Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biomolecules ; 14(2)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38397376

ABSTRACT

Zebrafish are now widely used to study skeletal development and bone-related diseases. To that end, understanding osteoblast differentiation and function, the expression of essential transcription factors, signaling molecules, and extracellular matrix proteins is crucial. We isolated Sp7-expressing osteoblasts from 4-day-old larvae using a fluorescent reporter. We identified two distinct subpopulations and characterized their specific transcriptome as well as their structural, regulatory, and signaling profile. Based on their differential expression in these subpopulations, we generated mutants for the extracellular matrix protein genes col10a1a and fbln1 to study their functions. The col10a1a-/- mutant larvae display reduced chondrocranium size and decreased bone mineralization, while in adults a reduced vertebral thickness and tissue mineral density, and fusion of the caudal fin vertebrae were observed. In contrast, fbln1-/- mutants showed an increased mineralization of cranial elements and a reduced ceratohyal angle in larvae, while in adults a significantly increased vertebral centra thickness, length, volume, surface area, and tissue mineral density was observed. In addition, absence of the opercle specifically on the right side was observed. Transcriptomic analysis reveals up-regulation of genes involved in collagen biosynthesis and down-regulation of Fgf8 signaling in fbln1-/- mutants. Taken together, our results highlight the importance of bone extracellular matrix protein genes col10a1a and fbln1 in skeletal development and homeostasis.


Subject(s)
Collagen Type X , Extracellular Matrix Proteins , Osteoblasts , Zebrafish , Animals , Cell Differentiation , Extracellular Matrix/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Homeostasis/genetics , Minerals/metabolism , Osteoblasts/metabolism , Transcriptome/genetics , Zebrafish/genetics , Zebrafish/growth & development , Collagen Type X/genetics , Collagen Type X/physiology
2.
Osteoarthritis Cartilage ; 23(8): 1307-15, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25891750

ABSTRACT

OBJECTIVE: In healthy joints, a zone of calcified cartilage (ZCC) provides the mechanical integration between articular cartilage and subchondral bone. Recapitulation of this architectural feature should serve to resist the constant shear force from the movement of the joint and prevent the delamination of tissue-engineered cartilage. Previous approaches to create the ZCC at the cartilage-substrate interface have relied on strategic use of exogenous scaffolds and adhesives, which are susceptible to failure by degradation and wear. In contrast, we report a successful scaffold-free engineering of ZCC to integrate tissue-engineered cartilage and a porous biodegradable bone substitute, using sheep bone marrow stromal cells (BMSCs) as the cell source for both cartilaginous zones. DESIGN: BMSCs were predifferentiated to chondrocytes, harvested and then grown on a porous calcium polyphosphate substrate in the presence of triiodothyronine (T3). T3 was withdrawn, and additional predifferentiated chondrocytes were placed on top of the construct and grown for 21 days. RESULTS: This protocol yielded two distinct zones: hyaline cartilage that accumulated proteoglycans and collagen type II, and calcified cartilage adjacent to the substrate that additionally accumulated mineral and collagen type X. Constructs with the calcified interface had comparable compressive strength to native sheep osteochondral tissue and higher interfacial shear strength compared to control without a calcified zone. CONCLUSION: This protocol improves on the existing scaffold-free approaches to cartilage tissue engineering by incorporating a calcified zone. Since this protocol employs no xenogeneic material, it will be appropriate for use in preclinical large-animal studies.


Subject(s)
Bone Marrow Cells/cytology , Calcification, Physiologic/physiology , Hyaline Cartilage/physiology , Stromal Cells/cytology , Tissue Engineering/methods , Animals , Cell Culture Techniques/methods , Cell Differentiation , Collagen Type II/physiology , Collagen Type X/physiology , Proteoglycans/physiology , Sheep , Triiodothyronine/pharmacology
3.
Osteoarthritis Cartilage ; 21(2): 269-78, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23159952

