Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 94
1.
Cells ; 13(9)2024 Apr 23.
Article En | MEDLINE | ID: mdl-38727265

Fibrous dysplasia (FD) is a rare bone disorder characterized by the replacement of normal bone with benign fibro-osseous tissue. Developments in our understanding of the pathophysiology and treatment options are impeded by the lack of suitable research models. In this study, we developed an in vitro organotypic model capable of recapitulating key intrinsic and phenotypic properties of FD. Initially, transcriptomic profiling of individual cells isolated from patient lesional tissues unveiled intralesional molecular and cellular heterogeneity. Leveraging these insights, we established patient-derived organoids (PDOs) using primary cells obtained from patient FD lesions. Evaluation of PDOs demonstrated preservation of fibrosis-associated constituent cell types and transcriptional signatures observed in FD lesions. Additionally, PDOs retained distinct constellations of genomic and metabolic alterations characteristic of FD. Histological evaluation further corroborated the fidelity of PDOs in recapitulating important phenotypic features of FD that underscore their pathophysiological relevance. Our findings represent meaningful progress in the field, as they open up the possibility for in vitro modeling of rare bone lesions in a three-dimensional context and may signify the first step towards creating a personalized platform for research and therapeutic studies.


Fibrous Dysplasia of Bone , Organoids , Phenotype , Humans , Organoids/pathology , Organoids/metabolism , Fibrous Dysplasia of Bone/pathology , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Male , Female , Transcriptome/genetics , Adult
2.
Cells ; 13(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38727310

Fibrous dysplasia (FD) is a mosaic skeletal disorder caused by somatic activating variants of GNAS encoding for Gαs and leading to excessive cyclic adenosine monophosphate signaling in bone-marrow stromal cells (BMSCs). The effect of Gαs activation in the BMSC transcriptome and how it influences FD lesion microenvironment are unclear. We analyzed changes induced by Gαs activation in the BMSC transcriptome and secretome. RNAseq analysis of differential gene expression of cultured BMSCs from patients with FD and healthy volunteers, and from an inducible mouse model of FD, was performed, and the transcriptomic profiles of both models were combined to build a robust FD BMSC genetic signature. Pathways related to Gαs activation, cytokine signaling, and extracellular matrix deposition were identified. To assess the modulation of several key secreted factors in FD pathogenesis, cytokines and other factors were measured in culture media. Cytokines were also screened in a collection of plasma samples from patients with FD, and positive correlations of several cytokines to their disease burden score, as well as to one another and bone turnover markers, were found. These data support the pro-inflammatory, pro-osteoclastic behavior of FD BMSCs and point to several cytokines and other secreted factors as possible therapeutic targets and/or circulating biomarkers for FD.


Fibrous Dysplasia of Bone , Mesenchymal Stem Cells , Transcriptome , Humans , Animals , Mesenchymal Stem Cells/metabolism , Transcriptome/genetics , Mice , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Male , Female , Cytokines/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Adult , Middle Aged
3.
Int J Mol Sci ; 25(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38732172

Fibrous dysplasia (FD) poses a therapeutic challenge due to the dysregulated extracellular matrix (ECM) accumulation within affected bone tissues. In this study, we investigate the therapeutic potential of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) in managing FD by examining its effects on FD-derived cells in vitro. Our findings demonstrate that 1,25(OH)2D3 treatment attenuates the pro-fibrotic phenotype of FD-derived cells by suppressing the expression of key pro-fibrotic markers and inhibiting cell proliferation and migration. Moreover, 1,25(OH)2D3 enhances mineralization by attenuating pre-osteoblastic cellular hyperactivity and promoting maturation towards an osteocytic phenotype. These results offer valuable insights into potential treatments for FD, highlighting the role of 1,25(OH)2D3 in modulating the pathological properties of FD-derived cells.


Cell Proliferation , Fibrous Dysplasia of Bone , Humans , Cell Proliferation/drug effects , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Fibrous Dysplasia of Bone/drug therapy , Phenotype , Vitamin D/pharmacology , Vitamin D/metabolism , Fibrosis , Osteoblasts/drug effects , Osteoblasts/metabolism , Cell Movement/drug effects , Cell Differentiation/drug effects , Calcitriol/pharmacology , Cells, Cultured
4.
Int J Mol Sci ; 24(8)2023 Apr 20.
Article En | MEDLINE | ID: mdl-37108778

