF was studied in ovariectomized (OVx) female Balb/c mice. BF (50 and 100mg/kg/day dose orally) was given for 8 weeks. Micro-architecture of long bones, biomechanical strength, formations of mineralized nodule by bone marrow osteoprogenitor cells, osteoid formation and bone turnover markers were studied. One way ANOVA was used to test the significance of effects of Passiflora foetida. RESULTS: OVx mice treated with BF represented with better micro-architectural parameters at various anatomical positions, better bone biomechanical strength and more osteoprogenitor cells in the bone marrow compared with OVx group. BF did not exhibit uterine estrogenicity. CONCLUSION: Oral administration of BF at both the doses (50 and 100mg/kg/day) derived from Passiflora Foetida, was found to afford anti-osteoporotic effect under estrogen deficiency by likely stimulation of osteoblast function and inhibition of osteoclast function.


Subject(s)
Bone Density Conservation Agents/pharmacology , Osteoporosis/drug therapy , Ovariectomy , Passiflora/chemistry , Animals , Biomechanical Phenomena , Bone Marrow Cells/drug effects , Bone and Bones/pathology , Butanols , Female , Mice , Mice, Inbred BALB C , Osteoporosis/etiology , Osteoporosis/pathology , Solvents , Stem Cells/drug effects , Trabecular Meshwork/pathology , Trabecular Meshwork/ultrastructure , Uterus/pathology
13.
Biomed Pharmacother ; 86: 16-22, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27936389

ABSTRACT

Leaves of Dalbergia sissoo is known to have protective actions against postmenopausal bone loss in rat. In this study, we have evaluated the fracture healing properties of ethanolic extract (EE) of Dalbergia sissoo leaves. To observe the fracture healing property in the drill-hole injury model, we randomly divided total 32 adult female Sprague Dawley rats (180±200g) into 4 groups: (i) Control operated group; (ii) EE (250mg/kg/day); (iii) EE (500mg/kg/day) and (iv) EE (1000mg/kg/day). The right femora were fractured at the mid-diaphysis region and each group of rats received their respective treatment for 15days. Ethanol extract dose dependently induced bone regeneration at the fracture site assessed by fluorochrome labeling. All of three doses, 250mg/kg/day dose significantly increased bone volume fraction, trabecular thickness, trabecular number, and connectivity density and decreased trabecular separation in bone. Furthermore, the extract induced the expression of osteogenic genes including BMP-2, BMP-4, RunX-2 and COL-1 compared to the control group. The EE improved fracture healing much earlier (day 15) than the normal healing process, as assessed by the increased callus volumes and mineralized nodule formation. This extract is found beneficial in fracture healing of rat.


Subject(s)
Bone Regeneration/drug effects , Cortical Bone/drug effects , Dalbergia , Ethanol/pharmacology , Fracture Healing/drug effects , Plant Extracts/pharmacology , Animals , Bone Regeneration/physiology , Cells, Cultured , Cortical Bone/injuries , Cortical Bone/physiology , Disease Models, Animal , Female , Femur/drug effects , Femur/injuries , Femur/physiology , Fracture Healing/physiology , Osteogenesis/drug effects , Osteogenesis/physiology , Plant Extracts/isolation & purification , Rats , Rats, Sprague-Dawley
14.
Eur J Pharmacol ; 788: 65-74, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27316792