ABSTRACT

OBJECTIVE: Increasing evidence points to a strong genetic component to osteoarthritis (OA) and that certain changes that occur in osteoarthritic cartilage recapitulate the developmental process of endochondral ossification. As zebrafish are a well validated model for genetic studies and developmental biology, our objective was to establish the spatiotemporal expression pattern of a number of OA susceptibility genes in the larval zebrafish providing a platform for functional studies into the role of these genes in OA. DESIGN: We identified the zebrafish homologues for Mcf2l, Gdf5, PthrP/Pthlh, Col9a2, and Col10a1 from the Ensembl genome browser. Labelled probes were generated for these genes and in situ hybridisations were performed on wild type zebrafish larvae. In addition, we generated transgenic reporter lines by modification of bacterial artificial chromosomes (BACs) containing full length promoters for col2a1 and col10a1. RESULTS: For the first time, we show the spatiotemporal expression pattern of Mcf2l. Furthermore, we show that all six putative OA genes are dynamically expressed during zebrafish larval development, and that all are expressed in the developing skeletal system. Furthermore, we demonstrate that the transgenic reporters we have generated for col2a1 and col10a1 can be used to visualise chondrocyte hypertrophy in vivo. CONCLUSION: In this study we describe the expression pattern of six OA susceptibility genes in zebrafish larvae and the generation of two new transgenic lines marking chondrocytes at different stages of maturation. Moreover, the tools used demonstrate the utility of the zebrafish model for functional studies on genes identified as playing a role in OA.


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Developmental/physiology , Genetic Predisposition to Disease/genetics , Osteoarthritis/genetics , Osteoarthritis/physiopathology , Zebrafish/genetics , Zebrafish/physiology , Animals , Animals, Genetically Modified , Chondrocytes/pathology , Chromosomes, Artificial, Bacterial/genetics , Collagen Type II/genetics , Collagen Type II/metabolism , Collagen Type II/physiology , Collagen Type IX/genetics , Collagen Type IX/metabolism , Collagen Type IX/physiology , Collagen Type X/genetics , Collagen Type X/metabolism , Collagen Type X/physiology , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/physiology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/physiology , Hypertrophy/genetics , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/physiology , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology
4.
Dev Cell ; 22(3): 597-609, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22421045

ABSTRACT

The transcription factor Sox9 is necessary for early chondrogenesis, but its subsequent roles in the cartilage growth plate, a highly specialized structure that drives skeletal growth and endochondral ossification, remain unclear. Using a doxycycline-inducible Cre transgene and Sox9 conditional null alleles in the mouse, we show that Sox9 is required to maintain chondrocyte columnar proliferation and generate cell hypertrophy, two key features of functional growth plates. Sox9 keeps Runx2 expression and ß-catenin signaling in check and thereby inhibits not only progression from proliferation to prehypertrophy, but also subsequent acquisition of an osteoblastic phenotype. Sox9 protein outlives Sox9 RNA in upper hypertrophic chondrocytes, where it contributes with Mef2c to directly activate the major marker of these cells, Col10a1. These findings thus reveal that Sox9 remains a central determinant of the lineage fate and multistep differentiation program of growth plate chondrocytes and thereby illuminate our understanding of key molecular mechanisms underlying skeletogenesis.


Subject(s)
Cell Differentiation , Chondrocytes/physiology , Growth Plate/physiology , Osteoblasts/physiology , SOX9 Transcription Factor/physiology , Animals , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen Type X/metabolism , Collagen Type X/physiology , Core Binding Factor Alpha 1 Subunit/biosynthesis , Female , Growth Plate/growth & development , Growth Plate/metabolism , MEF2 Transcription Factors , Male , Mice , Mice, Transgenic , Myogenic Regulatory Factors/metabolism , Myogenic Regulatory Factors/physiology , Osteoblasts/metabolism , Osteogenesis/physiology , SOX9 Transcription Factor/metabolism , beta Catenin/metabolism , beta Catenin/physiology
5.
Ann N Y Acad Sci ; 1237: 79-87, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22082369

ABSTRACT

The collagen X transgenic and null (ColX-Tg/KO) mice have revealed a link between endochondral ossification (EO) and hematopoiesis, and thus serve as model systems to study hematopoietic niches. The altered collagen X function in ColX-Tg/KO mice resulted not only in skeletal defects, which included changes in growth plate ultrastructure, altered localization of heparan sulfate proteoglycans (HSPG), and reduced trabecular bone, but also in hematopoietic defects, which included reduced B lymphocyte numbers throughout life without associated increases in B cell apoptosis. Consequently, the ColX-Tg/KO mice exhibited diminished in vitro and in vivo immune responses. Moreover, reduced expression of several hematopoietic and B lymphopoietic cytokines were measured from ColX-KO-derived hypertrophic chondrocyte and trabecular osteoblast cultures. Together, these data expand the current hematopoietic niche model by including the EO-derived extracellular matrix, for example, the collagen X/HSPG network, as well as the EO-derived hypertrophic chondrocytes and trabecular osteoblasts as hematopoietic signal mediating cells.