Fibrous dysplasia (FD) is a skeletal stem cell disease caused by mutations in the guanine nucleotide-binding protein, alpha-stimulating activity polypeptide (GNAS) gene, which results in the abnormal accumulation of cyclic adenosine monophosphate (cAMP) and hyperactivation of downstream signaling pathways. Parathyroid hormone-related protein (PTHrP) is secreted by the osteoblast lineage and is involved in various physiological and pathological activities of bone. However, the association between the abnormal expression of PTHrP and FD, as well as its underlying mechanism, remains unclear. In this study, we discovered that FD patient-derived bone marrow stromal cells (FD BMSCs) expressed significantly higher levels of PTHrP during osteogenic differentiation and exhibited greater proliferation capacity but impaired osteogenic ability compared to normal control patient-derived BMSCs (NC BMSCs). Continuous exogenous PTHrP exposure on the NC BMSCs promoted the FD phenotype in both in vitro and in vivo experiments. Through the PTHrP/cAMP/PKA axis, PTHrP could partially influence the proliferation and osteogenesis capacity of FD BMSCs via the overactivation of the Wnt/ß-Catenin signaling pathway. Furthermore, PTHrP not only directly modulated cAMP/PKA/CREB transduction but was also demonstrated as a transcriptional target of CREB. This study provides novel insight into the possible pathogenesis involved in the FD phenotype and enhances the understanding of its molecular signaling pathways, offering theoretical evidence for the feasibility of potential therapeutic targets for FD.


Craniofacial Fibrous Dysplasia , Fibrous Dysplasia of Bone , Humans , Osteogenesis/genetics , Parathyroid Hormone-Related Protein/genetics , Cell Differentiation/genetics , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Wnt Signaling Pathway , Cell Proliferation/genetics , Cells, Cultured
5.
Expert Opin Biol Ther ; 22(3): 397-405, 2022 Mar.
Article En | MEDLINE | ID: mdl-34964677

INTRODUCTION: Fibrous dysplasia (FD) is a rare bone disease that is associated with various endocrine conditions, such as McCune Albright syndrome. It manifests as abnormal osteolysis, multiple fractures, or deformities that are reported during disease course. The receptor activator of nuclear factor-kappa B (RANK)/RANK ligand (RANKL) pathway is upregulated in FD and can be targeted with denosumab, a blocking monoclonal antibody. AREAS COVERED: Preclinical and clinical data on the scientific rationale for using denosumab in FD and on the efficacy and safety of this therapy for this condition have been reviewed, in addition to other therapies. EXPERT OPINION: Denosumab is a potential therapeutic agent against FD. A combined synergic approach involving theranostics might increase its therapeutic potential.


Denosumab , Fibrous Dysplasia of Bone , Antibodies, Monoclonal/therapeutic use , Bone and Bones/metabolism , Denosumab/therapeutic use , Fibrous Dysplasia of Bone/drug therapy , Fibrous Dysplasia of Bone/metabolism , Humans , Receptor Activator of Nuclear Factor-kappa B
6.
Mol Med ; 27(1): 81, 2021 07 22.
Article En | MEDLINE | ID: mdl-34294046

BACKGROUND: Fibrous dysplasia (FD) is a bone marrow stromal cell (BMSC) disease caused by activating mutations of guanine nucleotide-binding protein alpha-stimulating activity polypeptide (GNAS) and is characterized by increased proliferative activity and disrupted osteogenesis of BMSCs. However, the molecular mechanisms regulating the pathophysiologic features of BMSCs in FD remain unknown. This study aimed to identify and verify the roles of the CREB1-miR-181a-5p regulatory loop in FD pathophysiology. METHODS: MicroRNA (miRNA) sequencing analysis was used to identify the possible miRNAs implicated in FD. The proliferation, apoptosis, and osteogenic differentiation of BMSCs, as well as the osteoclast-induced phenotype, were measured and compared after exogenous miR-181a-5p transfection into FD BMSCs or miR-181a-5p inhibitor transfection into normal BMSCs. Chromatin immunoprecipitation and luciferase reporter assays were performed to verify the interactions between CREB1 and miR-181a-5p and their effects on the FD pathological phenotype. RESULTS: Compared to normal BMSCs, FD BMSCs showed decreased miR-181a-5p levels and exhibited increased proliferative activity, decreased apoptotic capacity, and impaired osteogenesis. FD BMSCs also showed a stronger osteoclast activation effect. miR-181a-5p overexpression reversed the pathophysiologic features of FD BMSCs, whereas miR-181a-5p suppression induced an FD-like phenotype in normal BMSCs. Mechanistically, miR-181a-5p was the downstream target of CREB1, and CREB1 was posttranscriptionally regulated by miR-181a-5p. CONCLUSIONS: Our study identifies that the interaction loop between CREB1 and miR-181a-5p plays a crucial role in regulating the pathophysiologic features of FD BMSCs. MiR-181a-5p may be a potential therapeutic target for the treatment of FD.


