Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters










Publication year range
1.
Curr Top Dev Biol ; 133: 195-234, 2019.
Article in English | MEDLINE | ID: mdl-30902253

ABSTRACT

Fibroblast growth factors (FGFs) and their receptors (FGFRs) are expressed throughout all stages of skeletal development. In the limb bud and in cranial mesenchyme, FGF signaling is important for formation of mesenchymal condensations that give rise to bone. Once skeletal elements are initiated and patterned, FGFs regulate both endochondral and intramembranous ossification programs. In this chapter, we review functions of the FGF signaling pathway during these critical stages of skeletogenesis, and explore skeletal malformations in humans that are caused by mutations in FGF signaling molecules.


Subject(s)
Fibroblast Growth Factors/metabolism , Osteogenesis , Animals , Bone Diseases/genetics , Chondrocytes/metabolism , Humans , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
3.
J Bone Miner Res ; 33(2): 199-210, 2018 02.
Article in English | MEDLINE | ID: mdl-29206311

ABSTRACT

During the last three decades, important progress in bone cell biology and in human and mouse genetics led to major advances in our understanding of the life and functions of cells of the osteoblast lineage. Previously unrecognized sources of osteogenic cells have been identified. Novel cellular and molecular mechanisms controlling osteoblast differentiation and senescence have been determined. New mechanisms of communications between osteogenic cells, osteocytes, osteoclasts, and chondrocytes, as well as novel links between osteogenic cells and blood vessels have been identified. Additionally, cells of the osteoblast lineage were shown to be important components of the hematopoietic niche and to be implicated in hematologic dysfunctions and malignancy. Lastly, unexpected interactions were found between osteogenic cells and several soft tissues, including the central nervous system, gut, muscle, fat, and testis through the release of paracrine factors, making osteogenic cells multifunctional regulatory cells, in addition to their bone-making function. These discoveries considerably enlarged our vision of the life and functions of osteogenic cells, which may lead to the development of novel therapeutics with immediate applications in bone disorders. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Osteoblasts/cytology , Osteogenesis , Animals , Bone and Bones/physiology , Cell Communication , Humans , Signal Transduction
4.
Cell Death Dis ; 8(6): e2902, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28661485

ABSTRACT

Stimulating bone formation is an important challenge for bone anabolism in osteoporotic patients or to repair bone defects. The osteogenic properties of matrix glycosaminoglycans (GAGs) have been explored; however, the functions of GAGs at the surface of bone-forming cells are less documented. Syndecan-2 is a membrane heparan sulfate proteoglycan that is associated with osteoblastic differentiation. We used a transgenic mouse model with high syndecan-2 expression in osteoblasts to enrich the bone surface with cellular GAGs. Bone mass was increased in these transgenic mice. Syndecan-2 overexpression reduced the expression of receptor activator of NF-kB ligand (RANKL) in bone marrow cells and strongly inhibited bone resorption. Osteoblast activity was not modified in the transgenic mice, but bone formation was decreased in 4-month-old transgenic mice because of reduced osteoblast number. Increased proteoglycan expression at the bone surface resulted in decreased osteoblastic and osteoclastic precursors in bone marrow. Indeed, syndecan-2 overexpression increased apoptosis of mesenchymal precursors within the bone marrow. However, syndecan-2 specifically promoted the vasculature characterized by high expression of CD31 and Endomucin in 6-week-old transgenic mice, but this was reduced in 12-week-old transgenic mice. Finally, syndecan-2 functions as an inhibitor of Wnt-ß-catenin-T-cell factor signaling pathway, activating glycogen synthase kinase 3 and then decreasing the Wnt-dependent production of Wnt ligands and R-spondin. In conclusion, our results show that GAG supply may improve osteogenesis, but also interfere with the crosstalk between the bone surface and marrow cells, altering the supporting function of osteoblasts.


