Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
Add more filters










Publication year range
1.
J Bone Miner Metab ; 36(4): 447-453, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28664247

ABSTRACT

When children around 2 years of age show leg bowing and diseases are ruled out based on radiographic findings without conducting blood tests, they are classified as "physiologic" genu varum. Since whether or not physiologic genu varum is associated with bone metabolism is unclear, this study was conducted to clarify the association between genu varum and bone metabolism in children. Thirty-five pediatric patients with genu varm who visited our out-patient clinic were enrolled. While two of the 35 children had nutritional rickets, showing abnormalities on both blood test (ALP, ≥1000 IU/L; iPTH, >65 pg/mL and 25(OH)D, ≤20 ng/mL) and radiographs (such as cupping, fraying or splaying), five of 35 children showed abnormalities on blood tests but not radiographs. While metaphyseal-diaphyseal angle (MDA) correlated with serum 25-hydroxy vitamin D (r = -0.35, p = 0.04) and magnesium (r = -0.36, p = 0.04), MDA and femorotibial angle (FTA) correlated with alkaline phosphatase (r = 0.43, p = 0.01 and r = 0.51, p = 0.006, respectively). A ridge regression analysis adjusted for age and body mass index indicated that ALP was associated with MDA and FTA. A logistic regression analysis adjusted for age and BMI indicated that higher ALP influenced an MDA >11°, which indicates the risk for the progression of genu varum (odds ratio 1.002, 95% confidence interval 1.0003-1.003, p = 0.021). The higher ALP (+100 IU), the higher risk of an MDA >11° (odds ratio 1.22). In conclusion, genu varum is associated with the alkaline phosphatase level regardless of the presence of radiographic abnormalities in the growth plate in children.


Subject(s)
Alkaline Phosphatase/blood , Genu Varum/blood , Genu Varum/diagnostic imaging , Growth Plate/diagnostic imaging , Growth Plate/enzymology , Leg/diagnostic imaging , Leg/physiopathology , Biomechanical Phenomena , Body Mass Index , Child, Preschool , Diaphyses/physiopathology , Female , Genu Varum/enzymology , Genu Varum/physiopathology , Growth Plate/physiopathology , Humans , Male , Regression Analysis
2.
Acta Biochim Biophys Sin (Shanghai) ; 49(1): 25-32, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27864279

ABSTRACT

Nutrition deficiency is reported to induce apoptosis of chondrocytes and degeneration of cartilage endplate (CEP) in rabbit. Cartilage endplate stem cells (CESCs) are important for the integrity of structure and function of CEP. Bcl-2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3) has been reported to regulate apoptosis, autophagy, and cytoprotection. In this study, we aimed to determine whether nutrition deficiency induces apoptosis of CESCs, and whether or not the BNIP3-related pathway is activated in CESCs during nutrition deficiency. CESCs isolated from degenerated human CEP were cultured under normal or nutrition-deficient condition. Then, apoptosis was analyzed by flow cytometry. The expression and intracellular localization of BNIP3 were detected by quantitative real-time polymerase chain reaction, western blot analysis, and immunofluorescence assay, respectively. Mitochondrial membrane potential (MMP) and caspase-3 activity were measured by JC-1 staining and caspase-3 activity assay. Our results showed that nutrition deficiency promotes apoptosis and BNIP3 expression in CESCs. Notably, knockdown of BNIP3 could partially decrease nutrition deficiency-induced apoptosis of CESCs. In addition, nutrition deficiency could also induce upregulation of BNIP3, resulting in mitochondrial translocation of BNIP3 and loss of MMP in CESCs in a time-dependent manner. However, nutrition deficiency showed no effects on caspase-3 activity in CESCs. In summary, nutrition deficiency may promote CESC apoptosis partially through upregulating BNIP3, which might lead to activation of the BNIP3-related pathway and apoptosis of CESCs in a caspase-independent manner.


