Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
2.
JCI Insight ; 9(10)2024 May 22.
Article En | MEDLINE | ID: mdl-38713511

While sclerostin-neutralizing antibodies (Scl-Abs) transiently stimulate bone formation by activating Wnt signaling in osteoblast lineage cells, they exert sustained inhibition of bone resorption, suggesting an alternate signaling pathway by which Scl-Abs control osteoclast activity. Since sclerostin can activate platelet-derived growth factor receptors (PDGFRs) in osteoblast lineage cells in vitro and PDGFR signaling in these cells induces bone resorption through M-CSF secretion, we hypothesized that the prolonged anticatabolic effect of Scl-Abs could result from PDGFR inhibition. We show here that inhibition of PDGFR signaling in osteoblast lineage cells is sufficient and necessary to mediate prolonged Scl-Ab effects on M-CSF secretion and osteoclast activity in mice. Indeed, sclerostin coactivates PDGFRs independently of Wnt/ß-catenin signaling inhibition, by forming a ternary complex with LRP6 and PDGFRs in preosteoblasts. In turn, Scl-Ab prevents sclerostin-mediated coactivation of PDGFR signaling and consequent M-CSF upregulation in preosteoblast cultures, thereby inhibiting osteoclast activity in preosteoblast/osteoclast coculture assays. These results provide a potential mechanism explaining the dissociation between anabolic and antiresorptive effects of long-term Scl-Ab.


Adaptor Proteins, Signal Transducing , Bone Resorption , Osteoblasts , Osteoclasts , Receptors, Platelet-Derived Growth Factor , Signal Transduction , Animals , Osteoblasts/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antibodies, Neutralizing/pharmacology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Cell Lineage , Osteogenesis/drug effects , Cell Differentiation
3.
STAR Protoc ; 2(2): 100452, 2021 06 18.
Article En | MEDLINE | ID: mdl-33912848

Mesenchymal-derived osteoblasts play a key role in bone formation via synthesis and mineralization of the bone and bone remodeling. Osteoclasts are multinucleated cells of hematopoietic origin with a role in bone resorption. Here, we describe a protocol for generating primary cultures of these two cell types from bone tissue including the femur, tibia, and humerus of young mice. We describe methods for addressing their activity and/or differentiation, enabling studying the effects of various treatments during or following differentiation ex vivo. For further practical example of using these protocols, please refer to Chevalier et al. (2020).


Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Primary Cell Culture , Animals , Mice
4.
J Bone Miner Res ; 35(12): 2458-2469, 2020 12.
Article En | MEDLINE | ID: mdl-32777109

The physiological functions of platelet-derived growth factor receptors (PDGFRs) α and ß in osteoblast biology and bone metabolism remain to be established. Here, we show that PDGFRA and PDGFRB genes are expressed by osteoblast-lineage canopy and reversal cells in close proximity to PDGFB-expressing osteoclasts within human trabecular bone remodeling units. We also report that, although removal of only one of the two PDGFRs in Osterix-positive cells does not affect bone phenotype, suppression of both PDGFRs in those osteoblast lineage cells increases trabecular bone volume in male mice as well as in female gonad-intact and ovariectomized mice. Furthermore, osteoblast lineage-specific suppression of PDGFRs reduces Csf1 expression, bone marrow level of macrophage colony-stimulating factor (M-CSF), number of osteoclasts, and, therefore, bone resorption, but does not change bone formation. Finally, abrogation of PDGFR signaling in osteoblasts blocks PDGF-induced ERK1/2-mediated Csf1 expression and M-CSF secretion in osteoblast cultures and calcitriol-mediated osteoclastogenesis in co-cultures. In conclusion, our results indicate that PDGFR signaling in osteoblast lineage cells controls bone resorption through ERK1/2-mediated Csf1 expression. © 2020 American Society for Bone and Mineral Research (ASBMR).