ABSTRACT

Dalbergia sissoo Roxb. is a well known medicinal plant of India, enriched with various flavonoids used for treating multiple diseases. Earlier, we have shown that extract of Dalbergia sissoo Roxb. leaves mitigate ovariectomy induced bone loss and pure compounds (neoflavonoids) isolated from it, promote osteoblastogenesis in primary calvarial osteoblasts cells in vitro. Here, we hypothesize that dalsissooal (DSL), a novel neoflavonoid isolated from the heartwood of Dalbergia sissoo Roxb. is an important constituent of the extract that imparts bone forming effects. Treatment with DSL enhanced trabecular bone micro-architecture parameters, biomechanical strength, increased bone formation rate and mineral apposition rate in OVx mice comparable to 17ß-estradiol. It increased bone formation by enhancing osteoblast gene expression and reduced bone turnover by decreasing osteoclastic gene expressions. Interestingly, we observed that DSL has no uterine estrogenic effects. At cellular levels, DSL promoted differentiation of bone marrow cells as well as calvaria osteoblast cells towards osteoblast lineage by enhancing differentiation and mineralizing ability to form mineralizing nodules via stimulating BMP-2 and RunX-2 expressions. Overall, our data suggest that oral supplementation of a novel neoflavonoid dalsissooal isolated from heartwood of Dalbergia sissoo Roxb. exhibited bone anabolic action by improving structural property of bone, promoting new bone formation and reducing bone turnover rate in post-menopausal model for osteoporosis with no uterine hyperplasia.


Subject(s)
Acrolein/analogs & derivatives , Dalbergia/chemistry , Flavonoids/pharmacology , Osteogenesis/drug effects , Osteoporosis/physiopathology , Phenols/pharmacology , Acrolein/isolation & purification , Acrolein/pharmacology , Animals , Calcification, Physiologic/drug effects , Cancellous Bone/drug effects , Cancellous Bone/pathology , Cancellous Bone/physiopathology , Cell Differentiation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Estrogens/deficiency , Female , Flavonoids/isolation & purification , Gene Expression Regulation/drug effects , Mice , Osteoblasts/drug effects , Osteoblasts/pathology , Osteocalcin/blood , Osteoporosis/metabolism , Osteoporosis/pathology , Ovariectomy , Phenols/isolation & purification , Plant Leaves/chemistry , Uterus/drug effects , Uterus/metabolism
15.
Bioorg Med Chem Lett ; 26(15): 3719-24, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27317644

ABSTRACT

Terpenoids were isolated using chromatographic purification through solvent purification technique and identified as Azadirone (1), Epoxyazadiradione (2) Azadiradione (3) Gedunin (4) Nimbin (5) Salannin (6) Azadirachtin A (7) and Azadirachtin B (8) from Azadirachta indica. Out of eight compounds, only three compounds had osteogenic activity and enhanced osteoblast proliferation, differentiation and mineralization in osteoblast cells. Active compounds stimulated osteogenic genes ALP, RunX-2 and OCN expressions in vitro, but Azadirachtin A had a maximum ability to stimulate osteoblast differentiation and mineralization compared to other two active compounds. For in vivo study, Azadirachtin A injected subcutaneously in pups, which enhanced osteogenic gene expressions and promoted bone formation rate significantly. Here, we conclude that active compounds of Azadirachta indica have osteogenic activity and Azadirachtin A has a beneficial effects on bone.


Subject(s)
Azadirachta/chemistry , Osteoblasts/drug effects , Triterpenes/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Structure , Osteoblasts/cytology , Structure-Activity Relationship , Triterpenes/chemistry , Triterpenes/isolation & purification
16.
J Pharm Pharmacol ; 68(8): 1051-63, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27265669

ABSTRACT

OBJECTIVE: This study evaluates the effect of isoflavone cladrin on high-fat diet (HFD)-induced bone loss and adipogenesis. METHODS: Thirty-two 4-week-old male C57BL/6J mice were divided into four groups: a standard diet group, a HFD group and HFD group with cladrin (5 and 10 mg/kg per day orally) for 12 weeks. The effect of cladrin on bone micro-architecture, bone marrow cell lineages and hyperlipidaemia were assessed. For assessing anti-adipogenic activity of cladrin, 3T3-L1 cells were used. KEY FINDINGS: Cladrin attenuated HFD-induced hyperlipidaemia and bone loss by preserving bone micro-architecture and strength. Effect of cladrin was found at the level of bone marrow progenitor cells. Gene expression profile of cladrin-treated mice bone showed upregulation of osteoblast and downregulation of adipogenic transcription factors and increased OPG/RANKL ratio. Cladrin inhibited cellular lipid accumulation through downregulation of transcription factors such as PPAR-γ and C/EBP-α and modulated the expression of major adipokines involved behind obesity stimulation without eliciting cell cytotoxicity in 3T3-L1 adipocytes. CONCLUSION: We conclude that cladrin may improve obesity-induced bone loss and hyperlipidaemia in mice fed HFD and adipogenesis in 3T3-L1 cells by modifying adipokines and could offer clinical benefits as a supplement to treat obesity-induced disorders.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue/metabolism , Bone and Bones/drug effects , Diet, High-Fat/adverse effects , Isoflavones/therapeutic use , Obesity/metabolism , Osteoporosis , 3T3-L1 Cells , Adipogenesis/genetics , Adipokines/metabolism , Animals , Butea/chemistry , Isoflavones/pharmacology , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/genetics , Osteoporosis/etiology , Osteoporosis/prevention & control , Osteoprotegerin/metabolism , Phytoestrogens/pharmacology , Phytoestrogens/therapeutic use , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , RANK Ligand/metabolism , Transcription Factors/metabolism
17.
Life Sci ; 151: 207-217, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26956522