Subject(s)
Bone Remodeling/drug effects , Glycosaminoglycans/administration & dosage , Heparitin Sulfate/administration & dosage , Syndecan-2/genetics , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Remodeling/genetics , Bone Resorption/drug therapy , Bone Resorption/genetics , Bone Resorption/pathology , Cell Differentiation/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Mice , Mice, Transgenic , Osteoblasts/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics , RANK Ligand , Wnt Signaling Pathway/drug effects
5.
Hum Mol Genet ; 25(7): 1281-93, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26769674

ABSTRACT

Patients with cystic fibrosis (CF) display low bone mass and alterations in bone formation. Mice carrying the F508del genetic mutation in the cystic fibrosis conductance regulator (Cftr) gene display reduced bone formation and decreased bone mass. However, the underlying molecular mechanisms leading to these skeletal defects are unknown, which precludes the development of an efficient anti-osteoporotic therapeutic strategy. Here we report a key role for the intermediate filament protein keratin 8 (Krt8), in the osteoblast dysfunctions in F508del-Cftr mice. We found that murine and human osteoblasts express Cftr and Krt8 at low levels. Genetic studies showed that Krt8 deletion (Krt8(-/-)) in F508del-Cftr mice increased the levels of circulating markers of bone formation, corrected the expression of osteoblast phenotypic genes, promoted trabecular bone formation and improved bone mass and microarchitecture. Mechanistically, Krt8 deletion in F508del-Cftr mice corrected overactive NF-κB signaling and decreased Wnt-ß-catenin signaling induced by the F508del-Cftr mutation in osteoblasts. In vitro, treatment with compound 407, which specifically disrupts the Krt8-F508del-Cftr interaction in epithelial cells, corrected the abnormal NF-κB and Wnt-ß-catenin signaling and the altered phenotypic gene expression in F508del-Cftr osteoblasts. In vivo, short-term treatment with 407 corrected the altered Wnt-ß-catenin signaling and bone formation in F508del-Cftr mice. Collectively, the results show that genetic or pharmacologic targeting of Krt8 leads to correction of osteoblast dysfunctions, altered bone formation and osteopenia in F508del-Cftr mice, providing a therapeutic strategy targeting the Krt8-F508del-CFTR interaction to correct the abnormal bone formation and bone loss in cystic fibrosis.


Subject(s)
Bone Diseases, Metabolic/etiology , Cystic Fibrosis/complications , Gene Deletion , Keratin-8/genetics , Osteogenesis , Animals , Bone Diseases, Metabolic/metabolism , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Disease Models, Animal , Female , Humans , Male , Mice , NF-kappa B , Osteoblasts/metabolism , Signal Transduction , Young Adult , beta Catenin
7.
PLoS One ; 10(10): e0140848, 2015.
Article in English | MEDLINE | ID: mdl-26509533

ABSTRACT

The sympathetic nervous system controls bone remodeling by regulating bone formation and resorption. How nerves and bone cells influence each other remains elusive. Here we modulated the content or activity of the neuropeptide Vasoactive Intestinal Peptide to investigate nerve-bone cell interplays in the mandible periosteum by assessing factors involved in nerve and bone behaviors. Young adult rats were chemically sympathectomized or treated with Vasoactive Intestinal Peptide or Vasoactive Intestinal Peptide10-28, a receptor antagonist. Sympathectomy depleted the osteogenic layer of the periosteum in neurotrophic proNerve Growth Factor and neurorepulsive semaphorin3a; sensory Calcitonin-Gene Related Peptide-positive fibers invaded this layer physiologically devoid of sensory fibers. In the periosteum non-osteogenic layer, sympathectomy activated mast cells to release mature Nerve Growth Factor while Calcitonin-Gene Related Peptide-positive fibers increased. Vasoactive Intestinal Peptide treatment reversed sympathectomy effects. Treating intact animals with Vasoactive Intestinal Peptide increased proNerve Growth Factor expression and stabilized mast cells. Vasoactive Intestinal Peptide10-28 treatment mimicked sympathectomy effects. Our data suggest that sympathetic Vasoactive Intestinal Peptide modulate the interactions between nervous fibers and bone cells by tuning expressions by osteogenic cells of factors responsible for mandible periosteum maintenance while osteogenic cells keep nervous fibers at a distance from the bone surface.