Subject(s)
Chondrocytes/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Lymphopoiesis , Osteogenesis , Animals , Collagen Type X/physiology , Extracellular Matrix/genetics , Heparan Sulfate Proteoglycans/physiology , Humans , Lymphopoiesis/physiology , Osteogenesis/physiology
6.
Nat Med ; 16(6): 678-86, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20495570

ABSTRACT

Chondrocyte hypertrophy followed by cartilage matrix degradation and vascular invasion, characterized by expression of type X collagen (COL10A1), matrix metalloproteinase-13 (MMP-13) and vascular endothelial growth factor (VEGF), respectively, are central steps of endochondral ossification during normal skeletal growth and osteoarthritis development. A COL10A1 promoter assay identified hypoxia-inducible factor-2alpha (HIF-2alpha, encoded by EPAS1) as the most potent transactivator of COL10A1. HIF-2alpha enhanced promoter activities of COL10A1, MMP13 and VEGFA through specific binding to the respective hypoxia-responsive elements. HIF-2alpha, independently of oxygen-dependent hydroxylation, was essential for endochondral ossification of cultured chondrocytes and embryonic skeletal growth in mice. HIF-2alpha expression was higher in osteoarthritic cartilages versus nondiseased cartilages of mice and humans. Epas1-heterozygous deficient mice showed resistance to osteoarthritis development, and a functional single nucleotide polymorphism (SNP) in the human EPAS1 gene was associated with knee osteoarthritis in a Japanese population. The EPAS1 promoter assay identified RELA, a nuclear factor-kappaB (NF-kappaB) family member, as a potent inducer of HIF-2alpha expression. Hence, HIF-2alpha is a central transactivator that targets several crucial genes for endochondral ossification and may represent a therapeutic target for osteoarthritis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Bone Development/genetics , Chondrocytes/physiology , Osteoarthritis/genetics , Trans-Activators/genetics , Transcription, Genetic/genetics , Aged , Aged, 80 and over , Animals , Bone Development/physiology , Case-Control Studies , Cells, Cultured , Collagen Type X/genetics , Collagen Type X/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Humans , Mice , Mice, Mutant Strains , Middle Aged , Osteoarthritis/physiopathology , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/physiopathology , Osteogenesis/genetics , Osteogenesis/physiology , Trans-Activators/physiology , Transcription, Genetic/physiology
7.
J Cell Physiol ; 208(1): 77-86, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16575901

ABSTRACT

We investigated the molecular mechanisms underlying canonical Wnt-mediated regulation of chondrocyte hypertrophy using chick upper sternal chondrocytes. Replication competent avian sarcoma (RCAS) viral over-expression of Wnt8c and Wnt9a, upregulated type X collagen (col10a1) and Runx2 mRNA expression thereby inducing chondrocyte hypertrophy. Wnt8c and Wnt9a strongly inhibited mRNA levels of Sox9 and type II collagen (col2a1). Wnt8c further enhanced canonical bone morphogenetic proteins (BMP-2)-induced expression of Runx2 and col10a1 while Wnt8c and Wnt9a inhibited TGF-beta-induced expression of Sox9 and col2a1. Over-expression of beta-catenin mimics the effect of Wnt8c and Wnt9a by upregulating Runx2, col10a1, and alkaline phosphatase (AP) mRNA levels while it inhibits col2a1 transcription. Western blot analysis shows that Wnt8c and beta-catenin also induces Runx2 protein levels in chondrocytes. Thus, our results indicate that activation of the canonical beta-catenin Wnt signaling pathway induces chondrocyte hypertrophy and maturation. We further investigated the effects of beta-catenin-TCF/Lef on Runx2 promoter. Co-transfection of lymphoid enhancer factor (Lef1) and beta-catenin in chicken upper sternal chondrocytes together with deletion constructs of the Runx2 promoter shows that the proximal region spanning the first 128 base pairs of this promoter is responsible for the Wnt-mediated induction of Runx2. Mutation of the TCF/Lef binding site in the -128 fragment of the Runx2 promoter resulted in loss of its responsiveness to beta-catenin. Additionally, gel-shift assay analyses determined the DNA/protein interaction of the TCF/Lef binding sites on the Runx2 promoter. Finally, our site-directed mutagenesis data demonstrated that the Runx2 site on type X collagen promoter is required for canonical Wnt induction of col10a1. Altogether we demonstrate that Wnt/beta-catenin signaling is regulated by TGF-beta and BMP-2 in chick upper sternal chondrocytes, and mediates chondrocyte hypertrophy at least partly through activation of Runx2 which in turn may induce col10a1 expression.