Bone Resorption , Macrophage Colony-Stimulating Factor , Animals , Cell Differentiation , Female , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, Platelet-Derived Growth Factor , Up-Regulation
5.
Bone ; 113: 95-104, 2018 08.
Article En | MEDLINE | ID: mdl-29751129

Mice deficient in the non-receptor tyrosine kinase Src exhibit high bone mass due to impaired bone resorption and increased bone formation. Although several Src family kinase inhibitors inhibit bone resorption in vivo, they display variable effects on bone formation. SU6656 is a selective Src family kinase inhibitor with weaker activity towards the non-receptor tyrosine kinase Abl and receptor tyrosine kinases which are required for appropriate osteoblast proliferation, differentiation and function. Therefore, we sought to determine whether SU6656 could increase bone mass by inhibiting bone resorption and by stimulating bone formation, and to explore its mechanisms of action. Four-month-old female C57Bl/6J mice received intraperitoneal injections of either 25 mg/kg SU6656 or its vehicle every other day for 12 weeks. SU6656-treated mice exhibited increased bone mineral density, cortical thickness, cancellous bone volume and trabecular thickness. SU6656 inhibited bone resorption in mice as shown by reduced osteoclast number, and diminished expressions of Oscar, Trap5b and CtsK. SU6656 did not affect Rankl or Opg expressions. However, it blocked c-fms signaling, osteoclastogenesis and matrix resorption, and induced osteoclast apoptosis in vitro. In addition, SU6656 stimulated bone formation rates at trabecular, endosteal and periosteal bone envelopes, and increased osteoblast number in trabecular bone. SU6656 did not affect expressions of clastokines favoring bone formation in mice. However, it stimulated osteoblast differentiation and matrix mineralization by specifically facilitating BMP-SMAD signaling pathway in vitro. Knockdown of Src and Yes mimicked the stimulatory effect of SU6656 on osteoblast differentiation. In conclusion, SU6656 uncouples bone formation from resorption by inhibiting osteoclast development, function and survival, and by enhancing BMP-mediated osteoblast differentiation.


Indoles/pharmacology , Osteogenesis/drug effects , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Bone Density/drug effects , Bone Resorption/metabolism , Female , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoclasts/drug effects
6.
J Bone Miner Res ; 31(5): 985-93, 2016 05.
Article En | MEDLINE | ID: mdl-26643857

Intermittent parathyroid hormone administration (iPTH) increases bone mass and strength by stimulating osteoblast number and activity. PTH exerts its anabolic effects through cAMP/protein kinase A (PKA) signaling pathway in mature osteoblasts and osteocytes. Here, we show that inactivation of the p38α MAPK-encoding gene with the use of an osteocalcin-cre transgene prevents iPTH bone anabolic action. Indeed, iPTH fails to increase insulin-like growth factor 1 expression, osteoblast number and activity, and bone formation in mice lacking p38α in osteoblasts and osteocytes. Moreover, iPTH-induced expression of receptor activator of NF-κB ligand (RANKL) and subsequent increased bone resorption are suppressed in those mice. Finally, we found that PTH activates p38α MAPK downstream of cAMP/PKA signaling pathway in mature osteoblasts. Our findings identify p38α MAPK as a key component of PTH signaling in osteoblast lineage cells and highlight its requirement in iPTH osteoanabolic activity. © 2015 American Society for Bone and Mineral Research.


