Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Int J Mol Sci ; 24(11)2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37298702

ABSTRACT

The receptor activator of NF-κB ligand (RANKL)-binding peptide is known to accelerate bone morphogenetic protein (BMP)-2-induced bone formation. Cholesterol-bearing pullulan (CHP)-OA nanogel-crosslinked PEG gel (CHP-OA nanogel-hydrogel) was shown to release the RANKL-binding peptide sustainably; however, an appropriate scaffold for peptide-accelerated bone formation is not determined yet. This study compares the osteoconductivity of CHP-OA hydrogel and another CHP nanogel, CHP-A nanogel-crosslinked PEG gel (CHP-A nanogel-hydrogel), in the bone formation induced by BMP-2 and the peptide. A calvarial defect model was performed in 5-week-old male mice, and scaffolds were placed in the defect. In vivo µCT was performed every week. Radiological and histological analyses after 4 weeks of scaffold placement revealed that the calcified bone area and the bone formation activity at the defect site in the CHP-OA hydrogel were significantly lower than those in the CHP-A hydrogel when the scaffolds were impregnated with both BMP-2 and the RANKL-binding peptide. The amount of induced bone was similar in both CHP-A and CHP-OA hydrogels when impregnated with BMP-2 alone. In conclusion, CHP-A hydrogel could be an appropriate scaffold compared to the CHP-OA hydrogel when the local bone formation was induced by the combination of RANKL-binding peptide and BMP-2, but not by BMP-2 alone.


Subject(s)
Hydrogels , Peptides , Animals , Male , Mice , Bone Morphogenetic Protein 2/pharmacology , Cholesterol , Hydrogels/pharmacology , Nanogels , Peptides/pharmacology , RANK Ligand/chemistry , RANK Ligand/metabolism
2.
J Cell Physiol ; 236(4): 2800-2816, 2021 04.
Article in English | MEDLINE | ID: mdl-32964459

ABSTRACT

The tumor necrosis factor (TNF)-like core domain of receptor activator of nuclear factor-κB ligand (RANKL) is a functional domain critical for osteoclast differentiation. One of the missense mutations identified in patients with osteoclast-poor autosomal recessive osteopetrosis (ARO) is located in residue methionine 199 that is replaced with lysine (M199K) amid the TNF-like core domain. However, the structure-function relationship of this mutation is not clear. Sequence-based alignment revealed that the fragment containing human M199 is highly conserved and equivalent to M200 in rat. Using site-directed mutagenesis, we generated three recombinant RANKL mutants M200K/A/E (M200s) by replacing the methionine 200 with lysine (M200K), alanine (M200A), and glutamic acid (M200E), representative of distinct physical properties. TRAcP staining and bone pit assay showed that M200s failed to support osteoclast formation and bone resorption, accompanied by impaired osteoclast-related signal transduction. However, no antagonistic effect was found in M200s against wild-type rat RANKL. Analysis of the crystal structure of RANKL predicted that this methionine residue is located within the hydrophobic core of the protein, thus, likely to be crucial for protein folding and stability. Consistently, differential scanning fluorimetry analysis suggested that M200s were less stable. Western blot analysis analyses further revealed impaired RANKL trimerization by M200s. Furthermore, receptor-ligand binding assay displayed interrupted interaction of M200s to its intrinsic receptors. Collectively, our studies revealed the molecular basis of human M199-induced ARO and elucidated the indispensable role of rodent residue M200 (equivalent to human M199) for the RANKL function.


Subject(s)
Mutation, Missense , RANK Ligand/genetics , Animals , Bone Resorption , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Osteoclasts/metabolism , Osteogenesis , Protein Conformation , Protein Folding , Protein Stability , RANK Ligand/chemistry , RANK Ligand/metabolism , RAW 264.7 Cells , Rats , Signal Transduction , Structure-Activity Relationship
3.
J Nat Prod ; 83(5): 1585-1591, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32267694

ABSTRACT

Irijimasides A-E (1-5), a series of new 14-membered macrolide glycosides, were isolated from a marine cyanobacterium collected in Okinawa. The gross structures of 1-5 were established by spectroscopic analysis, including 2D NMR, while absolute stereostructures were determined based on NOESY spectra, chemical derivatization, and ECD data. All five macrolides suppressed receptor activator of nuclear factor-κB ligand (RANKL)-induced tartrate-resistant acid phosphatase (TRAP) activity in mouse RAW264 macrophage cells, indicating that these compounds inhibit osteoclast formation.


