Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Am Heart Assoc ; 9(17): e015731, 2020 09.
Article in English | MEDLINE | ID: mdl-32856519

ABSTRACT

Background Decreased extracellular matrix formation and few vascular smooth muscle cells (VSMCs) in cerebral vascular walls are the main characteristics of intracranial aneurysm (IA) pathogenesis. Recently, osteoprotegerin was reported to activate collagen biosynthesis and VSMC proliferation via the TGF-ß1 (transforming growth factor-ß1) signaling. This study aimed to investigate whether osteoprotegerin can prevent IA progression in rats through enhanced collagen expression and VSMC proliferation. Methods and Results IAs were surgically induced in 7-week-old male Sprague-Dawley rats; at 1-week post-operation, recombinant mouse osteoprotegerin or vehicle control was continuously infused for 4 weeks into the lateral ventricle using an osmotic pump. In the osteoprotegerin-treatment group, the aneurysmal size was significantly smaller (37.5 µm versus 60.0 µm; P<0.01) and the media of IA walls was thicker (57.1% versus 36.0%; P<0.01) than in the vehicle-control group. Type-I and type-III collagen, TGF-ß1, phosphorylated Smad2/3, and proliferating cell nuclear antigen were significantly upregulated in the IA walls of the osteoprotegerin group than that in the control group. No significant difference was found in the expression of proinflammatory genes between the groups. In mouse VSMC cultures, osteoprotegerin treatment upregulated the expression of collagen and TGF-ß1 genes, and activated VSMC proliferation; the inhibition of TGF-ß1 signaling nullified this effect. Conclusions Osteoprotegerin suppressed the IA progression by a unique mechanism whereby collagen biosynthesis and VSMC proliferation were activated via TGF-ß1 without altering proinflammatory gene expression. Osteoprotegerin may represent a novel therapeutic target for IAs.


Subject(s)
Collagen/biosynthesis , Intracranial Aneurysm/genetics , Muscle, Smooth, Vascular/metabolism , Osteoprotegerin/metabolism , Animals , Case-Control Studies , Cell Proliferation , Disease Progression , Infusion Pumps , Infusions, Intraventricular , Male , Models, Animal , Osteoprotegerin/administration & dosage , Osteoprotegerin/genetics , Osteoprotegerin/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
2.
Aging Male ; 23(3): 189-201, 2020 Sep.
Article in English | MEDLINE | ID: mdl-30146916

ABSTRACT

Aim: The aim of this work is to compare the effects of osteoprotegerin (OPG) and testosterone on bone quality in a model of orchidectomised (ORX) rats.Methods: Three-month-old ORX or SHAM operated groups (n = 15 each group) were used. The SHAM and ORX groups received saline. There were two ORX groups, receiving OPG-Fc (10 mg/kg twice weekly) (ORX + OPG-Fc) or testosterone cypionate (1.7 mg/kg/weekly) for 8 weeks. After sacrifice, bone analysis by femoral and lumbar dual-energy X-ray absorptiometry and micro-computed tomography in femora were performed. Histological sections of vertebrae were dyed with hematoxylin-eosin or safranin. Serum osteocalcin (BGP), total alkaline phosphatase (ALP), and C-terminal telopeptide of type I collagen (CTX) were analyzed.Results: ORX resulted in femoral and vertebral bone loss and in microarchitectural deterioration. Treatment with OPG-Fc and testosterone recovered lumbar (L) and femoral (F) bone mineral densitometry bone mineral density (BMD) to SHAM levels. Femoral BMD was significantly higher after treatment with OPG-Fc than after testosterone treatment due to the presence of osteopetrotic changes in the metaphyseal region of long bones. Serum levels of ALP and CTX increased, while OPG levels were unchanged in ORX rats. Treatment with OPG-Fc decreased the levels of BGP, ALP, and CTX. Treatment with testosterone maintained biochemical markers of bone turnover at levels similar to or higher than those of ORX rats.


Subject(s)
Bone Density , Osteoprotegerin/pharmacology , Testosterone/pharmacology , Animals , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Humans , Male , Orchiectomy , Osteoprotegerin/administration & dosage , Osteoprotegerin/blood , Random Allocation , Rats , Rats, Wistar , Spine/diagnostic imaging , Spine/drug effects , Spine/pathology , Testosterone/administration & dosage , Testosterone/blood
3.
Eur J Orthod ; 41(1): 1-8, 2019 01 23.
Article in English | MEDLINE | ID: mdl-29608684