MAP Kinase Signaling System/drug effects , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , MAP Kinase Signaling System/genetics , Mice, Transgenic , Osteocytes/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
7.
Bonekey Rep ; 4: 757, 2015.
Article En | MEDLINE | ID: mdl-26587226

Sclerostin inhibits bone formation mostly by antagonizing LRP5/6, thus inhibiting Wnt signaling. However, experiments with genetically modified mouse models suggest that a significant part of sclerostin-mediated inhibition of bone formation is due to interactions with other binding partners. The objective of the present work was to identify signaling pathways affected by sclerostin in relation with its inhibitory action on osteogenic differentiation of C3H10T1/2 cells, MC3T3-E1 cells and primary osteoblasts. Sclerostin inhibited BMP2-induced osteoblast differentiation without altering SMAD1/5 phosphorylation and transcriptional activity. Moreover, sclerostin prevented Wnt3a-mediated osteoblastogenesis without affecting LRP5/6 phosphorylation or ß-catenin transcriptional activity. In addition, sclerostin inhibited mineralization promoted by GSK3 inhibition, which mimics canonical Wnt signaling without activation of LRP5/6, suggesting that sclerostin can prevent osteoblast differentiation without antagonizing LRP5/6. Finally, we found that sclerostin could activate platelet-derived growth factor receptor (PDGFR) and its downstream signaling pathways PLCγ, PKC, Akt and ERK1/2. PDGFR inhibition could reverse sclerostin-mediated inhibitory activity on BMP2-induced osteoblast differentiation. Therefore, our data suggest that sclerostin can activate PDGFR signaling by itself, and this functional interaction may be involved in the negative effect of sclerostin on osteoblast differentiation.

8.
Endocrinology ; 156(12): 4377-87, 2015 Dec.
Article En | MEDLINE | ID: mdl-26441240

Estrogen deficiency causes bone loss by increasing the number of bone-resorbing osteoclasts. Selective p38α MAPK inhibitors prevent bone-wasting effects of estrogen withdrawal but implicated mechanisms remain to be identified. Here, we show that inactivation of the p38α-encoding gene in osteoblast lineage cells with the use of an osteocalcin-cre transgene protects mice from ovariectomy-induced bone loss (a murine model of postmenopausal osteoporosis). Ovariectomy fails to induce bone loss, increase bone resorption, and stimulate receptor activator of nuclear factor κB ligand and IL-6 expression in mice lacking p38α in osteoblasts and osteocytes. Finally, TNFα or IL-1, which are osteoclastogenic cytokines overproduced in the bone marrow under estrogen deficiency, can activate p38α signaling in osteoblasts, but those cytokines cannot enhance Rankl and Il6 expressions or increase osteoclast formation in p38a-deficient osteoblast cultures. These findings demonstrate that p38α MAPK signaling in osteoblast lineage cells mediates ovariectomy-induced bone loss by up-regulating receptor activator of nuclear factor κB ligand and IL-6 production.


Bone Resorption/genetics , Estrogens/deficiency , Interleukin-6/metabolism , Mitogen-Activated Protein Kinase 14/genetics , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis, Postmenopausal/genetics , RANK Ligand/metabolism , Absorptiometry, Photon , Animals , Blotting, Western , Bone Resorption/metabolism , Cells, Cultured , Coculture Techniques , Collagen Type I/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Humans , Interleukin-1/metabolism , Mice , Osteocytes/metabolism , Osteoporosis, Postmenopausal/metabolism , Ovariectomy , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , X-Ray Microtomography
9.
Bonekey Rep ; 4: 711, 2015.
Article En | MEDLINE | ID: mdl-26131361

The p38 mitogen-activated protein kinase (MAPK) signaling pathway can be activated in response to a wide range of extracellular signals. As a consequence, it can generate many different biological effects that depend on the stimulus and on the activated cell type. Therefore, this pathway has been found to regulate many aspects of tissue development and homeostasis. Recent work with the aid of genetically modified mice has highlighted the physiological functions of this pathway in skeletogenesis and postnatal bone maintenance. In this review, emphasis is given to the roles of the p38 MAPK pathway in chondrocyte, osteoblast and osteoclast biology. In particular, we describe the molecular mechanisms of p38 MAPK activation and downstream targets. The requirement of this pathway in physiological bone development and homeostasis is demonstrated by the ability of p38 MAPK to regulate master transcription factors controlling geneses and functions of chondrocytes, osteoblasts and osteoclasts.

