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










Publication year range
1.
JA Clin Rep ; 6(1): 73, 2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32989528

ABSTRACT

BACKGROUND: Excessive dynamic airway collapse (EDAC) is an uncommon cause of high airway pressure during mechanical ventilation. However, EDAC is not widely recognized by anesthesiologists, and therefore, it is often misdiagnosed as asthma. CASE PRESENTATION: A 70-year-old woman with a history of asthma received anesthesia with sevoflurane for a laparotomic cholecystectomy. Under general anesthesia, she developed wheezing, high inspiratory pressure, and a shark-fin waveform on capnography, which was interpreted as an asthma attack. However, treatment with a bronchodilator was ineffective. Bronchoscopy revealed the collapse of the trachea and main bronchi upon expiration. We reviewed the preoperative computed tomography scan and saw bulging of the posterior membrane into the airway lumen, leading to a diagnosis of EDAC. CONCLUSIONS: Although both EDAC and bronchospasm present as similar symptoms, the treatments are different. Bronchoscopy proved useful for distinguishing between these two entities. Positive end-expiratory pressure should be applied and bronchodilators avoided in EDAC.

3.
Sci Rep ; 8(1): 8475, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855498

ABSTRACT

Bone morphogenetic proteins (BMPs) activate the canonical Smad1/5/8 and non-canonical Tak1-MAPK pathways via BMP receptors I and II to regulate skeletal development and bone remodeling. Specific ablation of Bmpr1a in immature osteoblasts, osteoblasts, or osteocytes results in an increase in cancellous bone mass, yet opposite results have been reported regarding the underlying mechanisms. Moreover, the role for BMPRIA-mediated signaling in bone marrow mesenchymal stromal cells (BM-MSCs) has not been explored. Here, we specifically ablated Bmpr1a in BM-MSCs in adult mice to study the function of BMPR1A in bone remodeling and found that the mutant mice showed an increase in cancellous and cortical bone mass, which was accompanied by a decrease in bone formation rate and a greater decrease in bone resorption. Decreased bone formation was associated with a defect in BM-MSC osteogenic differentiation whereas decreased bone resorption was associated with a decrease in RANKL production and osteoclastogenesis. However, ablation of Tak1, a critical non-canonical signaling molecule downstream of BMP receptors, in BM-MSCs at adult stage did not affect bone remodeling. These results suggest that BMP signaling through BMPRIA controls BM-MSC osteogenic differentiation/bone formation and RANKL expression/osteoclastogenesis in adult mice independent of Tak1 signaling.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Differentiation , RANK Ligand/metabolism , Animals , Bone Marrow Cells/cytology , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Lineage , Homeodomain Proteins/metabolism , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , RANK Ligand/genetics , Signal Transduction
4.
Biochim Biophys Acta ; 1852(5): 839-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25558817

ABSTRACT

Aberrant fibroblast growth factor (FGF) signaling disturbs chondrocyte differentiation in skeletal dysplasia, but the mechanisms underlying this process remain unclear. Recently, FGF was found to activate canonical WNT/ß-catenin pathway in chondrocytes via Erk MAP kinase-mediated phosphorylation of WNT co-receptor Lrp6. Here, we explore the cellular consequences of such a signaling interaction. WNT enhanced the FGF-mediated suppression of chondrocyte differentiation in mouse limb bud micromass and limb organ cultures, leading to inhibition of cartilage nodule formation in micromass cultures, and suppression of growth in cultured limbs. Simultaneous activation of the FGF and WNT/ß-catenin pathways resulted in loss of chondrocyte extracellular matrix, expression of genes typical for mineralized tissues and alteration of cellular shape. WNT enhanced the FGF-mediated downregulation of chondrocyte proteoglycan and collagen extracellular matrix via inhibition of matrix synthesis and induction of proteinases involved in matrix degradation. Expression of genes regulating RhoA GTPase pathway was induced by FGF in cooperation with WNT, and inhibition of the RhoA signaling rescued the FGF/WNT-mediated changes in chondrocyte cellular shape. Our results suggest that aberrant FGF signaling cooperates with WNT/ß-catenin in suppression of chondrocyte differentiation.


