Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 10(1): 1354, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30902975

ABSTRACT

Osteoporosis is caused by increased bone resorption and decreased bone formation. Intermittent administration of a fragment of Parathyroid hormone (PTH) activates osteoblast-mediated bone formation and is used in patients with severe osteoporosis. However, the mechanisms by which PTH elicits its anabolic effect are not fully elucidated. Here we show that the absence of the homeodomain protein TG-interacting factor 1 (Tgif1) impairs osteoblast differentiation and activity, leading to a reduced bone formation. Deletion of Tgif1 in osteoblasts and osteocytes decreases bone resorption due to an increased secretion of Semaphorin 3E (Sema3E), an osteoclast-inhibiting factor. Tgif1 is a PTH target gene and PTH treatment failed to increase bone formation and bone mass in Tgif1-deficient mice. Thus, our study identifies Tgif1 as a novel regulator of bone remodeling and an essential component of the PTH anabolic action. These insights contribute to a better understanding of bone metabolism and the anabolic function of PTH.


Subject(s)
Anabolic Agents/pharmacology , Bone Remodeling/drug effects , Parathyroid Hormone/pharmacology , Repressor Proteins/deficiency , Adaptor Proteins, Signal Transducing , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Differentiation/drug effects , Gene Deletion , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Organ Size/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Repressor Proteins/metabolism , Semaphorins/pharmacology , Transcription Factor AP-1/metabolism , Wnt Signaling Pathway/drug effects
2.
J Bone Miner Res ; 32(11): 2207-2218, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28650106

ABSTRACT

Osteoclastogenesis is essential for bone remodeling and normal skeletal maintenance. Receptor activator of NF-κB ligand (RANKL) promotes osteoclast differentiation and function but requires costimulation of immunoreceptor tyrosine-based activation motif (ITAM)-coupled immunoreceptors. Triggering receptor expressed on myeloid cells-2 (TREM2) coupled to ITAM-adaptor protein DNAX activation protein 12kDA (DAP12) provides costimulation of intracellular calcium signaling during osteoclastogenesis. Previously, we found that downstream of kinase-3 (DOK3) physically associates with DAP12 to inhibit toll-like receptor (TLR)-induced inflammatory signaling in macrophages. However, whether and how DOK3 modulates DAP12-dependent osteoclastogenesis is unknown and the focus of this study. Bone microarchitecture and histology of sex- and age-matched wild-type (WT) and DOK3-deficient (DOK3-/- ) mice were evaluated. Male and female DOK3-/- mice have significantly reduced trabecular bone mass compared with WT mice with increased TRAP+ osteoclasts in vivo. In vitro, DOK3-/- bone marrow-derived macrophages (BMMs) have increased macrophage colony-stimulating factor (M-CSF)-induced proliferation and increased sensitivity to RANKL-induced osteoclastogenesis. Compared with WT, DOK3-/- osteoclasts are significantly larger with more nuclei and have increased resorptive capacity. Mechanistically, DOK3 limits osteoclastogenesis by inhibiting activation of Syk and ERK in response to RANKL and M-CSF. DOK3 is phosphorylated in a DAP12-dependent manner and associates with Grb2 and Cbl. Compared with DAP12-/- mice with high bone mass, DOK3- and DAP12- doubly deficient mice (DKO) have normalized bone mass, indicating that DOK3 also limits DAP12-independent osteoclastogenesis in vivo. In vitro osteoclasts derived from DKO mice are mononuclear with poor resorptive capacity similar to DAP12-/- osteoclasts. Histomorphometry reveals that DOK3-/- mice also have reduced osteoblast parameters. DOK3-/- osteoblasts have reduced in vitro osteoblastogenesis and increased osteoprotegerin (OPG) to RANKL expression ratio compared with WT osteoblasts. Co-culture of WT and DOK3-/- osteoblasts with pre-osteoclasts reveals a reduced capacity of DOK3-/- osteoblasts to support osteoclastogenesis. These data indicate that DOK3 regulates bone remodeling by negatively regulating M-CSF- and RANKL-mediated osteoclastogenesis and positively regulating osteoblastogenesis. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Remodeling , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , Adaptor Proteins, Signal Transducing/deficiency , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Remodeling/drug effects , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoporosis/metabolism , Osteoporosis/pathology , Phosphorylation/drug effects , RANK Ligand/metabolism , RAW 264.7 Cells , Receptors, Immunologic/metabolism , Signal Transduction/drug effects
3.
Sci Signal ; 6(289): ra72, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23962980

ABSTRACT

DNAX-activating protein of 12 kD (DAP12) is an immunoreceptor tyrosine-based activation motif (ITAM)-containing adaptor protein found in myeloid cells and natural killer cells, and it couples to various receptors that mediate either cellular activation or inhibition. DAP12 inhibits Toll-like receptor (TLR) signaling, such as that of TLR4 in response to its ligand lipopolysaccharide (LPS), as well as cytokine responses by coupling to TREM2 (triggering receptor expressed on myeloid cells 2) at the plasma membrane. Understanding the mechanisms that inhibit inflammatory responses in macrophages is important for the development of therapies to treat inflammatory diseases. We show that inhibition of LPS responses by DAP12 is mediated by the adaptor protein DOK3 (downstream of kinase 3). DOK3 physically associated with the ITAM of DAP12 through its phosphotyrosine-binding domain. In response to LPS, DOK3 was phosphorylated in a DAP12- and Src-dependent manner, which led to translocation of phosphorylated DOK3 to the plasma membrane. DOK3-deficient cells exhibited increased production of proinflammatory cytokines and activation of extracellular signal-regulated kinase (ERK). Compared to wild-type mice, DOK3-deficient mice had increased susceptibility to challenge with a sublethal dose of LPS and produced increased serum concentrations of the inflammatory cytokine tumor necrosis factor-α (TNF-α). Together, these data suggest the mechanism by which DAP12 and TREM2 inhibit LPS signaling in macrophages to prevent inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cell Membrane/immunology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/immunology , Macrophages/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Cell Membrane/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Lipopolysaccharides/adverse effects , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Mutant Strains , Phosphorylation/drug effects , Phosphorylation/immunology , Protein Transport/genetics , Protein Transport/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
4.
J Biol Chem ; 288(31): 22219-32, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23770672

ABSTRACT

Ca(2+) signaling is essential for bone homeostasis and skeletal development. Here, we show that the transient receptor potential canonical 1 (TRPC1) channel and the inhibitor of MyoD family, I-mfa, function antagonistically in the regulation of osteoclastogenesis. I-mfa null mice have an osteopenic phenotype characterized by increased osteoclast numbers and surface, which are normalized in mice lacking both Trpc1 and I-mfa. In vitro differentiation of pre-osteoclasts derived from I-mfa-deficient mice leads to an increased number of mature osteoclasts and higher bone resorption per osteoclast. These parameters return to normal levels in osteoclasts derived from double mutant mice. Consistently, whole cell currents activated in response to the depletion of intracellular Ca(2+) stores are larger in pre-osteoclasts derived from I-mfa knock-out mice compared with currents in wild type mice and normalized in cells derived from double mutant mice, suggesting a cell-autonomous effect of I-mfa on TRPC1 in these cells. A new splice variant of TRPC1 (TRPC1ε) was identified in early pre-osteoclasts. Heterologous expression of TRPC1ε in HEK293 cells revealed that it is unique among all known TRPC1 isoforms in its ability to amplify the activity of the Ca(2+) release-activated Ca(2+) (CRAC) channel, mediating store-operated currents. TRPC1ε physically interacts with Orai1, the pore-forming subunit of the CRAC channel, and I-mfa is recruited to the TRPC1ε-Orai1 complex through TRPC1ε suppressing CRAC channel activity. We propose that the positive and negative modulation of the CRAC channel by TRPC1ε and I-mfa, respectively, fine-tunes the dynamic range of the CRAC channel regulating osteoclastogenesis.


Subject(s)
Osteoclasts/cytology , TRPC Cation Channels/physiology , Animals , Base Sequence , Cell Division , Cell Line , Codon , DNA Primers , Humans , Mice , Mice, Knockout , Protein Biosynthesis , RNA, Messenger/genetics , TRPC Cation Channels/genetics
5.
Mater Sci Eng C Mater Biol Appl ; 33(1): 327-39, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23565038

ABSTRACT

Commercially pure titanium plates/coupons and pure titanium powders were soaked for 24 h in 5 M NaOH and 5 M KOH solutions, under identical conditions, over the temperature range of 37° to 90 °C. Wettability of the surfaces of alkali-treated cpTi coupons was studied by using contact angle goniometry. cpTi coupons soaked in 5 M NaOH or 5 M KOH solutions were found to have hydrophilic surfaces. Hydrous alkali titanate nanofibers and nanotubes were identified with SEM/EDXS and grazing incidence XRD. Surface areas of Ti powders increased > 50­220 times, depending on the treatment, when soaked in the above solutions. A solution was developed to coat amorphous calcium phosphate, instead of hydroxyapatite, on Ti coupon surfaces. In vitro cell culture tests were performed with osteoblast-like cells on the alkali-treated samples.


Subject(s)
Hydroxides/chemistry , Potassium Compounds/chemistry , Sodium Hydroxide/chemistry , Titanium/chemistry , Animals , Calcium Phosphates/chemistry , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Mice , Microscopy, Electron, Scanning , Nanofibers/chemistry , Nanofibers/ultrastructure , Nanotubes/chemistry , Nanotubes/ultrastructure , Solutions/chemistry , Spectrometry, X-Ray Emission , Surface Properties , Temperature , Titanium/pharmacology , Wettability , X-Ray Diffraction
6.
J Bone Miner Res ; 27(12): 2476-89, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22807064

ABSTRACT

Osteoclasts are bone-specific polykaryons derived from myeloid precursors under the stimulation of macrophage colony stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). E proteins are basic helix-loop-helix (bHLH) transcription factors that modulate lymphoid versus myeloid cell fate decisions. To study the role of E proteins in osteoclasts, myeloid-specific E protein gain-of-function transgenic mice were generated. These mice have high bone mass due to decreased osteoclast numbers and increased osteoclast apoptosis leading to overall reductions in resorptive capacity. The molecular mechanism of decreased osteoclast numbers and resorption is in part a result of elevated expression of CD38, a regulator of intracellular calcium pools with known antiosteoclastogenic properties, which increases sensitivity to apoptosis. In vivo, exogenous RANKL stimulation can overcome this inhibition to drive osteoclastogenesis and bone loss. In vitro-derived ET2 osteoclasts are more spread and more numerous with increases in RANK, triggering receptor expressed on myeloid cells 2 (TREM2), and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) compared to wild type. However, their resorptive capacity does not increase accordingly. Thus, E proteins participate in osteoclast maturation and survival in homeostatic bone remodeling.


Subject(s)
Cell Differentiation/drug effects , Osteoclasts/physiology , Transcription Factor 3/physiology , ADP-ribosyl Cyclase 1/biosynthesis , Animals , Apoptosis , Bone Remodeling/genetics , Bone Resorption , Bone and Bones/anatomy & histology , Female , Mice , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/pathology , RANK Ligand/pharmacology
7.
J Cell Physiol ; 227(6): 2622-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21882189

ABSTRACT

CD731 is a GPI-anchored cell surface protein with ecto-5'-nucleotidase enzyme activity that plays a crucial role in adenosine production. While the roles of adenosine receptors (AR) on osteoblasts and osteoclasts have been unveiled to some extent, the roles of CD73 and CD73-generated adenosine in bone tissue are largely unknown. To address this issue, we first analyzed the bone phenotype of CD73-deficient (cd73(-/-)) mice. The mutant male mice showed osteopenia, with significant decreases of osteoblastic markers. Levels of osteoclastic markers were, however, comparable to those of wild-type mice. A series of in vitro studies revealed that CD73 deficiency resulted in impairment in osteoblast differentiation but not in the number of osteoblast progenitors. In addition, over expression of CD73 on MC3T3-E1 cells resulted in enhanced osteoblastic differentiation. Moreover, MC3T3-E1 cells expressed adenosine A(2A) receptors (A(2A)AR) and A(2B) receptors (A(2B)AR) and expression of these receptors increased with osteoblastic differentiation. Enhanced expression of osteocalcin (OC) and bone sialoprotein (BSP) observed in MC3T3-E1 cells over expressing CD73 were suppressed by treatment with an A(2B)AR antagonist but not with an A(2A) AR antagonist. Collectively, our results indicate that CD73 generated adenosine positively regulates osteoblast differentiation via A(2B)AR signaling.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine/metabolism , Cell Differentiation , Femur/enzymology , Osteoblasts/enzymology , Tibia/enzymology , 3T3 Cells , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Biomarkers/metabolism , Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/enzymology , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/pathology , Cell Differentiation/drug effects , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Genotype , Humans , Integrin-Binding Sialoprotein/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/drug effects , Osteoblasts/pathology , Osteocalcin/metabolism , Osteogenesis , Phenotype , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , Signal Transduction , Tibia/diagnostic imaging , Tibia/drug effects , Tibia/pathology , Time Factors , Transfection , X-Ray Microtomography
8.
Bonekey Rep ; 12012 Apr 18.
Article in English | MEDLINE | ID: mdl-23789115

ABSTRACT

The study of bone and immunology (termed osteoimmunology) has led to the discovery of many important similarities between the two systems including shared niches, mechanisms, cytokines and receptors. The bone marrow provides a niche for hematopoietic cells including those of the lymphoid and myeloid lineage. Osteoclasts, specialized polykarons arising from myeloid precursors, bind to bone and resorb the organic and inorganic components through secretion of acid and proteases. Osteoclasts are differentiated and activated by cytokines that can be produced by immune cells and osteoclast activity can be dysregulated in states of autoimmunity or high inflammation. Similar to B and T cells, osteoclasts require coordinated co-stimulation of signaling pathways provided in the form of receptor-associated immunoreceptor tyrosine-based activation motif adaptor proteins, DAP12 and FcRγ, to drive differentiation and activation. In this review, we will cover the differentiation process of osteoclasts from the earliest precursors shown to have differentiation potential and the signals needed to drive these cells into osteoclast commitment and activation.

SELECTION OF CITATIONS
SEARCH DETAIL
...