Subject(s)
Apoptosis , Caspases/metabolism , Growth Plate/cytology , Malnutrition/pathology , Membrane Proteins/metabolism , Stem Cells/cytology , Up-Regulation , Animals , Cells, Cultured , Growth Plate/enzymology , Growth Plate/metabolism , Matrix Metalloproteinases/metabolism , Membrane Proteins/genetics , Rabbits , Stem Cells/enzymology , Stem Cells/metabolism
3.
J Orthop Res ; 34(7): 1121-9, 2016 07.
Article in English | MEDLINE | ID: mdl-26694636

ABSTRACT

Yes-associated protein (YAP) and extracellular signal-regulated kinase (ERK) have been considered as key regulators in tissue homeostasis, organ development, and tumor formation. However, the roles of YAP and ERK in the mediating strain mechanosensing in the growth plate cartilage have not been determined. In this study, chondrocytes obtained from the growth plate cartilage of 2-week-old Sprague-Dawley rats were subjected to the mechanical strain with different magnitudes and durations at a frequency of 0.5 Hz. We found that YAP and ERK activation in response to mechanical strain was time and magnitude dependent. Pretreatment with a RhoA inhibitor (C3 toxin) or a microfilament cytoskeleton disrupting reagent (cytochalasin D) could suppress their activation. In addition, activated YAP and ERK were able to induce cell cycle progression by up-regulating the expression of cell cycle-related genes. These results shed new light on the function of YAP and ERK in mechanical strain-promoted growth plate development. Our results also provided evidence that RhoA and cytoskeletal dynamics are required for this mechanotransduction. © 2015 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1121-1129, 2016.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Cycle , Chondrocytes/physiology , MAP Kinase Signaling System , Mechanotransduction, Cellular , Animals , Cytoskeleton/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Growth Plate/cytology , Growth Plate/enzymology , Primary Cell Culture , Rats, Sprague-Dawley , Stress, Mechanical , YAP-Signaling Proteins , rho GTP-Binding Proteins/metabolism
4.
J Bone Miner Res ; 30(8): 1481-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25704664

ABSTRACT

Long-bone growth by endochondral ossification is cooperatively accomplished by chondrocyte proliferation, hypertrophic differentiation, and appropriate secretion of collagens, glycoproteins, and proteoglycans into the extracellular matrix (ECM). Before folding and entering the secretory pathway, ECM macromolecules in general are subject to extensive posttranslational modification, orchestrated by chaperone complexes in the endoplasmic reticulum (ER). ERp57 is a member of the protein disulfide isomerase (PDI) family and facilitates correct folding of newly synthesized glycoproteins by rearrangement of native disulfide bonds. Here, we show that ERp57-dependent PDI activity is essential for postnatal skeletal growth, especially during the pubertal growth spurt characterized by intensive matrix deposition. Loss of ERp57 in growth plates of cartilage-specific ERp57 knockout mice (ERp57 KO) results in ER stress, unfolded protein response (UPR), reduced proliferation, and accelerated apoptotic cell death of chondrocytes. Together this results in a delay of long-bone growth with the following characteristics: (1) enlarged growth plates; (2) expanded hypertrophic zones; (3) retarded osteoclast recruitment; (4) delayed remodeling of the proteoglycan-rich matrix; and (5) reduced numbers of bone trabeculae. All the growth plate and bone abnormalities, however, become attenuated after the pubertal growth spurt, when protein synthesis is decelerated and, hence, ERp57 function is less essential.


Subject(s)
Cartilage/enzymology , Chondrocytes/enzymology , Endoplasmic Reticulum Stress , Growth Plate/enzymology , Protein Disulfide-Isomerases/metabolism , Sexual Maturation , Animals , Cartilage/pathology , Chondrocytes/pathology , Growth Plate/pathology , Mice , Mice, Knockout , Protein Disulfide-Isomerases/genetics , Unfolded Protein Response
5.
Endocrinology ; 156(2): 499-510, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406016

ABSTRACT

Prostaglandins, particularly PGE2, are important to adult bone and joint health, but how prostaglandins act on growth plate cartilage to affect bone growth is unclear. We show that growth plate cartilage is distinct from articular cartilage with respect to cyclooxygenase (COX)-2 mRNA expression; although articular chondrocytes express very little COX-2, COX-2 expression is high in growth plate chondrocytes and is increased by IGF-I. In bovine primary growth plate chondrocytes, ATDC5 cells, and human metatarsal explants, inhibition of COX activity with nonsteroidal antiinflammatory drugs (NSAIDs) inhibits chondrocyte proliferation and ERK activation by IGF-I. This inhibition is reversed by prostaglandin E2 and prostacyclin (PGI2) but not by prostaglandin D2 or thromboxane B2. Inhibition of COX activity in young mice by ip injections of NSAIDs causes dwarfism. In growth plate chondrocytes, inhibition of proliferation and ERK activation by NSAIDs is reversed by forskolin, 8-bromoadenosine, 3',5'-cAMP and a prostacyclin analog, iloprost. The inhibition of proliferation and ERK activation by celecoxib is also reversed by 8CPT-2Me-cAMP, an activator of Epac, implicating the small G protein Rap1 in the pathway activated by iloprost. These results imply that prostacyclin is required for proper growth plate development and bone growth.