Subject(s)
Cartilage/drug effects , Cell Differentiation/drug effects , Chondrocytes/drug effects , Fibroblast Growth Factors/pharmacology , Receptors, Fibroblast Growth Factor/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Blotting, Western , Cartilage/cytology , Cartilage/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cells, Cultured , Chondrocytes/metabolism , Drug Synergism , Fibroblast Growth Factor 2/pharmacology , HEK293 Cells , Humans , Limb Buds/drug effects , Limb Buds/embryology , Limb Buds/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Microscopy, Confocal , Models, Biological , Rats , Receptors, Fibroblast Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Transcriptome/genetics , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt3A Protein/pharmacology , beta Catenin/genetics
5.
J Bone Miner Res ; 30(5): 765-74, 2015 May.
Article in English | MEDLINE | ID: mdl-25401279

ABSTRACT

Chondrocytes in the epiphyseal cartilage undergo terminal differentiation prior to their removal through apoptosis. To examine the role of ERK1 and ERK2 in chondrocyte terminal differentiation, we generated Osterix (Osx)-Cre; ERK1(-/-) ; ERK2(flox/flox) mice (conditional knockout Osx [cKOosx]), in which ERK1 and ERK2 were deleted in hypertrophic chondrocytes. These cKOosx mice were grossly normal in size at birth, but by 3 weeks of age exhibited shorter long bones. Histological analysis in these mice revealed that the zone of hypertrophic chondrocytes in the growth plate was markedly expanded. In situ hybridization and quantitative real-time PCR analyses demonstrated that Matrix metalloproteinase-13 (Mmp13) and Osteopontin expression was significantly decreased, indicating impaired chondrocyte terminal differentiation. Moreover, Egr1 and Egr2, transcription factors whose expression is restricted to the last layers of hypertrophic chondrocytes in wild-type mice, were also strongly downregulated in these cKOosx mice. In transient transfection experiments in the RCS rat chondrosarcoma cell line, the expression of Egr1, Egr2, or a constitutively active mutant of MEK1 increased the activity of an Osteopontin promoter, whereas the MEK1-induced activation of the Osteopontin promoter was inhibited by the coexpression of Nab2, an Egr1 and Egr2 co-repressor. These results suggest that MEK1-ERK signaling activates the Osteopontin promoter in part through Egr1 and Egr2. Finally, our histological analysis of cKOosx mice demonstrated enchondroma-like lesions in the bone marrow that are reminiscent of human metachondromatosis, a skeletal disorder caused by mutations in PTPN11. Our observations suggest that the development of enchondromas in metachondromatosis may be caused by reduced extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK MAPK) signaling.


Subject(s)
Cell Differentiation , Chondrocytes/enzymology , Chondrocytes/pathology , Chondrogenesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteogenesis , Animals , Biomarkers/metabolism , Calcification, Physiologic , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 2/metabolism , Enzyme Activation , Gene Deletion , Hypertrophy , In Situ Hybridization , Integrases/metabolism , Mice, Knockout , Osteoblasts/enzymology , Osteoblasts/pathology , Sp7 Transcription Factor , Transcription Factors/metabolism , Transgenes
6.
J Cell Physiol ; 229(11): 1607-17, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24604556

ABSTRACT

The transcriptional cofactor Jab1 controls cell proliferation, apoptosis, and differentiation in diverse developmental processes by regulating the activity of various transcription factors. To determine the role of Jab1 during early limb development, we developed a novel Jab1(flox/flox) ; Prx1-Cre conditional Knockout (cKO) mutant mouse model in which Jab1 was deleted in the osteochondral progenitor cells of the limb buds. Jab1 cKO mutant mice displayed drastically shortened limbs at birth. The short-limb defect became apparent in Jab1 cKO mutants at E15.5 and increasingly worsened thereafter. By E18.5, Jab1 cKO mutant mice exhibited significantly shorter limbs with: very few hypertrophic chondrocytes, disorganized chondrocyte columns, much smaller primary ossification centers, and significantly increased apoptosis. Real-time RT-PCR analysis showed decreased expression of Sox9, Col2a1, Ihh, and Col10a1 in Jab1 cKO mutant long bones, indicating impaired chondrogenesis. Furthermore, in a micromass culture model of early limb mesenchyme cells, alcian blue staining showed a significant decrease in chondrogenesis in Jab1 cKO limb bud cells. The expression of Sox9 and its downstream targets Col2a1 and Aggrecan, as well as BMP signaling downstream targets, Noggin, Id1, and Ihh, were significantly decreased in Jab1 cKO micromass cultures. Moreover, over-expression of SOX9 in Jab1 cKO micromass cultures partially restored Col2a1and Aggrecan expression. Jab1-deficient micromass cultures also exhibited decreased BMP signaling response and reduced BMP-specific reporter activity ex vivo. In summary, our study demonstrates that Jab1 is an essential regulator of early embryonic limb development in vivo, likely in part by co-activating Sox9 and BMP signaling.