ABSTRACT

Background: Because orthodontic tooth movement is dependent upon osteoclast-mediated resorption of alveolar bone adjacent to the pressure side of tooth roots, biologic mediators that regulate osteoclasts can be utilized to control tooth movement. Objectives: To develop a novel method to locally enhance orthodontic anchorage. Methods: We encapsulated osteoprotegerin (OPG) in polymer microspheres and tested the effectiveness of microsphere encapsulated versus non-encapsulated OPG for enhancing orthodontic anchorage in a rodent model of tooth movement. A single injection of 1 mg/kg non-encapsulated or microsphere encapsulated OPG was delivered into the palatal mucosa mesial to the first maxillary molar 1 day prior to tooth movement. A positive control group received injections of 5 mg/kg non-encapsulated OPG every 3 days during tooth movement. After 28 days of tooth movement, hemi-maxillae and femurs were dissected. Molar mesial and incisor distal tooth movement was measured using stone casts that were scanned and magnified. Local alveolar, distant femur bone, and tooth root volumes were analyzed by micro computed tomography. Serum OPG levels were measured by ELISA. Osteoclast numbers were quantified by histomorphometry. Results: The single injection of microsphere encapsulated OPG significantly enhanced orthodontic anchorage, while the single injection of non-encapsulated OPG did not. Injection of encapsulated OPG inhibited molar mesial movement but did not inhibit incisor tooth movement, and did not alter alveolar or femur bone volume fraction, density, or mineral content. Multiple injections of 5 mg/kg non-encapsulated OPG enhanced orthodontic anchorage, but also inhibited incisor retraction and altered alveolar and femur bone quality parameters. Increased OPG levels were found only in animals receiving multiple injections of non-encapsulated 5 mg/kg OPG. Osteoclast numbers were higher upon tooth movement in animals that did not receive OPG. Osteoclast numbers in OPG injected animals were variable within groups. Conclusions: Microsphere encapsulation of OPG allows for controlled drug release, and enhances site-specific orthodontic anchorage without systemic side effects. With additional refinements, this drug delivery system could be applicable to a broad array of potential biologic orthodontic therapeutics.


Subject(s)
Bone Resorption/prevention & control , Orthodontic Anchorage Procedures/methods , Osteoprotegerin/administration & dosage , Tooth Movement Techniques/methods , Animals , Bone Resorption/diagnostic imaging , Drug Delivery Systems , Drug Evaluation, Preclinical , Femur/diagnostic imaging , Femur/drug effects , Incisor/diagnostic imaging , Incisor/drug effects , Male , Microspheres , Molar/diagnostic imaging , Molar/drug effects , Osteoclasts/drug effects , Osteoprotegerin/therapeutic use , Rats, Sprague-Dawley , X-Ray Microtomography
4.
Bone ; 116: 162-170, 2018 11.
Article in English | MEDLINE | ID: mdl-30077758

ABSTRACT

Serum calcium (Ca) is maintained in a narrow range through regulation of Ca metabolism in the intestine, kidney, and bone. Calcium is incorporated and resorbed from bone during bone remodeling via cellular processes as well as by exchange. Both routes contribute to calcium homeostasis. To assess the magnitude of bone turnover contribution to calcium homeostasis we labeled bone with a Ca tracer and measured Ca release following stimulation or suppression of bone resorption. Young growing male rats (n = 162) were dosed with 45Ca to label skeletal Ca. After a one-month period to allow the label to incorporate into the skeleton, rats were treated with a bone resorption antagonist (OPG), a bone resorption agonist (RANKL), or vehicle control (PBS). Serum and urine 45Ca and total Ca, and serum TRACP5b (a bone resorption biomarker), were monitored for 45 days following treatment. Tracer data were analyzed by a compartmental model using WinSAAM to quantify dynamic changes in Ca metabolism and identify sites of change following treatment. In RANKL treated rats, both serum 45Ca and serum TRACP5b were increased by >70% due to a 25-fold increase in bone resorption. In OPG treated rats, both serum 45Ca and serum TRACP5b were suppressed by >70% due to a 75% decrease in bone resorption, a 3-fold increase in bone formation, and a 50% increase in absorption. Because TRACP5b and 45Ca responded similarly, we conclude that Ca release from bone into serum occurs mostly via osteoclast-mediated bone resorption. However, because serum Ca concentration did not change with altered resorption in response to either RANKL or OPG treatment, we also conclude that serum Ca concentration under normal dietary conditions in young growing male rats is maintained by processes in addition to cellular bone resorption.


Subject(s)
Bone Resorption/blood , Calcium/blood , Growth and Development , Osteoprotegerin/metabolism , Animals , Body Weight/drug effects , Bone Resorption/urine , Calcium/urine , Male , Models, Biological , Osteoprotegerin/administration & dosage , Osteoprotegerin/pharmacology , RANK Ligand/administration & dosage , RANK Ligand/pharmacology , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase/metabolism
5.
Eur J Orthod ; 38(4): 379-85, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26293288

ABSTRACT

BACKGROUND AND OBJECTIVES: Anchorage is one of the most challenging sides in orthodontics. The use of biological modulators that inhibit osteoclasts could be a solution to address these problems and provide new adjunctive approaches. The aim of this study was to assess the effectiveness of recombinant osteoprotegerin fusion protein (OPG-Fc) in orthodontic anchorage. MATERIALS AND METHODS: Two groups of male Sprague-Dawley rats were utilized. The animals in the experimental group received twice-weekly injections with high dose of OPG-Fc (5.0mg/kg) in mesial and distal mucosa of the first molars, and those in the control group received no drugs. Right first maxillary molars were mesialized using a calibrated nickel-titanium spring connected to an anterior mini-screw. Tooth movement was measured by two blinded observers using scanned and magnified stone casts. Receptor activator of nuclear factor κB (RANK), run-related transcription factor 2 (Runx2), type I collagen, vimentin, matrix metalloproteinases 2 and 9, S100 protein and the putative mechanoproteins acid-sensing ion channel (ASIC2) and transient receptor potential vainilloid 4 (TRPV4) were evaluated using immunohistochemistry. RESULTS: OPG-Fc group showed an important decreased in mesial molar movement with only 52%, 31%, and 22% of the total mesial molar movement compared with control group at Days 7, 14, and 21, respectively (P < 0.001). RANK ligand and Runx2 positive cells were severely reduced after OPG-Fc treatment. Periodontal ligament architecture, cell arrangement, and immunohistochemical patter for vimentin, type I collagen and the mechanoproteins TRPV4 and ASIC2 were altered by tooth movement and all these parameters altered by the applied treatment. CONCLUSIONS: OPG-Fc effectively inhibits osteoclastogenesis resulting in improved bone quantity and orthodontic anchorage. Based on present results, OPG-Fc could have clinical utility in preventing undesired tooth movements.