Subject(s)
Mandible/innervation , Nerve Fibers/metabolism , Osteoblasts/metabolism , Periosteum/metabolism , Animals , Male , Mandible/drug effects , Nerve Fibers/drug effects , Nerve Growth Factors/metabolism , Osteoblasts/drug effects , Periosteum/cytology , Periosteum/drug effects , Rats , Rats, Wistar , Vasoactive Intestinal Peptide/pharmacology
8.
Genes Dev ; 29(14): 1463-86, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26220993

ABSTRACT

Fibroblast growth factor (FGF) signaling pathways are essential regulators of vertebrate skeletal development. FGF signaling regulates development of the limb bud and formation of the mesenchymal condensation and has key roles in regulating chondrogenesis, osteogenesis, and bone and mineral homeostasis. This review updates our review on FGFs in skeletal development published in Genes & Development in 2002, examines progress made on understanding the functions of the FGF signaling pathway during critical stages of skeletogenesis, and explores the mechanisms by which mutations in FGF signaling molecules cause skeletal malformations in humans. Links between FGF signaling pathways and other interacting pathways that are critical for skeletal development and could be exploited to treat genetic diseases and repair bone are also explored.


Subject(s)
Bone Diseases/genetics , Bone and Bones/embryology , Bone and Bones/physiopathology , Fibroblast Growth Factors/physiology , Signal Transduction , Animals , Bone Diseases/therapy , Bone Regeneration/genetics , Chondrogenesis , Fibroblast Growth Factors/genetics , Humans , Minerals/metabolism , Mutation , Osteogenesis
9.
J Biol Chem ; 290(29): 18009-18017, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26060255

ABSTRACT

The prevalent human ΔF508 mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) is associated with reduced bone formation and bone loss in mice. The molecular mechanisms by which the ΔF508-CFTR mutation causes alterations in bone formation are poorly known. In this study, we analyzed the osteoblast phenotype in ΔF508-CFTR mice and characterized the signaling mechanisms underlying this phenotype. Ex vivo studies showed that the ΔF508-CFTR mutation negatively impacted the differentiation of bone marrow stromal cells into osteoblasts and the activity of osteoblasts, demonstrating that the ΔF508-CFTR mutation alters both osteoblast differentiation and function. Treatment with a CFTR corrector rescued the abnormal collagen gene expression in ΔF508-CFTR osteoblasts. Mechanistic analysis revealed that NF-κB signaling and transcriptional activity were increased in mutant osteoblasts. Functional studies showed that the activation of NF-κB transcriptional activity in mutant osteoblasts resulted in increased ß-catenin phosphorylation, reduced osteoblast ß-catenin expression, and altered expression of Wnt/ß-catenin target genes. Pharmacological inhibition of NF-κB activity or activation of canonical Wnt signaling rescued Wnt target gene expression and corrected osteoblast differentiation and function in bone marrow stromal cells and osteoblasts from ΔF508-CFTR mice. Overall, the results show that the ΔF508-CFTR mutation impairs osteoblast differentiation and function as a result of overactive NF-κB and reduced Wnt/ß-catenin signaling. Moreover, the data indicate that pharmacological inhibition of NF-κB or activation of Wnt/ß-catenin signaling can rescue the abnormal osteoblast differentiation and function induced by the prevalent ΔF508-CFTR mutation, suggesting novel therapeutic strategies to correct the osteoblast dysfunctions in cystic fibrosis.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/immunology , NF-kappa B/immunology , Osteoblasts/cytology , Wnt Signaling Pathway , Animals , Cell Differentiation , Cells, Cultured , Male , Mice , Osteoblasts/immunology , Osteoblasts/pathology , beta Catenin/immunology
10.
Bonekey Rep ; 4: 666, 2015.
Article in English | MEDLINE | ID: mdl-25848534