Subject(s)
Chondrocytes/metabolism , Embryonic Development , Extremities/embryology , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Bone Morphogenetic Proteins/metabolism , COP9 Signalosome Complex , Calcification, Physiologic , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Chondrogenesis , Gene Deletion , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Osteoblasts/cytology , Osteogenesis , Peptide Hydrolases/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
7.
Bone ; 58: 136-145, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24513582

ABSTRACT

Bones consist of a number of cell types including osteoblasts and their precursor cells at various stages of differentiation. To analyze cellular organization within the bone, we generated Col1a1CreER-DsRed transgenic mice that express, in osteoblasts, CreER and DsRed under the control of a mouse 3.2kb Col1a1 promoter. We further crossed Col1a1CreER-DsRed mice with Prx1CreER-GFP mice that express CreER and GFP in osteochondro progenitor cells under the control of a 2.4kb Prx1 promoter. Since the 3.2kb Col1a1 promoter becomes active in osteoblasts at early stages of differentiation, and Prx1CreER-GFP-expressing periosteal cells show endogenous Col1a1 expression, we expected to find a cell population in which both the 2.4kb Prx1 promoter and the 3.2kb Col1a1 promoter are active. However, our histological and flow cytometric analyses demonstrated that these transgenes are expressed in distinct cell populations. In the periosteum of long bones, Col1a1CreER-DsRed is expressed in the innermost layer directly lining the bone surface, while Prx1CreER-GFP-expressing cells are localized immediately outside of the Col1a1CreER-DsRed-expressing osteoblasts. In the calvaria, Prx1CreER-GFP-expressing cells are also localized in the cranial suture mesenchyme. Our experiments further showed that Col1a1CreER-DsRed-expressing cells lack chondrogenic potential, while the Prx1CreER-GFP-expressing cells show both chondrogenic and osteogenic potential. Our results indicate that Col1a1CreER-DsRed-expressing cells are committed osteoblasts, while Prx1CreER-GFP-expressing cells are osteochondro progenitor cells. The Prx1CreER-GFP and Col1a1CreER-DsRed transgenes will offer novel approaches for analyzing lineage commitment and early stages of osteoblast differentiation under physiologic and pathologic conditions.

8.
Arthritis Rheumatol ; 66(1): 78-89, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24449577

ABSTRACT

OBJECTIVE: The C-type natriuretic peptide (CNP) signaling pathway is a major contributor to postnatal skeletal growth in humans. This study was undertaken to investigate whether CNP signaling could prevent growth delay and cartilage damage in an animal model of inflammatory arthritis. METHODS: We generated transgenic mice that overexpress CNP (B6.SJL-Col2a1-NPPC) in chondrocytes. We introduced the CNP transgene into mice with experimental systemic inflammatory arthritis (K/BxN T cell receptor [TCR]) and determined the effect of CNP overexpression in chondrocytes on the severity of arthritis, cartilage damage, and linear growth. We also examined primary chondrocyte cultures for changes in gene and protein expression resulting from CNP overexpression. RESULTS: K/BxN TCR mice exhibited linear growth delay (P < 0.01) compared to controls, and this growth delay was correlated with the severity of arthritis. Diminished chondrocyte proliferation and matrix production was also seen in K/BxN TCR mice. Compared to non-CNP-transgenic mice, K/BxN TCR mice with overexpressed CNP had milder arthritis, no growth delay, and less cartilage damage. Primary chondrocytes from mice overexpressing CNP were less sensitive to inflammatory cytokines than wild-type mouse chondrocytes. CONCLUSION: CNP overexpression in chondrocytes can prevent endochondral growth delay and protect against cartilage damage in a mouse model of inflammatory arthritis. Pharmacologic or biologic modulation of the CNP signaling pathway may prevent growth retardation and protect cartilage in patients with inflammatory joint diseases, such as juvenile idiopathic arthritis.