10.
Mol Cell Endocrinol ; 382(1): 120-130, 2014 Jan 25.
Article En | MEDLINE | ID: mdl-24060635

Bone morphogenic proteins (BMPs) promote mesenchymal stem cell (MSC) osteogenic differentiation, whereas platelet derived growth factor (PDGF) and fibroblast growth factor (FGF) activate their proliferation through receptors tyrosine kinase (RTK). The effects of PDGF or FGF receptor signaling pathway on BMP2-induced osteoblastic differentiation was investigated in human MSC (HMSC). Inhibition of PDGF or/and FGF receptors enhanced BMP2-induced alkaline phosphatase (ALP) activity, expression of Osterix, ALP and Bone sialoprotein, and matrix calcification. These effects were associated with increased Smad-1 activity, indicating that mitogenic factors interfere with Smad signaling in HMSC differentiation. RTK activate MAPK and inhibit GSK3 through the PI3K/Akt pathway. Biochemical analysis indicated that MAPK JNK and GSK3 especially are potential signaling molecules regulating BMP-induced osteoblastic HMSC differentiation. These observations highlight that the osteogenic effects of BMP2 are modulated by mitogenic factors acting through RTK.


Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Glycogen Synthase Kinase 3/metabolism , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Receptor Cross-Talk , Receptor Protein-Tyrosine Kinases/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism , Models, Biological , Osteoblasts/drug effects , Osteoblasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor Cross-Talk/drug effects , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Young Adult
11.
J Bone Miner Res ; 28(2): 260-70, 2013 Feb.
Article En | MEDLINE | ID: mdl-22927028

Previous studies have shown that Wnt3a enhances the proliferation and inhibits the osteogenic differentiation of human mesenchymal stem cells (hMSCs). In this study, we investigated the signaling pathways involved in Wnt3a-induced osteoblastic cell proliferation. Experiments with DKK1, a natural antagonist of Lrp5/6, indicated that Wnt/ß-catenin did not play a major role in Wnt3a-induced osteoblastic cell proliferation. The use of selective inhibitors of known mitogenic pathways implicates Src family kinases (SFKs) and a protein kinase C (PKC) in this cellular response. Time-dependent analysis of signaling molecules activated by Wnt3a in MC3T3-E1 cells revealed parallel activation of the canonical pathway and of several tyrosine kinases, including SFKs and PDGF receptors (PDGF-Rs). Functional analysis with specific inhibitors suggested a major role of PDGF-Rs in mediating Wnt3a-induced cell proliferation. Further investigation with an si-RNA approach confirmed a predominant role of this receptor in this cellular response. The use of soluble decoy PDGF-Rs that can sequester extracellular PDGFs excluding that part of the increased PDGF receptor phosphorylation by Wnt3a was the result of autocrine production of PDGFs. A selective SFK inhibitor blunted the enhanced PDGF-R phosphorylation and cell proliferation induced by Wnt3a. Studies of initial events involved in the regulation of this pathway suggest a role of dishevelled. In conclusion, data presented in this study indicate that cell proliferation induced by Wnt3a in osteoblastic cells is mediated by a dishevelled-dependent and ß-catenin-independent pathway, which involves the transactivation of PDGF receptors.


Osteoblasts/cytology , Osteoblasts/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Transcriptional Activation/drug effects , Wnt3A Protein/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Dishevelled Proteins , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Phosphoproteins/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/genetics , beta Catenin/metabolism
12.
Biochem Biophys Res Commun ; 427(4): 737-42, 2012 Nov 02.
Article En | MEDLINE | ID: mdl-23044416