Subject(s)
Osteoprotegerin/pharmacology , Tooth Mobility/prevention & control , Tooth Movement Techniques/methods , Animals , Drug Administration Schedule , Drug Evaluation, Preclinical/methods , Male , Maxilla , Molar/drug effects , Molar/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoprotegerin/administration & dosage , Periodontal Ligament/drug effects , RANK Ligand/metabolism , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Tooth Mobility/physiopathology
6.
Arthritis Res Ther ; 17: 251, 2015 Sep 12.
Article in English | MEDLINE | ID: mdl-26373710

ABSTRACT

INTRODUCTION: We designed OP3-4 (YCEIEFCYLIR), a cyclic peptide, to mimic the soluble osteoprotegerin (OPG), and was proven to bind to RANKL (receptor activator of NF-κB ligand), thereby inhibiting osteoclastogenesis. We recently found that another RANKL binding peptide, W9, could accelerate bone formation by affecting RANKL signaling in osteoblasts. We herein demonstrate the effects of OP3-4 on bone formation and bone loss in a murine model of rheumatoid arthritis. METHODS: Twenty-four seven-week-old male DBA/1J mice were used to generate a murine model of collagen-induced arthritis (CIA). Then, vehicle or OP3-4 (9 mg/kg/day or 18 mg/kg/day) was subcutaneously infused using infusion pumps for three weeks beginning seven days after the second immunization. The arthritis score was assessed, and the mice were sacrificed on day 49. Thereafter, radiographic, histological and biochemical analyses were performed. RESULTS: The OP3-4 treatment did not significantly inhibit the CIA-induced arthritis, but limited bone loss. Micro-CT images and quantitative measurements of the bone mineral density revealed that 18 mg/kg/day OP3-4 prevented the CIA-induced bone loss at both articular and periarticular sites of tibiae. As expected, OP3-4 significantly reduced the CIA-induced serum CTX levels, a marker of bone resorption. Interestingly, the bone histomorphometric analyses using undecalcified sections showed that OP3-4 prevented the CIA-induced reduction of bone formation-related parameters at the periarticular sites. CONCLUSION: The peptide that mimicked OPG prevented inflammatory bone loss by inhibiting bone resorption and stimulating bone formation. It could therefore be a useful template for the development of small molecule drugs for inflammatory bone loss.


Subject(s)
Arthritis, Experimental/drug therapy , Bone Resorption/prevention & control , Cartilage, Articular/drug effects , Oligopeptides/pharmacology , RANK Ligand/metabolism , Amino Acid Sequence , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Bone Density/drug effects , Bone Resorption/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Differentiation/drug effects , Cells, Cultured , Collagen Type I/blood , Enzyme-Linked Immunosorbent Assay , Infusions, Subcutaneous , Male , Mice, Inbred DBA , Oligopeptides/administration & dosage , Oligopeptides/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteoprotegerin/administration & dosage , Osteoprotegerin/metabolism , Osteoprotegerin/pharmacology , Peptides/blood , Protein Binding , Tibia/drug effects , Tibia/metabolism , Tibia/pathology , X-Ray Microtomography
7.
Oncol Rep ; 34(6): 3007-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26398902

ABSTRACT

Receptor activator for nuclear factor κB ligand (RANKL) is a member of the tumor necrosis factor (TNF) family. The interaction between RANKL and its receptor RANK plays an important role in the development and function of diverse tissues. However, the expression and role of RANKL in cervical cancer are still unknown. In the present study, we found that RANKL and RANK were highly co-expressed in cervical cancer. HeLa and SiHa cells secreted soluble RANKL (sRANKL), expressed member RANKL (mRANKL) and RANK. Recombinant human RANKL protein had no effect on the viability of HeLa and SiHa cells. Yet, blocking RANKL with an anti-human RANKL neutralizing antibody (α-RANKL) or recombinant human osteoprotegrin (OPG) protein resulted in the downregulation of Ki-67 and B-cell lymphoma 2 (Bcl-2) expression and an increase in Fas and Fas ligand (FasL) expression, as well as a high level of viability and a low level of apoptosis in the HeLa and SiHa cells. In addition, α-RANKL led to a decrease in IL-8 secretion. Recombinant human IL-8 protein reversed the effect of α-RANKL on the expression of proliferation- and apoptosis­related molecules, and proliferation and apoptosis in the HeLa and SiHa cells. The present study suggests that a high level of mRANKL/RANK expression in cervical cancer lesions plays an important role in the rapid growth of cervical cancer cells possibly through strengthening the dialogue between cervical cancer cells and regulation of IL-8 secretion, which may be a possible target for cervical cancer therapy.