Subject(s)
Arthritis, Experimental/physiopathology , Bone Development/physiology , Cartilage, Articular/growth & development , Chondrocytes/physiology , Natriuretic Peptide, C-Type/physiology , Animals , Arthritis, Experimental/metabolism , Arthritis, Juvenile/metabolism , Arthritis, Juvenile/physiopathology , Cartilage, Articular/metabolism , Cell Differentiation , Cell Proliferation , Chondrocytes/metabolism , Mice , Mice, Transgenic , Natriuretic Peptide, C-Type/metabolism , Signal Transduction/physiology
9.
ISME J ; 8(1): 40-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23924784

ABSTRACT

Deep-sea vents harbor dense populations of various animals that have their specific symbiotic bacteria. Scaly-foot gastropods, which are snails with mineralized scales covering the sides of its foot, have a gammaproteobacterial endosymbiont in their enlarged esophageal glands and diverse epibionts on the surface of their scales. In this study, we report the complete genome sequencing of gammaproteobacterial endosymbiont. The endosymbiont genome displays features consistent with ongoing genome reduction such as large proportions of pseudogenes and insertion elements. The genome encodes functions commonly found in deep-sea vent chemoautotrophs such as sulfur oxidation and carbon fixation. Stable carbon isotope ((13)C)-labeling experiments confirmed the endosymbiont chemoautotrophy. The genome also includes an intact hydrogenase gene cluster that potentially has been horizontally transferred from phylogenetically distant bacteria. Notable findings include the presence and transcription of genes for flagellar assembly, through which proteins are potentially exported from bacterium to the host. Symbionts of snail individuals exhibited extreme genetic homogeneity, showing only two synonymous changes in 19 different genes (13 810 positions in total) determined for 32 individual gastropods collected from a single colony at one time. The extremely low genetic individuality in endosymbionts probably reflects that the stringent symbiont selection by host prevents the random genetic drift in the small population of horizontally transmitted symbiont. This study is the first complete genome analysis of gastropod endosymbiont and offers an opportunity to study genome evolution in a recently evolved endosymbiont.


Subject(s)
Gammaproteobacteria/physiology , Genome, Bacterial/genetics , Snails/microbiology , Animals , Carbon Isotopes/metabolism , DNA Transposable Elements/genetics , Flagella/genetics , Gammaproteobacteria/classification , Gammaproteobacteria/genetics , Gammaproteobacteria/metabolism , Genetic Variation , Hydrothermal Vents , Molecular Sequence Data , Oxidation-Reduction , Phylogeny , Symbiosis
10.
Stem Cells ; 31(12): 2789-99, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23963683

ABSTRACT

The protein kinase inhibitor (Pki) gene family inactivates nuclear protein kinase A (PKA) and terminates PKA-induced gene expression. We previously showed that Pkig is the primary family member expressed in osteoblasts and that Pkig knockdown increases the effects of parathyroid hormone and isoproterenol on PKA activation, gene expression, and inhibition of apoptosis. Here, we determined whether endogenous levels of Pkig regulate osteoblast differentiation. Pkig is the primary family member in murine embryonic fibroblasts (MEFs), murine marrow-derived mesenchymal stem cells, and human mesenchymal stem cells. Pkig deletion increased forskolin-dependent nuclear PKA activation and gene expression and Pkig deletion or knockdown increased osteoblast differentiation. PKA signaling is known to stimulate adipogenesis; however, adipogenesis and osteogenesis are often reciprocally regulated. We found that the reciprocal regulation predominates over the direct effects of PKA since adipogenesis was decreased by Pkig deletion or knockdown. Pkig deletion or knockdown also simultaneously increased osteogenesis and decreased adipogenesis in mixed osteogenic/adipogenic medium. Pkig deletion increased PKA-induced expression of leukemia inhibitory factor (Lif) mRNA and LIF protein. LIF neutralizing antibodies inhibited the effects on osteogenesis and adipogenesis of either Pkig deletion in MEFs or PKIγ knockdown in both murine and human mesenchymal stem cells. Collectively, our results show that endogenous levels of Pkig reciprocally regulate osteoblast and adipocyte differentiation and that this reciprocal regulation is mediated in part by LIF. Stem Cells 2013;31:2789-2799.


Subject(s)
Adipocytes/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia Inhibitory Factor/metabolism , Osteoblasts/cytology , Adipocytes/metabolism , Animals , Cell Differentiation/physiology , Down-Regulation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia Inhibitory Factor/genetics , Male , Mice , Osteoblasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
11.
J Neurosurg Pediatr ; 12(1): 13-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23641963