Understanding the interactions between growth factors and bone morphogenic proteins (BMPs) signaling remains a crucial issue to optimize the use of human mesenchymal stem cells (HMSCs) and BMPs in therapeutic perspectives and bone tissue engineering. BMPs are potent inducers of osteoblastic differentiation. They exert their actions via BMP receptors (BMPR), including BMPR1A, BMPR1B and BMPR2. Fibroblast growth factor 2 (FGF2) is expressed by cells of the osteoblastic lineage, increases their proliferation and is secreted during the healing process of fractures or in surgery bone sites. We hypothesized that FGF2 might influence HMSC osteoblastic differentiation by modulating expressions of BMPs and their receptors. BMP2, BMP4, BMPR1A and mainly BMPR1B expressions were up-regulated during this differentiation. FGF2 inhibited HMSCs osteoblastic differentiation and the up-regulation of BMPs and BMPR. This effect was prevented by inhibiting the ERK or JNK mitogen-activated protein kinases which are known to be activated by FGF2. These data provide a mechanism explaining the inhibitory effect of FGF2 on osteoblastic differentiation of HMSCs. These crosstalks between growth and osteogenic factors should be considered in the use of recombinant BMPs in therapeutic purpose of fracture repair or skeletal bioengineering.


Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Proteins/antagonists & inhibitors , Cell Differentiation/physiology , Fibroblast Growth Factor 2/physiology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Bone Morphogenetic Protein Receptors/biosynthesis , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Cell Differentiation/drug effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Osteoblasts/drug effects , Up-Regulation
13.
Cell Mol Life Sci ; 69(18): 3115-25, 2012 Sep.
Article En | MEDLINE | ID: mdl-22527716

Bone continuously remodels throughout life by coordinated actions of osteoclasts and osteoblasts. Abnormalities in either osteoclast or osteoblast functions lead to bone disorders. The p38 MAPK pathway has been shown to be essential in controlling osteoblast differentiation and skeletogenesis. Although p38α is the most abundant p38 member in osteoblasts, its specific individual contribution in regulating postnatal osteoblast activity and bone metabolism is unknown. To elucidate the specific role of p38α in regulating osteoblast function and bone homeostasis, we generated mice lacking p38α in differentiated osteoblasts. Osteoblast-specific p38a knockout mice were of normal weight and size. Despite non-significant bone alterations until 5 weeks of age, mutant mice demonstrated significant and progressive decrease in bone mineral density from that age. Adult mice deficient in p38a in osteoblasts displayed a striking reduction in cancellous bone volume at both axial and appendicular skeletal sites. At 6 months of age, trabecular bone volume was reduced by 62% in those mice. Mutant mice also exhibited progressive decrease in cortical thickness of long bones. These abnormalities correlated with decreased endocortical and trabecular bone formation rate and reduced expressions of type 1 collagen, alkaline phosphatase, osteopontin and osteocalcin whereas bone resorption and osteoclasts remained unaffected. Finally, osteoblasts lacking p38α showed impaired marker gene expressions and defective mineralization in vitro. These findings indicate that p38α is an essential positive regulator of osteoblast function and postnatal bone formation in vivo.


Bone Development/physiology , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Osteoblasts/physiology , Osteogenesis/physiology , Age Factors , Alkaline Phosphatase/genetics , Animals , Animals, Newborn , Bone Density/genetics , Bone Resorption/genetics , Collagen Type I/genetics , Female , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Osteocalcin/genetics , Osteopontin/genetics
14.
Cell Physiol Biochem ; 27(3-4): 243-50, 2011.
Article En | MEDLINE | ID: mdl-21471713