Cyclic AMP Response Element-Binding Protein/metabolism , Fibrous Dysplasia of Bone/etiology , Fibrous Dysplasia of Bone/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Apoptosis , Biomarkers , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Disease Susceptibility , Fibrous Dysplasia of Bone/pathology , Humans , Models, Biological , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis/genetics
7.
Mol Cell Biochem ; 476(2): 1221-1232, 2021 Feb.
Article En | MEDLINE | ID: mdl-33201382

The mammalian Na+/H+ exchanger isoform one (NHE1) is a plasma membrane protein that is ubiquitously present in human cells. It functions to regulate intracellular pH removing an intracellular proton in exchange for one extracellular sodium and is involved in heart disease and in promoting metastasis in cancer. It is made of a 500 amino acid membrane domain plus a 315 amino acid, regulatory cytosolic tail. The membrane domain is thought to have 12 transmembrane segments and a large membrane-associated extracellular loop. Early studies demonstrated that in mice, disruption of the NHE1 gene results in locomotor ataxia and a phenotype of slow-wave epilepsy. Defects included a progressive neuronal degeneration. Growth and reproductive ability were also reduced. Recent studies have identified human autosomal homozygous recessive mutations in the NHE1 gene (SLC9A1) that result in impaired development, ataxia and other severe defects, and explain the cause of the human disease Lichtenstein-Knorr syndrome. Other human mutations have been identified that are stop codon polymorphisms. These cause short non-functional NHE1 proteins, while other genetic polymorphisms in the NHE1 gene cause impaired expression of the NHE1 protein, reduced activity, enhanced protein degradation or altered kinetic activation of the protein. Since NHE1 plays a key role in many human physiological functions and in human disease, genetic polymorphisms of the protein that significantly alter its function and are likely play significant roles in varying human phenotypes and be involved in disease.


Fibrous Dysplasia of Bone/pathology , Immunologic Deficiency Syndromes/pathology , Mutation , Neurodegenerative Diseases/pathology , Neutropenia/pathology , Sodium-Hydrogen Exchanger 1/genetics , Amino Acid Sequence , Animals , Facies , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neutropenia/genetics , Neutropenia/metabolism , Protein Transport , Proteolysis , Sodium-Hydrogen Exchanger 1/metabolism
8.
J Bone Miner Res ; 34(12): 2171-2182, 2019 12.
Article En | MEDLINE | ID: mdl-31295366

Fibrous dysplasia of bone/McCune-Albright syndrome (Polyostotic FD/MAS; OMIM#174800) is a crippling skeletal disease caused by gain-of-function mutations of Gs α. Enhanced bone resorption is a recurrent histological feature of FD and a major cause of fragility of affected bones. Previous work suggests that increased bone resorption in FD is driven by RANKL and some studies have shown that the anti-RANKL monoclonal antibody, denosumab, reduces bone turnover and bone pain in FD patients. However, the effect of RANKL inhibition on the histopathology of FD and its impact on the natural history of the disease remain to be assessed. In this study, we treated the EF1α-Gs αR201C mice, which develop an FD-like phenotype, with an anti-mouse RANKL monoclonal antibody. We found that the treatment induced marked radiographic and microscopic changes at affected skeletal sites in 2-month-old mice. The involved skeletal segments became sclerotic due to the deposition of new, highly mineralized bone within developing FD lesions and showed a higher mechanical resistance compared to affected segments from untreated transgenic mice. Similar changes were also detected in older mice with a full-blown skeletal phenotype. The administration of anti-mouse RANKL antibody arrested the growth of established lesions and, in young mice, prevented the appearance of new ones. However, after drug withdrawal, the newly formed bone was remodelled into FD tissue and the disease progression resumed in young mice. Taken together, our results show that the anti-RANKL antibody significantly affected the bone pathology and natural history of FD in the mouse. Pending further work on the prevention and management of relapse after treatment discontinuation, our preclinical study suggests that RANKL inhibition may be an effective therapeutic option for FD patients. © 2019 American Society for Bone and Mineral Research.


