Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bone ; 97: 20-28, 2017 04.
Article in English | MEDLINE | ID: mdl-27939957

ABSTRACT

Sclerostin antibodies increase bone mass by stimulating bone formation. However, human and animal studies show that bone formation increases transiently and returns to pre-treatment level despite ongoing antibody treatment. To understand its mechanism of action, we studied the time course of bone formation, correlating the rate and extent of accrual of bone mass and strength after sclerostin antibody treatment. Ovariectomized (OVX) rats were treated with a sclerostin-antibody (Scle-ab) at 20mg/kg sc once weekly and sacrificed at baseline and 2, 3, 4, 6, and 8weeks post-treatment. In Scle-ab treated rats, serum PINP and OCN rapidly increased at week 1, peaked around week 3, and returned to OVX control levels by week 6. Transcript analyses from the distal femur revealed an early increase in bone formation followed by a sustained decrease in bone resorption genes. Lumbar vertebral (LV) osteoblast surface increased 88% by week 2, and bone formation rate (BFR/BS) increased 138% by week 4. Both parameters were below OVX control by week 8. Bone formation was primarily a result of modeling based formation. Endocortical and periosteal BFR/BS peaked around week 4 at 313% and 585% of OVX control, respectively. BFR/BS then declined but remained higher than OVX control on both surfaces through week 8. Histomorphometric analyses showed LV-BV/TV did not further increase after week 4, while BMD continued to increase at LV, mid femur (MF), and femoral neck (FN) through week 8. Biomechanical tests showed a similar improvement in bone strength through 8weeks in MF and FN, but bone strength plateaued between weeks 6 and 8 for LV. Our data suggest that bone formation with Scle-ab treatment is rapid and modeling formation dominated in OVX rats. Although transient, the bone formation response persists longer in cortical than trabecular bone.


Subject(s)
Antibodies/pharmacology , Bone Morphogenetic Proteins/immunology , Bone and Bones/pathology , Bone and Bones/physiopathology , Genetic Markers/immunology , Osteogenesis/drug effects , Ovariectomy , Animals , Biomarkers/blood , Biomechanical Phenomena , Bone Resorption/blood , Bone Resorption/pathology , Bone and Bones/drug effects , Cancellous Bone/drug effects , Cancellous Bone/pathology , Densitometry , Female , Femur/drug effects , Femur/pathology , Femur/physiopathology , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Lumbar Vertebrae/physiopathology , Organ Size/drug effects , Rats, Sprague-Dawley , Time Factors , Wnt Proteins/genetics , Wnt Proteins/metabolism
2.
J Bone Miner Res ; 31(6): 1225-34, 2016 06.
Article in English | MEDLINE | ID: mdl-26763740

ABSTRACT

Sclerostin is a potent inhibitor of osteoblastogenesis. Interestingly, newly diagnosed multiple myeloma (MM) patients have high levels of circulating sclerostin that correlate with disease stage and fractures. However, the source and impact of sclerostin in MM remains to be defined. Our goal was to determine the role of sclerostin in the biology of MM and its bone microenvironment as well as investigate the effect of targeting sclerostin with a neutralizing antibody (scl-Ab) in MM bone disease. Here we confirm increased sclerostin levels in MM compared with precursor disease states like monoclonal gammopathy of undetermined significance (MGUS) and smoldering MM. Furthermore, we found that a humanized MM xenograft mouse model bearing human MM cells (NOD-SCID.CB17 male mice injected intravenously with 2.5 million of MM1.S-Luc-GFP cells) demonstrated significantly higher concentrations of mouse-derived sclerostin, suggesting a microenvironmental source of sclerostin. Associated with the increased sclerostin levels, activated ß-catenin expression levels were lower than normal in MM mouse bone marrow. Importantly, a high-affinity grade scl-Ab reversed osteolytic bone disease in this animal model. Because scl-Ab did not demonstrate significant in vitro anti-MM activity, we combined it with the proteasome inhibitor carfilzomib. Our data demonstrated that this combination therapy significantly inhibited tumor burden and improved bone disease in our in vivo MM mouse model. In agreement with our in vivo data, sclerostin expression was noted in marrow stromal cells and osteoblasts of MM patient bone marrow samples. Moreover, MM cells stimulated sclerostin expression in immature osteoblasts while inhibiting osteoblast differentiation in vitro. This was in part regulated by Dkk-1 secreted by MM cells and is a potential mechanism contributing to the osteoblast dysfunction noted in MM. Our data confirm the role of sclerostin as a potential therapeutic target in MM bone disease and provides the rationale for studying scl-Ab combined with proteasome inhibitors in MM. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Diseases/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Osteoblasts/metabolism , Tumor Microenvironment , Adaptor Proteins, Signal Transducing , Animals , Bone Diseases/genetics , Bone Diseases/pathology , Female , Glycoproteins/genetics , Heterografts , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Neoplasm Transplantation , Osteoblasts/pathology
3.
J Orthop Res ; 32(3): 471-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24243768