ABSTRACT

Kaempferol (KEM) has been observed to stimulate Krt-14 protein which subsequently contributes to matrix maturation and mineralization in rat primary osteoblast cells. Incorporation of Krt-14 siRNA results in reduced mRNA and protein expression after 48h post transfection and remained low for 9days. At day 9 Krt-14 siRNA significantly reduced mineralization without concomitant change in the cell number. ColI and OCN gene expression was reduced in Krt-14 siRNA-treated osteoblast cells. Soluble osteocalcin and collagen levels were markedly decreased in conditioned medium as well as in acid-salt soluble cell-ECM layer treated with Krt-14 siRNA compared to control siRNA treated cells corroborated at the ultrastructral level by AFM. Further, knockdown of Krt-14 and inhibitors against AMPK and mTOR, repressed the activation of mTOR and mineralization attenuated by KEM confirmed the role of Krt-14 in mineralization. These findings strongly suggest that Krt-14 regulates osteoblast mineralization by organizing osteoblast derived ECM.


Subject(s)
Calcification, Physiologic/drug effects , Kaempferols/pharmacology , Keratin-14/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Animals , Cell Count , Cells, Cultured , Collagen/metabolism , Extracellular Matrix/metabolism , Gene Expression/drug effects , Gene Knockdown Techniques , Keratin-14/biosynthesis , Keratin-14/genetics , Osteoblasts/ultrastructure , Osteocalcin/metabolism , Pyrazoles/antagonists & inhibitors , Pyrimidines/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Rats , Sirolimus/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
18.
J Biol Chem ; 291(8): 3959-66, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26663087

ABSTRACT

Embryonic skeletogenesis and postnatal bone development require the transfer of calcium from the mother to the offspring during pregnancy and lactation. Therefore, bone resorption in the mother becomes elevated during these periods, resulting in significant maternal skeletal loss. There follows an anabolic phase around weaning during which there is a remarkable recovery of the maternal skeleton. However, the mechanism(s) of this anabolic response remain(s) largely unknown. We identified eight differentially expressed miRNAs by array profiling, of which miR-874-3p was highly expressed at weaning, a time when bone loss was noted to recover. We report that this weaning-associated miRNA is an anabolic target. Therefore, an agomir of miR-874-3p induced osteoblast differentiation and mineralization. These actions were mediated through the inhibition of Hdac1 expression and enhanced Runx2 transcriptional activation. When injected in vivo, the agomir significantly increased osteoblastogenesis and mineralization, reversed bone loss caused by ovariectomy, and increased bone strength. We speculate that elevated miR-874-3p expression during weaning enhances bone formation and that this miRNA may become a therapeutic target for conditions of bone loss.