Subject(s)
Cyanobacteria/chemistry , Glycosides/chemistry , Macrolides/chemistry , Osteoclasts/drug effects , Tartrate-Resistant Acid Phosphatase/metabolism , Animals , Mice , Molecular Structure , RANK Ligand/chemistry , RANK Ligand/metabolism , Tartrate-Resistant Acid Phosphatase/chemistry
4.
J Nat Prod ; 83(4): 1005-1017, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32233482

ABSTRACT

Eighteen new jatrophane diterpenoids, euphoesulatins A-R (1-18), and three known diterpenoids (19-21) were isolated from Euphorbia esula. Compounds 1-7, 14, and 18 represent a rare type of jatrophane-type diterpenoid containing a nicotinoyloxy group. The absolute configuration of 1 was determined by X-ray crystallography. The compounds were assayed for their antiosteoporotic activity in a bone-marrow-derived macrophage cell line, and compounds 1, 8, and 10 significantly inhibited the formation of osteoclasts, with IC50 values of 1.2, 3.5, and 2.3 µM, respectively. These three compounds also dose-dependently reduced the activity of nuclear factor activated T-cell cytoplasmic 1. This study reveals the antiosteoporotic effects of jatrophane diterpenoids for the first time.


Subject(s)
Diterpenes/pharmacology , Euphorbia/chemistry , Macrophages/drug effects , Osteogenesis/drug effects , RANK Ligand/metabolism , Cell Line , Crystallography, X-Ray , Diterpenes/chemistry , Molecular Structure , RANK Ligand/chemistry , Structure-Activity Relationship
5.
J Nat Prod ; 83(4): 1174-1182, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32237724

ABSTRACT

The increase of bone-resorbing osteoclast activity in bone remodeling is the major characteristic of various bone diseases. Thus, inhibiting osteoclastogenesis and bone-resorbing function may be an effective therapeutic target for bone diseases. Betulinic acid (BA), a natural plant-derived pentacyclic triterpenoid compound, is known to possess numerous pharmacological and biochemical properties including anti-inflammatory, anticancer, and antiadipogenic activity. However, the effect of BA on osteoclast differentiation and function in bone metabolism has not been demonstrated so far. In this study, we investigated whether BA could suppress RANKL-induced osteoclastogenesis and bone resorption. Interestingly, BA significantly suppressed osteoclastogenesis by decreasing the phosphorylation of Akt and IκB, as well as PLCγ2-Ca2+ signaling, in pathways involved in early osteoclastogenesis as well as through the subsequent suppression of c-Fos and NFATc1. The inhibition of these pathways by BA was once more confirmed by retrovirus infection of constitutively active (CA)-Akt and CA-Ikkß retrovirus and measurement of Ca2+ influx. BA also significantly inhibited the expression of osteoclastogenesis-specific marker genes. Moreover, we found that BA administration restored the bone loss induced through acute lipopolysaccharide injection in mice by a micro-CT and histological analysis. Our findings suggest that BA is a potential therapeutic candidate for bone diseases involving osteoclasts.


Subject(s)
Bone Marrow Cells/drug effects , Bone Resorption/metabolism , NF-kappa B/antagonists & inhibitors , Osteogenesis/drug effects , Pentacyclic Triterpenes/pharmacology , Phospholipase C gamma/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction/drug effects , Animals , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Mice , Molecular Structure , Osteoclasts/drug effects , Pentacyclic Triterpenes/chemistry , Phospholipase C gamma/chemistry , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/chemistry , RANK Ligand/metabolism , Betulinic Acid
6.
Molecules ; 25(1)2020 Jan 03.
Article in English | MEDLINE | ID: mdl-31947859