Fibrous Dysplasia of Bone/metabolism , RANK Ligand/antagonists & inhibitors , Animals , Biomechanical Phenomena , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calcification, Physiologic , Denosumab/pharmacology , Disease Models, Animal , Disease Progression , Fibrous Dysplasia of Bone/complications , Fibrous Dysplasia of Bone/diagnostic imaging , Fibrous Dysplasia of Bone/pathology , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Mice, Transgenic , Osteolysis/blood , Osteolysis/complications , Peptide Elongation Factor 1/metabolism , Phenotype , RANK Ligand/metabolism , Rats
9.
Pathol Res Pract ; 215(7): 152450, 2019 Jul.
Article En | MEDLINE | ID: mdl-31109869

Differential diagnosis among fibrous dysplasias, cemento-ossifying fibromas and cemento-osseous dysplasias is difficult, since there is considerable overlap of histologic features, but also extremely important, since they differ greatly in etiology, clinical behaviour, prognosis and terapeuthic approach. There is no data about the use of immunohistochemistry, a viable and accessible technique, for this purpose. The objective of this study was to investigate, comparatively, the immunohistochemical expression of major non-collagenous proteins (osteonectin [ON], osteopontin [OP], bone sialoprotein [BSP] and osteocalcin [OC]) of mineralized tissue extracellular matrix in 22 cases of fibrous dysplasias, 16 of cemento-ossifying fibromas and 16 of cemento-osseous dysplasias. ON maintained the same expression profile in all cases; the staining for OP was negative in fusiform cells producing cementoid globules and weak, as well as heterogeneous, in high mineralized matrixes; there was negativity for BSP in cementoid globules and in the fusiform cells that produce them, differently from the strong positive expression found in the majority of bone trabeculae and their peripheral cuboidal osteoblasts; and finally, the immuno-reactivity for OC was weak, except in cuboidal osteoblasts and osteocytes. We can conclude that the nature of mineralized structure and the cellular phenotype are much more responsible for variability in immunohistochemical profile than the type of lesion (fibrous dysplasias, cemento-ossifying fibromas and cemento-osseous dysplasias) which makes difficult, at least for a while, the use of these proteins with diagnosis purpose.


Cementoma/diagnosis , Fibroma, Ossifying/diagnosis , Fibrous Dysplasia of Bone/diagnosis , Integrin-Binding Sialoprotein/metabolism , Osteocalcin/metabolism , Osteonectin/metabolism , Osteopontin/metabolism , Bone and Bones/pathology , Cementoma/metabolism , Cementoma/pathology , Diagnosis, Differential , Fibroma, Ossifying/metabolism , Fibroma, Ossifying/pathology , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Humans
10.
Clin Nucl Med ; 44(10): e593-e594, 2019 Oct.
Article En | MEDLINE | ID: mdl-31058693

Fibrous dysplasia is an uncommon benign bone pathology with only rare potential for malignant transformation. We describe the heterogeneous uptake of fibrous dysplasia of a whole rib on Ga-labeled prostate-specific membrane antigen (PSMA) PET/CT. Ga-PSMA binds to PSMA-expressing prostate cancer cells, but it can also bind to the neovasculature of various solid tumors, as well as to some benign lesions. Ga-PSMA expression in fibrous dysplasia should not automatically be equated with malignant transformation or confused with prostate cancer metastases.


Fibrous Dysplasia of Bone/diagnostic imaging , Fibrous Dysplasia of Bone/metabolism , Membrane Glycoproteins/metabolism , Organometallic Compounds/metabolism , Positron Emission Tomography Computed Tomography , Aged , Biological Transport , Diagnosis, Differential , Gallium Isotopes , Gallium Radioisotopes , Humans , Male
11.
Clin Nucl Med ; 44(6): e396-e397, 2019 Jun.
Article En | MEDLINE | ID: mdl-30932978

Prostate cancer is one of the most common malignancies. Imaging tools play an important role throughout the entire process of the disease. The scenario, however, is going to change. Thanks to a higher sensitivity and specificity, the use of the prostate-specific membrane antigen (PSMA) PET is of increasing importance, particularly at the time of diagnosis and in case of biochemical recurrence. Nevertheless, previous reports have described false-positive findings, as tracer-avid physiological findings or benign processes, potential pitfalls for interpretation of Ga-PSMA PET/CT. Here we report a case of PSMA uptake in a histologically proven fibrous dysplasia.


Fibrous Dysplasia of Bone/diagnostic imaging , Membrane Glycoproteins , Organometallic Compounds , Aged , Biological Transport , False Positive Reactions , Fibrous Dysplasia of Bone/metabolism , Gallium Isotopes , Gallium Radioisotopes , Humans , Male , Membrane Glycoproteins/metabolism , Organometallic Compounds/metabolism , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/pathology , Sensitivity and Specificity
12.
J Bone Miner Res ; 34(4): 653-660, 2019 04.
Article En | MEDLINE | ID: mdl-30645769

