Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 36(10): 1999-2016, 2021 10.
Article in English | MEDLINE | ID: mdl-34101894

ABSTRACT

Parathyroid hormone-related protein (PTHrP, gene name Pthlh) is a pleiotropic regulator of tissue homeostasis. In bone, Dmp1Cre-targeted PTHrP deletion in osteocytes causes osteopenia and impaired cortical strength. We report here that this outcome depends on parental genotype. In contrast to our previous report using mice bred from heterozygous (flox/wild type) Dmp1Cre.Pthlhf/w parents, adult (16-week-old and 26-week-old) flox/flox (f/f) Dmp1Cre.Pthlhf/f mice from homozygous parents (Dmp1Cre.Pthlhf/f(hom) ) have stronger bones, with 40% more trabecular bone mass and 30% greater femoral width than controls. This greater bone size was observed in Dmp1Cre.Pthlhf/f(hom) mice as early as 12 days of age, when greater bone width was also found in male and female Dmp1Cre.Pthlhf/f(hom) mice compared to controls, but not in gene-matched mice from heterozygous parents. This suggested a maternal influence on skeletal size prior to weaning. Although Dmp1Cre has previously been reported to cause gene recombination in mammary gland, milk PTHrP protein levels were normal. The wide-bone phenotype was also noted in utero: Dmp1Cre.Pthlhf/f(hom) embryonic femurs were more mineralized and wider than controls. Closer examination revealed that Dmp1Cre caused PTHrP recombination in placenta, and in the maternal-derived decidual layer that resides between the placenta and the uterus. Decidua from mothers of Dmp1Cre.Pthlhf/f(hom) mice also exhibited lower PTHrP levels by immunohistochemistry and were smaller than controls. We conclude that Dmp1Cre leads to gene recombination in decidua, and that decidual PTHrP might, through an influence on decidual cells, limit embryonic bone radial growth. This suggests a maternal-derived developmental origin of adult bone strength. © 2021 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Osteocytes , Parathyroid Hormone-Related Protein , Animals , Bone Development/genetics , Bone and Bones , Decidua , Female , Male , Mice , Parathyroid Hormone-Related Protein/genetics , Pregnancy
2.
Biochem Pharmacol ; 169: 113627, 2019 11.
Article in English | MEDLINE | ID: mdl-31476292

ABSTRACT

Parathyroid hormone (PTH)-related protein (PTHrP) (gene name Pthlh) was discovered as the factor responsible for the humoral hypercalcemia of malignancy. It shares such sequence similarity with PTH in the amino-terminal region that the two are equally able to act through a single G protein-coupled receptor, PTH1R. A number of biological activities are ascribed to domains of PTHrP beyond the amino-terminal domain. PTH functions as a circulating hormone, but PTHrP is generated locally in many tissues including bone, where it acts as a paracrine factor on osteoblasts and osteocytes. The present study compares how PTH and PTHrP influence cyclic AMP (cAMP) formation through adenylyl cyclase, the first event in cell activation through PTH1R. Brief exposure to full length PTHrP(1-141) in several osteoblastic cell culture systems was followed by sustained adenylyl cyclase activity for more than an hour after ligand washout. This effect was dose-dependent and was not found with shorter PTHrP or PTH peptides even though they were fully able to activate adenylyl cyclase with acute treatment. The persistent activation response to PTHrP(1-141) was seen also with later events in the cAMP/PKA pathway, including persistent activation of CRE-luciferase and sustained regulation of several CREB-responsive mRNAs, up to 24 h after the initial exposure. Pharmacologic blockade of endocytosis prevented the persistent activation of cAMP and gene responses. We conclude that full length PTHrP, the likely local physiological effector in bone, differs in intracellular action to PTH by undergoing endosomal translocation to induce a prolonged adenylyl cyclase activation in its target cells.


