Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Cell ; 184(5): 1330-1347.e13, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636130

ABSTRACT

Osteoclasts are large multinucleated bone-resorbing cells formed by the fusion of monocyte/macrophage-derived precursors that are thought to undergo apoptosis once resorption is complete. Here, by intravital imaging, we reveal that RANKL-stimulated osteoclasts have an alternative cell fate in which they fission into daughter cells called osteomorphs. Inhibiting RANKL blocked this cellular recycling and resulted in osteomorph accumulation. Single-cell RNA sequencing showed that osteomorphs are transcriptionally distinct from osteoclasts and macrophages and express a number of non-canonical osteoclast genes that are associated with structural and functional bone phenotypes when deleted in mice. Furthermore, genetic variation in human orthologs of osteomorph genes causes monogenic skeletal disorders and associates with bone mineral density, a polygenetic skeletal trait. Thus, osteoclasts recycle via osteomorphs, a cell type involved in the regulation of bone resorption that may be targeted for the treatment of skeletal diseases.


Subject(s)
Bone Resorption/pathology , Osteoclasts/pathology , RANK Ligand/metabolism , Animals , Apoptosis , Bone Resorption/metabolism , Cell Fusion , Cells, Cultured , Humans , Macrophages/cytology , Mice , Osteochondrodysplasias/drug therapy , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/pathology , Osteoclasts/metabolism , Signal Transduction
3.
Blood ; 129(26): 3452-3464, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28515094

ABSTRACT

Multiple myeloma (MM) is a plasma cell cancer that develops in the skeleton causing profound bone destruction and fractures. The bone disease is mediated by increased osteoclastic bone resorption and suppressed bone formation. Bisphosphonates used for treatment inhibit bone resorption and prevent bone loss but fail to influence bone formation and do not replace lost bone, so patients continue to fracture. Stimulating bone formation to increase bone mass and fracture resistance is a priority; however, targeting tumor-derived modulators of bone formation has had limited success. Sclerostin is an osteocyte-specific Wnt antagonist that inhibits bone formation. We hypothesized that inhibiting sclerostin would prevent development of bone disease and increase resistance to fracture in MM. Sclerostin was expressed in osteocytes from bones from naive and myeloma-bearing mice. In contrast, sclerostin was not expressed by plasma cells from 630 patients with myeloma or 54 myeloma cell lines. Mice injected with 5TGM1-eGFP, 5T2MM, or MM1.S myeloma cells demonstrated significant bone loss, which was associated with a decrease in fracture resistance in the vertebrae. Treatment with anti-sclerostin antibody increased osteoblast numbers and bone formation rate but did not inhibit bone resorption or reduce tumor burden. Treatment with anti-sclerostin antibody prevented myeloma-induced bone loss, reduced osteolytic bone lesions, and increased fracture resistance. Treatment with anti-sclerostin antibody and zoledronic acid combined increased bone mass and fracture resistance when compared with treatment with zoledronic acid alone. This study defines a therapeutic strategy superior to the current standard of care that will reduce fractures for patients with MM.


Subject(s)
Bone Density/drug effects , Bone Morphogenetic Proteins/antagonists & inhibitors , Fractures, Bone/prevention & control , Osteocytes/chemistry , Osteogenesis/drug effects , Adaptor Proteins, Signal Transducing , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Bone Morphogenetic Proteins/immunology , Cell Line, Tumor , Diphosphonates/therapeutic use , Genetic Markers/immunology , Humans , Imidazoles/therapeutic use , Mice , Multiple Myeloma/complications , Tumor Cells, Cultured , Zoledronic Acid
4.
J Bone Miner Res ; 39(4): 484-497, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38477789

ABSTRACT

Rebound bone loss following denosumab discontinuation is an important clinical challenge. Current treatment strategies to prevent this fail to suppress the rise and overshoot in osteoclast-mediated bone resorption. In this study, we use a murine model of denosumab treatment and discontinuation to show the temporal changes in osteoclast formation and activity during RANKL inhibition and withdrawal. We show that the cellular processes that drive the formation of osteoclasts and subsequent bone resorption following withdrawal of RANKL inhibition precede the rebound bone loss. Furthermore, a rise in serum TRAP and RANKL levels is detected before markers of bone turnover used in current clinical practice. These mechanistic advances may provide insight into a more defined window of opportunity to intervene with sequential therapy following denosumab discontinuation.