Subject(s)
Chondrocytes/pathology , Core Binding Factor Alpha 1 Subunit/physiology , Gene Expression Regulation/physiology , Wnt Proteins/physiology , Alkaline Phosphatase/analysis , Alkaline Phosphatase/genetics , Alkaline Phosphatase/physiology , Animals , Avian Sarcoma Viruses/genetics , Blotting, Western , Bone Morphogenetic Proteins/analysis , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Cells, Cultured , Chick Embryo , Chondrocytes/chemistry , Chondrocytes/physiology , Collagen Type II/analysis , Collagen Type II/genetics , Collagen Type II/physiology , Collagen Type X/genetics , Collagen Type X/physiology , Core Binding Factor Alpha 1 Subunit/genetics , Electrophoretic Mobility Shift Assay , Gene Expression Regulation/genetics , High Mobility Group Proteins/analysis , High Mobility Group Proteins/genetics , High Mobility Group Proteins/physiology , Hypertrophy/pathology , Hypertrophy/physiopathology , Mutation , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor , Signal Transduction/genetics , Signal Transduction/physiology , TCF Transcription Factors/analysis , TCF Transcription Factors/genetics , TCF Transcription Factors/physiology , Transcription Factors/analysis , Transcription Factors/genetics , Transcription Factors/physiology , Transfection , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/physiology , Wnt Proteins/analysis , Wnt Proteins/genetics , beta Catenin/analysis , beta Catenin/physiology
8.
J Cell Physiol ; 208(1): 87-96, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16575902

ABSTRACT

Canonical Wnt signaling (beta-catenin/TCF) has emerged as a key regulator of skeletogenesis. In this study, chondrogenesis is examined in a mouse model in which the Wnt antagonist secreted frizzled related protein 1 (sFRP1) is non-functional and results in a high bone mass phenotype and activation through the canonical pathway of the Runx2 transcription factor that is essential for bone formation. We find during the period of rapid post-natal growth, shortened height of the growth plate and increased calcification of the hypertrophic zone (HZ) in the sFRP1-/- mouse, indicating accelerated endochondral ossification. Using mouse embryo fibroblasts (MEFs) induced into the chondrogenic lineage, increased chondrogenesis and accelerating differentiation of hypertrophic chondrocytes in the sFRP1-/- MEFs was observed compared to WT cells. The induced maturation of hypertrophic chondrocytes in sFRP1(-/-) MEFs was inversely correlated to phospho-beta-catenin levels, indicating involvement of activated canonical Wnt signaling characterized by an increased expression of collagen type 2a1 and Sox 9. However, an absence of Indian hedgehog expression which occurs in WT cells was found. SFRP1-/- cells also exhibited an early induction of collagen type 10a1. Thus, these modifications in gene expression are contributing mechanism(s) for increased chondrocyte differentiation in SFRP1-/- cells. These studies have identified sFRP1 as a critical negative regulator of Wnt signaling for the normal progression of chondrocyte differentiation. Microarray gene profiling provided additional novel insights into the regulatory factors for appropriate Wnt signaling necessary for the control of chondrocyte maturation.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/physiology , Chondrocytes/pathology , Chondrocytes/physiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Wnt Proteins/physiology , Animals , Blotting, Western , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/analysis , Bone Morphogenetic Proteins/genetics , Cell Differentiation/genetics , Cells, Cultured , Chondrocytes/chemistry , Collagen Type II/analysis , Collagen Type II/genetics , Collagen Type II/physiology , Collagen Type X/analysis , Collagen Type X/genetics , Collagen Type X/physiology , Core Binding Factor Alpha 1 Subunit/analysis , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/physiology , Fibroblasts/chemistry , Fibroblasts/pathology , Fibroblasts/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Growth Plate/chemistry , Growth Plate/pathology , Growth Plate/physiopathology , Hedgehog Proteins , High Mobility Group Proteins/analysis , High Mobility Group Proteins/genetics , High Mobility Group Proteins/physiology , Hypertrophy/pathology , Hypertrophy/physiopathology , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Knockout , Osteogenesis/genetics , Osteogenesis/physiology , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor , Signal Transduction/genetics , Trans-Activators/analysis , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/analysis
9.
Cell Signal ; 18(6): 761-70, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16386877