Fibrous dysplasia (FD) is a mosaic disease in which bone is replaced with fibro-osseous tissue. Lesions expand during childhood, reaching final burden by age 15 years. In vitro data suggest that disease activity decreases in adulthood; however, there is no clinical data to support this concept. Bone turnover markers (BTMs) have been used as markers of disease activity in FD; however, the natural history of BTM changes, the effects of antiresorptive treatment, and their association to clinical outcomes have not been described. The goals of this study are to describe 1) the natural history of FD disease activity and its association with pain; 2) the impact of bisphosphonates on the natural history of BTMs; and 3) the effect of bisphosphonates on progression of FD burden during childhood. Disease burden scores and alkaline phosphatase, osteocalcin, NTx, FGF23, and RANKL levels from 178 subjects in an FD/MAS natural history study were reviewed, including 73 subjects treated with bisphosphonates. BTMs, RANKL, and FGF23 demonstrated a sustained reduction with age. Bisphosphonate treatment did not significantly impact this age-dependent decrease in BTMs. Pain was more prevalent and severe in adults compared with children and was not associated with BTMs. In children, the progression of disease burden was not affected by bisphosphonates. In conclusion, FD is associated with an age-dependent decline in bone turnover and other markers of disease activity. Pain, in contrast, is more frequent and severe in adults with FD and is not related to bone turnover. Bisphosphonate treatment does not significantly impact the age-dependent decrease in bone turnover, nor does it prevent the progression of FD disease burden in children. These findings, in association with the established adverse effects of antiresorptives, should be considered when evaluating use and response to bisphosphonates in patients being treated for FD and in any study using BTMs as surrogate endpoints. © 2019 American Society for Bone and Mineral Research.


Aging/metabolism , Bone Remodeling/drug effects , Diphosphonates/administration & dosage , Fibrous Dysplasia of Bone/drug therapy , Fibrous Dysplasia of Bone/metabolism , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Aging/pathology , Biomarkers/metabolism , Child , Child, Preschool , Female , Fibroblast Growth Factor-23 , Fibrous Dysplasia of Bone/epidemiology , Fibrous Dysplasia of Bone/pathology , Humans , Male , Middle Aged , Pain/epidemiology , Pain/metabolism , Pain/pathology , Prevalence
13.
J Bone Miner Res ; 34(2): 290-294, 2019 02.
Article En | MEDLINE | ID: mdl-30496606

Fibrous dysplasia of bone (FD) is a mosaic disease caused by mutations in GNAS. Constitutive activation of the α-subunit of the Gs stimulatory protein (Gαs) leads to dysregulated proliferation of bone marrow stromal cells (BMSCs), generating expansile lesions of fibrotic tissue and abnormal bone. Local bone remodeling regulation by BMSCs is also altered, and FD tissue is characterized by abundant osteoclast-like cells that may be essential for lesion expansion. Animal models show local expression of RANKL in bone lesions, and treatment with the RANKL neutralizing antibody denosumab decreased lesion expansion rate in a patient with aggressive FD. However, the role of RANKL/osteoprotegerin (OPG) in FD pathophysiology is not yet understood. We measured serum levels of RANKL, OPG, and inactive RANKL-OPG complexes in FD patients of known disease burden and in healthy volunteers (HVs). RANK, RANKL, and Ki67 immunohistochemistry were assessed in FD tissue. Cultured FD and HV BMSCs were stimulated with prostaglandin E2 (PGE2 ) and 1,25 vitamin D3 to increase RANKL expression, and media levels of RANKL and OPG were measured. Osteoclastogenic induction by FD or HV BMSCs was assessed in co-cultures with HV peripheral monocytes. FD patients showed a 16-fold increase in serum RANKL compared to HVs. OPG was moderately increased (24%), although RANKL/OPG ratio was 12-fold higher in FD patients than in HVs. These measurements were positively correlated with the skeletal burden score (SBS), a validated marker of overall FD burden. No differences in serum inactive RANKL-OPG complexes were observed. In FD tissue, RANKL+ and Ki67+ fibroblastic cells were observed near RANK+ osteoclasts. High levels of RANKL were released by FD BMSCs cultures, but were undetectable in HV cultures. FD BMSC released less OPG than HV BMSCs. FD, but not HV BMSCs, induced osteoclastogenesis in monocyte co-cultures, which was prevented by denosumab addition. These data are consistent with the role of RANKL as a driver in FD-induced osteoclastogenesis. © 2018 American Society for Bone and Mineral Research.