Subject(s)
Cyclic AMP/biosynthesis , Endocytosis/physiology , Parathyroid Hormone-Related Protein/pharmacology , Peptide Fragments/pharmacology , Adenylyl Cyclases/metabolism , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Mice , Mice, Inbred C57BL , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/physiology
3.
J Biol Chem ; 294(19): 7850-7863, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30923130

ABSTRACT

Interleukin 6 (IL-6) supports development of bone-resorbing osteoclasts by acting early in the osteoblast lineage via membrane-bound (cis) or soluble (trans) receptors. Here, we investigated how IL-6 signals and modifies gene expression in differentiated osteoblasts and osteocytes and determined whether these activities can promote bone formation or support osteoclastogenesis. Moreover, we used a genetically altered mouse with circulating levels of the pharmacological IL-6 trans-signaling inhibitor sgp130-Fc to determine whether IL-6 trans-signaling is required for normal bone growth and remodeling. We found that IL-6 increases suppressor of cytokine signaling 3 (Socs3) and CCAAT enhancer-binding protein δ (Cebpd) mRNA levels and promotes signal transducer and activator of transcription 3 (STAT3) phosphorylation by both cis- and trans-signaling in cultured osteocytes. In contrast, RANKL (Tnfsf11) mRNA levels were elevated only by trans-signaling. Furthermore, we observed soluble IL-6 receptor release and ADAM metallopeptidase domain 17 (ADAM17) sheddase expression by osteocytes. Despite the observation that IL-6 cis-signaling occurs, IL-6 stimulated bone formation in vivo only via trans-signaling. Although IL-6 stimulated RANKL (Tnfsf11) mRNA in osteocytes, these cells did not support osteoclast formation in response to IL-6 alone; binucleated TRAP+ cells formed, and only in response to trans-signaling. Finally, pharmacological, sgp130-Fc-mediated inhibition of IL-6 trans-signaling did not impair bone growth or remodeling unless mice had circulating sgp130-Fc levels > 10 µg/ml. At those levels, osteopenia and impaired bone growth occurred, reducing bone strength. We conclude that high sgp130-Fc levels may have detrimental off-target effects on the skeleton.


Subject(s)
Cytokine Receptor gp130/metabolism , Interleukin-6/metabolism , Osteoclasts/metabolism , Osteocytes/metabolism , Osteogenesis , Signal Transduction , ADAM17 Protein/metabolism , Animals , CCAAT-Enhancer-Binding Protein-delta/metabolism , Mice , RANK Ligand/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism
4.
Cell Death Dis ; 9(9): 844, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154459

ABSTRACT

The loss of p53 function is a central event in the genesis of osteosarcoma (OS). How mutation of p53 enables OS development from osteoblastic lineage cells is poorly understood. We and others have reported a key role for elevated and persistent activation of the cAMP/PKA/Creb1 pathway in maintenance of OS. In view of the osteoblast lineage being the cell of origin of OS, we sought to determine how these pathways interact within the context of the normal osteoblast. Normal osteoblasts (p53 WT) rapidly underwent apoptosis in response to acute elevation of cAMP levels or activity, whereas p53-deficient osteoblasts tolerated this aberrant cAMP/Creb level and activity. Using the p53 activating small-molecule Nutlin-3a and cAMP/Creb1 activator forskolin, we addressed the question of how p53 responds to the activation of cAMP. We observed that p53 acts dominantly to protect cells from excessive cAMP accumulation. We identify a Creb1-Cbp complex that functions together with and interacts with p53. Finally, translating these results we find that a selective small-molecule inhibitor of the Creb1-Cbp interaction demonstrates selective toxicity to OS cells where this pathway is constitutively active. This highlights the cAMP/Creb axis as a potentially actionable therapeutic vulnerability in p53-deficient tumors such as OS. These results define a mechanism through which p53 protects normal osteoblasts from excessive or abnormal cAMP accumulation, which becomes fundamentally compromised in OS.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Osteoblasts/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Membrane Proteins/metabolism , Mice , Osteosarcoma/metabolism , Signal Transduction/physiology
5.
Article in English | MEDLINE | ID: mdl-29867773