ABSTRACT

We compared the effect of a sclerostin antibody to that of a clinically relevant dose of parathyroid hormone (PTH) in a rat model for metaphyseal bone healing. Screws of steel or poly methyl methacrylate (PMMA) were inserted bilaterally into the proximal tibia of young male rats. During 4 weeks the animals then received injections of either phosphate buffered saline (control), sclerostin antibody (25 mg/kg, twice weekly) or PTH (5 µg/kg, daily). The healing response around the screws was then assessed by mechanical testing and X-ray microtomography (µCT). To distinguish between effects on healing and general effects on the skeleton, other untraumatized bone sites and serum biomarkers were also assessed. After 4 weeks of treatment, PTH yielded a 48% increase in screw pull-out force compared to control (p = 0.03), while the antibody had no significant effect. In contrast, the antibody increased femoral cortical and vertebral strength where PTH had no significant effect. µCT showed only slight changes that were statistically significant for the antibody mainly at cortical sites. The results suggest that a relatively low dose of PTH stimulates metaphyseal repair (screw fixation) specifically, whereas the sclerostin antibody has wide-spread effects, mainly on cortical bone, with less influence on metaphyseal healing.


Subject(s)
Antibodies/therapeutic use , Bone Density Conservation Agents/therapeutic use , Bone Morphogenetic Proteins/immunology , Fracture Healing/drug effects , Fractures, Bone/drug therapy , Genetic Markers/immunology , Teriparatide/therapeutic use , Animals , Bone Density Conservation Agents/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Screws , Drug Evaluation, Preclinical , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Teriparatide/pharmacology
4.
Bone ; 53(2): 487-96, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23333523

ABSTRACT

Over-expression of the protein Dickkopf-1 (Dkk1) has been associated with multiple myeloma bone disease. Previous reports with the use of anti-Dkk1 neutralizing Ab directed strategies have demonstrated a pro-anabolic effect with associated anti-myeloma activity in 2 in vivo mouse models. However new insights on the role of the wnt pathway in osteoclasts (OC) are emerging and the potential effect of a neutralizing Ab to Dkk1 in osteoclastogenesis remains to be elucidated. In order to better define the effect of an anti-Dkk1 neutralizing Ab on osteoclastogenesis and myeloma, we studied a novel anti-Dkk1 monoclonal Ab in our preclinical models. In vivo data confirmed the pro-anabolic and anti-MM effect. In vitro data in part confirmed the in vivo observation, suggesting an indirect anti-MM effect secondary to inhibition of osteoclastogenesis and thus the interaction between MM and bone microenvironment. However, when studies on osteoclastogenesis were extended to samples derived from MM patients, we observed a variable response to anti-Dkk1 treatment without correlation to expression of surface receptors for Dkk1 in OCs suggesting potential heterogeneity in the efficacy of such a strategy. In conclusion, Dkk1 is a promising target for the treatment of both MM and bone disease, and ongoing clinical studies will help elucidate its efficacy.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Intercellular Signaling Peptides and Proteins/immunology , Multiple Myeloma/drug therapy , Animals , Cell Line , Cells, Cultured , Humans , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, SCID , Multiple Myeloma/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , X-Ray Microtomography
5.
Spine (Phila Pa 1976) ; 37(15): 1275-82, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22281479

ABSTRACT

STUDY DESIGN: Controlled animal experiments. OBJECTIVE: To test the dose and efficacy of teriparatide in a rat spinal fusion model. SUMMARY OF BACKGROUND DATA: Teriparatide was shown to enhance spinal fusion in rats and rabbits previously, but the dose-dependent effect of teriparatide in spinal fusion in rats was not well characterized. METHODS: A 0.5 × 0.5 cm trabecular bone graft was taken and implanted onto the L5 and L6 transverse processes of the same rat. Rats were randomly assigned into 3 groups: saline vehicle control (Vehicle), teriparatide 4 µg/kg per day (PTH4), and teriparatide 23 µg/kg per day (PTH23) subcutaneous injections for 4 weeks (5 d per wk). The L5-L6 spinal segments were harvested at week 4, and assessments included radiography, micro-computed tomography, manual palpation, and histomorphometry. L3 vertebra, femurs, and serum bone markers were examined. RESULTS: The average radiographical score of L5-L6 fusion in Vehicle, PTH4, and PTH23 groups was 1.53, 2.87, and 4.11, respectively, with the PTH23 being significantly higher (P = 0.001 vs. Vehicle). The average micro-computed tomographic score of L5-L6 fusion in Vehicle, PTH4, and PTH23 groups was 1.53, 2.40, and 3.74, respectively (P = 0.001, PTH23 vs. Vehicle and PTH4). Manual palpation showed that fusion rate was 20%, 50%, and 67.7% in Vehicle, PTH4, and PTH23 groups, respectively. The bone mineralization apposition rate at the fusion site was significantly increased in a dose-dependent manner among the groups. Teriparatide significantly increased vertebral and femoral bone mineral density, bone mineral content, and trabecular area in a dose-dependent manner relative to Vehicle. No difference was found between the circulating Procollagen type I N-terminal propeptide and intact osteocalcin levels in the serum at 4 weeks after treatments. CONCLUSION: Teriparatide at 23 µg/kg per day for 4 weeks showed anabolic skeletal effects and significantly enhanced spinal fusion rate in rats, whereas teriparatide at 4 µg/kg per day had also anabolic effects but did not significantly enhance spinal fusion rate. Higher doses of teriparatide may be needed to promote spinal fusion in short-term application.


Subject(s)
Lumbar Vertebrae/drug effects , Lumbar Vertebrae/surgery , Spinal Fusion/methods , Teriparatide/pharmacology , Animals , Bone Density/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacology , Bone Transplantation/methods , Dose-Response Relationship, Drug , Injections, Subcutaneous , Lumbar Vertebrae/diagnostic imaging , Male , Osteocalcin/blood , Random Allocation , Rats , Rats, Sprague-Dawley , Teriparatide/administration & dosage , Time Factors , Transplantation, Autologous , Treatment Outcome , X-Ray Microtomography
6.
Bone ; 48(5): 988-96, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21329773

ABSTRACT

The secreted protein Dickkopf-1 (Dkk1) is an antagonist of canonical Wnt signaling, expressed during fracture healing. It is unclear how it is involved in the mechanical control of bone maintenance. We investigated the response to administration of a Dkk1 neutralizing antibody (Dkk1-ab) in metaphyseal bone under different loading conditions, with or without trauma. In this three part experiment, 120 rats had a screw or bone chamber inserted either unilaterally or bilaterally in the proximal tibia. Mechanical (pull-out) testing, µCT and histology were used for evaluation. The animals were injected with either 10mg/kg Dkk1-ab or saline every 14days for 14, 28, or 42days. Antibody treatment increased bone formation around the screws and improved their fixation. After 28days, the pull-out force was increased by over 100%. In cancellous bone, the bone volume fraction was increased by 50%. In some animals, one hind limb was paralyzed with Botulinum toxin A (Botox) to create a mechanically unloaded environment. This did not increase the response to antibody treatment with regard to screw fixation, but in cancellous bone, the bone volume fraction increased by 233%. Thus, the response in unloaded, untraumatized bone was proportionally larger, suggesting that Dkk1 may be up-regulated in unloaded bone. There was also an increase in thickness of the metaphyseal cortex. In bone chambers, the antibody treatment increased the bone volume fraction. The results suggest that antibodies blocking Dkk1 might be used to stimulate bone formation especially during implant fixation, fracture repair, or bone disuse. It also seems that Dkk1 is up-regulated both after metaphyseal trauma and after unloading, and that Dkk1 is involved in mechano-transduction.


Subject(s)
Antibodies, Neutralizing/pharmacology , Bone and Bones/drug effects , Fracture Fixation , Implants, Experimental , Intercellular Signaling Peptides and Proteins/immunology , Animals , Biomechanical Phenomena/drug effects , Bone Screws , Bone and Bones/diagnostic imaging , Mice , Organ Size/drug effects , Osteogenesis/drug effects , Rats , Rats, Sprague-Dawley , Tibia/diagnostic imaging , Tibia/pathology , Weight-Bearing/physiology , X-Ray Microtomography
7.
Cancer Res ; 70(23): 9916-26, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21098705