Bone Marrow Cells/metabolism , Fibrous Dysplasia of Bone/metabolism , Mesenchymal Stem Cells/metabolism , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction , Bone Marrow Cells/pathology , Cells, Cultured , Female , Fibrous Dysplasia of Bone/pathology , Humans , Male , Mesenchymal Stem Cells/pathology
14.
Proc Natl Acad Sci U S A ; 115(3): E428-E437, 2018 01 16.
Article En | MEDLINE | ID: mdl-29282319

Fibrous dysplasia (FD) is a disease caused by postzygotic activating mutations of GNAS (R201C and R201H) that encode the α-subunit of the Gs stimulatory protein. FD is characterized by the development of areas of abnormal fibroosseous tissue in the bones, resulting in skeletal deformities, fractures, and pain. Despite the well-defined genetic alterations underlying FD, whether GNAS activation is sufficient for FD initiation and the molecular and cellular consequences of GNAS mutations remains largely unresolved, and there are no currently available targeted therapeutic options for FD. Here, we have developed a conditional tetracycline (Tet)-inducible animal model expressing the GαsR201C in the skeletal stem cell (SSC) lineage (Tet-GαsR201C/Prrx1-Cre/LSL-rtTA-IRES-GFP mice), which develops typical FD bone lesions in both embryos and adult mice in less than 2 weeks following doxycycline (Dox) administration. Conditional GαsR201C expression promoted PKA activation and proliferation of SSCs along the osteogenic lineage but halted their differentiation to mature osteoblasts. Rather, as is seen clinically, areas of woven bone admixed with fibrous tissue were formed. GαsR201C caused the concomitant expression of receptor activator of nuclear factor kappa-B ligand (Rankl) that led to marked osteoclastogenesis and bone resorption. GαsR201C expression ablation by Dox withdrawal resulted in FD-like lesion regression, supporting the rationale for Gαs-targeted drugs to attempt FD cure. This model, which develops FD-like lesions that can form rapidly and revert on cessation of mutant Gαs expression, provides an opportunity to identify the molecular mechanism underlying FD initiation and progression and accelerate the development of new treatment options.


Fibrous Dysplasia of Bone/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Anti-Bacterial Agents/toxicity , Bone Development/drug effects , Bone and Bones/pathology , Cell Differentiation , Doxycycline/toxicity , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Regulation , Gene Expression Regulation, Developmental/drug effects , Mice , Mutation
15.
Article En | MEDLINE | ID: mdl-29056287

OBJECTIVE: The aim of this study was to compare the expression of proliferative markers and apoptosis-associated proteins in the oral mucosa adjacent to bone sequestrum associated with osteonecrosis (ON) of the jaws and florid osseous dysplasia (OD). STUDY DESIGN: Oral mucosal samples derived from surgical procedures performed for treatment of ON and OD (10 cases each) were retrieved. Histologic analysis was done on hematoxylin and eosin-stained slides; immunohistochemical reactions against epidermal growth factor (EGF), keratinocyte growth factor (KGF), vascular endothelial growth factor (VEGF), Bcl-2 protein, interleukin-6 (IL-6), and Ki-67 antigen were performed with the immunoperoxidase technique. RESULTS: The epithelium was hyperplastic in 60% and 22% of ON and OD samples, respectively. Cytoplasmic EGF and KGF expression; Bcl-2, VEGF, and IL-6 expressions; and the mean epithelial proliferative index were not statistically different between the 2 groups. Membranous EGF expression was more evident in samples showing hyperplastic epithelium. CONCLUSIONS: Exposure to bisphosphonates did not alter the expression of KGF, EGF, VEGF, Bcl-2, IL-6, and the epithelial proliferating index in comparison with inflamed oral mucosa not exposed to bisphosphonates.


Bisphosphonate-Associated Osteonecrosis of the Jaw/metabolism , Epidermal Growth Factor/metabolism , Epithelium/metabolism , Fibroblast Growth Factor 7/metabolism , Fibrous Dysplasia of Bone/metabolism , Ki-67 Antigen/metabolism , Mouth Mucosa/metabolism , Osteomyelitis/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , Aged , Biomarkers/metabolism , Bisphosphonate-Associated Osteonecrosis of the Jaw/surgery , Female , Fibrous Dysplasia of Bone/surgery , Humans , Immunoenzyme Techniques , Male , Middle Aged , Osteomyelitis/surgery
16.
Biomaterials ; 107: 61-73, 2016 11.
Article En | MEDLINE | ID: mdl-27614159

Fibrous dysplasia (FD) caused by a mosaic somatic mutation of GNAS is characterized by replacement of the affected bone with abnormal fibrous tissue. Herein, we present novel disease models for FD developed with pairs of isogenic wild-type and GNAS(R201H)-mutated induced pluripotent stem cells (iPSCs) and their derivative mesenchymal stem cells (MSCs). Both 2D and 3D MSC culture models for FD successfully reflect FD's typical molecular characteristics, such as enhanced cAMP level, PKA activity, CREB1 phosphorylation and the pathologic fibrotic phenotype. The fibrotic features of GNAS(R201H) FD model cells were closely linked to augmented glycolysis and depended on glycolytic PFKFB4 and the activation of pro-fibrotic TGFß signalling. Either depletion of PFKFB4 or inhibition of glycolysis or TGFß signalling potentially blocked fibrosis progression in GNAS(R201H) FD model cells. Our FD models could facilitate a better mechanistic understanding of FD and help develop effective therapeutics for FD and other fibrosis diseases.