ABSTRACT

Parathyroid hormone-related protein (PTHrP) expression in breast cancer is enriched in bone metastases compared to primary tumors. Human MCF7 breast cancer cells "home" to the bones of immune deficient mice following intracardiac inoculation, but do not grow well and stain negatively for Ki67, thus serving as a model of breast cancer dormancy in vivo. We have previously shown that PTHrP overexpression in MCF7 cells overcomes this dormant phenotype, causing them to grow as osteolytic deposits, and that PTHrP-overexpressing MCF7 cells showed significantly lower expression of genes associated with dormancy compared to vector controls. Since early work showed a lack of cyclic AMP (cAMP) response to parathyroid hormone (PTH) in MCF7 cells, and cAMP is activated by PTH/PTHrP receptor (PTHR1) signaling, we hypothesized that the effects of PTHrP on dormancy in MCF7 cells occur through non-canonical (i.e., PTHR1/cAMP-independent) signaling. The data presented here demonstrate the lack of cAMP response in MCF7 cells to full length PTHrP(1-141) and PTH(1-34) in a wide range of doses, while maintaining a response to three known activators of adenylyl cyclase: calcitonin, prostaglandin E2 (PGE2), and forskolin. PTHR1 mRNA was detectable in MCF7 cells and was found in eight other human breast and murine mammary carcinoma cell lines. Although PTHrP overexpression in MCF7 cells changed expression levels of many genes, RNAseq analysis revealed that PTHR1 was unaltered, and only 2/32 previous PTHR1/cAMP responsive genes were significantly upregulated. Instead, PTHrP overexpression in MCF7 cells resulted in significant enrichment of the calcium signaling pathway. We conclude that PTHR1 in MCF7 breast cancer cells is not functionally linked to activation of the cAMP pathway. Gene expression responses to PTHrP overexpression must, therefore, result from autocrine or intracrine actions of PTHrP independent of PTHR1, through signals emanating from other domains within the PTHrP molecule.

7.
Development ; 143(4): 648-57, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26755702

ABSTRACT

The majority of the skeleton arises by endochondral ossification, whereby cartilaginous templates expand and are resorbed by osteoclasts then replaced by osteoblastic bone formation. Ephrin B2 is a receptor tyrosine kinase expressed by osteoblasts and growth plate chondrocytes that promotes osteoblast differentiation and inhibits osteoclast formation. We investigated the role of ephrin B2 in endochondral ossification using Osx1Cre-targeted gene deletion. Neonatal Osx1Cre.Efnb2(Δ/Δ) mice exhibited a transient osteopetrosis demonstrated by increased trabecular bone volume with a high content of growth plate cartilage remnants and increased cortical thickness, but normal osteoclast numbers within the primary spongiosa. Osteoclasts at the growth plate had an abnormal morphology and expressed low levels of tartrate-resistant acid phosphatase; this was not observed in more mature bone. Electron microscopy revealed a lack of sealing zones and poor attachment of Osx1Cre.Efnb2(Δ/Δ) osteoclasts to growth plate cartilage. Osteoblasts at the growth plate were also poorly attached and impaired in their ability to deposit osteoid. By 6 months of age, trabecular bone mass, osteoclast morphology and osteoid deposition by Osx1Cre.Efnb2(Δ/Δ) osteoblasts were normal. Cultured chondrocytes from Osx1Cre.Efnb2(Δ/Δ) neonates showed impaired support of osteoclastogenesis but no significant change in Rankl (Tnfsf11) levels, whereas Adamts4 levels were significantly reduced. A population of ADAMTS4(+) early hypertrophic chondrocytes seen in controls was absent from Osx1Cre.Efnb2(Δ/Δ) neonates. This suggests that Osx1Cre-expressing cells, including hypertrophic chondrocytes, are dependent on ephrin B2 for their production of cartilage-degrading enzymes, including ADAMTS4, and this might be required for attachment of osteoclasts and osteoblasts to the cartilage surface during endochondral ossification.


Subject(s)
Cartilage/pathology , Chondrocytes/metabolism , Ephrin-B2/metabolism , Osteoclasts/pathology , Osteogenesis , ADAM Proteins/metabolism , ADAMTS4 Protein , Animals , Animals, Newborn , Cartilage/metabolism , Cell Adhesion , Cell Differentiation , Chondrocytes/pathology , Female , Gene Expression Regulation , Immunohistochemistry , Integrases/metabolism , Mice, Inbred C57BL , Models, Biological , Organ Size , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/ultrastructure , Osteogenesis/genetics , Osteopetrosis/genetics , Osteopetrosis/pathology , Phenotype , Procollagen N-Endopeptidase/metabolism , Tibia/metabolism , Tibia/pathology
8.
J Endocrinol ; 223(2): 181-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25228504