ABSTRACT

True ossification within benign brain tumors is rare, and the molecular mechanism for this process is poorly understood. The authors report a case of ossified choroid plexus papilloma (CPP) and analyze it to help elucidate the underlying molecular basis of osteogenesis in benign brain tumors. A 21-year-old man presented with headache and depression that progressed over years. Computed tomography, MRI, and angiography demonstrated a large heavily calcified fourth ventricular tumor with a vascular blush and no hydrocephalus. The tumor was resected and was found to be an ossified CPP. Immunohistochemical staining for VEGF, Sox2, BMP-2, osterix, osteopontin, and osteocalcin was performed in an attempt to elucidate the mechanism of bone formation. The tumor was extensively ossified with mature bone trabeculae. Immunostaining for VEGF was positive. Additional staining showed the presence of osteocalcin in this ossified tumor but not in samples of nonossified CPPs collected from other patients. Staining for osterix and osteopontin was equivocally positive in the ossified CPP but also in the nonossified CPPs examined. The presence of osteocalcin in the ossified CPP demonstrates that there is true bone formation rather than simple calcification. Its appearance within cells around the trabeculae suggests the presence of osteoblasts. The presence of osterix suggests that a pluripotent cell, or one that is already partially differentiated, may be differentiated into an osteoblast through this pathway. This represents the first systematic immunohistochemical analysis of osteogenesis within choroid plexus tumors.


Subject(s)
Fourth Ventricle , Ossification, Heterotopic , Papilloma, Choroid Plexus/pathology , Papilloma, Choroid Plexus/surgery , Adult , Biomarkers/analysis , Depression/etiology , Fourth Ventricle/pathology , Fourth Ventricle/surgery , Headache/etiology , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Ossification, Heterotopic/surgery , Osteocalcin/analysis , Osteogenesis , Papilloma, Choroid Plexus/complications , Papilloma, Choroid Plexus/diagnosis , Tomography, X-Ray Computed
12.
J Bone Miner Metab ; 30(1): 19-30, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21678127

ABSTRACT

To examine the roles of FGF and ERK MAPK signaling in osteocyte differentiation and function, we performed microarray analyses using the osteocyte cell line MLO-Y4. This experiment identified a number of mineralization-related genes that were regulated by FGF2 in an ERK MAPK-dependent manner. Real-time PCR analysis indicated that FGF2 upregulates Ank, Enpp1, Mgp, Slc20a1, and Dmp1 in MLO-Y4 cells. Consistent with this observation, the selective FGF receptor inhibitor PD173074 decreased Ank, Enpp1, Slc20a1, and Dmp1 mRNA expression in mouse calvaria in organ culture. Since Dmp1 plays a central role in osteocyte differentiation and mineral homeostasis, we further analyzed FGF regulation of Dmp1. Similar to FGF2, FGF23 upregulated Dmp1 expression in MLO-Y4 cells in the presence of Klotho. Furthermore, increased extracellular phosphate levels partially inhibited FGF2-induced upregulation of Dmp1 mRNA expression, suggesting a coordinated regulation of Dmp1 expression by FGF signaling and extracellular phosphate. In MLO-Y4 osteocytes and in MC3T3E1 and primary calvaria osteoblasts, U0126 strongly inhibited both basal expression of Dmp1 mRNA and FGF2-induced upregulation. Consistent with the in vitro observations, real-time PCR and immunohistochemical analysis showed a strong decrease in Dmp1 expression in the skeletal elements of ERK1(-/-); ERK2(flox/flox); Prx1-Cre mice. Furthermore, scanning electron microscopic analysis revealed that no osteocytes with characteristic dendritic processes develop in the limbs of ERK1(-/-); ERK2 (flox/flox); Prx1-Cre mice. Collectively, our observations indicate that FGF signaling coordinately regulates mineralization-related genes in the osteoblast lineage and that ERK signaling is essential for Dmp1 expression and osteocyte differentiation.


Subject(s)
Calcification, Physiologic/genetics , Cell Differentiation/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/genetics , Osteocytes/cytology , Animals , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Line , Cycloheximide/pharmacology , Enzyme Activation/drug effects , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor-23 , Humans , Immunohistochemistry , MAP Kinase Signaling System/drug effects , Mice , Oligonucleotide Array Sequence Analysis , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocytes/drug effects , Osteocytes/enzymology , Osteocytes/ultrastructure , Phosphates/pharmacology , Protein Biosynthesis/drug effects , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Skull/cytology , Skull/drug effects , Skull/metabolism , Up-Regulation/drug effects
13.
J Orthop Res ; 29(3): 375-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20922792

ABSTRACT

Activating mutations in FGFR3 cause the most common forms of human dwarfism: achondroplasia and thanatophoric dysplasia. In mouse models of achondroplasia, recent studies have implicated the ERK MAPK pathway, a pathway activated by FGFR3, in creating reduced bone growth. Our recent studies have indicated that increased Fgfr3 and ERK MAPK signaling in chondrocytes also causes premature synchondrosis closure in the cranial base and vertebrae, accounting for the sometimes fatal stenosis of the foramen magnum and spinal canal in achondroplasia. Conversely, whether the decrease--or inactivation--of ERK1 and ERK2 promotes bone growth and delays synchondrosis closure remains to be investigated. In this study, we inactivated ERK2 in the chondrocytes of ERK1-null mice using the Col2a1-Cre and Col2a1-CreER transgenes. We found that the genetic inactivation of ERK1 and ERK2 in chondrocytes enhances the growth of cartilaginous skeletal elements. We also found that the postnatal inactivation of ERK1 and ERK2 in chondrocytes delays synchondrosis closure and enlarges the spinal canal. These observations make ERK1 and ERK2 an attractive target for the treatment of achondroplasia and other FGFR3-related skeletal syndromes.