Bone and Bones/metabolism , Bone and Bones/pathology , Cellular Reprogramming , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Glycolysis , Phosphofructokinase-2/metabolism , Cell Differentiation , Cells, Cultured , Female , Fibrosis , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology
17.
PLoS One ; 11(5): e0154680, 2016.
Article En | MEDLINE | ID: mdl-27163152

The receptor-activator of nuclear kappaB ligand (RANKL) signaling pathway plays an important role in the regulation of bone growth and mediates the formation and activation of osteoclasts. Osteoclasts are involved in significant bone resorption and destruction. Denosumab is a fully human monoclonal antibody against RANKL that specifically inhibits osteoclast differentiation and bone resorption. It has been approved for use for multiple myeloma and bone metastases, as well as for giant cell tumor of bone. However, there is no previous report quantitatively, comparing RANKL expression in histologically varied bone tumors. Therefore, we analyzed the mRNA level of various bone tumors and investigated the possibility of these tumors as a new therapeutic target for denosumab. We examined RANKL mRNA expression in 135 clinical specimens of primary and metastatic bone tumors using real-time PCR. The relative quantification of mRNA expression levels was performed via normalization with RPMI8226, a human multiple myeloma cell line that is recognized to express RANKL. Of 135 cases, 64 were also evaluated for RANKL expression by using immunohistochemistry. Among all of the tumors investigated, RANKL expression and the RANKL/osteoprotegerin ratio were highest in giant cell tumor of bone. High RANKL mRNA expression was observed in cases of aneurysmal bone cyst, fibrous dysplasia, osteosarcoma, chondrosarcoma, and enchondroma, as compared to cases of multiple myeloma and bone lesions from metastatic carcinoma. RANKL-positive stromal cells were detected in six cases: five cases of GCTB and one case of fibrous dysplasia. The current study findings indicate that some primary bone tumors present new therapeutic targets for denosumab, particularly those tumors expressing RANKL and those involving bone resorption by osteoclasts.


Antineoplastic Agents/therapeutic use , Bone Cysts, Aneurysmal/genetics , Bone Neoplasms/genetics , Denosumab/therapeutic use , Giant Cell Tumor of Bone/genetics , RANK Ligand/genetics , Bone Cysts, Aneurysmal/drug therapy , Bone Cysts, Aneurysmal/metabolism , Bone Cysts, Aneurysmal/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Resorption/prevention & control , Chondroma/drug therapy , Chondroma/genetics , Chondroma/metabolism , Chondroma/pathology , Chondrosarcoma/drug therapy , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Female , Fibrous Dysplasia of Bone/drug therapy , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Gene Expression Regulation, Neoplastic , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/metabolism , Giant Cell Tumor of Bone/pathology , Humans , Male , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Organ Specificity , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Hum Mol Genet ; 24(2): 463-70, 2015 Jan 15.
Article En | MEDLINE | ID: mdl-25205112

Lichtenstein-Knorr syndrome is an autosomal recessive condition that associates sensorineural hearing loss and cerebellar ataxia. Here, we report the first identification of a gene involved in Lichtenstein-Knorr syndrome. By using a combination of homozygosity mapping and whole-exome sequencing, we identified the homozygous p.Gly305Arg missense mutation in SLC9A1 that segregates with the disease in a large consanguineous family. Mutant glycine 305 is a highly conserved amino acid present in the eighth transmembrane segment of all metazoan orthologues of NHE1, the Na(+)/H(+) exchanger 1, encoded by SLC9A1. We demonstrate that the p.Gly305Arg mutation causes the near complete de-glycosylation, mis-targeting and loss of proton pumping activity of NHE1. The comparison of our family with the phenotypes of spontaneous and knockout Slc9a1 murine models demonstrates that the association between ataxia and hearing loss is caused by complete or near complete loss of function of NHE1 and altered regulation of pHi in the central nervous system.