Subject(s)
Interleukin-8/biosynthesis , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Uterine Cervical Neoplasms/genetics , Antibodies, Neutralizing/administration & dosage , Apoptosis/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Fas Ligand Protein/biosynthesis , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Interleukin-8/genetics , Osteoprotegerin/administration & dosage , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RANK Ligand/antagonists & inhibitors , RANK Ligand/biosynthesis , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Uterine Cervical Neoplasms/pathology
8.
Biomacromolecules ; 16(8): 2374-81, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26151628

ABSTRACT

Polymers of similar molecular weights and chemical constitution but varying in their macromolecular architectures were conjugated to osteoprotegerin (OPG) to determine the effect of polymer topology on protein activity in vitro and in vivo. OPG is a protein that inhibits bone resorption by preventing the formation of mature osteoclasts from the osteoclast precursor cell. Accelerated bone loss disorders, such as osteoporosis, rheumatoid arthritis, and metastatic bone disease, occur as a result of increased osteoclastogenesis, leading to the severe weakening of the bone. OPG has shown promise as a treatment in bone disorders; however, it is rapidly cleared from circulation through rapid liver uptake, and frequent, high doses of the protein are necessary to achieve a therapeutic benefit. We aimed to improve the effectiveness of OPG by creating OPG-polymer bioconjugates, employing reversible addition-fragmentation chain transfer polymerization to create well-defined polymers with branching densities varying from linear, loosely branched to densely branched. Polymers with each of these architectures were conjugated to OPG using a "grafting-to" approach, and the bioconjugates were characterized by sodium dodecyl sulfate polyacrylamide gel electrophoresis. The OPG-polymer bioconjugates showed retention of activity in vitro against osteoclasts, and each bioconjugate was shown to be nontoxic. Preliminary in vivo studies further supported the nontoxic characteristics of the bioconjugates, and measurement of the bone mineral density in rats 7 days post-treatment via peripheral quantitative computed tomography suggested a slight increase in bone mineral density after administration of the loosely branched OPG-polymer bioconjugate.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Bone Resorption/drug therapy , Osteoporosis/drug therapy , Osteoprotegerin/chemistry , Animals , Arthritis, Rheumatoid/pathology , Bone Density/drug effects , Bone Resorption/pathology , Humans , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoporosis/pathology , Osteoprotegerin/administration & dosage , Polymers/administration & dosage , Polymers/chemistry , Rats
9.
Orthod Craniofac Res ; 18 Suppl 1: 187-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25865548

ABSTRACT

OBJECTIVES: To determine minimal dose levels required for local inhibition of orthodontic relapse by recombinant OPG protein (OPG-Fc), while also determining effects of injected OPG-Fc on alveolar bone and long bone. SETTING AND SAMPLE POPULATION: The Department of Orthodontics and Pediatric Dentistry at the University of Michigan. Eighteen male Sprague Dawley rats. MATERIALS & METHODS: Maxillary molars were moved with nickel-titanium springs and then allowed to relapse in Sprague Dawley rats. Upon appliance removal, animals were injected with a single dose of 1.0 mg/kg OPG-Fc, 0.1 mg/kg OPG-Fc, or phosphate-buffered saline (vehicle) just distal to the molar teeth. Tooth movement measurements were made from stone casts, which were scanned and digitally measured. Alveolar tissues were examined by histology. Micro-computed tomography was used to quantify changes in alveolar and femur bone. RESULTS: Local injection of OPG-Fc inhibited molar but not incisor relapse, when compared to vehicle-injected animals. No significant differences in alveolar or femur bone were seen between the three treatment groups after 24 days of relapse. CONCLUSIONS: Our results demonstrate that a single local injection of OPG-Fc effectively inhibits orthodontic relapse, with minimal systemic bone metabolic effects. Our results also show that a single injection of OPG-Fc will influence tooth movement only in teeth close to the injection site. These findings indicate that OPG-Fc has potential as a safe and effective pharmacological means to locally control osteoclasts, for uses such as maintaining anchorage during orthodontic tooth movement and preventing orthodontic relapse in humans.


Subject(s)
Alveolar Process/drug effects , Bone Resorption/prevention & control , Osteoprotegerin/therapeutic use , Tooth Movement Techniques/methods , Alveolar Process/pathology , Animals , Bone Density/drug effects , Bone Remodeling/drug effects , Bone Resorption/pathology , Femur/drug effects , Femur/pathology , Incisor/drug effects , Injections , Male , Maxilla/drug effects , Maxilla/pathology , Models, Dental , Molar/drug effects , Orthodontic Wires , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoprotegerin/administration & dosage , Pharmaceutical Vehicles , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Recurrence , Tooth Movement Techniques/instrumentation , X-Ray Microtomography/methods
10.
Mol Med Rep ; 12(1): 631-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25738879