BACKGROUND/AIMS: Strontium ranelate (SrRan) is an anti-osteoporotic treatment that reduces the risk of vertebral and hip fractures. Recent in vitro studies suggest that the effect of strontium ranelate on osteoblastic cell growth likely involves two processes including activation of the calcium sensing receptor (CaSR) and a yet undefined mechanism. In the present study, we investigated the CaSR-independent molecular mechanism by which SrRan stimulates osteoblast growth. METHODS: MC3T3-E1 and primary osteoblastic cells, specific inhibitors of receptor tyrosine kinases (RTK) and western blot analysis were used to characterize the CaSR-independent mechanism in osteoblastic cells. RESULTS: A selective inhibitor of FGF receptor but not other RTK inhibitors markedly blunted cell growth induced by SrRan in osteoblastic cells. Associated with this observation, SrRan induced rapid activation of FGFR signaling pathways such as PLCγ, FRS2, Akt, ERK1,2 and p38. FGFR-dependent stimulation of osteogenic cell growth was also observed with other cations but not with neomycin, a selective CaSR agonist. Also, in cultured conditions used in this study, MC3T3-E1 cells and primary osteoblasts did not express the CaSR. CONCLUSION: data presented in this study suggest that activation of FGFRs is a new potential mechanism by which strontium can stimulate osteoblastic cell growth. Activation of FGFR-dependent cell growth is also observed in response to other cations suggesting that activation of FGF receptors is a new cation sensing mechanism in osteoblasts.


Bone Density Conservation Agents/pharmacology , Organometallic Compounds/pharmacology , Osteoblasts/drug effects , Receptors, Fibroblast Growth Factor/metabolism , Thiophenes/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
J Proteomics ; 74(7): 1123-34, 2011 Jun 10.
Article En | MEDLINE | ID: mdl-21515422

Matrix vesicles (MVs), released by budding from apical microvilli of osteoblasts during bone formation and development, are involved in the initiation of mineralization by promoting the formation of hydroxyapatite in their lumen. To gain additional insights into MV biogenesis and functions, MVs and apical microvilli were co-isolated from mineralizing osteoblast-like Saos-2 cells and their proteomes were characterized using LC-ESI-MS/MS and compared. In total, 282 MV and 451 microvillar proteins were identified. Of those, 262 were common in both preparations, confirming that MVs originate from apical microvilli. The occurrence of vesicular trafficking molecules (e.g. Rab proteins) and of the on-site protein synthetic machinery suggests that cell polarization and apical targeting are required for the incorporation of specific lipids and proteins at the site of MV formation. MV release from microvilli may be driven by actions of actin-severing proteins (gelsolin, cofilin 1) and contractile motor proteins (myosins). In addition to the already known proteins involved in MV-mediated mineralization, new MV residents were detected, such as inorganic pyrophosphatase 1, SLC4A7 sodium bicarbonate cotransporter or sphingomyelin phosphodiesterase 3, providing additional insights into MV functions.


Cytoplasmic Vesicles/physiology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/physiology , Microvilli/physiology , Osteoblasts/physiology , Bone Matrix/physiology , Calcification, Physiologic/physiology , Cell Differentiation , Humans , Osteogenesis/physiology , Proteome/analysis , Spectrometry, Mass, Electrospray Ionization , Tumor Cells, Cultured
16.
J Cell Biochem ; 106(1): 127-38, 2009 Jan 01.
Article En | MEDLINE | ID: mdl-19009559

In bone, mineralization is tightly regulated by osteoblasts and hypertrophic chondrocytes which release matrix vesicles (MVs) and control extracellular ionic conditions and matrix composition. MVs are the initial sites of hydroxyapatite (HA) mineral formation. Despite growing knowledge about their morphology and function, their biogenesis is not well understood. The purpose of this work was to determine the source of MVs in osteoblast lineage, Saos-2 cells, and to check whether MVs originated from microvilli. Microvilli were isolated from the apical plasma membrane of Saos-2 cells. Their morphology, structure, and function were compared with those of MVs. The role of actin network in MV release was investigated by using microfilament perturbing drugs. When examined by electron microscopy MVs and microvillar vesicles were found to exhibit similar morphology with trilaminar membranes and diameters in the same range. Both types of vesicles were able to induce HA formation. Their electrophoretic profiles displayed analogous enrichment in alkaline phosphatase, Na(+)/K(+) ATPase, and annexins A2 and A6. MVs and microvillar vesicles exhibited almost the same lipid composition with a higher content of cholesterol, sphingomyelin, and phosphatidylserine as compared to plasma membrane. Finally, cytochalasin D, which inhibits actin polymerization, was found to stimulate release of MVs. Our findings were consistent with the hypothesis that MVs originated from cell microvilli and that actin filament disassembly was involved in their biogenesis.