ABSTRACT

Syndecans 1-4 are a family of transmembrane proteins composed of a core protein and glycosaminoglycan chains. Although the four syndecans have common functions, they appear to be connected to different signaling pathways, and their expression occurs in a cell- and development-specific pattern. In contrast to other syndecans, syndecan-2 expression increases during osteoblast differentiation. Mechanistically, syndecan-2 exerts multiple functions in cells of the osteoblast lineage as it serves as a co-receptor for fibroblast growth factors and Wnt proteins and controls cell adhesion, proliferation, differentiation and apoptosis. Recent studies indicate that syndecan-2 also contributes to osteosarcoma cell response to cytotoxic agents through interactions with Wnt/ß-catenin signaling. Here we summarize our current understanding of the role of syndecan-2 in the control of osteoblast biology and pathology and discuss how syndecan-2 acts as a modulator of the bone cell microenvironment.

11.
J Biol Chem ; 290(11): 6903-12, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25631051

ABSTRACT

The α5ß1 integrin is a key fibronectin (FN) receptor that binds to RGD-containing peptides to mediate cell adhesion. We previously reported that α5ß1 integrin promotes osteogenic differentiation in mesenchymal skeletal cells (MSCs), but the underlying mechanisms are not fully understood. In this study, we determined the signaling mechanisms induced by α5ß1 integrin interaction with its high-affinity ligand CRRETAWAC in murine and human MSCs and in vivo. We show that cyclized CRRETAWAC fully displaced MSC adhesion to FN, whereas related peptides lacking the full RRET sequence produced a partial displacement, indicating that RRET acts as an RGD-like sequence that is required to antagonize FN-mediated cell adhesion. However, all peptides increased focal adhesion kinase phosphorylation, OSE2 transcriptional activity, osteoblast gene expression, and matrix mineralization in MSCs, indicating that peptide-induced α5ß1 integrin priming can promote osteogenic differentiation independently of the RRET sequence. Biochemical analyses showed that peptide-induced α5ß1 integrin priming transiently increased PI3K/Akt phosphorylation and promoted Wnt/ß-catenin transcriptional activity independently of RRET. Consistently, pharmacological inhibition of PI3K activity reduced osteoblast differentiation and abolished Wnt regulatory gene expression induced by α5ß1 integrin priming. In vivo, systemic delivery of cyclized GACRETAWACGA linked to (DSS)6 to allow delivery to bone-forming sites for 6 weeks increased serum osteocalcin levels and improved long bone mass and microarchitecture in SAMP-6 senescent osteopenic mice. The results support a mechanism whereby α5ß1 integrin priming by high-affinity ligands integrates Wnt/ß-catenin signaling to promote osteoblast differentiation independently of cell adhesion, which could be used to improve bone mass and microarchitecture in the aging skeleton.


Subject(s)
Bone Diseases, Metabolic/drug therapy , Integrin alpha5beta1/metabolism , Mesenchymal Stem Cells/cytology , Oligopeptides/therapeutic use , Osteoblasts/drug effects , Wnt Signaling Pathway/drug effects , Amino Acid Sequence , Animals , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Differentiation/drug effects , Cell Line , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Oligopeptides/chemistry , Oligopeptides/pharmacology , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , Phosphatidylinositol 3-Kinases/metabolism , beta Catenin/metabolism
12.
Cell Mol Life Sci ; 72(7): 1347-61, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25487608

ABSTRACT

Several metabolic, genetic and oncogenic bone diseases are characterized by defective or excessive bone formation. These abnormalities are caused by dysfunctions in the commitment, differentiation or survival of cells of the osteoblast lineage. During the recent years, significant advances have been made in our understanding of the cellular and molecular mechanisms underlying the osteoblast dysfunctions in osteoporosis, skeletal dysplasias and primary bone tumors. This led to suggest novel therapeutic approaches to correct these abnormalities such as the modulation of WNT signaling, the pharmacological modulation of proteasome-mediated protein degradation, the induction of osteoprogenitor cell differentiation, the repression of cancer cell proliferation and the manipulation of epigenetic mechanisms. This article reviews our current understanding of the major cellular and molecular mechanisms inducing osteoblastic cell abnormalities in age-related bone loss, genetic skeletal dysplasias and primary bone tumors, and discusses emerging therapeutic strategies to counteract the osteoblast abnormalities in these disorders of bone formation.