Subject(s)
Bone Development , Guanine Nucleotide Exchange Factors/metabolism , Prostaglandins I/physiology , rap1 GTP-Binding Proteins/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal , Cattle , Celecoxib , Cells, Cultured , Chondrocytes/enzymology , Cyclooxygenase 2/metabolism , Epoprostenol/metabolism , Female , Growth Plate/drug effects , Growth Plate/enzymology , Humans , Iloprost , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Pyrazoles , Signal Transduction , Sulfonamides
6.
Biochim Biophys Acta ; 1840(7): 2112-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24637075

ABSTRACT

BACKGROUND: Choline kinase has three isoforms encoded by the genes Chka and Chkb. Inactivation of Chka in mice results in embryonic lethality, whereas Chkb(-/-) mice display neonatal forelimb bone deformations. METHODS: To understand the mechanisms underlying the bone deformations, we compared the biology and biochemistry of bone formation from embryonic to young adult wild-type (WT) and Chkb(-/-) mice. RESULTS: The deformations are specific to the radius and ulna during the late embryonic stage. The radius and ulna of Chkb(-/-) mice display expanded hypertrophic zones, unorganized proliferative columns in their growth plates, and delayed formation of primary ossification centers. The differentiation of chondrocytes of Chkb(-/-) mice was impaired, as was chondrocyte proliferation and expression of matrix metalloproteinases 9 and 13. In chondrocytes from Chkb(-/-) mice, phosphatidylcholine was slightly lower than in WT mice whereas the amount of phosphocholine was decreased by approximately 75%. In addition, the radius and ulna from Chkb(-/-) mice contained fewer osteoclasts along the cartilage/bone interface. CONCLUSIONS: Chkb has a critical role in the normal embryogenic formation of the radius and ulna in mice. GENERAL SIGNIFICANCE: Our data indicate that choline kinase beta plays an important role in endochondral bone formation by modulating growth plate physiology.


Subject(s)
Cell Differentiation/genetics , Choline Kinase/genetics , Growth Plate/growth & development , Osteogenesis/genetics , Animals , Choline Kinase/metabolism , Chondrocytes/enzymology , Embryo, Mammalian/enzymology , Embryonic Development/genetics , Forelimb/embryology , Forelimb/enzymology , Forelimb/growth & development , Growth Plate/enzymology , Humans , Mice , Mice, Knockout , Phosphatidylcholines/metabolism
8.
Biomed Res ; 34(3): 119-28, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23782745

ABSTRACT

We have histologically examined vascular invasion and calcification of the hypertrophic zone during endochondral ossification in matrix metalloproteinase (MMP)-9 deficient (MMP-9-/-) mice and in their littermates at 3 days, 3 weeks and 6 weeks after birth. Capillaries and osteoclasts at the chondro-osseous junction showed an intense MMP-9 immunopositivity, suggesting that they recognize chemical properties of cartilaginous matrices, and then release MMP-9 for cartilage degradation. CD31-positive capillaries and tartrate-resistant acid phosphatase-reactive osteoclasts could be found in the close proximity in the region of chondro-osseous junction in MMP-9-/- mice, while in wild-type mice, vascular invasion preceded osteoclastic migration into the epiphyseal cartilage. Although MMP-9-/- mice revealed larger hypertrophic zones, the index of calcified area was significantly smaller in MMP-9-/- mice. Interestingly, the lower layer of the MMP-9-/- hypertrophic zone showed intense MMP-13 staining, which could not be observed in wild-type mice. This indicates that MMP-13 may compensate for MMP-9 deficiency at that specific region, but not to a point at which the deficiency could be completely rescued. In conclusion, it seems that MMP-9 is the optimal enzyme for cartilage degradation during endochondral ossification by controlling vascular invasion and subsequent osteoclastic migration.