ABSTRACT

The C1q-globular domain (gC1qD) is a highly conserved oligomerization motif that distinguishes a superfamily of proteins, which includes circulating factors like C1q (the first component of the complement cascade) and adiponectin. The compact structure resulting from gC1qD trimerization is well known for its versatility in establishing highly specific interactions with different ligands. Among the many binding targets are a large number of extracellular membrane-associated receptors involved in cell development, apoptosis, and immunological processes. Interestingly, proteins interacting with the prototypical globular domain of C1q have been described also inside the cell where they were shown to recognize signal transducers such as G protein coupled receptors and their downstream effectors. Afterward, it was shown that variants of the gC1qD have been adopted by intracellular proteins involved in signal transduction. This review summarizes the evidence supporting the presence of the gC1qD inside the cell and explores the possibility that the domain might play novel signaling functions in this context, such as determining highly specific protein-protein interactions aimed to organize signaling complexes on the cytosolic side of cellular membranes.


Subject(s)
Complement C1q/physiology , Signal Transduction , Adiponectin/classification , Adiponectin/genetics , Adiponectin/physiology , Amino Acid Sequence , Collagen Type X/physiology , Evolution, Molecular , Humans , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Sequence Alignment
10.
Genesis ; 42(1): 33-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15830383

ABSTRACT

In order to investigate the physiological control of hypertrophic chondrocytes which present the terminally differentiated form of chondrocytes, we generated a mouse line expressing the Cre recombinase under the control of the mouse type X collagen (Col10a1) promoter. In situ hybridization analysis demonstrated the expression of Col10a1-Cre transgene in hypertrophic chondrocytes of femur at postnatal day 2 (P2). In order to test the excision activity of the Cre recombinase, the Col10a1-Cre transgenic line was crossed with the mouse strain carrying the Smad4 conditional alleles (Smad4co/co) and the reporter line ROSA26. Multiple tissue PCR of Col10a1-Cre;Smad4co/+ mice revealed the restricted Cre activity in tissues containing hypertrophic chondrocytes. LacZ staining revealed that the Cre activity was observed in the cartilage primordia of ribs at E14.5 and only detected in the lower hypertrophic region of ribs at P1. These data suggest that the Col10a1-Cre mouse line described here could be used to achieve conditional gene targeting in hypertrophic chondrocytes.


Subject(s)
Chondrocytes/physiology , Integrases/biosynthesis , Integrases/genetics , Mice, Transgenic/genetics , Animals , Cell Culture Techniques , Cell Differentiation , Chondrocytes/pathology , Collagen Type X/genetics , Collagen Type X/physiology , Female , Genes, Reporter , In Situ Hybridization , Male , Mice
11.
Orthod Craniofac Res ; 8(1): 11-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15667640

ABSTRACT

UNLABELLED: AUTHOR: Shen G Objective -This review was compiled to explore the role of type X collagen in growth, development and remodeling of articular cartilage by elucidating the linkage between the synthesis of this protein and the phenotypic changes in chondrogenesis and the onset of endochondral ossification. DESIGN: The current studies closely dedicated to elucidating the role of type X collagen incorporating into chondrogenesis and endochondral ossification of articular cartilage were assessed and analyzed to allow for obtaining the mainstream consensus on the bio-molecular mechanism with which type X collagen functions in articular cartilage. RESULTS: There are spatial and temporal correlations between synthesis of type X collagen and occurrence of endochondral ossification. The expression of type X collagen is confined within hypertrophic condrocytes and precedes the embark of endochondral bone formation. Type X collagen facilitates endochondral ossification by regulating matrix mineralization and compartmentalizing matrix components. CONCLUSION: Type X collagen is a reliable marker for new bone formation in articular cartilage. The future clinical application of this collagen in inducing or mediating endochondral ossification is perceived, e.g. the fracture healing of synovial joints and adaptive remodeling of madibular condyle.