ABSTRACT

Parathyroid hormone (PTH) treatment stimulates osteoblast differentiation and bone formation, and is the only currently approved anabolic therapy for osteoporosis. In cells of the osteoblast lineage, PTH also stimulates the expression of members of the interleukin 6 (IL-6) cytokine superfamily. Although the similarity of gene targets regulated by these cytokines and PTH suggest cooperative action, the dependence of PTH anabolic action on IL-6 cytokine signaling is unknown. To determine whether cytokine signaling in the osteocyte through glycoprotein 130 (gp130), the common IL-6 superfamily receptor subunit, is required for PTH anabolic action, male mice with conditional gp130 deletion in osteocytes (Dmp1Cre.gp130(f/f)) and littermate controls (Dmp1Cre.gp130(w/w)) were treated with hPTH(1-34) (30 µg/kg 5× per week for 5 weeks). PTH dramatically increased bone formation in Dmp1Cre.gp130(w/w) mice, as indicated by elevated osteoblast number, osteoid surface, mineralizing surface, and increased serum N-terminal propeptide of type 1 collagen (P1NP). However, in mice with Dmp1Cre-directed deletion of gp130, PTH treatment changed none of these parameters. Impaired PTH anabolic action was associated with a 50% reduction in Pth1r mRNA levels in Dmp1Cre.gp130(f/f) femora compared with Dmp1Cre.gp130(w/w). Furthermore, lentiviral-Cre infection of gp130(f/f) primary osteoblasts also lowered Pth1r mRNA levels to 16% of that observed in infected C57/BL6 cells. In conclusion, osteocytic gp130 is required to maintain PTH1R expression in the osteoblast lineage, and for the stimulation of osteoblast differentiation that occurs in response to PTH.


Subject(s)
Cytokine Receptor gp130/physiology , Osteoblasts/drug effects , Osteocytes/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/physiology , Osteocytes/cytology , Osteocytes/physiology , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/genetics
9.
FASEB J ; 28(10): 4482-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24982128

ABSTRACT

Cells that form bone (osteoblasts) express both ephrinB2 and EphB4, and previous work has shown that pharmacological inhibition of the ephrinB2/EphB4 interaction impairs osteoblast differentiation in vitro and in vivo. The purpose of this study was to determine the role of ephrinB2 signaling in the osteoblast lineage in the process of bone formation. Cultured osteoblasts from mice with osteoblast-specific ablation of ephrinB2 showed delayed expression of osteoblast differentiation markers, a finding that was reproduced by ephrinB2, but not EphB4, RNA interference. Microcomputed tomography, histomorphometry, and mechanical testing of the mice lacking ephrinB2 in osteoblasts revealed a 2-fold delay in bone mineralization, a significant reduction in bone stiffness, and a 50% reduction in osteoblast differentiation induced by anabolic parathyroid hormone (PTH) treatment, compared to littermate sex- and age-matched controls. These defects were associated with significantly lower mRNA levels of late osteoblast differentiation markers and greater levels of osteoblast and osteocyte apoptosis, indicated by TUNEL staining and transmission electron microscopy of bone samples, and a 2-fold increase in annexin V staining and 7-fold increase in caspase 8 activation in cultured ephrinB2 deficient osteoblasts. We conclude that osteoblast differentiation and bone strength are maintained by antiapoptotic actions of ephrinB2 signaling within the osteoblast lineage.


Subject(s)
Apoptosis , Calcification, Physiologic , Osteoblasts/metabolism , Osteogenesis , Receptor, EphB2/metabolism , Animals , Annexin A5/genetics , Annexin A5/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Receptor, EphB2/genetics , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Signal Transduction
10.
Mol Cell Oncol ; 1(4): e965624, 2014.
Article in English | MEDLINE | ID: mdl-27308361

ABSTRACT

In osteosarcoma, knockdown of the parathyroid hormone-related protein (PTHrP) receptor reduces activation through cyclic AMP-dependent protein kinase A (PKA) and substantially decreases tumor differentiation, invasion, and proliferation in vivo. These findings complement other evidence supporting a central role of the PKA pathway in osteosarcoma biology and pathogenesis.