Subject(s)
Endothelial Cells/cytology , Growth Plate/blood supply , Growth Plate/cytology , Matrix Metalloproteinase 9/genetics , Osteoclasts/cytology , Osteogenesis , Animals , Animals, Newborn , Calcification, Physiologic , Cell Movement , Chondrocytes/cytology , Chondrocytes/enzymology , Endothelial Cells/enzymology , Gene Expression , Growth Plate/enzymology , Growth Plate/growth & development , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/deficiency , Mice , Mice, Knockout , Neovascularization, Physiologic , Osteoclasts/enzymology
9.
Injury ; 44(7): 914-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23427856

ABSTRACT

INTRODUCTION: Injured growth plate cartilage is often repaired by bony tissue, causing bone growth defects in children. Currently, mechanisms for the undesirable repair remain unclear and there are no biological treatments available to prevent the associated bone growth defects. Osterix is known as a vital transcription factor for osteoblast differentiation which is critical for normal bone formation and bone repair, and osterix is known to be regulated by protein kinase-D; however it is unknown whether protein kinase-D-osterix signalling plays any roles in the bony repair of injured growth plate. METHODS: Using a rat model, this study investigated potential roles of protein kinase-D (PKD) in regulating expression of osteogenic transcription factor osterix and the growth plate bony repair. 4 days post injury at the proximal tibial growth plate, rats received four once-daily injections of vehicle or 2.35 mg/kg gö6976 (a PKD inhibitor), and growth plate tissues collected at day 10 were examined histologically and molecularly. In addition, effects of PKD inhibition on osteogenic and chondrogenic differentiation were examined in vitro using rat bone marrow mesenchymal stromal cells. RESULTS: Compared to vehicle control, PKD inhibition caused a decrease in bone volume (p<0.05), an increase in % of mesenchymal tissue (p<0.01), and an increase in cartilaginous tissue within the injury site. Consistently, gö6976 treatment tended to decrease expression of bone-related genes (osterix, osteocalcin) and increase levels of cartilage-related genes (Sox9, collagen-2a, collagen-10a1). In support, in vitro experiments showed that gö6976 presence in the primary rat marrow stromal cell culture resulted in a decrease of alkaline phosphatase(+) CFU-f colonies formed (p<0.05) and an increase in collagen-2a expression in chondrogenic pellet culture (p<0.05). CONCLUSION: These studies suggest that PKD is important for growth plate bony repair and its inhibition after growth plate injury may result in less bone formation and potentially more cartilage repair.


Subject(s)
Cartilage/injuries , Cartilage/metabolism , Growth Plate/enzymology , Protein Kinase C/biosynthesis , Salter-Harris Fractures , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cartilage/pathology , Disease Models, Animal , Gene Expression , Growth Plate/drug effects , Growth Plate/pathology , Male , Osteoblasts , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism
10.
J Biol Chem ; 286(33): 29227-29240, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21652717

ABSTRACT

Site-1 protease (S1P) is a proprotein convertase with essential functions in lipid homeostasis and unfolded protein response pathways. We previously studied a mouse model of cartilage-specific knock-out of S1P in chondroprogenitor cells. These mice exhibited a defective cartilage matrix devoid of type II collagen protein (Col II) and displayed chondrodysplasia with no endochondral bone formation even though the molecular program for endochondral bone development appeared intact. To gain insights into S1P function, we generated and studied a mouse model in which S1P is ablated in postnatal chondrocytes. Postnatal ablation of S1P results in chondrodysplasia. However, unlike early embryonic ablations, the growth plates of these mice exhibit a lack of Ihh, PTHrP-R, and Col10 expression indicating a loss of chondrocyte hypertrophic differentiation and thus disruption of the molecular program required for endochondral bone development. S1P ablation results in rapid growth plate disruption due to intracellular Col II entrapment concomitant with loss of chondrocyte hypertrophy suggesting that these two processes are related. Entrapment of Col II in the chondrocytes of the prospective secondary ossification center precludes its development. Trabecular bone formation is dramatically diminished in the primary spongiosa and is eventually lost. The primary growth plate is eradicated by apoptosis but is gradually replaced by a fully functional new growth plate from progenitor stem cells capable of supporting new bone growth. Our study thus demonstrates that S1P has fundamental roles in the preservation of postnatal growth plate through chondrocyte differentiation and Col II deposition and functions to couple growth plate maturation to trabecular bone development in growing mice.