ABSTRACT

The aim of the present study was to compare the osteoclast-inhibiting ability of recombinant osteoprotegerin (OPG) protein (rhOPG-Fc) and recombinant receptor activator of nuclear factor κB (rhRANK) in vitro and in vivo. Osteoclasts were cultured with either rhOPG-Fc or rhRANK for 9 days. The number of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells and resorption pits in bone slices were then counted. In the in vivo investigation, female mice were bilaterally ovariectomized (OVX) and intraperitoneally injected with 3 mg/kg rhOPG-Fc or rhRANK for 12 weeks, respectively. Bone metabolism, bone mineral density and microstructure changes were then evaluated. The number of TRAP-positive cells and bone resorption pits decreased significantly following culture with either rhOPG-Fc or rhRANK, and this was more marked following culture with rhRANK compared with rhOPG-Fc. The levels of calcium and alkaline phosphatase in the serum were similar pre-OVX and after 12 weeks of treatment, while the levels of phosphorus in the serum were higher following treatment with rhRANK compared with rhOPG. The bone mineral density (BMD) of the whole body, femoral neck and L4 lumbar vertebral body in the mice treated with either rhOPG-Fc or rhRANK increased markedly. In addition, the mice treated with rhRANK exhibited significantly higher BMD in the femoral neck and lumbar vertebral body compared with those treated with rhOPG-Fc. Microcomputed tomography analysis demonstrated that the mice treated with rhRANK exhibited an increased bone volume and structure model index, and decreased trabecular spacing compared with those treated with rhOPG-Fc. rhRANK increased the inhibition of osteoclast differentiation and bone resorption, and rescued OVX-induced osteoporosis more effectively compared with rhOPG-Fc.


Subject(s)
Osteoporosis/genetics , Osteoprotegerin/administration & dosage , Receptor Activator of Nuclear Factor-kappa B/administration & dosage , Recombinant Proteins/administration & dosage , Animals , Bone Density , Bone Resorption/drug therapy , Bone Resorption/genetics , Bone Resorption/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Humans , Mice , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoporosis/drug therapy , Osteoporosis/pathology , Osteoprotegerin/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Recombinant Proteins/genetics
11.
Int J Cancer ; 137(4): 968-77, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25603921

ABSTRACT

Dormant disseminated tumour cells can be detected in the bone marrow of breast cancer patients several years after resection of the primary tumour. The majority of these patients will remain asymptomatic, however, ∼ 15% will go on to develop overt bone metastases and this condition is currently incurable. The reason why these dormant cells are stimulated to proliferate and form bone tumours in some patients and not others remains to be elucidated. We have recently shown that in an in vivo model, increasing bone turnover by ovariectomy stimulated proliferation of disseminated tumour cells, resulting in formation of bone metastasis. We now show for the first time that osteoclast mediated mechanisms induce growth of tumours from dormant MDA-MB-231 cells disseminated in the bone. We also show that disruption of RANK-RANKL interactions following administration of OPG-Fc inhibits growth of these dormant tumour cells in vivo. Our data support early intervention with anti-resorptive therapy in a low-oestrogen environment to prevent development of bone metastases.


Subject(s)
Bone Neoplasms/drug therapy , Breast Neoplasms/pathology , Osteoprotegerin/administration & dosage , Ovariectomy/adverse effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Bone Resorption/drug therapy , Bone Resorption/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/immunology , Mice , Osteoclasts , Osteoprotegerin/immunology , RANK Ligand/metabolism
12.
Exp Hematol ; 43(2): 110-4.e1-2, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25461255

ABSTRACT

The contribution of osteoclasts to hematopoietic stem/progenitor cell (HSPC) retention in the bone marrow is controversial. Studies of HSPC trafficking in osteoclast-deficient mice are limited by osteopetrosis. Here, we employed two non-osteopetrotic mouse models to assess the contribution of osteoclasts to basal and granulocyte colony-stimulating factor (G-CSF)-induced HSPC mobilization. We generated Rank(-/-) fetal liver chimeras using Csf3r(-/-) recipients to produce mice lacking G-CSF receptor expression in osteoclasts. Basal and G-CSF-induced HSPC mobilization was normal in these chimeras. We next acutely depleted osteoclasts in wild-type mice using the RANK ligand inhibitor osteoprotegerin. Marked suppression of osteoclasts was observed after a single injection of osteoprotegerin-Fc. Basal and G-CSF-induced HSPC mobilization in osteoprotegerin-Fc-treated mice was comparable to that in control mice. Together, these data indicate that osteoclasts are not required for the efficient retention of HSPCs in the bone marrow and are dispensable for HSPC mobilization by G-CSF.


Subject(s)
Bone Marrow Cells/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Osteoclasts/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Chimera/genetics , Fetus , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Knockout , Osteoclasts/cytology , Osteoclasts/metabolism , Osteoprotegerin/administration & dosage , Osteoprotegerin/genetics , RANK Ligand/antagonists & inhibitors , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/deficiency , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, Colony-Stimulating Factor/deficiency , Receptors, Colony-Stimulating Factor/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics
13.
Mol Cell Endocrinol ; 394(1-2): 13-20, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-24998520

ABSTRACT

BACKGROUND: Inflammation is believed to link obesity to insulin resistance, as in the setting of metabolic syndrome (MetS). Osteoprotegerin (OPG) is a soluble protein that seems to exert proatherogenic and prodiabetogenic effects. This study aims at determining OPG levels in MetS and whether OPG might contribute to MetS development and progression. METHODOLOGY/PRINCIPAL FINDINGS: Circulating OPG was measured in 46 patients with MetS and 63 controls, and was found significantly elevated in those with MetS. In addition, circulating and tissue OPG was significantly increased in high-fat diet (HFD) fed C57BL6 mice, which is one of the animal models for the study of MetS. To evaluate the consequences of OPG elevation, we delivered this protein to C57BL6 mice, finding that it promoted systemic and adipose tissue proinflammatory changes in association with metabolic abnormalities. CONCLUSIONS/SIGNIFICANCE: These data suggest that OPG may trigger adipose tissue proinflammatory changes in MetS/HFD-induced obesity.