11.
Bone ; 55(1): 166-78, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23486187

ABSTRACT

Osteosarcoma is the most common primary cancer of bone and one that predominantly affects children and adolescents. Osteoblastic osteosarcoma represents the major subtype of this tumor, with approximately equal representation of fibroblastic and chondroblastic subtypes. We and others have previously described murine models of osteosarcoma based on osteoblast-restricted Cre:lox deletion of Trp53 (p53) and Rb1 (Rb), resulting in a phenotype most similar to fibroblastic osteosarcoma in humans. We now report a model of the most prevalent form of human osteosarcoma, the osteoblastic subtype. In contrast to other osteosarcoma models that have used Cre:lox mediated gene deletion, this model was generated through shRNA-based knockdown of p53. As is the case with the human disease the shRNA tumors most frequently present in the long bones and preferentially disseminate to the lungs; feature less consistently modeled using Cre:lox approaches. Our approach allowed direct comparison of the in vivo consequences of targeting the same genetic drivers using two different technologies, Cre:lox and shRNA. This demonstrated that the effects of Cre:lox and shRNA mediated knock-down are qualitatively different, at least in the context of osteosarcoma, and yielded distinct subtypes of osteosarcoma. Through the use of complementary genetic modification strategies we have established a model of the most common clinical subtype of osteosarcoma that was not previously represented and more fully recapitulated the clinical spectrum of this cancer.


Subject(s)
Cell Lineage/genetics , Integrases/metabolism , Models, Biological , Osteosarcoma/classification , Osteosarcoma/genetics , RNA, Small Interfering/metabolism , Transgenes/genetics , Animals , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Membrane/metabolism , Chromosomes, Mammalian/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Karyotyping , Mice , Mice, Transgenic , Osteoblasts/metabolism , Osteoblasts/pathology , Osteosarcoma/diagnostic imaging , Osteosarcoma/pathology , Penetrance , Phenotype , Radiography , Signal Transduction , Survival Analysis , Tumor Suppressor Protein p53/metabolism
12.
J Bone Miner Res ; 28(4): 912-25, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23165727

ABSTRACT

Previous reports indicate that ephrinB2 expression by osteoblasts is stimulated by parathyroid hormone (PTH) and its related protein (PTHrP) and that ephrinB2/EphB4 signaling between osteoblasts and osteoclasts stimulates osteoblast differentiation while inhibiting osteoclast differentiation. To determine the role of the ephrinB2/EphB4 interaction in the skeleton, we used a specific inhibitor, soluble EphB4 (sEphB4), in vitro and in vivo. sEphB4 treatment of cultured osteoblasts specifically inhibited EphB4 and ephrinB2 phosphorylation and reduced mRNA levels of late markers of osteoblast/osteocyte differentiation (osteocalcin, dentin matrix protein-1 [DMP-1], sclerostin, matrix-extracellular phosphoglycoprotein [MEPE]), while substantially increasing RANKL. sEphB4 treatment in vivo in the presence and absence of PTH increased osteoblast formation and mRNA levels of early osteoblast markers (Runx2, alkaline phosphatase, Collagen 1α1, and PTH receptor [PTHR1]), but despite a substantial increase in osteoblast numbers, there was no significant change in bone formation rate or in late markers of osteoblast/osteocyte differentiation. Rather, in the presence of PTH, sEphB4 treatment significantly increased osteoclast formation, an effect that prevented the anabolic effect of PTH, causing instead a decrease in trabecular number. This enhancement of osteoclastogenesis by sEphB4 was reproduced in vitro but only in the presence of osteoblasts. These data indicate that ephrinB2/EphB4 signaling within the osteoblast lineage is required for late stages of osteoblast differentiation and, further, restricts the ability of osteoblasts to support osteoclast formation, at least in part by limiting RANKL production. This indicates a key role for the ephrinB2/EphB4 interaction within the osteoblast lineage in osteoblast differentiation and support of osteoclastogenesis.