Subject(s)
Cell Differentiation , Chondrocytes/enzymology , Growth Plate/enzymology , Osteochondrodysplasias/enzymology , Osteogenesis , Proprotein Convertases/biosynthesis , Serine Endopeptidases/biosynthesis , Stem Cells/enzymology , Animals , Chondrocytes/pathology , Collagen Type II/genetics , Collagen Type II/metabolism , Growth Plate/pathology , Mice , Mice, Knockout , Organ Specificity/genetics , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Proprotein Convertases/genetics , Serine Endopeptidases/genetics , Stem Cells/pathology
11.
J Biol Chem ; 286(7): 5803-12, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21148564

ABSTRACT

Chondroitin sulfate (CS) is a glycosaminoglycan, consisting of repeating disaccharide units of N-acetylgalactosamine and glucuronic acid residues, and plays important roles in development and homeostasis of organs and tissues. Here, we generated and analyzed mice lacking chondroitin sulfate N-acetylgalactosaminyltransferase 1 (CSGalNAcT-1). Csgalnact1(-/-) mice were viable and fertile but exhibited slight dwarfism. Biochemically, the level of CS in Csgalnact1(-/-) cartilage was reduced to ∼50% that of wild-type cartilage, whereas its chain length was similar to wild-type mice, indicating that CSGalNAcT-1 participates in the CS chain initiation as suggested in the previous study (Sakai, K., Kimata, K., Sato, T., Gotoh, M., Narimatsu, H., Shinomiya, K., and Watanabe, H. (2007) J. Biol. Chem. 282, 4152-4161). Histologically, the growth plate of Csgalnact1(-/-) mice contained shorter and slightly disorganized chondrocyte columns with a reduced volume of the extracellular matrix principally in the proliferative layer. Immunohistochemical analysis revealed that the level of both aggrecan and link protein 1 were decreased in Csgalnact1(-/-) cartilage. Western blot analysis demonstrated an increase in processed forms of aggrecan core protein. These results suggest that CSGalNAcT-1 is required for normal levels of CS biosynthesis in cartilage. Our observations suggest that CSGalNAcT-1 is necessary for normal levels of endochondral ossification, and the decrease in CS amount in the growth plate by its absence causes a rapid catabolism of aggrecan.


Subject(s)
Aggrecans/metabolism , Cartilage/enzymology , Chondrocytes/enzymology , Chondroitin Sulfates/biosynthesis , Growth Plate/enzymology , N-Acetylgalactosaminyltransferases/metabolism , Osteogenesis/physiology , Aggrecans/genetics , Animals , Cartilage/metabolism , Chondrocytes/cytology , Chondroitin Sulfates/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Growth Plate/cytology , Mice , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , Proteoglycans/genetics , Proteoglycans/metabolism
12.
Development ; 138(2): 359-70, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21177348

ABSTRACT

For tissues that develop throughout embryogenesis and into postnatal life, the generation of differentiated cells to promote tissue growth is at odds with the requirement to maintain the stem cell/progenitor cell population to preserve future growth potential. In the growth plate cartilage, this balance is achieved in part by establishing a proliferative phase that amplifies the number of progenitor cells prior to terminal differentiation into hypertrophic chondrocytes. Here, we show that endogenous calcium/calmodulin-dependent protein kinase II (CamkII, also known as Camk2) activity is upregulated prior to hypertrophy and that loss of CamkII function substantially blocks the transition from proliferation to hypertrophy. Wnt signaling and Pthrp-induced phosphatase activity negatively regulate CamkII activity. Release of this repression results in activation of multiple effector pathways, including Runx2- and ß-catenin-dependent pathways. We present an integrated model for the regulation of proliferation potential by CamkII activity that has important implications for studies of growth control and adult progenitor/stem cell populations.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Chondrocytes/cytology , Chondrocytes/enzymology , Growth Plate/embryology , Growth Plate/enzymology , Animals , Animals, Genetically Modified , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Enlargement , Cell Proliferation , Chick Embryo , Core Binding Factor Alpha 1 Subunit/metabolism , DNA Primers/genetics , Frizzled Receptors/metabolism , Growth Plate/cytology , Mice , Mice, Mutant Strains , Models, Biological , Parathyroid Hormone-Related Protein/metabolism , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
13.
Biochem J ; 432(1): 47-55, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20812917

ABSTRACT

CS (chondroitin sulfate) is a glycosaminoglycan species that is widely distributed in the extracellular matrix. To understand the physiological roles of enzymes involved in CS synthesis, we produced CSGalNAcT1 (CS N-acetylgalactosaminyltransferase 1)-null mice. CS production was reduced by approximately half in CSGalNAcT1-null mice, and the amount of short-chain CS was also reduced. Moreover, the cartilage of the null mice was significantly smaller than that of wild-type mice. Additionally, type-II collagen fibres in developing cartilage were abnormally aggregated and disarranged in the homozygous mutant mice. These results suggest that CSGalNAcT1 is required for normal CS production in developing cartilage.