Extracellular Matrix/metabolism , Osteoblasts/metabolism , Calcification, Physiologic , Cell Line, Tumor , Cell Lineage , Chondrocytes/cytology , Chondrocytes/metabolism , Humans , Microscopy, Electron , Microvilli/metabolism , Osteosarcoma/metabolism
17.
Proteomics ; 8(1): 192-205, 2008 Jan.
Article En | MEDLINE | ID: mdl-18095356

Matrix vesicles (MVs) are extracellular organelles that initiate mineral formation, accumulating inorganic phosphate (P(i)) and calcium leading to the formation of hydroxyapatite (HA) crystals, the main mineral component of bones. MVs are produced during bone formation, as well as during the endochondral calcification of cartilage. MVs are released into the extracellular matrix from osseous cells such as osteoblasts and hypertrophic chondrocytes. In this report, using 1-D SDS-PAGE, in-gel tryptic digestion and an LC-MS-MS/MS protein identification protocol, we characterized the proteome of MVs isolated from chicken embryo (Gallus gallus) bones and cartilage. We identified 126 gene products, including proteins related to the extracellular matrix and ion transport, as well as enzymes, cytoskeletal, and regulatory proteins. Among the proteins recognized for the first time in MVs were aquaporin 1, annexin A1 (AnxA1), AnxA11, glycoprotein HT7, G(i) protein alpha2, and scavenger receptor type B. The pathways for targeting the identified proteins into MVs and their particular functions in the biomineralization process are discussed. Obtaining a knowledge of the functions and roles of these proteins during embryonic mineralization is a prerequisite for the overall understanding of the initial mineral formation mechanisms.


Bone Matrix/chemistry , Chickens/metabolism , Cytoplasmic Vesicles/chemistry , Femur/chemistry , Proteome , Animals , Bone Matrix/embryology , Bone Matrix/ultrastructure , Calcification, Physiologic , Chick Embryo , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix/chemistry , Extracellular Matrix/ultrastructure , Femur/embryology , Femur/ultrastructure
18.
Postepy Biochem ; 53(2): 159-63, 2007.
Article Pl | MEDLINE | ID: mdl-17969876

Annexins are necessary for mineralization process. They seem to play a major role in regulation of cells competent in mineralization as well as in direct formation of mineral phase in the extracellular matrix. Their ability to accommodate to different functions in different cellular compartments is associated with their property to bind to biological membranes in a lipid- and Ca2+-dependent and independent manners. The aim of this review is to describe potential functions of the annexin family of proteins in a mineralization process with special emphasis to structure-function relationships of annexins.


Annexins/physiology , Bone and Bones/physiology , Calcification, Physiologic/physiology , Chondrocytes/physiology , Membrane Lipids/physiology , Membrane Proteins/chemistry , Alkaline Phosphatase/metabolism , Animals , Bone Matrix/metabolism , Calcium/metabolism , Extracellular Matrix/metabolism , Growth Plate/metabolism , Humans , Membrane Proteins/isolation & purification , Microscopy, Electron , Phosphorus/metabolism
19.
Plant Cell Physiol ; 48(6): 792-803, 2007 Jun.
Article En | MEDLINE | ID: mdl-17452342