Subject(s)
Bone Diseases, Developmental/physiopathology , Bone Neoplasms/physiopathology , Osteoblasts/physiology , Osteoporosis/physiopathology , Signal Transduction , Apoptosis , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Differentiation , Humans , Models, Biological , Osteoblasts/metabolism , Osteoporosis/genetics , Osteoporosis/metabolism
13.
J Endocrinol ; 223(1): 25-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25056116

ABSTRACT

Intermittent administration of parathyroid hormone (PTH) 1-34 at a standard dose has been shown to induce anabolic effects in bone. However, whether low-dose PTH promotes bone formation during senescence is unknown. To address this issue, we determined the effects of low-dose PTH and analysed the underlying mechanisms in prematurely senescent mice that display osteopenia. Treatment of 9-week-old Samp6 mice for 6 weeks with PTH at a standard dose (100 µg/kg per day) increased vertebral and femoral bone mass and improved bone microarchitecture as a result of increased bone-forming surfaces and mineral apposition rate (MAR). At a tenfold lower dose (10 µg/kg per day), PTH increased axial bone volume and trabecular thickness, as detected by bone histomorphometry but not by micro-computed tomography analysis. This anabolic effect resulted from increased osteoblast activity, as reflected by increased serum N-terminal propeptide of type 1 procollagen (P1NP) levels and MAR, with unchanged bone-forming surface or osteoblast surface. Mechanistically, low-dose PTH increased the expression of osteoblast markers in bone marrow stromal cells and mature osteoblasts, which was associated with increased expression of the Wnt effector Wisp1. Moreover, low-dose PTH decreased the expression of the Mef2c transcription factor, resulting in decreased Sost expression in osteoblasts/osteocytes. These results indicate that PTH at a low dose is effective at promoting bone formation and increased bone volume in senescent osteopenic mice through increased osteoblast activity and modulation of specific Wnt effectors, which raises the potential therapeutic use of intermittent PTH at low dose to increase bone forming activity and bone mass in skeletal senescence.


Subject(s)
Aging , Bone Diseases, Metabolic/genetics , Glycoproteins/genetics , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Collagen Type I/blood , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Dose-Response Relationship, Drug , Gene Expression/drug effects , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , X-Ray Microtomography
14.
Trends Endocrinol Metab ; 25(11): 567-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25034128

ABSTRACT

Cell-cell and cell-matrix interactions mediated by cell adhesion molecules are important mechanisms controlling cell fate and function. Here, we review recent advances in the implication of the cell adhesion molecules integrins and cadherins in the control of osteoblastogenesis and bone formation. We discuss emerging evidence indicating that signaling pathways mediated by integrins and cadherins and their crosstalk with the Wnt/ß-catenin signaling pathway regulate osteogenic differentiation and mechanotransduction. We also offer a comprehensive view of the mechanisms by which some integrins and cadherins control the differentiation of cells of the osteoblast lineage in bone marrow niches. Understanding how specific integrins or cadherins may promote osteogenic cell differentiation, bone formation, and repair may lead to novel therapeutic strategies.


Subject(s)
Bone Resorption/therapy , Bone and Bones/metabolism , Cadherins/metabolism , Integrins/metabolism , Molecular Targeted Therapy , Animals , Cadherins/physiology , Cell Differentiation/physiology , Humans , Integrins/physiology , Molecular Targeted Therapy/methods , Osteoblasts/physiology , Osteogenesis/physiology , Signal Transduction , Wnt Signaling Pathway
15.
J Cell Physiol ; 229(11): 1765-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24664975