Subject(s)
Chondrogenesis , Chondroitin Sulfates/biosynthesis , Growth Plate/enzymology , N-Acetylgalactosaminyltransferases/metabolism , Animals , Blotting, Western , Cell Line , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/enzymology , Chondrocytes/metabolism , Collagen Type II/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Growth Plate/embryology , Growth Plate/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , Proliferating Cell Nuclear Antigen/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Dis Model Mech ; 3(3-4): 224-35, 2010.
Article in English | MEDLINE | ID: mdl-20142327

ABSTRACT

Long bone development depends on endochondral bone formation, a complex process requiring exquisite balance between hypertrophic cartilage (HC) formation and its ossification. Dysregulation of this process may result in skeletal dysplasias and heterotopic ossification. Endochondral ossification requires the precise orchestration of HC vascularization, extracellular matrix remodeling, and the recruitment of osteoclasts and osteoblasts. Matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF) and osteoclasts have all been shown to regulate endochondral ossification, but how their function interrelates is not known. We have investigated the functional relationship among these regulators of endochondral ossification, demonstrating that they have complementary but non-overlapping functions. MMP-9, VEGF and osteoclast deficiency all cause impaired growth plate ossification resulting in the accumulation of HC. VEGF mRNA and protein expression are increased at the MMP-9-/- growth plate, and VEGF activity contributes to endochondral ossification since sequestration of VEGF by soluble receptors results in further inhibition of growth plate vascularization and ossification. However, VEGF bioavailability is still limited in MMP-9 deficiency, as exogenous VEGF is able to rescue the MMP-9-/- phenotype, demonstrating that MMP-9 may partially, but not fully, regulate VEGF bioavailability. The organization of the HC extracellular matrix at the MMP-9-/- growth plate is altered, supporting a role for MMP-9 in HC remodeling. Inhibition of VEGF impairs osteoclast recruitment, whereas MMP-9 deficiency leads to an accumulation of osteoclasts at the chondro-osseous junction. Growth plate ossification in osteoclast-deficient mice is impaired in the presence of normal MMP-9 expression, indicating that other osteoclastic functions are also necessary. Our data delineate the complementary interplay between MMP-9, VEGF and osteoclast function that is necessary for normal endochondral bone formation and provide a molecular framework for investigating the molecular defects contributing to disorders of endochondral bone formation.


Subject(s)
Chondrocytes/pathology , Matrix Metalloproteinase 9/metabolism , Osteoclasts/enzymology , Osteoclasts/pathology , Osteogenesis , Vascular Endothelial Growth Factor A/metabolism , Acid Phosphatase/metabolism , Animals , Animals, Newborn , Bone and Bones/drug effects , Bone and Bones/enzymology , Bone and Bones/pathology , Bone and Bones/ultrastructure , Cartilage/pathology , Cartilage/ultrastructure , Chondrocytes/drug effects , Chondrocytes/enzymology , Chondrocytes/ultrastructure , Collagen Type II/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Growth Plate/drug effects , Growth Plate/enzymology , Growth Plate/pathology , Growth Plate/ultrastructure , Humans , Hypertrophy , Isoenzymes/metabolism , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase Inhibitors , Mice , Models, Biological , Osteoclasts/drug effects , Osteoclasts/ultrastructure , Osteogenesis/drug effects , Phenotype , Protease Inhibitors/pharmacology , Tartrate-Resistant Acid Phosphatase , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/pharmacology
15.
PLoS One ; 5(1): e8866, 2010 Jan 25.
Article in English | MEDLINE | ID: mdl-20111593

ABSTRACT

BACKGROUND: Endochondral ossification, the process through which long bones are formed, involves chondrocyte proliferation and hypertrophic differentiation in the cartilage growth plate. In a previous publication we showed that pharmacological inhibition of the PI3K signaling pathway results in reduced endochondral bone growth, and in particular, shortening of the hypertrophic zone in a tibia organ culture system. In this current study we aimed to investigate targets of the PI3K signaling pathway in hypertrophic chondrocytes. METHODOLOGY/PRINCIPAL FINDINGS: Through the intersection of two different microarray analyses methods (classical single gene analysis and GSEA) and two different chondrocyte differentiation systems (primary chondrocytes treated with a pharmacological inhibitor of PI3K and microdissected growth plates), we were able to identify a high number of genes grouped in GSEA functional categories regulated by the PI3K signaling pathway. Genes such as Phlda2 and F13a1 were down-regulated upon PI3K inhibition and showed increased expression in the hypertrophic zone compared to the proliferative/resting zone of the growth plate. In contrast, other genes including Nr4a1 and Adamts5 were up-regulated upon PI3K inhibition and showed reduced expression in the hypertrophic zone. Regulation of these genes by PI3K signaling was confirmed by quantitative RT-PCR. We focused on F13a1 as an interesting target because of its known role in chondrocyte hypertrophy and osteoarthritis. Mouse E15.5 tibiae cultured with LY294002 (PI3K inhibitor) for 6 days showed decreased expression of factor XIIIa in the hypertrophic zone compared to control cultures. CONCLUSIONS/SIGNIFICANCE: Discovering targets of signaling pathways in hypertrophic chondrocytes could lead to targeted therapy in osteoarthritis and a better understanding of the cartilage environment for tissue engineering.