Cation Transport Proteins/genetics , Cerebellar Ataxia/genetics , Deafness/genetics , Fibrous Dysplasia of Bone/genetics , Immunologic Deficiency Syndromes/genetics , Mutation, Missense , Neutropenia/genetics , Sodium-Hydrogen Exchangers/genetics , Animals , Cation Transport Proteins/metabolism , Cerebellar Ataxia/metabolism , Deafness/metabolism , Facies , Female , Fibrous Dysplasia of Bone/metabolism , Humans , Immunologic Deficiency Syndromes/metabolism , Male , Mice , Mice, Knockout , Neutropenia/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/metabolism
19.
J Bone Miner Res ; 30(6): 1030-43, 2015 Jun.
Article En | MEDLINE | ID: mdl-25487351

We recently reported the generation and initial characterization of the first direct model of human fibrous dysplasia (FD; OMIM #174800), obtained through the constitutive systemic expression of one of the disease-causing mutations, Gsα(R201C) , in the mouse. To define the specific pathogenetic role(s) of individual cell types within the stromal/osteogenic system in FD, we generated mice expressing Gsα(R201C) selectively in mature osteoblasts using the 2.3kb Col1a1 promoter. We show here that this results in a striking high bone mass phenotype but not in a mimicry of human FD. The high bone mass phenotype involves specifically a deforming excess of cortical bone and prolonged and ectopic cortical bone remodeling. Expression of genes characteristic of late stages of bone cell differentiation/maturation is profoundly altered as a result of expression of Gsα(R201C) in osteoblasts, and expression of the Wnt inhibitor Sost is reduced. Although high bone mass is, in fact, a feature of some types/stages of FD lesions in humans, it is marrow fibrosis, localized loss of adipocytes and hematopoietic tissue, osteomalacia, and osteolytic changes that together represent the characteristic pathological profile of FD, as well as the sources of specific morbidity. None of these features are reproduced in mice with osteoblast-specific expression of Gsα(R201C) . We further show that hematopoietic progenitor/stem cells, as well as more mature cell compartments, and adipocyte development are normal in these mice. These data demonstrate that effects of Gsα mutations underpinning FD-defining tissue changes and morbidity do not reflect the effects of the mutations on osteoblasts proper.


Bone and Bones , Fibrous Dysplasia of Bone , GTP-Binding Protein alpha Subunits, Gs , Mutation, Missense , Osteoblasts , Amino Acid Substitution , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Chromogranins , Disease Models, Animal , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Organ Size , Osteoblasts/metabolism , Osteoblasts/physiology
20.
Am J Surg Pathol ; 38(11): 1510-5, 2014 Nov.
Article En | MEDLINE | ID: mdl-25118809

The most commonly encountered fibro-osseous lesions of the skull bone are fibrous dysplasia and ossifying fibroma. Two cases of a unique "protuberant fibro-osseous lesion of the temporal bone" were first described by Selesnick and colleagues in 1999, and 2 further cases were reported in 2010 under the name "Bullough lesion". We recently found 2 new cases of this rare entity. Two Korean female patients aged 70 and 54 years presented with slow growing postauricular masses without pain or tenderness for 6 and 7 years, respectively. Computed tomography revealed a 2.9 cm calcified mass in the temporal bone of the first patient, and a 5.5 cm enhancing mass with internal cartilaginous matrix in the temporal bone of the second patient. Intramedullary or intracranial extension was not found in either case, and en bloc removals were performed. Microscopically, multiple round to oval osseous islands were scattered throughout the bland fibrous stroma in both cases. The osseous islands varied in size and were lamellar or woven, without osteoblastic rimming, and surrounded by fibroblastic bands. Neither patient has shown evidence of postoperative recurrence for 18 months. The location, histology, and clinical course of these 2 cases were identical to the 4 cases previously reported, although age and sex varied. The lesions were tested for the R201H mutation in the GNAS gene, which is present in fibrous dysplasia. No mutations were found, suggesting a different genetic background for these lesions.


Fibroma, Ossifying , Fibrous Dysplasia of Bone , Skull Neoplasms , Temporal Bone , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Chromogranins , DNA Mutational Analysis , Female , Fibroma, Ossifying/chemistry , Fibroma, Ossifying/genetics , Fibroma, Ossifying/pathology , Fibroma, Ossifying/surgery , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Fibrous Dysplasia of Bone/surgery , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Immunohistochemistry , Middle Aged , Mutation , Skull Neoplasms/chemistry , Skull Neoplasms/genetics , Skull Neoplasms/pathology , Skull Neoplasms/surgery , Temporal Bone/chemistry , Temporal Bone/pathology , Temporal Bone/surgery , Tomography, X-Ray Computed , Treatment Outcome , Tumor Burden
...