ABSTRACT

Age-related bone loss is characterized by reduced osteoblastogenesis and excessive bone marrow adipogenesis. The mechanisms governing bone marrow mesenchymal stromal cell (BMSC) differentiation into adipocytes or osteoblasts during aging are unknown. We show here that overexpressing N-cadherin (Cadh2) in osteoblasts increased BMSC adipocyte differentiation and reduced osteoblast differentiation in young transgenic (Tg) mice whereas this phenotype was fully reversed with aging. The reversed phenotype with age was associated with enhanced Wnt5a and Wnt10b expression in osteoblasts and a concomitant increase in BMSC osteogenic differentiation. Consistent with this mechanism, conditioned media from young wild type osteoblasts inhibited adipogenesis and promoted osteoblast differentiation in BMSC from old Cadh2 Tg mice, and this response was abolished by Wnt5a and Wnt10b silencing. Transplantation of BMSC from old Cadh2 Tg mice into young Tg recipients increased Wnt5a and Wnt10b expression and rescued BMSC osteogenic differentiation. In senescent osteopenic mice, blocking the CADH2-Wnt interaction using an antagonist peptide increased Wnt5a and Wnt10b expression, bone formation, and bone mass. The data indicate that Cadh2/Wnt interaction in osteoblasts regulates BMSC lineage determination, bone formation, and bone mass and suggest a therapeutic target for promoting bone formation in the aging skeleton.


Subject(s)
Aging/metabolism , Bone Marrow Cells/cytology , Cadherins/metabolism , Cell Lineage , Mesenchymal Stem Cells/cytology , Wnt Proteins/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis , Animals , Bone Marrow Cells/metabolism , Bone Resorption/pathology , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Organ Size , Osteoblasts/metabolism , Osteoblasts/pathology , Osteogenesis , Protein Binding , Signal Transduction , Stem Cell Transplantation , Wnt-5a Protein
16.
Am J Pathol ; 184(4): 1132-1141, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24529904

ABSTRACT

In patients with cystic fibrosis (CF), rib and thoracic vertebral fractures can have adverse effects on lung health because the resulting pain and debilitation can impair airway clearance. The F508del mutation in the CF transmembrane conductance regulator (Cftr) gene induces an osteopenic phenotype in humans and mice. N-butyldeoxynojyrimicin (miglustat), an approved drug for treating type 1 Gaucher disease, was found to normalize CFTR-dependent chloride transport in human F508del CFTR lung cells and in nasal mucosa of F508del CF mice. Herein, we investigated whether targeting F508del-CFTR may rescue the skeletal osteopenic phenotype in murine CF. We found that oral administration of low-dose miglustat (120 mg/kg once a day for 28 days) improved bone mass and microarchitecture in the lumbar spine and femur in F508del mice. The increased bone density was associated with an increased bone formation rate and reduced bone resorption. This effect was associated with increased 17ß-estradiol but not with insulin-like growth factor 1 serum levels in miglustat-treated F508del mice. Exposure of primary F508del osteoblasts to miglustat partially restored the deficient CFTR-dependent chloride transport in these bone-forming cells. This study provides evidence that reversal of CFTR-dependent chloride transport in osteoblasts normalizes bone mass and microarchitecture in murine CF. These findings may provide a potential therapeutic strategy to prevent or correct the bone disease in patients with CF.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Bone and Bones/drug effects , Bone and Bones/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/complications , Enzyme Inhibitors/pharmacology , 1-Deoxynojirimycin/pharmacology , Animals , Cells, Cultured , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Male , Mice, Inbred CFTR , Mutation , Osteoblasts/metabolism
17.
J Bone Miner Res ; 29(6): 1311-21, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24496911

ABSTRACT

Age-related bone loss is in large part the consequence of senescence mechanisms that impact bone cell number and function. In recent years, progress has been made in the understanding of the molecular mechanisms underlying bone cell senescence that contributes to the alteration of skeletal integrity during aging. These mechanisms can be classified as intrinsic senescence processes, alterations in endogenous anabolic factors, and changes in local support. Intrinsic senescence mechanisms cause cellular dysfunctions that are not tissue specific and include telomere shortening, accumulation of oxidative damage, impaired DNA repair, and altered epigenetic mechanisms regulating gene transcription. Aging mechanisms that are more relevant to the bone microenvironment include alterations in the expression and signaling of local growth factors and altered intercellular communications. This review provides an integrated overview of the current concepts and interacting mechanisms underlying bone cell senescence during aging and how they could be targeted to reduce the negative impact of senescence in the aging skeleton.