ABSTRACT

Osteoblastic bone metastases are the most common metastases produced by human prostate cancers (PCa). Deregulated activity of Wnt growth factors resulting from overexpression of the Wnt inhibitor Dickkopf-1 (DKK-1) is known to contribute to formation of the osteoblastic component of PCa skeletal bone metastases. In this study, we report that DKK-1 knockdown in osteolytic human PCa cells unexpectedly delays the development of both soft tissue and osseous lesions. PCa cells deficient in DKK-1 expression did not increase canonical Wnt signaling in target osteoblast cell lines; however, DKK-1 knockdown PCa cells exhibited increased expression of the CDK inhibitor p21(CIP1/WAF1) and a 32% increase in G(1) arrest compared with control cells. Ablating p21(CIP1/WAF1) in PCa cells deficient in DKK-1 was sufficient to rescue tumor growth. Collectively, our findings demonstrate that DKK-1 overexpression supports tumor growth in part by restricting expression of p21(CIP1/WAF1) through a mechanism independent of canonical Wnt signaling.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms, Experimental/metabolism , Prostatic Neoplasms/metabolism , Animals , Blotting, Western , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , G1 Phase , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transplantation, Heterologous , Tumor Burden , Wnt Proteins/metabolism
8.
Cell Metab ; 11(2): 161-71, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-20142103

ABSTRACT

Parathyroid hormone (PTH) suppresses Dickkopf 1 (Dkk1) expression in osteoblasts. To determine whether this suppression is essential for PTH-mediated Wnt signaling and bone formation, we examined mice that overexpress Dkk1 in osteoblasts (Dkk1 mice). Dkk1 mice were osteopenic due to abnormal osteoblast and osteoclast activity. When fed a low-calcium diet, and in two other models of hyperparathyroidism, these mice failed to develop the peritrabecular stromal cell response ("osteitis fibrosis") and new bone formation seen in wild-type mice. Despite these effects of Dkk1 overexpression, PTH still activated Wnt signaling in Dkk1 mice and in osteoblastic cells cultured from these mice. In cultured MC3T3E1 preosteoblastic cells, PTH dramatically suppressed Dkk1 expression, induced PKA-mediated phosphorylation of beta-catenin, and significantly enhanced Lef1 expression. Our findings indicate that the full actions of PTH require intact Wnt signaling but that PTH can activate the Wnt pathway despite overexpression of Dkk1.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Osteogenesis , Parathyroid Hormone/metabolism , Stromal Cells/metabolism , Wnt Proteins/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Line , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoclasts/metabolism , RNA, Messenger/genetics , Signal Transduction
9.
Biochemistry ; 44(33): 11106-14, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16101294

ABSTRACT

Hu007, a humanized IgG1 monoclonal antibody, binds and neutralizes human, cynomolgus, and rabbit IL-1beta but only weakly binds to mouse and rat IL-1beta. Biacore experiments demonstrated that Hu007 and the type-I IL-1 receptor competed for binding to IL-1beta. Increasing salt concentrations decrease the association rate with only moderate effects on the dissociation rate, suggesting that long-range electrostatics are critical for formation of the initial complex. To understand the ligand-binding specificity of Hu007, we have mapped the critical residues involved in the recognition of IL-1beta. Selected residues in cynomolgus IL-1beta were mutated to the corresponding residues in mouse IL-1beta, and the effects of the changes on binding were evaluated by surface plasmon resonance measurements using Biacore. Specifically, substitution of F150S decreased binding affinity by 100-fold, suggesting the importance of hydrophobic interactions in stabilizing the antibody/antigen complex. Substitution of three amino acids near the N- and C-terminal regions of cIL-1beta with those found in mouse IL-1beta (V3I/S5Q/F150S) decreased the binding affinity of Hu007 to IL-1beta by about 1000-fold. Conversely, mutating the corresponding residues in mouse IL-1beta to the human sequence resulted in an increase in binding affinity of about 1000-fold. Hydrogen-deuterium exchange/mass spectrometry analysis confirmed that these regions of IL-1beta were protected from exchange because of antibody binding. The results from this study demonstrate that Hu007 binds to a region located in the open end of the beta-barrel structure of IL-1beta and blocks binding of IL-1beta to its receptor.


Subject(s)
Antibodies, Monoclonal/chemistry , Binding Sites, Antibody , Deuterium/chemistry , Epitope Mapping , Interleukin-1/chemistry , Models, Molecular , Amino Acid Substitution/genetics , Animals , Antibodies, Monoclonal/immunology , Antibody Affinity , Binding Sites, Antibody/physiology , Epitope Mapping/methods , Humans , Interleukin-1/genetics , Interleukin-1/immunology , Macaca fascicularis , Mass Spectrometry/methods , Mice , Mutagenesis, Site-Directed , Point Mutation , Protein Conformation , Rabbits , Rats , Species Specificity , Surface Plasmon Resonance/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...