Subject(s)
Adipose Tissue/metabolism , Metabolic Syndrome/blood , Obesity/blood , Osteoprotegerin/blood , Adipose Tissue/pathology , Adult , Animals , Blood Glucose/metabolism , Body Mass Index , C-Reactive Protein/metabolism , Case-Control Studies , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diet, High-Fat/adverse effects , Female , Humans , Inflammation/blood , Inflammation/etiology , Inflammation/pathology , Insulin/blood , Insulin Resistance , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/etiology , Obesity/pathology , Osteoprotegerin/administration & dosage , Triglycerides/blood
14.
Chin J Traumatol ; 16(5): 277-80, 2013.
Article in English | MEDLINE | ID: mdl-24103822

ABSTRACT

OBJECTIVE: To observe the effect of recombinant interleukin-6 (IL-6) and osteoprotegerin (OPG) on inhibiting bone absorption induced by receptor activator for nuclear factor-kB ligand (RANKL) in murine osteoclast precursor cells (OCPs) model. METHODS: RAW 264.7 cells were solely treated with 50 ng/ml RANKL for 1 day, and then they were divided into three groups: RANKL (control group), RANKL+IL-6 (IL-6 group) and RANKL+IL-6+OPG (combination group). These cells were harvested and investigated by means of HE staining under light microscope after consecutive 9 days. Furthermore, staining tartrate-resistant acid phosphatase(TRAP)-positive multinucleated cells were detected by inverted phase contrast microscope. The absorption pits of bone slices were observed under scanning electron microscope. RESULTS: The number of mature osteoclast cells in control group was more than that in IL-6 alone or IL-6 combined with OPG group (P less than 0.05). Interestingly, this experiment has also demonstrated that there was a large number of TRAP-positive multinucleated osteoclasts (more than 3 nuclei) and several bone absorption formation in the control group, whereas the outcome was completely different in both IL-6 group and IL-6+OPG group (P less than 0.05). CONCLUSION: IL-6 can suppress the differentiation of mature osteoclasts as directly adding it into the RAW 264.7 cells induced by 50 ng/ml RANKL, and further the effect of osteolysis is remarkably reduced. When treatment with IL-6 combined with OPG, a more effective strategy for the treatment of osteoporosis is reached.


Subject(s)
Cell Differentiation/drug effects , Interleukin-6/administration & dosage , Osteoclasts/drug effects , Osteoprotegerin/administration & dosage , Animals , Cells, Cultured , Interleukin-6/pharmacology , Mice , Osteoclasts/cytology , Osteoprotegerin/pharmacology , RANK Ligand/pharmacology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
15.
Pharm Res ; 29(11): 3143-55, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22729371

ABSTRACT

PURPOSE: Recombinant osteoprotegerin (OPG) has been proven to be useful for treating various bone disorders such as osteoporosis. To improve its in vivo pharmacological effect, OPG was conjugated to novel comb-shaped co-polymers of polyethylene glycol (PEG) allylmethylether and maleamic acid (poly(PEG), 5 kDa). Biodistribution and bioactivity were evaluated. METHODS: OPG was conjugated via lysine to poly(PEG) and to linear PEG (0.5 kDa and 5 kDa). Poly(PEG)-OPG was compared with linear PEG0.5k-OPG and PEG5k-OPG in terms of in vitro and in vivo efficacy and bone distribution. RESULTS: The in vitro receptor binding study showed that poly(PEG)-OPG could be the most bioactive among the three PEG-OPG derivatives. Pharmacokinetic studies in ovariectomized (OVX) rats showed that serum half-life and AUC of poly(PEG)-OPG were comparable with those of linear PEG-OPG derivatives. For in vivo pharmacological effect, poly(PEG)-OPG showed the strongest inhibitory effect on bone resorption activity in OVX rats. Poly(PEG)-OPG demonstrated enhanced bone marrow distribution with higher selectivity than linear PEG5k-OPG. CONCLUSION: Poly(PEG) modification could provide longer residence time in serum and higher bone-marrow specific delivery of OPG, leading to a higher in vivo pharmacological effect.


Subject(s)
Bone and Bones/drug effects , Bone and Bones/metabolism , Osteoclasts/drug effects , Osteoprotegerin/administration & dosage , Osteoprotegerin/chemistry , Polyethylene Glycols/administration & dosage , Administration, Intravenous , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Resorption/drug therapy , Bone Resorption/metabolism , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems/methods , Female , Humans , Maleates/administration & dosage , Maleates/chemistry , Osteoclasts/metabolism , Osteoprotegerin/pharmacokinetics , Ovariectomy/methods , Polyethylene Glycols/chemistry , Polymers/administration & dosage , Polymers/chemistry , Rats , Rats, Inbred F344 , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Structure-Activity Relationship , Tissue Distribution
16.
Calcif Tissue Int ; 90(4): 330-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22382900