ABSTRACT

Receptor activator of nuclear factor-κB ligand (RANKL) is a cytokine responsible for bone resorption. It binds its receptor RANK, which activates osteoporosis. High levels of osteoprotegerin (OPG) competitively binding RANKL limit formation of ligand-receptor complexes and enable bone mass maintenance. The new approach to prevent osteoporosis is searching for therapeutics that can bind RANKL and support OPG function. The aim of the study was to verify the hypothesis that isoflavones can form complexes with RANKL limiting binding of the cytokine to its receptor. Interactions of five isoflavones with RANKL were investigated by isothermal titration calorimetry (ITC), by in silico docking simulation and on Saos-2 cells. Daidzein and biochanin A showed the highest affinity for RANKL. Among studied isoflavones coumestrol, formononetin and biochanin A showed the highest potential for Saos-2 mineralization and were able to regulate the expression of RANKL and OPG at the mRNA levels, as well as osteogenic differentiation markers: alkaline phosphatase (ALP), collagen type 1, and Runt-related transcription factor 2 (Runx2). Comparison of the osteogenic activities of isoflavones showed that the use of physicochemical techniques such as ITC or in silico docking are good tools for the initial selection of substances showing a specific bioactivity.


Subject(s)
Bone Density Conservation Agents , Isoflavones , Molecular Docking Simulation , Osteogenesis/drug effects , Osteoporosis , RANK Ligand , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacology , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , Isoflavones/chemistry , Isoflavones/pharmacology , Osteoporosis/drug therapy , Osteoporosis/metabolism , Osteoporosis/pathology , RANK Ligand/agonists , RANK Ligand/chemistry , RANK Ligand/metabolism
7.
Sci Rep ; 9(1): 15434, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659208

ABSTRACT

We recently developed a partial peptide of receptor activator of nuclear factor-кB ligand (RANKL) known as microglial healing peptide 1 (MHP1-AcN), that inhibits Toll-like receptor (TLR)-related inflammation through RANKL/RANK signaling in microglia and macrophages without promoting osteoclast activation. The abnormal activation of TLRs contributes to the initiation and maintenance of psoriasis, which is a chronic inflammatory skin disease that involves the aberrant expression of proinflammatory cytokines and the subsequent dermal γδ T cell and T helper 17 (Th17) cell responses. The inhibition of TLR-mediated inflammation provides an important strategy to treat psoriasis. Here, we examined the preventative effects of MHP1-AcN in a mouse model of imiquimod (a TLR 7/8 agonist)-induced psoriasis. Topical imiquimod application induced psoriasis-like skin lesions on the ear and dorsal skin. Systemic administration of MHP1-AcN by daily subcutaneous injection significantly prevented the development of skin lesions, including erythema, scaling and thickening. Mice treated with MHP1-AcN showed reduced levels of skin Il6 mRNA at 32 h and reduced levels of Il23 and Il17a mRNA at d9. Serum levels of IL-6 and IL-23 were reduced at 32 h, and IL-17A was reduced at d9. These results indicated that MHP1-AcN could decrease imiquimod-induced IL-6, IL-23 and IL-17A production. MHP1-AcN is potentially an alternative treatment for psoriasis.


Subject(s)
Cytokines/immunology , Imiquimod/toxicity , Peptides/pharmacology , Psoriasis , RANK Ligand/chemistry , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Peptides/chemistry , Psoriasis/chemically induced , Psoriasis/immunology , Psoriasis/pathology , Psoriasis/prevention & control
8.
FEBS J ; 286(18): 3582-3593, 2019 09.
Article in English | MEDLINE | ID: mdl-31081236

ABSTRACT

Fibrosis is characterized by the progressive alteration of the tissue structure due to the excessive production of extracellular matrix (ECM). The signaling system encompassing Receptor Activator of Nuclear factor NF-κB Ligand (RANKL)/RANK/Osteoprotegerin (OPG) was discovered to play an important role in the regulation of ECM formation and degradation in bone tissue. However, whether and how this signaling pathway plays a role in liver or pulmonary ECM degradation is unclear up to now. Interestingly, increased decoy receptor OPG levels are found in fibrotic tissues. We hypothesize that RANKL can stimulate RANK on macrophages and initiate the process of ECM degradation. This process may be inhibited by highly expressed OPG in fibrotic conditions. In this case, RANKL mutants that can bind to RANK without binding to OPG might become promising therapeutic candidates. In this study, we built a structure-based library containing 44 RANKL mutants and found that the Q236 residue of RANKL is important for OPG binding. We show that RANKL_Q236D can activate RAW cells to initiate the process of ECM degradation and is able to escape from the obstruction by exogenous OPG. We propose that the generation of RANKL mutants with reduced affinity for OPG is a promising strategy for the exploration of new therapeutics against fibrosis.