Plant annexins, Ca(2+)- and membrane-binding proteins, are probably implicated in the cellular response to stress resulting from acidification of cytosol. To understand how annexins can contribute to cellular ion homeostasis, we investigated the pH-induced changes in the structure and function of recombinant annexin AnnAt1 from Arabidopsis thaliana. The decrease of pH from 7.0 to 5.8 reduced the time of the formation of ion channels by AnnAt1 in artificial lipid membranes from 3.5 h to 15-20 min and increased their unitary conductance from 32 to 63 pS. These changes were accompanied by an increase in AnnAt1 hydrophobicity as revealed by hydrophobicity predictions, by an increase in fluorescence of 2-(p-toluidino)naphthalene-6-sulfonic acid (TNS) bound to AnnAt1 and fluorescence resonance energy transfer from AnnAt1 tryptophan residues to TNS. Concomitant lipid partition of AnnAt1 at acidic pH resulted in its partial protection from proteolytic digestion. Secondary structures of AnnAt1 determined by circular dichroism and infrared spectroscopy were also affected by lowering the pH from 7.2 to 5.2. These changes were characterized by an increase in beta-sheet content at the expense of alpha-helical structures, and were accompanied by reversible formation of AnnAt1 oligomers as probed by ultracentrifugation in a sucrose gradient. A further decrease of pH from 5.2 to 4.5 or lower led to the formation of irreversible aggregates and loss of AnnAt1 ionic conductance. Our findings suggest that AnnAt1 can sense changes of the pH milieu over the pH range from 7 to 5 and respond by changes in ion channel conductance, hydrophobicity, secondary structure of the protein and formation of oligomers. Further acidification irreversibly inactivated AnnAt1. We suggest that the pH-sensitive ion channel activity of AnnAt1 may play a role in intracellular ion homeostasis.


Annexins/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Annexins/chemistry , Annexins/genetics , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Environment , Gene Expression Regulation, Plant , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Lipids/chemistry , Membranes, Artificial , Protein Structure, Secondary
20.
J Biol Chem ; 280(44): 37289-96, 2005 Nov 04.
Article En | MEDLINE | ID: mdl-16147995

Inorganic pyrophosphate is a potent inhibitor of bone mineralization by preventing the seeding of calcium-phosphate complexes. Plasma cell membrane glycoprotein-1 and tissue nonspecific alkaline phosphatase were reported to be antagonistic regulators of mineralization toward inorganic pyrophosphate formation (by plasma cell membrane glycoprotein-1) and degradation (by tissue nonspecific alkaline phosphatase) under physiological conditions. In addition, they possess broad overlapping enzymatic functions. Therefore, we examined the roles of tissue nonspecific alkaline phosphatase within matrix vesicles isolated from femurs of 17-day-old chick embryos, under conditions where these both antagonistic and overlapping functions could be evidenced. Addition of 25 microM ATP significantly increased duration of mineralization process mediated by matrix vesicles, while supplementation of mineralization medium with levamisole, an alkaline phosphatase inhibitor, reduces the ATP-induced retardation of mineral formation. Phosphodiesterase activity of tissue nonspecific alkaline phosphatase for bis-p-nitrophenyl phosphate was confirmed, the rate of this phosphodiesterase activity is in the same range as that of phosphomonoesterase activity for p-nitrophenyl phosphate under physiological pH. In addition, tissue nonspecific alkaline phosphatase at pH 7.4 can hydrolyze ADPR. On the basis of these observations, it can be concluded that tissue nonspecific alkaline phosphatase, acting as a phosphomonoesterase, could hydrolyze free phosphate esters such as pyrophosphate and ATP, while as phosphodiesterase could contribute, together with plasma cell membrane glycoprotein-1, in the production of pyrophosphate from ATP.


Adenosine Triphosphate/pharmacology , Alkaline Phosphatase/metabolism , Bone Matrix/metabolism , Calcification, Physiologic/physiology , Phosphates/metabolism , Phosphoric Diester Hydrolases/metabolism , Animals , Bone Matrix/cytology , Bone Matrix/ultrastructure , Carrier Proteins/metabolism , Chick Embryo , Nitrophenols/metabolism , Organophosphorus Compounds/metabolism , Pyrophosphatases/metabolism
...