Stopping denosumab, a medication commonly used to improve bone mass by blocking formation of bone resorbing osteoclasts, leads to a rebound loss in the bone which was gained during treatment. Current strategies to prevent this bone loss fail in most cases as they are unable to prevent the rise and overshoot in bone resorption by osteoclasts. Thie stems from an incomplete understanding of how osteoclasts behave during denosumab treatment and after treatment is discontinued. We use a mouse model of this phenomenon to show how osteoclast formation and activity changes throughout this process. We show that increases in the processes that drive the formation of osteoclasts can be detected in the circulation before bone loss occurs. These findings could therefore provide insight into a targeted 'window of opportunity' to intervene and prevent the rebound bone loss following stopping denosumab in patients.


Subject(s)
Bone Resorption , Denosumab , Osteoclasts , RANK Ligand , Animals , Osteoclasts/metabolism , Osteoclasts/drug effects , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Denosumab/pharmacology , Mice , Bone Resorption/pathology , Bone Resorption/drug therapy , Bone Resorption/blood , Time Factors , Tartrate-Resistant Acid Phosphatase/metabolism , Female , Mice, Inbred C57BL , Biomarkers/metabolism , Biomarkers/blood
5.
J Bone Miner Res ; 38(6): 814-828, 2023 06.
Article in English | MEDLINE | ID: mdl-36987921

ABSTRACT

An imbalance between bone resorption and bone formation underlies the devastating osteolytic lesions and subsequent fractures seen in more than 90% of multiple myeloma (MM) patients. Currently, Wnt-targeted therapeutic agents that prevent soluble antagonists of the Wnt signaling pathway, sclerostin (SOST) and dickkopf-1 (DKK1), have been shown to prevent bone loss and improve bone strength in preclinical models of MM. In this study, we show increasing Wnt signaling via a novel anti-low-density lipoprotein receptor-related protein 6 (LRP6) antibody, which potentiates Wnt1-class ligand signaling through binding the Wnt receptor LRP6, prevented the development of myeloma-induced bone loss primarily through preventing bone resorption. When combined with an agent targeting the soluble Wnt antagonist DKK1, we showed more robust improvements in bone structure than anti-LRP6 treatment alone. Micro-computed tomography (µCT) analysis demonstrated substantial increases in trabecular bone volume in naïve mice given the anti-LRP6/DKK1 combination treatment strategy compared to control agents. Mice injected with 5TGM1eGFP murine myeloma cells had significant reductions in trabecular bone volume compared to naïve controls. The anti-LRP6/DKK1 combination strategy significantly improved bone volume in 5TGM1-bearing mice by 111%, which was also superior to anti-LRP6 single treatment; with similar bone structural changes observed within L4 lumbar vertebrae. Consequently, this combination strategy significantly improved resistance to fracture in lumbar vertebrae in 5TGM1-bearing mice compared to their controls, providing greater protection against fracture compared to anti-LRP6 antibody alone. Interestingly, these improvements in bone volume were primarily due to reduced bone resorption, with significant reductions in osteoclast numbers and osteoclast surface per bone surface demonstrated in 5TGM1-bearing mice treated with the anti-LRP6/DKK1 combination strategy. Importantly, Wnt stimulation with either single or combined Wnt-targeted agents did not exacerbate tumor activity. This work provides a novel approach of targeting both membrane-bound and soluble Wnt pathway components to provide superior skeletal outcomes in patients with multiple myeloma and other bone destructive cancers. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Intercellular Signaling Peptides and Proteins , Low Density Lipoprotein Receptor-Related Protein-6 , Multiple Myeloma , Osteolysis , Animals , Mice , Mice, Inbred C57BL , Antibodies/administration & dosage , Low Density Lipoprotein Receptor-Related Protein-6/antagonists & inhibitors , Bone and Bones/drug effects , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Wnt Signaling Pathway/drug effects , Osteolysis/drug therapy , Intercellular Signaling Peptides and Proteins/metabolism , Female , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL