Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Bone Miner Res ; 37(4): 764-775, 2022 04.
Article in English | MEDLINE | ID: mdl-35080046

ABSTRACT

Vertebrate lonesome kinase (Vlk) is a secreted tyrosine kinase important for normal skeletogenesis during embryonic development. Vlk null mice (Vlk-/- ) are born with severe craniofacial and limb skeletal defects and die shortly after birth. We used a conditional deletion model to remove Vlk in limb bud mesenchyme (Vlk-Prx1 cKO) to assess the specific requirement for Vlk expression by skeletal progenitor cells during endochondral ossification, and an inducible global deletion model (Vlk-Ubq iKO) to address the role of Vlk during fracture repair. Deletion of Vlk with Prx1-Cre recapitulated the limb skeletal phenotype of the Vlk-/- mice and enabled us to study the postnatal skeleton as Vlk-Prx1 cKO mice survived to adulthood. In Vlk-Prx1 cKO adult mice, limbs remained shorter with decreased trabecular and cortical bone volumes. Both Vlk-Prx1 cKO and Vlk-Ubq iKO mice had a delayed fracture repair response but eventually formed bridging calluses. Furthermore, levels of phosphorylated osteopontin (OPN) were decreased in tibias of Vlk-Ubq iKO, establishing OPN as a Vlk substrate in bone. In summary, our data indicate that Vlk produced by skeletal progenitor cells influences the timing and extent of chondrogenesis during endochondral bone formation and fracture repair. © 2022 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Chondrogenesis , Osteogenesis , Animals , Bone and Bones , Chondrogenesis/genetics , Extremities , Mice , Mice, Knockout , Osteogenesis/genetics , Protein-Tyrosine Kinases
2.
Blood ; 139(1): 104-117, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34329392

ABSTRACT

Tyrosine phosphorylation of extracellular proteins is observed in cell cultures and in vivo, but little is known about the functional roles of tyrosine phosphorylation of extracellular proteins. Vertebrate lonesome kinase (VLK) is a broadly expressed secretory pathway tyrosine kinase present in platelet α-granules. It is released from platelets upon activation and phosphorylates substrates extracellularly. Its role in platelet function, however, has not been previously studied. In human platelets, we identified phosphorylated tyrosines mapped to luminal or extracellular domains of transmembrane and secreted proteins implicated in the regulation of platelet activation. To determine the role of VLK in extracellular tyrosine phosphorylation and platelet function, we generated mice with a megakaryocyte/platelet-specific deficiency of VLK. Platelets from these mice are normal in abundance and morphology but have significant changes in function both in vitro and in vivo. Resting and thrombin-stimulated VLK-deficient platelets exhibit a significant decrease in several tyrosine phosphobands. Results of functional testing of VLK-deficient platelets show decreased protease-activated receptor 4-mediated and collagen-mediated platelet aggregation but normal responses to adenosine 5'-diphosphate. Dense granule and α-granule release are reduced in these platelets. Furthermore, VLK-deficient platelets exhibit decreased protease-activated receptor 4-mediated Akt (S473) and Erk1/2 (T202/Y204) phosphorylation, indicating altered proximal signaling. In vivo, mice lacking VLK in megakaryocytes/platelets display strongly reduced platelet accumulation and fibrin formation after laser-induced injury of cremaster arterioles compared with control mice but with normal bleeding times. These studies show that the secretory pathway tyrosine kinase VLK is critical for stimulus-dependent platelet activation and thrombus formation, providing the first evidence that a secreted protein kinase is required for normal platelet function.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Protein-Tyrosine Kinases/metabolism , Thrombosis/metabolism , Animals , Blood Platelets/pathology , Gene Deletion , HEK293 Cells , Humans , Mice, Transgenic , Protein-Tyrosine Kinases/genetics , Thrombosis/pathology
4.
J Bone Miner Res ; 32(6): 1332-1342, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28240364

ABSTRACT

N-cadherin inhibits osteogenic cell differentiation and canonical Wnt/ß-catenin signaling in vitro. However, in vivo both conditional Cdh2 ablation and overexpression in osteoblasts lead to low bone mass. We tested the hypothesis that N-cadherin has different effects on osteolineage cells depending upon their differentiation stage. Embryonic conditional osteolineage Cdh2 deletion in mice results in defective growth, low bone mass, and reduced osteoprogenitor number. These abnormalities are prevented by delaying Cdh2 ablation until 1 month of age, thus targeting only committed and mature osteoblasts, suggesting they are the consequence of N-cadherin deficiency in osteoprogenitors. Indeed, diaphyseal trabecularization actually increases when Cdh2 is ablated postnatally. The sclerostin-insensitive Lrp5A214V mutant, associated with high bone mass, does not rescue the growth defect, but it overrides the low bone mass of embryonically Cdh2-deleted mice, suggesting N-cadherin interacts with Wnt signaling to control bone mass. Finally, bone accrual and ß-catenin accumulation after administration of an anti-Dkk1 antibody are enhanced in N-cadherin-deficient mice. Thus, although lack of N-cadherin in embryonic and perinatal age is detrimental to bone growth and bone accrual, in adult mice loss of N-cadherin in osteolineage cells favors bone formation. Hence, N-cadherin inhibition may widen the therapeutic window of osteoanabolic agents. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Cadherins/metabolism , Cell Lineage , Homeostasis , Osteogenesis , Animals , Animals, Newborn , Bone and Bones/pathology , Cell Count , Embryo, Mammalian/cytology , Gain of Function Mutation , Gene Deletion , Intercellular Signaling Peptides and Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mesenchymal Stem Cells/metabolism , Mice, Knockout , Organ Size , Osteoblasts/metabolism , Phenotype , Sp7 Transcription Factor/metabolism
5.
J Bone Miner Res ; 30(2): 274-85, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25088803

ABSTRACT

Interaction between parathyroid hormone/parathyroid hormone-related peptide receptor 1 (PTHR1) and low-density lipoprotein receptor-related protein 6 (Lrp6) is important for parathyroid hormone (PTH) signaling and anabolic action. Because N-cadherin has been shown to negatively regulate canonical Wnt/ß-catenin signaling, we asked whether N-cadherin alters PTH signaling and stimulation of bone formation. Ablation of the N-cadherin gene (Cdh2) in primary osteogenic lineage cells resulted in increased Lrp6/PTHR1 interaction in response to PTH1-34 , associated with enhanced PTH-induced PKA signaling and PKA-dependent ß-catenin C-terminus phosphorylation, which promotes ß-catenin transcriptional activity. ß-catenin C-terminus phosphorylation was abolished by Lrp6 knockdown. Accordingly, PTH1-34 stimulation of Tcf/Lef target genes, Lef1 and Axin2, was also significantly enhanced in Cdh2-deficient cells. This enhanced responsiveness to PTH extends to the osteo-anabolic effect of PTH, as mice with a conditional Cdh2 deletion in Osx+ cells treated with intermittent doses of PTH1-34 exhibited significantly larger gains in trabecular bone mass relative to control mice, the result of accentuated osteoblast activity. Therefore, N-cadherin modulates Lrp6/PTHR1 interaction, restraining the intensity of PTH-induced ß-catenin signaling, and ultimately influencing bone formation in response to intermittent PTH administration.


Subject(s)
Anabolic Agents/pharmacology , Cadherins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Signal Transduction/drug effects , beta Catenin/metabolism , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Gene Expression Regulation/drug effects , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Osteoblasts/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Receptor, Parathyroid Hormone, Type 1/metabolism , Sp7 Transcription Factor , TCF Transcription Factors/metabolism , Transcription Factors/metabolism , beta Catenin/chemistry
6.
Calcif Tissue Int ; 94(1): 46-54, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23657489

ABSTRACT

Direct cell-to-cell interactions via cell adhesion molecules, in particular cadherins, are critical for morphogenesis, tissue architecture, and cell sorting and differentiation. Partially overlapping, yet distinct roles of N-cadherin (cadherin-2) and cadherin-11 in the skeletal system have emerged from mouse genetics and in vitro studies. Both cadherins are important for precursor commitment to the osteogenic lineage, and genetic ablation of Cdh2 and Cdh11 results in skeletal growth defects and impaired bone formation. While Cdh11 defines the osteogenic lineage, persistence of Cdh2 in osteoblasts in vivo actually inhibits their terminal differentiation and impairs bone formation. The action of cadherins involves both cell-cell adhesion and interference with intracellular signaling, and in particular the Wnt/ß-catenin pathway. Both cadherin-2 and cadherin-11 bind to ß-catenin, thus modulating its cytoplasmic pools and transcriptional activity. Recent data demonstrate that cadherin-2 also interferes with Lrp5/6 signaling by sequestering these receptors in inactive pools via axin binding. These data extend the biologic action of cadherins in bone forming cells, and provide novel mechanisms for development of therapeutic strategies aimed at enhancing bone formation.


Subject(s)
Bone and Bones/cytology , Bone and Bones/metabolism , Cadherins/metabolism , Cell Adhesion/physiology , Cell Communication/physiology , Signal Transduction/physiology , Animals , Humans , Osteoblasts/metabolism
7.
Blood ; 120(2): 295-302, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22323481

ABSTRACT

There is evidence suggesting that N-cadherin expression on osteoblast lineage cells regulates hematopoietic stem cell (HSC) function and quiescence. To test this hypothesis, we conditionally deleted N-cadherin (Cdh2) in osteoblasts using Cdh2(flox/flox) Osx-Cre mice. N-cadherin expression was efficiently ablated in osteoblast lineage cells as assessed by mRNA expression and immunostaining of bone sections. Basal hematopoiesis is normal in these mice. In particular, HSC number, cell cycle status, long-term repopulating activity, and self-renewal capacity were normal. Moreover, engraftment of wild-type cells into N-cadherin-deleted recipients was normal. Finally, these mice responded normally to G-CSF, a stimulus that mobilizes HSCs by inducing alterations to the stromal micro-environment. In conclusion, N-cadherin expression in osteoblast lineage cells is dispensable for HSC maintenance in mice.


Subject(s)
Cadherins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Animals , Base Sequence , Bone Diseases, Metabolic/etiology , Cadherins/deficiency , Cadherins/genetics , Cell Lineage/genetics , Cell Lineage/physiology , Female , Fluorouracil/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...