Subject(s)
Achondroplasia/genetics , Bone Development/physiology , Chondrocytes/enzymology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Spinal Canal/physiology , Achondroplasia/physiopathology , Animals , Chondrocytes/cytology , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Spinal Canal/growth & development , Spine/growth & development , Spine/physiology , Tibia/growth & development , Tibia/physiology
14.
J Biol Chem ; 285(27): 20644-53, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20439987

ABSTRACT

The FGFR3 receptor tyrosine kinase represents an attractive target for therapy due to its role in several human disorders, including skeletal dysplasias, multiple myeloma, and cervical and bladder carcinomas. By using molecular library screening, we identified a compound named NF449 with inhibitory activity toward FGFR3 signaling. In cultured chondrocytes and murine limb organ culture, NF449 rescued FGFR3-mediated extracellular matrix loss and growth inhibition, which represent two major cellular phenotypes of aberrant FGFR3 signaling in cartilage. Similarly, NF449 antagonized FGFR3 action in the multiple myeloma cell lines OPM2 and KMS11, as evidenced by NF449-mediated reversal of ERK MAPK activation and transcript accumulation of CCL3 and CCL4 chemokines, both of which are induced by FGFR3 activation. In cell-free kinase assays, NF449 inhibited the kinase activity of both wild type and a disease-associated FGFR3 mutant (K650E) in a fashion that appeared non-competitive with ATP. Our data identify NF449 as a novel antagonist of FGFR3 signaling, useful for FGFR3 inhibition alone or in combination with inhibitors that target the ATP binding site.


Subject(s)
Benzenesulfonates/pharmacology , Chondrocytes/physiology , Multiple Myeloma/physiopathology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Animals , Bone and Bones/cytology , Bone and Bones/drug effects , CHO Cells , Cell Line, Tumor , Chondrocytes/drug effects , Cricetinae , Cricetulus , Female , Humans , Mice , Protein Kinases/drug effects , Protein Kinases/metabolism , RNA/drug effects , RNA/genetics , RNA, Neoplasm/drug effects , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfates/metabolism , Urinary Bladder Neoplasms/physiopathology , Uterine Cervical Neoplasms/physiopathology
15.
Mol Cell Biol ; 29(21): 5843-57, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19737917

ABSTRACT

Osteoblasts and chondrocytes arise from common osteo-chondroprogenitor cells. We show here that inactivation of ERK1 and ERK2 in osteo-chondroprogenitor cells causes a block in osteoblast differentiation and leads to ectopic chondrogenic differentiation in the bone-forming region in the perichondrium. Furthermore, increased mitogen-activated protein kinase signaling in mesenchymal cells enhances osteoblast differentiation and inhibits chondrocyte differentiation. These observations indicate that extracellular signal-regulated kinase 1 (ERK1) and ERK2 play essential roles in the lineage specification of mesenchymal cells. The inactivation of ERK1 and ERK2 resulted in reduced beta-catenin expression, suggesting a role for canonical Wnt signaling in ERK1 and ERK2 regulation of skeletal lineage specification. Furthermore, inactivation of ERK1 and ERK2 significantly reduced RANKL expression, accounting for a delay in osteoclast formation. Thus, our results indicate that ERK1 and ERK2 not only play essential roles in the lineage specification of osteo-chondroprogenitor cells but also support osteoclast formation in vivo.