Subject(s)
Fibrosis/genetics , Osteoprotegerin/chemistry , RANK Ligand/chemistry , Receptor Activator of Nuclear Factor-kappa B/chemistry , Animals , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/ultrastructure , Fibrosis/pathology , Humans , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Macrophages/chemistry , Macrophages/metabolism , Macrophages/pathology , Mice , NF-kappa B/genetics , Osteoprotegerin/genetics , Osteoprotegerin/ultrastructure , Protein Binding/genetics , Protein Conformation , RANK Ligand/ultrastructure , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/ultrastructure , Signal Transduction/genetics
9.
J Med Chem ; 62(11): 5370-5381, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31082234

ABSTRACT

Targeting RANKL/RANK offers the possibility of developing novel therapeutic approaches to treat bone metabolic diseases. Multiple efforts have been made to inhibit RANKL. For example, marketed monoclonal antibody drug Denosumab could inhibit the maturation of osteoclasts by binding to RANKL. This study is an original approach aimed at discovering small-molecule inhibitors impeding RANKL/RANK protein interaction. We identified compound 34 as a potent and selective RANKL/RANK inhibitor by performing structure-based virtual screening and hit optimization. Disruption of the RANKL/RANK interaction by 34 effectively inhibits RANKL-induced osteoclastogenesis and bone resorption. The expression of osteoclast marker genes was also suppressed by treatment of 34. Furthermore, 34 markedly blocked the NFATc1/c-fos pathway. Thus, our current work demonstrates that the chemical tractability of the difficult PPI (RANKL/RANK) target by a small-molecule compound 34 offers a potential lead compound to facilitate the development of new medications for bone-related diseases.


Subject(s)
RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Bone Resorption , Drug Evaluation, Preclinical , Molecular Docking Simulation , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Protein Binding/drug effects , Protein Conformation , RANK Ligand/chemistry , Small Molecule Libraries/metabolism , User-Computer Interface
10.
J Biol Chem ; 293(45): 17523-17535, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30254073

ABSTRACT

Previous structural studies of osteoprotegerin (OPG), a crucial negative regulator of bone remodeling and osteoclastogenesis, were mostly limited to the N-terminal ligand-binding domains. It is now known that the three C-terminal domains of OPG also play essential roles in its function by mediating OPG dimerization, OPG-heparan sulfate (HS) interactions, and formation of the OPG-HS-receptor activator of nuclear factor κB ligand (RANKL) ternary complex. Employing hydrogen-deuterium exchange MS methods, here we investigated the structure of full-length OPG in complex with HS or RANKL in solution. Our data revealed two noteworthy aspects of the OPG structure. First, we found that the interconnection between the N- and C-terminal domains is much more rigid than previously thought, possibly because of hydrophobic interactions between the fourth cysteine-rich domain and the first death domain. Second, we observed that two hydrophobic clusters located in two separate C-terminal domains directly contribute to OPG dimerization, likely by forming a hydrophobic dimerization interface. Aided by site-directed mutagenesis, we further demonstrated that an intact dimerization interface is essential for the biological activity of OPG. Our study represents an important step toward deciphering the structure-function relationship of the full-length OPG protein.


Subject(s)
Deuterium Exchange Measurement , Mass Spectrometry , Osteoprotegerin/chemistry , Protein Multimerization , Animals , Heparitin Sulfate/chemistry , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Mice , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Protein Domains , RANK Ligand/chemistry , RANK Ligand/genetics , RANK Ligand/metabolism
11.
Nature ; 561(7722): 195-200, 2018 09.
Article in English | MEDLINE | ID: mdl-30185903

ABSTRACT

Receptor activator of nuclear factor-kappa B (RANK) ligand (RANKL) binds RANK on the surface of osteoclast precursors to trigger osteoclastogenesis. Recent studies have indicated that osteocytic RANKL has an important role in osteoclastogenesis during bone remodelling; however, the role of osteoblastic RANKL remains unclear. Here we show that vesicular RANK, which is secreted from the maturing osteoclasts, binds osteoblastic RANKL and promotes bone formation by triggering RANKL reverse signalling, which activates Runt-related transcription factor 2 (Runx2). The proline-rich motif in the RANKL cytoplasmic tail is required for reverse signalling, and a RANKL(Pro29Ala) point mutation reduces activation of the reverse signalling pathway. The coupling of bone resorption and formation is disrupted in RANKL(Pro29Ala) mutant mice, indicating that osteoblastic RANKL functions as a coupling signal acceptor that recognizes vesicular RANK. RANKL reverse signalling is therefore a potential pharmacological target for avoiding the reduced bone formation associated with inhibition of osteoclastogenesis.