ABSTRACT

Relapse after orthodontic tooth movement is a significant problem in orthodontics. The purpose of this study was to examine the efficacy of the osteoclast inhibitor osteoprotegerin-Fc (OPG-Fc) for inhibiting postorthodontic relapse. Rat maxillary molars were moved mesially and allowed to relapse for 24 days. Low-dose (1 mg/kg) or high-dose (5 mg/kg) OPG-Fc or saline was injected adjacent to the molars during relapse. Tooth movement, micro-CT, histologic bone quality, and serum OPG and TRAP-5b were measured. OPG-Fc injections significantly diminished postorthodontic relapse from 63% (0.78/1.20 mm) of total movement in vehicle control rats to 31% (0.31/1.00 mm) in low-dose and 24% (0.28/1.16 mm) in high-dose OPG-Fc groups 24 days after appliance removal. Normalization of bone and periodontal tissues occurred as early as 8 and 16 days in the high- and low-dose OPG-Fc-treated groups, respectively, while the vehicle-treated group showed only partial tissue recovery 24 days following tooth movement. After 24 days of relapse, there was complete recovery to pre-tooth-movement values for bone volume fraction (BVF) and tissue mineral density (TMD) in both the low- and high-dose OPG-Fc groups, while BVF recovered only partially and TMD did not recover in the vehicle control group. Greatly elevated serum OPG levels and reduced serum TRAP-5b levels in OPG-Fc-treated animals indicated systemic exposure to locally injected drug. The profound decrease in postorthodontic relapse by local OPG-Fc administration indicates that osteoclasts are critical to bone maturation following tooth movement and points to the potential pharmacologic use of OPG-Fc or other RANKL inhibitors for orthodontic retention.


Subject(s)
Osteoprotegerin/therapeutic use , Recombinant Proteins/therapeutic use , Tooth Mobility/drug therapy , Tooth/drug effects , Animals , Male , Osteoprotegerin/administration & dosage , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recurrence , Tooth/physiology , Tooth Movement Techniques
17.
Aust Orthod J ; 28(2): 219-24, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23304971

ABSTRACT

BACKGROUND: Osteoprotegerin (OPG), as an osteoclast antagonist, limits mineralised tissue resorption under physiological conditions. Previous work investigating OPG in a rat periodontal ligament (PDL) ankylosis model found no inhibitory effect on osteoclasts when OPG was administered at a dosage of 2.5mg/kg. AIMS: The object of this study was to determine whether dosages higher than 2.5 mg/kg of OPG were required to limit osteoclastic activity in an aseptic inflammatory model in rats. MATERIALS AND METHODS: Dry ice was applied for 15 minutes to the upper right first molar crown of eighteen, 8-week-old, male Sprague-Dawley rats. Three groups of 3 were injected with OPG at dosages of 2.5, 5.0 and 7.5 mg/kg of body weight immediately following the thermal insult. After 7 days, the rats were sacrificed and each maxilla processed for histological examination and stained for osteoclastic activity using tartrate-resistant acid phosphatase (TRAP). Osteoclast population numbers were estimated via light microscopy and results were analysed using a comparative mixed model statistical analysis. RESULTS: Results showed OPG inhibited osteoclastic activity in a dose-dependent manner. From 2.5 mg/kg to 7.5 mg/kg, osteoclast populations were linearly reduced by 39.78% (p < 0.05). OPG did not appear to affect the inflammatory process and had varied efficacy in different regions of individual teeth. CONCLUSION: Although osteoclastic activity reduced, it was not completely eliminated, perhaps because dosages were still inadequate, or additional factors might influence OPG and osteoclast activation in the aseptic inflammatory model.


Subject(s)
Osteoclasts/drug effects , Osteoprotegerin/pharmacology , Acid Phosphatase/analysis , Animals , Biomarkers/analysis , Cell Count , Disease Models, Animal , Dose-Response Relationship, Drug , Dry Ice/adverse effects , Freezing/adverse effects , Inflammation/pathology , Isoenzymes/analysis , Male , Maxilla/pathology , Molar/injuries , Necrosis , Odontoblasts/drug effects , Odontoblasts/pathology , Osteoclasts/pathology , Osteoprotegerin/administration & dosage , Rats , Rats, Sprague-Dawley , Root Resorption/pathology , Tartrate-Resistant Acid Phosphatase , Tooth Crown/injuries
18.
Mol Imaging Biol ; 14(4): 500-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21853370

ABSTRACT

PURPOSE: The goal of this study was to determine the specificity of 64Cu-CB-TE2A-c(RGDyK) (64Cu-RGD) for osteoclast-related diseases, such as Paget's disease or rheumatoid arthritis. PROCEDURES: C57BL/6 mice were treated systemically with osteoprotegerin (OPG) for 15 days or RANKL for 11 days to suppress and stimulate osteoclastogenesis, respectively. The mice were then imaged by positron emission tomography/computed tomography using 64Cu-RGD, followed by determination of serum TRAP5b and bone histology. Standard uptake values were determined to quantify 64Cu-RGD in bones and other tissues. RESULTS: Mice treated with OPG showed decreased bone uptake of 64Cu-RGD at 1, 2, and 24 h post-injection of the tracer (p < 0.01 for all time points) compared to vehicle controls, which correlated with a post-treatment decrease in serum TRAP5b. In contrast, mice treated with RANKL showed significantly increased bone uptake at 2 h post-injection of (64Cu-RGD (p < 0.05) compared to the vehicle control group, corresponding to increased serum TRAP5b and OC numbers as determined by bone histology. CONCLUSIONS: These data demonstrate that 64Cu-RGD localizes to areas in bone with increased osteoclast numbers and support the use of 64Cu-RGD as an imaging biomarker for osteoclast number that could be used to monitor osteoclast-related pathologies and their treatments.