Subject(s)
Chondrocytes/enzymology , Gene Expression Regulation, Developmental , Growth Plate/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Blotting, Western , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Chromones/pharmacology , Down-Regulation/drug effects , Growth Plate/cytology , Mice , Morpholines/pharmacology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Up-Regulation/drug effects
16.
Pediatr Nephrol ; 25(4): 633-42, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19830459

ABSTRACT

The goal of the study is to examine the relationship between the sensor molecules, Hypoxia Inducible Factor-1 (HIF-1), AMP activated Protein Kinase (AMPK) and mammalian Target of Rapamycin (mTOR) in chondrocyte survival and autophagy. We showed that chondrocytes expressed the energy sensor AMPK-1 and that activation increased with maturation. In addition, we showed that thapsigargin treatment activated AMPK and autophagy in a HIF-1-dependent manner. Using serum-starved AMPK-silenced cells, we demonstrated that AMPK was required for the induction of the autophagic response. We also noted a change in chondrocyte sensitivity to apoptogens, due to activation of caspase-8 and cleavage and activation of the pro-apoptotic protein, BID. To test the hypothesis that AMPK signaling directly promoted autophagy, we inhibited AMPK activity in mTOR silenced cells and showed that while mTOR suppression induced autophagy, AMPK inhibition did not block this activity. Based on these findings, it is concluded that because of the micro-environmental changes experienced by the chondrocyte, autophagy is activated by AMPK in a HIF-1-dependent manner.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/physiology , Chondrocytes/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Autophagy/drug effects , Calcium/metabolism , Cell Line , Cell Survival , Chondrocytes/cytology , Chondrocytes/drug effects , Enzyme Inhibitors/pharmacology , Gene Silencing , Growth Plate/cytology , Growth Plate/enzymology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases , Thapsigargin/pharmacology
17.
J Nutr Biochem ; 21(4): 297-303, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19369052

ABSTRACT

The nutritional influence of zinc on markers of bone extracellular matrix resorption and mineralization was investigated in growing rats. Thirty male weanling rats were randomly assigned to consume AIN-93G based diets containing 2.5, 5, 7.5, 15 or 30 microg Zn/g diet for 24 days. Femur zinc increased substantially as zinc increased from 5 to 15 microg/g diet and modestly between 15 and 30 microg/g (P<.05). By morphological assessment, trabecular bone increased steadily as dietary zinc increased to 30 microg/g. Increasing dietary zinc tended to decrease Zip2 expression nonsignificantly and elevated the relative expression of metallothionen-I at 15 but not 30 microg Zn/g diet. Femur osteoclastic resorption potential, indicated by matrix metalloproteinases (MMP-2 and MMP-9) and carbonic anhydrase-2 activities decreased with increasing dietary zinc. In contrast to indicators of extracellular matrix resorption, femur tartrate-resistant acid and alkaline phosphatase activities increased fourfold as dietary zinc increased from 2.5 to 30 microg Zn/g. Likewise, 15 or 30 microg Zn/g diet resulted in maximum relative expression of osteocalcin, without influencing expression of core-binding factor alpha-1, collagen Type 1 alpha-1, or nuclear factor of activated T cells c1. In conclusion, increased trabecular bone with additional zinc suggests that previous requirement estimates of 15 microg Zn/g diet may not meet nutritional needs for optimal bone development. Overall, the up-regulation of extracellular matrix modeling indexes and concomitant decrease in resorption activities as dietary zinc increased from 2.5 to 30 microg/g provide evidence of one or more physiological roles for zinc in modulating the balance between bone formation and resorption.