Subject(s)
Bone Resorption/metabolism , Osteogenesis , RANK Ligand/metabolism , Signal Transduction , Amino Acid Substitution , Animals , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Cross-Linking Reagents/chemistry , Cytoplasmic Vesicles/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , RANK Ligand/chemistry , RANK Ligand/deficiency , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism
12.
Biochem Biophys Res Commun ; 499(3): 648-654, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29604276

ABSTRACT

Bone homeostasis is maintained by a dynamic balance between osteoblastic bone formation and osteoclastic bone resorption. The receptor activator of nuclear-κB ligand (RANKL) is essential for the function of the bone-resorbing osteoclasts, and targeting RANKL has been proved highly successful in osteoporosis patients. This study aimed to design a novel vaccine targeting RANKL and evaluate its therapeutic effects in OVX-induced bone loss model. Anti-RANKL vaccine was generated by incorporating the unnatural amino acid p-nitrophenylalanine (pNO2Phe) into selected sites in the murine RANKL (mRANKL) molecule. Specifically, mutation of a single tyrosine residue Tyr234 (Y234) or Tyr240 (Y240) of mRANKL to pNO2Phe (thereafter named as Y234pNO2Phe or Y240pNO2Phe) induced a high titer antibody response in mice, whereas no significant antibody response was observed for the wild type mRANKL (WT mRANKL). The antiserum induced by Y234pNO2Phe or Y240pNO2Phe could efficiently prevent osteoclastogenesis in vitro. Moreover, immunization with Y234pNO2Phe or Y240pNO2Phe could also prevent OVX-induced bone loss in mice, suggesting that selected pNO2Phe-substituted mRANKL may pave the way for creating a novel vaccine to treat osteoporosis.


Subject(s)
Bone Resorption/immunology , Bone Resorption/prevention & control , Ovariectomy/adverse effects , Phenylalanine/analogs & derivatives , RANK Ligand/chemistry , Vaccines/immunology , Amino Acid Sequence , Animals , Bone Resorption/etiology , Bone Resorption/pathology , Cell Differentiation , Female , Immunization , Immunoglobulin G/metabolism , Mice, Inbred C57BL , Osteoclasts/metabolism , Phenylalanine/chemistry
13.
J Phys Chem B ; 121(39): 9141-9148, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28945380

ABSTRACT

Recent studies have revealed the importance and the active contribution of the RANKL/OPG/RANK pathway in many bone diseases including different forms of common osteoporosis. In this study, we present an extensive atomistic molecular dynamic study of the OPG/RANKL system. Within the molecular models, we varied the number of OPG molecules bound to the RANKL trimer and carried out a study to determine how the binding affinity of the OPG/RANKL system changes as a function of OPG concentration. The molecular mechanics Poisson-Boltzmann surface area method was used to analyze binding free energies. It is shown that the binding affinity decreases with increasing numbers of OPG molecules. Additionally, conformational changes of RANKL, interactions between the N-terminus outlier module of OPG with RANKL, and residues that play an important role in the binding of OPG to RANKL trimer were investigated. A probable cause for unfavorable binding for a third OPG molecule was found. Along with the currently available experimental studies, this computational study will be valuable for the comprehensive understanding of OPG/RANKL at the atomistic level.