Subject(s)
Calcification, Physiologic/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation, Enzymologic/physiology , Histone Deacetylase 1/metabolism , MicroRNAs/metabolism , Osteoblasts/metabolism , Animals , Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Female , Histone Deacetylase 1/genetics , Mice , MicroRNAs/genetics , Osteoblasts/cytology , Pregnancy , Weaning
19.
Menopause ; 22(11): 1246-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25850356

ABSTRACT

OBJECTIVE: Dalbergiphenol (DGP) is a neoflavonoid isolated from heartwood of Dalbergia sissoo. Effects of DGP on skeletal health remain to be elucidated. The objective of the present study was to investigate the biological effects of DGP on bone loss in ovariectomized mice. METHODS: Adult BALB/c mice were ovariectomized and administered DGP (1 and 5  mg/kg/d) or 17ß-estradiol (E2) orally for 6 weeks. The sham group and the ovariectomy (OVX) + vehicle group served as controls. Eight female BALB/c mice were taken for each group. Uterine estrogenicity, bone microarchitecture, biomechanical strength, new bone formation (based on bone formation rate and mineral apposition rate), and skeletal expression of osteogenic and resorptive gene markers were studied. RESULTS: OVX resulted in a marked increase in body weight and a decrease in femoral and vertebral trabecular bone volume that were prevented by DGP or E2 treatment. DGP treatment increased bone biomechanical strength and new bone formation rate in ovariectomized mice, comparable with E2 treatment. However, increase in uterine weight and estrogenicity were observed in E2-treated ovariectomized mice, but not in response to DGP treatment. Treatment with DGP increased messenger RNA expression of runt-related transcription factor 2, osterix, and collagen type I, and decreased messenger RNA expression of tartrate-resistant acid phosphatase and the osteoprotegerin-to-receptor activator of nuclear factor-κB ligand ratio in the femur of ovariectomized mice. CONCLUSIONS: Overall findings suggest that DGP treatment can effectively prevent OVX-induced increase in bone loss and decrease in bone strength possibly by increasing osteoblastic activities and by decreasing osteoclastic activities.


Subject(s)
Bone Remodeling/drug effects , Bone Resorption/prevention & control , Dalbergia/chemistry , Osteoporosis, Postmenopausal/prevention & control , Plant Extracts/administration & dosage , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Ovariectomy , Plant Leaves/chemistry
20.
Nutrition ; 31(1): 205-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25466667

ABSTRACT

OBJECTIVE: Bone protective effects of withaferin A (WFA) from leaves of Withania somnifera (L.) were evaluated in preventive model of Balb/c mice with 17 ß-estradiol (E2) and alendronate (ALD). METHODS: Adult female Balb/c mice, 7 to 9 wk, were bilaterally ovariectomized (OVx) to mimic the state of E2 deficiency. Immediately after surgery mice were administrated WFA at doses of 1, 5, 10 mg/kg/d while other two OVx groups received ALD or E2 for 2 mo. Sham and OVx groups with vehicle and no treatment served as controls. RESULTS: WFA administration increased new bone formation, as well as improving microarchitecture and biomechanical strength of the bones. It prevented bone loss by reducing expression of osteoclastic genes tartrate resistant acid phosphatase (TRAP) and receptor activator of nuclear factor κ B (RANK). Increase in bone turnover marker, osteocalcin (OCN) and inflammatory cytokine tumor necrosis factor-alpha (TNF-α) because of ovariectomy were reduced with WFA treatment, with effects comparable to E2 administration. Histomorphometric analysis of uterus shows that WFA was not fraught with estrogenic or antiestrogenic effects. At cellular level, WFA promoted differentiation of bone marrow cells (BMCs) and increased mineralization by inducing expression of osteogenic genes. WFA has bone protective potential as its treatment prevents bone loss that is comparable to ALD and E2. CONCLUSIONS: It is surmised that WFA in preclinical setting is effective in preserving bone loss by both inhibition of resorption and stimulation of new bone formation before onset of osteoporosis with no uterine hyperplasia.


Subject(s)
Alendronate/pharmacology , Estradiol/pharmacology , Osteoporosis/prevention & control , Plants, Medicinal/chemistry , Withania/chemistry , Withanolides/pharmacology , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Biomarkers/blood , Bone and Bones/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred BALB C , Osteocalcin/blood , Osteoclasts/drug effects , Osteoclasts/metabolism , Ovariectomy , Plant Leaves/chemistry , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tartrate-Resistant Acid Phosphatase , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...