Subject(s)
Bone Development/physiology , Bone Matrix/physiology , Bone Resorption/prevention & control , Calcification, Physiologic/physiology , Osteoclasts/cytology , Osteoclasts/physiology , Zinc/administration & dosage , Animals , Biomarkers , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Differentiation , Collagen Type I, alpha 1 Chain , Diet , Femur/chemistry , Femur/growth & development , Femur/metabolism , Gene Expression Regulation , Growth Plate/anatomy & histology , Growth Plate/enzymology , Growth Plate/growth & development , Male , Metallothionein/genetics , Metallothionein/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , RNA, Messenger/metabolism , Random Allocation , Rats , Tibia/anatomy & histology , Tibia/growth & development , Zinc/analysis , Zinc/physiology
18.
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
19.
J Anim Sci ; 87(11): 3544-55, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19648491

ABSTRACT

Tibial dyschondroplasia (TD) is a prevalent skeletal abnormality associated with rapid growth rate in many avian species. It is characterized by the presence of a nonvascularized, nonmineralized lesion that extends from the epiphyseal growth plate into the metaphysis of the proximal tibiotarsal bones. In this study, we examined the expression of 4 members of the matrix metalloproteinase (MMP) family (MMP-2, -3, -9, and -13) in thiram-induced TD lesions and in the process of recovery from TD, by in situ hybridization analysis and quantitative real-time PCR. A model for the induction and recovery of TD was established, consisting of 3 groups of broilers: (1) thiram group, chicks fed a thiram-enriched diet to induce TD; (2) recovery group, chicks fed a thiram-enriched diet during the first week of the experiment and a normal diet from the second week on; and (3) control group, chicks fed a normal diet throughout the experimental period. In agreement with our previous data, the 4 MMP were diminished in the TD lesion (P < 0.05); however, in the current study we show that the growth plate was able to repair itself and that the MMP reappeared during the process of recovery from TD. Our results strengthen the link between MMP expression and growth-plate impairment, and we suggest that gelatinase activity (MMP-2 and 9) facilitates this process.


Subject(s)
Chickens/growth & development , Growth Plate/enzymology , Matrix Metalloproteinases/biosynthesis , Osteochondrodysplasias/veterinary , Poultry Diseases/enzymology , Animals , Gene Expression Regulation, Enzymologic , Growth Plate/growth & development , In Situ Hybridization , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , Mutagens , Osteochondrodysplasias/enzymology , Polymerase Chain Reaction , Thiram/pharmacology , Tibia/enzymology , Tibia/growth & development
20.
Anat Histol Embryol ; 38(2): 154-60, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19183348

ABSTRACT

A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) have been reported to play a role in the degradation of aggrecan, a major component of cartilage. This study was performed to examine the effects of alendronate on the expression of ADAMTS in developing femoral epiphyseal cartilage. Primary cultured chondrocytes from this cartilage were treated with alendronate in vitro and postnatal day 1 rats were injected subcutaneously with alendronate (1 mg/kg) every second day in vivo. The number of cultured chondrocytes and their aggrecan mRNA levels were unaffected by the alendronate treatment at 10(-6) to 10(-4) M concentrations. The mRNA levels of ADAMTS-1, -2 and -9 in chondrocytes were also unaffected. However, the levels of ADAMTS-5 and -4 were reduced significantly by the same treatment. The thickness of the proliferating chondrocyte layers and the aggrecan mRNA levels in the epiphysis were unaffected by the alendronate treatment in vivo. However, the hypertrophied chondrocyte layers became significantly thicker, and the size of the secondary ossification centre was reduced significantly by the same treatment (P < 0.05). Both ADAMTS-4 and -5 mRNA expressions were also reduced significantly in vivo. The immunoreactivity against ADAMTS-4 was seen in hypertrophied chondrocytes and reduced significantly by the alendronate treatment. These results suggested that alendronate can inhibit the degradation of aggrecan in the articular cartilage by downregulating the expression of matrix enzymes such as ADAMTS-4 and -5.


Subject(s)
Alendronate/pharmacology , Disintegrins/drug effects , Growth Plate/growth & development , Metalloproteases/drug effects , Aggrecans/metabolism , Animals , Animals, Newborn , Bone Density Conservation Agents/pharmacology , Cells, Cultured , Chondrocytes/enzymology , Disintegrins/metabolism , Gene Expression Regulation, Enzymologic , Growth Plate/enzymology , Metalloendopeptidases/metabolism , Metalloproteases/metabolism , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Thrombospondins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...