Subject(s)
Bone and Bones/cytology , Cellular Senescence , Aging/physiology , Cellular Microenvironment , Humans
18.
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
19.
Gene ; 521(1): 55-61, 2013 May 25.
Article in English | MEDLINE | ID: mdl-23541807

ABSTRACT

Osteosarcoma is the most common primary bone tumor in children and adults. Despite improved prognosis, resistance to chemotherapy remains responsible for failure of osteosarcoma treatment. The identification of the molecular signals that contribute to the aberrant osteosarcoma cell growth may provide clues to develop new therapeutic strategies for chemoresistant osteosarcoma. Here we show that the expression of ErbB3 is increased in human osteosarcoma cells in vitro. Tissue microarray analysis of tissue cores from osteosarcoma patients further showed that the ErbB3 protein expression is higher in bone tumors compared to normal bone tissue, and is further increased in patients with recurrent disease or soft tissue metastasis. In murine osteosarcoma cells, silencing ErbB3 using shRNA decreased cell replication, cell migration and invasion, indicating that ErbB3 contributes to tumor cell growth and invasiveness. Furthermore, ErbB3 silencing markedly reduced tumor growth in a murine allograft model in vivo. Immunohistochemal analysis showed that the reduced tumor growth induced by ErbB3 silencing in this model resulted from decreased cell osteosarcoma cell proliferation, supporting a role of ErbB3 in bone tumor growth in vivo. Taken together, the results reveal that ErbB3 expression in human osteosarcoma correlates with tumor grade. Furthermore, silencing ErbB3 in a murine osteosarcoma model results in decreased cell growth and invasiveness in vitro, and reduced tumor growth in vivo, which supports the potential therapeutic interest of targeting ErbB3 in osteosarcoma.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/pathology , Osteosarcoma/pathology , Receptor, ErbB-3/genetics , Animals , Base Sequence , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Osteosarcoma/genetics , Osteosarcoma/metabolism , RNA, Small Interfering , Receptor, ErbB-3/metabolism , Xenograft Model Antitumor Assays
20.
Stem Cells ; 31(7): 1340-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23533197

ABSTRACT

The identification of the molecular mechanisms controlling the degradation of regulatory proteins in mesenchymal stromal cells (MSC) may provide clues to promote MSC osteogenic differentiation and bone regeneration. Ubiquitin ligase-dependent degradation of proteins is an important process governing cell fate. In this study, we investigated the role of the E3 ubiquitin ligase c-Cbl in MSC osteoblast differentiation and identified the mechanisms involved in this effect. Using distinct shRNA targeting c-Cbl, we showed that c-Cbl silencing promotes osteoblast differentiation in murine and human MSC, as demonstrated by increased alkaline phosphatase activity, expression of phenotypic osteoblast marker genes (RUNX2, ALP, type 1 collagen), and matrix mineralization in vitro. Coimmunoprecipitation analyses showed that c-Cbl interacts with the transcription factor STAT5, and that STAT5 forms a complex with RUNX2, a master transcription factor controlling osteoblastogenesis. Silencing c-Cbl decreased c-Cbl-mediated STAT5 ubiquitination, increased STAT5 protein level and phosphorylation, and enhanced STAT5 and RUNX2 transcriptional activity. The expression of insulin like growth factor-1 (IGF-1), a target gene of STAT5, was increased by c-Cbl silencing in MSC and in bone marrow stromal cells isolated from c-Cbl deficient mice, suggesting that IGF-1 contributes to osteoblast differentiation induced by c-Cbl silencing in MSC. Consistent with these findings, pharmacological inhibition of STAT5 activity, or neutralization of IGF-1 activity, abrogated the positive effect of c-Cbl knockdown on MSC osteogenic differentiation. Taken together, the data provide a novel functional mechanism by which the ubiquitin ligase c-Cbl regulates the osteoblastic differentiation program in mesenchymal cells by controlling Cbl-mediated STAT5 degradation and activity.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , STAT5 Transcription Factor/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , STAT5 Transcription Factor/genetics , Signal Transduction , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...