Subject(s)
Molecular Dynamics Simulation , Osteoprotegerin/chemistry , RANK Ligand/chemistry , Humans , Models, Biological , Protein Binding , Thermodynamics
14.
ChemMedChem ; 12(20): 1697-1702, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28885764

ABSTRACT

Receptor activator of NF-κB (RANK), a member of the TNF-receptor superfamily, plays an important role in bone resorption and stimulates immune and epithelial cell activation. Denosumab, a human monoclonal antibody that blocks the RANK ligand (RANKL), is approved for the treatment of osteoporosis and bone metastasis. However, a small molecule that inhibits the RANK-RANKL interaction would be beneficial to decrease cost and to facilitate treatments with orally available therapeutic agents. Herein we report the discovery of the first nonpeptidic inhibitors of RANK-RANKL interactions. In screening a chemical library by competitive ELISA, the porphyrin verteporfin was identified as a hit. Derivatives were screened, and the chlorin-macrocycle-containing pheophorbide A and purpurin 18 were found to bind recombinant RANKL, to inhibit RANK-RANKL interactions in the ELISA, and to suppress the RANKL-dependent activation of model cells and the differentiation of RANK-expressing precursors into osteoclasts. This discovery of a family of small molecules that inhibit RANK activation presents an initial basis for further development of nonpeptidic therapeutic agents targeting the interaction between RANK and RANKL.


Subject(s)
Porphyrins/pharmacology , RANK Ligand/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Animals , Cell Survival , Humans , Jurkat Cells , Mice , Molecular Structure , Osteogenesis/drug effects , Porphyrins/chemistry , Protein Binding , RANK Ligand/chemistry , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/chemistry , Receptor Activator of Nuclear Factor-kappa B/metabolism , Structure-Activity Relationship
15.
FEBS J ; 284(15): 2501-2512, 2017 08.
Article in English | MEDLINE | ID: mdl-28627025

ABSTRACT

Bone is a dynamic tissue that is maintained by continuous renewal. An imbalance in bone resorption and bone formation can lead to a range of disorders, such as osteoporosis. The receptor activator of NF-κB (RANK)-RANK-ligand (RANKL) pathway plays a major role in bone remodeling. Here, we investigated the effect of mutations at position I248 in the DE-loop of murine RANKL on the interaction of RANKL with RANK, and subsequent activation of osteoclastogenesis. Two single mutants, RANKL I248Y and I248K, were found to maintain binding and have the ability to reduce wild-type RANKL-induced osteoclastogenesis. The generation of RANK-antagonists is a promising strategy for the exploration of new therapeutics against osteoporosis.


Subject(s)
Mutation , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Amino Acid Substitution , Animals , Computational Biology , Energy Transfer , Expert Systems , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Kinetics , Mice , Mutagenesis, Site-Directed , Osteoclasts/cytology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Multimerization , RANK Ligand/chemistry , RANK Ligand/genetics , RAW 264.7 Cells , Receptor Activator of Nuclear Factor-kappa B/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
16.
Appl Immunohistochem Mol Morphol ; 25(5): 299-307, 2017.
Article in English | MEDLINE | ID: mdl-28248730

ABSTRACT

The expression and tissue distribution of RANK (Receptor Activator of Nuclear Factor κ B) and RANK Ligand (RANKL) are of critical interest in relation to efficacy and safety of antibodies against RANK or RANKL that are approved or under consideration as potential therapeutic agents. Data from the literature using protein or mRNA analyses of rodent and human tissues or immunohistochemical (IHC) studies with a variety of antibodies and methods have provided some background of the distribution of RANK and RANKL but have yielded inconsistent findings. The present study reports the generation of carefully validated antibodies to RANK and RANKL and the development of an optimized IHC method, with confirmatory data from 2 well-validated alternative protocols that were developed and performed in separate laboratories at USC and at Amgen. Tissue expression of RANK and RANKL is reported for the optimized IHC assay.


Subject(s)
Antibodies/metabolism , Immunohistochemistry/methods , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Animals , Humans , Immunohistochemistry/standards , Mice , RANK Ligand/chemistry , Receptor Activator of Nuclear Factor-kappa B/chemistry , Tissue Distribution
17.
Acta Biomater ; 49: 306-315, 2017 02.
Article in English | MEDLINE | ID: mdl-27940164