Subject(s)
Anabolic Agents/pharmacology , Cell Lineage/drug effects , Ephrin-B2/antagonists & inhibitors , Osteoblasts/cytology , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Receptor, EphB4/antagonists & inhibitors , Animals , Biomarkers/metabolism , Cell Count , Cells, Cultured , Ephrin-B2/metabolism , Femur/drug effects , Humans , Male , Mice, Inbred C57BL , Organ Size/drug effects , Osteoblasts/drug effects , Osteocytes/drug effects , Osteocytes/metabolism , RANK Ligand/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, EphB4/metabolism , Receptor, EphB4/pharmacology , Skull/cytology , Solubility
13.
J Bone Miner Res ; 27(4): 902-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22190112

ABSTRACT

Parathyroid hormone (PTH) is the only approved anabolic agent for osteoporosis treatment. It acts via osteoblasts to stimulate both osteoclast formation and bone formation, with the balance between these two activities determined by the mode of administration. Oncostatin M (OSM), a gp130-dependent cytokine expressed by osteoblast lineage cells, has similar effects and similar gene targets in the osteoblast lineage. In this study, we investigated whether OSM might participate in anabolic effects of PTH. Microarray analysis and quantitative real-time polymerase chain reaction (qPCR) of PTH-treated murine stromal cells and primary calvarial osteoblasts identified significant regulation of gp130 and gp130-dependent coreceptors and ligands, including a significant increase in OSM receptor (OSMR) expression. To determine whether OSMR signaling is required for PTH anabolic action, 6-week-old male Osmr(-/-) mice and wild-type (WT) littermates were treated with hPTH(1-34) for 3 weeks. In WT mice, PTH increased trabecular bone volume and trabecular thickness. In contrast, the same treatment had a catabolic effect in Osmr(-/-) mice, reducing both trabecular bone volume and trabecular number. This was not explained by any alteration in the increased osteoblast formation and mineral apposition rate in response to PTH in Osmr(-/-) compared with WT mice. Rather, PTH treatment doubled osteoclast surface in Osmr(-/-) mice, an effect not observed in WT mice. Consistent with this finding, when osteoclast precursors were cultured in the presence of osteoblasts, more osteoclasts were formed in response to PTH when Osmr(-/-) osteoblasts were used. Neither PTH1R mRNA levels nor cAMP response to PTH were modified in Osmr(-/-) osteoblasts. However, RANKL induction in PTH-treated Osmr(-/-) osteoblasts was sustained at least until 24 hours after PTH exposure, an effect not observed in WT osteoblasts. These data indicate that the transient RANKL induction by intermittent PTH administration, which is associated with its anabolic action, is changed to a prolonged induction in OSMR-deficient osteoblasts, resulting in bone destruction.


Subject(s)
Anabolic Agents/pharmacology , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , RANK Ligand/metabolism , Receptors, Oncostatin M/deficiency , Animals , Cell Line , Cytokine Receptor gp130/metabolism , Cytokines/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Organ Size/drug effects , Osteoblasts/drug effects , Protein Binding/drug effects , RANK Ligand/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
14.
Bioorg Med Chem Lett ; 21(23): 7089-93, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22030030

ABSTRACT

PDE4 inhibitors have been identified as therapeutic targets for a variety of conditions, particularly inflammatory diseases. We have serendipitously identified a novel class of phosphodiesterase 4 (PDE4) inhibitor during a study to discover antagonists of the parathyroid hormone receptor. X-ray crystallographic studies of PDE4D2 complexed to four potent inhibitors reveal the atomic details of how they inhibit the enzyme and a notable contrast to another recently reported thiophene-based inhibitor.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Models, Molecular , Phosphodiesterase 4 Inhibitors/chemistry , Thiophenes/chemical synthesis , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Phosphodiesterase 4 Inhibitors/chemical synthesis , Protein Binding , Thiophenes/chemistry , Thiophenes/pharmacology
15.
J Biol Chem ; 284(23): 15557-63, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19346515

ABSTRACT

Parathyroid hormone-related protein (PTHrP) plays a vital role in the embryonic development of the skeleton and other tissues. When it is produced in excess by cancers it can cause hypercalcemia, and its local production by breast cancer cells has been implicated in the pathogenesis of bone metastasis formation in that disease. Antibodies have been developed that neutralize the action of PTHrP through its receptor, parathyroid hormone receptor 1, without influencing parathyroid hormone action through the same receptor. Such neutralizing antibodies against PTHrP are therapeutically effective in animal models of the humoral hypercalcemia of malignancy and of bone metastasis formation. We have determined the crystal structure of the complex between PTHrP (residues 1-108) and a neutralizing monoclonal anti-PTHrP antibody that reveals the only point of contact is an alpha-helical structure extending from residues 14-29. Another striking feature is that the same residues that interact with the antibody also interact with parathyroid hormone receptor 1, showing that the antibody and the receptor binding site on the hormone closely overlap. The structure explains how the antibody discriminates between the two hormones and provides information that could be used in the development of novel agonists and antagonists of their common receptor.