Subject(s)
Cell Differentiation , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoclasts/enzymology , Osteogenesis , Animals , Base Pairing/genetics , Bone and Bones/abnormalities , Bone and Bones/enzymology , Chondrocytes/cytology , Chondrocytes/enzymology , Chondrogenesis , Choristoma/complications , Choristoma/enzymology , Enzyme Activation , Growth Plate/cytology , Growth Plate/enzymology , Homeodomain Proteins/genetics , Limb Deformities, Congenital/complications , Limb Deformities, Congenital/enzymology , MAP Kinase Kinase 1/metabolism , Mesoderm/cytology , Mesoderm/enzymology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 3/deficiency , Mutation/genetics , Osteochondrodysplasias/complications , Osteochondrodysplasias/enzymology , Osteoclasts/cytology , Promoter Regions, Genetic/genetics , RANK Ligand/metabolism
16.
Biochem Biophys Res Commun ; 386(3): 477-82, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19538944

ABSTRACT

We generated Prx1CreER-GFP transgenic mice that express tamoxifen-inducible Cre recombinase and GFP under the control of a 2.4 kb Prx1 promoter. The transgene is expressed in osteochondro progenitor cells in the developing limb buds and in a subpopulation of periosteal cells that is closely associated with the cortical bone. GFP-expressing cells isolated from the diaphyses of long bones by cell sorting express multiple markers of periosteal cells, including Prx1, Fgf18, Tenascin-W, Periostin, and Thrombospondin 2. In addition, these cells undergo chondrogenic and osteogenic differentiation in culture upon induction. Cell fate analysis using the Rosa26 LacZ reporter indicated that transgene-expressing cells give rise to some of the chondrocytes and osteoblasts in the fracture callus. Collectively, these observations strongly suggest that the transgene-expressing cells are osteochondro progenitor cells in the periosteum. The established Prx1CreER-GFP mice would offer novel approaches for analyzing the functions of periosteal cells in vitro and in vivo.


Subject(s)
Chondrocytes/physiology , Green Fluorescent Proteins/biosynthesis , Osteoblasts/physiology , Periosteum/physiology , Stem Cells/physiology , Animals , Biomarkers/metabolism , Chondrocytes/metabolism , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Integrases/genetics , Mice , Mice, Transgenic , Osteoblasts/metabolism , Periosteum/cytology , Proteins/genetics , RNA, Untranslated , Stem Cells/metabolism , beta-Galactosidase/genetics
17.
Hum Mol Genet ; 18(2): 227-40, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18923003

ABSTRACT

Activating mutations in FGFR3 cause achondroplasia and thanatophoric dysplasia, the most common human skeletal dysplasias. In these disorders, spinal canal and foramen magnum stenosis can cause serious neurologic complications. Here, we provide evidence that FGFR3 and MAPK signaling in chondrocytes promote synchondrosis closure and fusion of ossification centers. We observed premature synchondrosis closure in the spine and cranial base in human cases of homozygous achondroplasia and thanatophoric dysplasia as well as in mouse models of achondroplasia. In both species, premature synchondrosis closure was associated with increased bone formation. Chondrocyte-specific activation of Fgfr3 in mice induced premature synchondrosis closure and enhanced osteoblast differentiation around synchondroses. FGF signaling in chondrocytes increases Bmp ligand mRNA expression and decreases Bmp antagonist mRNA expression in a MAPK-dependent manner, suggesting a role for Bmp signaling in the increased bone formation. The enhanced bone formation would accelerate the fusion of ossification centers and limit the endochondral bone growth. Spinal canal and foramen magnum stenosis in heterozygous achondroplasia patients, therefore, may occur through premature synchondrosis closure. If this is the case, then any growth-promoting treatment for these complications of achondroplasia must precede the timing of the synchondrosis closure.


Subject(s)
Achondroplasia/metabolism , Bone Development , MAP Kinase Signaling System , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Thanatophoric Dysplasia/metabolism , Achondroplasia/genetics , Achondroplasia/physiopathology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cells, Cultured , Chondrocytes/metabolism , Chondrogenesis , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Thanatophoric Dysplasia/genetics , Thanatophoric Dysplasia/physiopathology
18.
Anat Rec A Discov Mol Cell Evol Biol ; 288(3): 316-22, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16463380

ABSTRACT

Achondroplasia, the most common short-limbed dwarfism in humans, results from a single nucleotide substitution in the gene for fibroblast growth factor receptor 3 (FGFR3). FGFR3 regulates bone growth in part via the mitogen-activated protein kinase pathway (MAPK). To examine the role of this pathway in chondrocyte differentiation, a transgenic mouse was generated that expresses a constitutively active mutant of MEK1 in chondrocytes and exhibits dwarfing characteristics typical of human achondroplasia, i.e., shortened axial and appendicular skeletons, mid-facial hypoplasia, and dome-shaped cranium. In this study, cephalometrics of the MEK1 mutant skulls were assessed to determine if the MEK1 mice are a good model of achondroplasia. Skull length, arc of the cranial vault, and area, maximum and minimum diameters of the brain case were measured on digitized radiographs of skulls of MEK1 and control mice. Cranial base and nasal bone length and foramen magnum diameter were measured on midsagittal micro-CT sections. Data were normalized by dividing by the cube root of each animal's weight. Transgenic mice exhibited a domed skull, deficient midface, and (relatively) prognathic mandible and had a shorter cranial base and nasal bone than the wild-type. Skull length was significantly less in transgenic mice, but cranial arc was significantly greater. The brain case was larger and more circular and minimum diameter of the brain case was significantly greater in transgenic mice. The foramen magnum was displaced anteriorly but not narrowed. MEK1 mouse cephalometrics confirm these mice as a model for achondroplasia, demonstrating that the MAP kinase signaling pathway is involved in FGF signaling in skeletal development.


Subject(s)
Achondroplasia/pathology , Disease Models, Animal , Mice, Transgenic/abnormalities , Skull/pathology , Achondroplasia/diagnostic imaging , Achondroplasia/genetics , Animals , Cephalometry , Mice , Radiography , Skull/diagnostic imaging
19.
Proc Natl Acad Sci U S A ; 103(2): 365-70, 2006 Jan 10.
Article in English | MEDLINE | ID: mdl-16387856

ABSTRACT

Accumulating in vitro evidence suggests that the p38 mitogen-activated protein kinase (MAPK) pathway is involved in endochondral ossification. To investigate the role of this pathway in endochondral ossification, we generated transgenic mice with expression in chondrocytes of a constitutively active mutant of MKK6, a MAPK kinase that specifically activates p38. These mice had a dwarf phenotype characterized by reduced chondrocyte proliferation, inhibition of hypertrophic chondrocyte differentiation, and a delay in the formation of primary and secondary ossification centers. Histological analysis with in situ hybridization showed reduced expression of Indian hedgehog, PTH/PTH-related peptide receptor (PTH, parathyroid hormone), cyclin D1, and increased expression of p21 in chondrocytes. In addition, both in vivo and in transfected cells, p38 signaling increased the transcriptional activity of Sox9, a transcription factor essential for chondrocyte differentiation. In agreement with this observation, transgenic mice that express a constitutively active mutant of MKK6 in chondrocytes showed phenotypes similar to those of mice that overexpress SOX9 in chondrocytes. These observations are consistent with the notion that increased activity of Sox9 accounts at least in part for the phenotype caused by constitutive activation of MKK6 in chondrocytes. Therefore, our study provides in vivo evidence for the role of p38 in endochondral ossification and suggests that Sox9 is a likely downstream target of the p38 MAPK pathway.


Subject(s)
Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , MAP Kinase Kinase 6/metabolism , Osteogenesis , Animals , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Enzyme Activation/genetics , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , MAP Kinase Kinase 6/genetics , Mice , Mice, Transgenic , Mutation/genetics , SOX9 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Genes Dev ; 18(3): 290-305, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871928

ABSTRACT

We generated transgenic mice that express a constitutively active mutant of MEK1 in chondrocytes. These mice showed a dwarf phenotype similar to achondroplasia, the most common human dwarfism, caused by activating mutations in FGFR3. These mice displayed incomplete hypertrophy of chondrocytes in the growth plates and a general delay in endochondral ossification, whereas chondrocyte proliferation was unaffected. Immunohistochemical analysis of the cranial base in transgenic embryos showed reduced staining for collagen type X and persistent expression of Sox9 in chondrocytes. These observations indicate that the MAPK pathway inhibits hypertrophic differentiation of chondrocytes and negatively regulates bone growth without inhibiting chondrocyte proliferation. Expression of a constitutively active mutant of MEK1 in chondrocytes of Fgfr3-deficient mice inhibited skeletal overgrowth, strongly suggesting that regulation of bone growth by FGFR3 is mediated at least in part by the MAPK pathway. Although loss of Stat1 restored the reduced chondrocyte proliferation in mice expressing an achondroplasia mutant of Fgfr3, it did not rescue the reduced hypertrophic zone, the delay in formation of secondary ossification centers, and the achondroplasia-like phenotype. These observations suggest a model in which Fgfr3 signaling inhibits bone growth by inhibiting chondrocyte differentiation through the MAPK pathway and by inhibiting chondrocyte proliferation through Stat1.


Subject(s)
Achondroplasia/genetics , Chondrocytes/metabolism , DNA-Binding Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/genetics , Trans-Activators/metabolism , Animals , Cell Differentiation , MAP Kinase Kinase 1 , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , Osteogenesis , Phenotype , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/deficiency , STAT1 Transcription Factor , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...