ABSTRACT

PURPOSE: To develop a nanofiber hydrogel (NF-hydrogel) for sustained and controlled release of the recombinant receptor activator of NF-kB ligand; (RANKL) and to characterize the release kinetics and bioactivity of the released RANKL. METHODS: Various concentrations of fluorescently-labelled RANKL protein were added to NF-hydrogels, composed of Acetyl-(Arg-Ala-Asp-Ala)4-CONH2 [(RADA)4] of different concentrations, to investigate the resulting in vitro release rates. The nano-structures of NF-hydrogel, with and without RANKL, were determined using atomic force microscopy (AFM). Released RANKL was further analyzed for changes in secondary and tertiary structure using CD spectroscopy and fluorescent emission spectroscopy, respectively. Bioactivity of released RANKL protein was determined using NFATc1 gene expression and tartrate resistant acid phosphatase (TRAP) activity of osteoclast cells as biomarkers. RESULTS: NF-hydrogel concentration dependent sustained release of RANKL protein was measured at concentrations between 0.5 and 2%(w/v). NF-hydrogel at 2%(w/v) concentration exhibited a sustained and slow-release of RANKL protein up to 48h. Secondary and tertiary structure analyses confirmed no changes to the RANKL protein released from NF-hydrogel in comparison to native RANKL. The results of NFATc1 gene mRNA expression and TRAP activities of osteoclast, showed that the release process did not affect the bioactivity of released RANKL. CONCLUSIONS: This novel study is the first of its kind to attempt in vitro characterization of NF-hydrogel based delivery of RANKL protein to induce osteoclastogenesis. We have shown the self-assembling NF-hydrogel peptide system is amenable to the sustained and controlled release of RANKL locally; that could in turn increase local concentration of RANKL to induce osteoclastogenesis, for application to the controlled mobilization of tooth movement in orthodontic procedures. STATEMENT OF SIGNIFICANCE: Orthodontic tooth movement (OTM) occurs through controlled application of light forces to teeth, facilitating the required changes in the surrounding alveolar bone through the process of bone remodelling. The RANKL system regulates alveolar bone remodelling and controls root resorption during OTM. The use of exogenous RANKL to accelerate OTM has not been attempted to date because large quantities of RANKL for systemic therapy may subsequently cause serious systemic loss of skeletal bone. The controlled and sustained local release of RANKL from a carrier matrix could maximize its therapeutic benefit whilst minimizing systemic side effects. In this study a NF-hydrogel was used for sustained and controlled release of RANKL and the release kinetics and biofunctionality of the released RANKL was characterized. Our results provide fundamental insight for further investigating the role of RANKL NF-hydrogel release systems for inducing osteoclastogenesis in vivo.


Subject(s)
Hydrogels/pharmacology , Nanofibers/chemistry , Osteoclasts/cytology , Osteogenesis/drug effects , RANK Ligand/pharmacology , Animals , Circular Dichroism , Drug Liberation , Humans , Kinetics , Mice , Microscopy, Atomic Force , Nanofibers/ultrastructure , Osteoclasts/drug effects , Osteoclasts/metabolism , Protein Structure, Secondary , RANK Ligand/chemistry , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Tartrate-Resistant Acid Phosphatase/metabolism
18.
J Nat Prod ; 80(1): 90-95, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28026175

ABSTRACT

Six new spongian diterpene derivatives, ceylonins A-F (1-6), were isolated from the Indonesian marine sponge Spongia ceylonensis along with spongia-13(16),14-dien-19-oic acid (7). They contained three additional carbons in ring D to supply an ether-bridged bicyclic ring system. Their structures were elucidated by analyzing NMR spectroscopic data and calculated ECD spectra in comparison to experimental ECD spectra. The bicyclic ring system may be derived from the major metabolite 7 and a C3 unit (an acrylic acid equivalent) through an intermolecular Diels-Alder reaction, which was experimentally supported by the formation of 1-6 from 7 and acrylic acid. The inhibitory effects of the isolated compounds on the RANKL-induced formation of multinuclear osteoclasts in RAW264 macrophages were examined.


Subject(s)
Diterpenes/isolation & purification , Diterpenes/pharmacology , Osteoclasts/drug effects , Porifera/chemistry , RANK Ligand/pharmacology , Animals , Diterpenes/chemistry , Indonesia , Magnetic Resonance Spectroscopy , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Osteoclasts/chemistry , RANK Ligand/chemistry
19.
J Biol Chem ; 291(46): 24160-24171, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27697839

ABSTRACT

Osteoprotegerin (OPG), a decoy receptor secreted by osteoblasts, is a major negative regulator of bone resorption. It functions by neutralizing the receptor activator of nuclear factor κB ligand (RANKL), which plays a central role in promoting osteoclastogenesis. OPG is known to be a high-affinity heparan sulfate (HS)-binding protein. Presumably, HS could regulate the function of OPG and affect how it inhibits RANKL. However, the molecular detail of HS-OPG interaction remains poorly understood, which hinders our understanding of how HS functions in osteoclastogenesis. Here we report mapping of the HS-binding site of OPG. The HS-binding site, identified by mutagenesis study, consists of eight basic residues that are located mostly at the junction of the second death domain and the C-terminal domain. We further show that heparin-derived dodecasaccharide is able to induce dimerization of OPG monomers with a stoichiometry of 1:1. Small-angle X-ray scattering analysis revealed that upon binding of HS, OPG undergoes a dramatic conformational change, resulting in a more compact and less flexible structure. Importantly, we present here three lines of evidence that HS, OPG, and RANKL form a stable ternary complex. Using a HS binding-deficient OPG mutant, we further show that in an osteoblast/bone marrow macrophage co-culture system, immobilization of OPG by HS at the osteoblast cell surface substantially lowers the inhibitory threshold of OPG toward RANKL. These discoveries strongly suggest that HS plays an active role in regulating OPG-RANKL interaction and osteoclastogenesis.


Subject(s)
Heparitin Sulfate/metabolism , Macrophages/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoprotegerin/metabolism , Animals , Binding Sites , Cell Line , Coculture Techniques , Heparitin Sulfate/chemistry , Heparitin Sulfate/genetics , Macrophages/cytology , Mice , Osteoblasts/cytology , Osteoclasts/cytology , Osteoprotegerin/chemistry , Osteoprotegerin/genetics , RANK Ligand/chemistry , RANK Ligand/genetics , RANK Ligand/metabolism
20.
Orthod Craniofac Res ; 19(4): 198-208, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27647720

ABSTRACT

OBJECTIVES: To test the following two hypotheses: 1) different types of retainers result in distinct levels of biomarkers in gingival crevicular fluid (GCF) and 2) the retainer bonded to all mandibular anterior teeth induces more detrimental outcomes to the periodontium. SETTING AND SAMPLE POPULATION: The Department of Orthodontics at the University of Florida. The population consisted of individuals in the retention phase of orthodontic treatment. MATERIAL AND METHODS: This was a cross-sectional study that enrolled 36 individuals. Subjects in group 1 had retainers bonded to the mandibular canines only. Group 2 consisted of individuals having retainers bonded to all mandibular anterior teeth. Group 3 included patients using mandibular removable retainers. After clinical examination, GCF was collected from the mandibular incisor and biomarker levels were compared between the groups. RESULTS: Plaque accumulation and gingivitis differed significantly among groups, with the highest median values in group 2 subjects. Pairwise comparison of the groups with respect to gingivitis showed significant differences between groups 1 and 2. Significant differences among groups were detected for RANKL, OPG, OPN, M-CSF, MMP-3, and MMP-9. The ratio RANKL/OPG was significantly higher in group 2 subjects, with pairwise comparisons indicating that groups 1 and 2 differed from group 3. CONCLUSION: An association was found between orthodontic retention groups and GCF biomarker levels, which should be further explored in longitudinal studies. The presence of retainers bonded to all anterior teeth seems to increase plaque accumulation and gingivitis.


Subject(s)
Biomarkers/chemistry , Dental Bonding/adverse effects , Dental Bonding/methods , Dental Plaque/etiology , Gingival Crevicular Fluid/chemistry , Gingival Recession/etiology , Gingivitis/etiology , Incisor/pathology , Incisor/physiopathology , Orthodontic Retainers/adverse effects , Adolescent , Adult , Cross-Sectional Studies , Cuspid , Dental Plaque Index , Female , Humans , Interleukin 1 Receptor Antagonist Protein/chemistry , Interleukin-1beta/chemistry , Interleukin-6/chemistry , Interleukin-8/chemistry , Macrophage Colony-Stimulating Factor/chemistry , Male , Mandible , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase 9/chemistry , Middle Aged , Orthodontic Appliance Design , Osteopontin/chemistry , Osteoprotegerin/chemistry , Periodontal Index , RANK Ligand/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...