Subject(s)
Antibody Specificity , Cyclic AMP/metabolism , Receptor, Parathyroid Hormone, Type 1/immunology , Animals , Binding Sites , Bone Neoplasms , Cell Line, Tumor , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Neutralization Tests , Osteosarcoma , Protein Conformation , RNA, Messenger/genetics , Rats , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Surface Properties , X-Ray Diffraction
16.
J Cell Physiol ; 216(1): 144-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18247361

ABSTRACT

Bone morphogenetic protein-2 (BMP-2) is strongly involved in the induction of osteoblast differentiation from mesenchymal cell precursors, as well as in enhancing bone matrix production by osteoblastic cells. Likewise, the osteoporotic phenotype of PTHrP deficient mice makes clear the importance of this paracrine regulator in bone physiology. Here, we report that BMP-2 rapidly down-regulated PTHrP gene expression through a transcriptional mechanism in pluripotent mesenchymal C2C12 cells, whereas BMP-2 increased expression of PTHrP receptor. PTHrP did not significantly alter the BMP-dependent Smad transcriptional pathway. Similarly, PTHrP did not significantly modify the BMP-regulated expression of RANKL or OPG, cytokines involved in osteoclastogenesis. More importantly, addition of PTHrP, through the PKA signaling pathway, partially prevented the BMP-dependent induction of some osteogenic markers such as Runx2 and Osterix in C2C12 cells. Our data suggest that BMP-2 down-regulation of PTHrP could facilitate terminal differentiation of osteoblasts.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Osteogenesis/physiology , Parathyroid Hormone-Related Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/genetics , Bone and Bones/cytology , Bone and Bones/physiology , Cell Line , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Osteoblasts/cytology , Osteoclasts/cytology , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Parathyroid Hormone-Related Protein/genetics , Promoter Regions, Genetic , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta/genetics
17.
J Cell Biochem ; 90(1): 158-69, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12938165

ABSTRACT

In order to study osteoblast differentiation we subcloned a cell derived from a mouse a bone marrow stromal cell line, Kusa O, and obtained a number of clones representative of three different phenotypes. One that neither differentiated into osteoblasts nor into adipocytes, a second that differentiated into osteoblasts but not adipocytes, and a third that differentiated into both osteoblasts and adipocytes. Four subclones were selected for further characterization according to their ability to mineralize and/or differentiate into adipocytes. The non-mineralizing clone had no detectable alkaline phosphatase activity although some alkaline phosphatase mRNA was detected after 21 days in osteoblast differentiating medium. Alkaline phosphatase activity and mRNA in the three mineralizing clones were comparable with the parent clones. Osteocalcin mRNA and protein levels in the non-mineralizing clone were low and non-detectable, respectively, while both were elevated in the parent cells and mineralizing subclones after 21 days in differentiating medium. PTH receptor mRNA and activity increased in the four subclones and parent cells with differentiation. mRNA for two other osteoblast phenotypic markers, osteopontin and bone sialoprotein, were similarly expressed in the parent cells and subclones while mRNAs for the transcription factors, Runx2 and osterix, were detectable in both parent and subclone cells. Runx2 was unchanged with differentiation while osterix was increased. Interestingly, PPARgamma mRNA expression did not correlate with cell line potential to differentiate into adipocytes. Indian hedgehog mRNA and its receptor (patched) mRNA levels both increased with differentiation while mRNA levels of the Wnt pathway components beta-catenin and dickkopf also increased with differentiation. Although we have focussed on characterizing these clones from the osteoblast perspective it is clear that they may be useful for studying both osteoblast and adipocyte differentiation as well as their transdifferentiation.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Stromal Cells/physiology , Zebrafish Proteins , Adipocytes/physiology , Animals , Bone Marrow Cells/physiology , Cell Line , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA Primers , Hedgehog Proteins , Mice , Osteoblasts/cytology , Osteoblasts/physiology , Phenotype , Polymerase Chain Reaction , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Stromal Cells/cytology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...