Subject(s)
Integrin alphaVbeta3/metabolism , Models, Animal , Osteoclasts/metabolism , Osteoclasts/pathology , Positron-Emission Tomography/methods , Animals , Biomarkers/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Count , Male , Mice , Mice, Inbred C57BL , Multimodal Imaging , Organ Specificity/drug effects , Organometallic Compounds , Osteoclasts/drug effects , Osteoprotegerin/administration & dosage , Osteoprotegerin/pharmacology , RANK Ligand/administration & dosage , RANK Ligand/pharmacology , Reproducibility of Results , Tomography, X-Ray Computed
19.
J Pharm Pharmacol ; 62(8): 985-94, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20663032

ABSTRACT

OBJECTIVES: Our aim was to investigate the effect of PEGylation on the uptake of osteoprotegerin/osteoclastogenesis inhibitory factor (OPG/OCIF) into rat liver, kidney and spleen, and human liver. METHODS: Copolymer of polyethyleneglycol allylmethylether and maleamic acid sodium salt with OCIF (poly(PEG)-OCIF) (0.5 mg/kg) was administered to rats and the concentrations of poly(PEG)-OCIF in the liver, kidney and spleen at 15 min after administration were measured by ELISA. For human liver uptake, the liver perfusion of OCIF and (3)H-labelled poly(PEG)-OCIF was conducted using fresh human liver block. KEY FINDINGS: The tissue uptake of poly(PEG)-OCIF in rats was significantly lower compared with that of OCIF. In fresh human liver perfusion, (3)H-poly(PEG)-OCIF was rarely taken up into the liver. On the other hand, more than 50% of the perfused OCIF was taken up. CONCLUSIONS: PEGylation of OCIF using poly(PEG) dramatically suppressed the uptake of OCIF into human liver as well as into rat liver and could be a promising approach for improving the pharmacokinetic and pharmacological effects of OCIF in the clinical setting.


Subject(s)
Bone Density Conservation Agents/pharmacokinetics , Liver/metabolism , Osteoprotegerin/pharmacokinetics , Polyethylene Glycols/chemistry , Animals , Biological Transport , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/blood , Bone Density Conservation Agents/chemistry , Cells, Cultured , Chemistry, Pharmaceutical , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Heparin/metabolism , Humans , Injections, Intravenous , Kidney/metabolism , Maleates/chemistry , Mice , Osteoclasts/drug effects , Osteoprotegerin/administration & dosage , Osteoprotegerin/blood , Osteoprotegerin/chemistry , Ovariectomy , Perfusion , Rats , Rats, Sprague-Dawley , Spleen/metabolism , Tissue Distribution
20.
J Bone Miner Res ; 25(11): 2427-37, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20533302

ABSTRACT

Sustained parathyroid hormone (PTH) elevation stimulates bone remodeling (ie, both resorption and formation). The former results from increased RANKL synthesis, but the cause of the latter has not been established. Current hypotheses include release of osteoblastogenic factors from osteoclasts or from the bone matrix during resorption, modulation of the production and activity of osteoblastogenic factors from cells of the osteoblast lineage, and increased angiogenesis. To dissect the contribution of these mechanisms, 6-month-old Swiss-Webster mice were infused for 5 days with 470 ng/h PTH(1-84) or 525 ng/h soluble RANKL (sRANKL). Both agents increased osteoclasts and osteoblasts in vertebral cancellous bone, but the ratio of osteoblasts to osteoclasts and the increase in bone formation was greater in PTH-treated mice. Cancellous bone mass was maintained in mice receiving PTH but lost in mice receiving sRANKL, indicating that maintenance of balanced remodeling requires osteoblastogenic effects beyond those mediated by osteoclasts. Consistent with this contention, PTH, but not sRANKL, decreased the level of the Wnt antagonist sclerostin and increased the expression of the Wnt target genes Nkd2, Wisp1, and Twist1. Furthermore, PTH, but not sRANKL, increased the number of blood vessels in the bone marrow. Weekly injections of the RANKL antagonist osteoprotegerin at 10 µg/g for 2 weeks prior to PTH infusion eliminated osteoclasts and osteoblasts and prevented the PTH-induced increase in osteoclasts, osteoblasts, and blood vessels. These results indicate that PTH stimulates osteoclast-dependent as well as osteoclast-independent (Wnt signaling) pro-osteoblastogenic pathways, both of which are required for balanced focal bone remodeling in cancellous bone.


Subject(s)
Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Parathyroid Hormone/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Body Weight/drug effects , Bone Marrow/blood supply , Bone Marrow/drug effects , Bone Morphogenetic Proteins/metabolism , Bone Remodeling/drug effects , Calcium/blood , Cell Count , Female , Genetic Markers , Glycoproteins , Humans , Intercellular Signaling Peptides and Proteins , Mice , Models, Biological , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/administration & dosage , Osteoprotegerin/pharmacology , Parathyroid Hormone/administration & dosage , RANK Ligand/administration & dosage , RANK Ligand/pharmacology , Signal Transduction/drug effects , Solubility/drug effects , Spine/drug effects , Spine/pathology , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...