Subject(s)
Bone Development/physiology , Cartilage, Articular/physiology , Collagen Type X/physiology , Animals , Bone Matrix/metabolism , Calcification, Physiologic/physiology , Chondrocytes/metabolism , Chondrogenesis/physiology , Collagen Type X/biosynthesis , Collagen Type X/genetics , Humans , Mandibular Condyle/metabolism
12.
J Cell Physiol ; 198(3): 428-40, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14755548

ABSTRACT

Growth plate chondrocytes integrate a multitude of growth factor signals during maturation. PTHrP inhibits maturation through stimulation of PKA/CREB signaling while the bone morphogenetic proteins (BMPs) stimulate maturation through Smad mediated signaling. In this manuscript, we show that interactions between CREB and the BMP associated Smads are promoter specific, and demonstrate for the first time the requirement of CREB signaling for Smad mediated activation of a BMP responsive region of the Smad6 promoter. The 28 base pairs (bp) BMP responsive element of the Smad6 promoter contains an 11 bp Smad binding region and an adjacent 17 bp region in which we characterize a putative CRE site. PKA/CREB gain of function enhanced BMP stimulation of this reporter, while loss of CREB function diminished transcriptional activity. In contrast, ATF-2 and AP-1 transcription factors had minimal effects. Electrophoretic mobility shift assay (EMSA) confirmed CREB binding to the Smad6 promoter element. Mutations eliminating binding resulted in loss of transcriptional activity, while mutations that maintained CREB binding had continued reporter activation by CREB and BMP-2. The Smad6 gene was similarly regulated by CREB. Dominant negative CREB reduced BMP-2 stimulated Smad6 gene transcription by 50%, but markedly increased BMP-2 mediated stimulation of colX and Ihh expression. In contrast, PTHrP which activates CREB signaling, blocked the stimulatory effect of BMP-2 on colX and Ihh, but minimally inhibited the stimulatory effect of BMP on Smad6. These findings are the first to demonstrate a cooperative association between CREB and BMP regulated Smads in cells from vertebrates and demonstrate that promoter-specific rather than generalized interactions between PKA/CREB and BMP signaling regulate gene expression in chondrocytes.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Chondrocytes/physiology , Cyclic AMP Response Element-Binding Protein/physiology , DNA-Binding Proteins/genetics , Trans-Activators/genetics , Transcriptional Activation/physiology , Transforming Growth Factor beta , Animals , Base Sequence , Bone Morphogenetic Protein 2 , Cell Differentiation/physiology , Cells, Cultured , Collagen Type X/drug effects , Collagen Type X/physiology , Cyclic AMP-Dependent Protein Kinases , DNA-Binding Proteins/drug effects , Electrophoretic Mobility Shift Assay , Hedgehog Proteins , Molecular Sequence Data , Parathyroid Hormone-Related Protein/pharmacology , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Smad6 Protein , Trans-Activators/drug effects , Trans-Activators/physiology , Transcription, Genetic/physiology
13.
Am J Pathol ; 160(6): 2019-34, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12057907

ABSTRACT

Each skeletal element where marrow develops is first defined by a hypertrophic cartilage blueprint. Through programmed tissue substitution, the cartilaginous skeletal model is replaced by trabecular bone and marrow, with accompanying longitudinal tissue growth. During this process of endochondral ossification, hypertrophic cartilage expresses a unique matrix molecule, collagen X. Previously we reported that transgenic mice with dominant interference collagen X mutations develop variable skeleto-hematopoietic abnormalities, manifested as growth plate compressions, diminished trabecular bone, and reduced lymphatic organs (Nature 1993, 365:56). Here, histology and flow cytometry reveal marrow hypoplasia and impaired hematopoiesis in all collagen X transgenic mice. A subset of mice with perinatal lethality manifested the most severe skeletal defects and a reduction of marrow hematopoiesis, highlighted by a lymphocyte decrease. Thymic reduction is accompanied by a paucity of cortical immature T cells, consistent with the marrow's inability to replenish maturing cortical lymphocytes. Diminished spleens exhibit indistinct lymphatic nodules and red pulp depletion; the latter correlates with erythrocyte-filled vascular sinusoids in marrows. All mice display reduced B cells in marrows and spleens, and elevated splenic T cells. These hematopoietic defects underscore an unforeseen link between hypertrophic cartilage, endochondral ossification, and establishment of the marrow microenvironment required for blood cell differentiation.


Subject(s)
Collagen Type X/physiology , Hematopoiesis/physiology , Osteogenesis/physiology , Animals , B-Lymphocytes , Bone Marrow/pathology , Cell Differentiation , Collagen Type X/genetics , Flow Cytometry , Hematopoiesis/genetics , Lymphocyte Count , Mice , Mice, Transgenic , Osteogenesis/genetics , Phenotype , Spleen/pathology , T-Lymphocytes , Thymus Gland/pathology
14.
Am J Pathol ; 159(6): 2257-69, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733375

ABSTRACT

Collagen X transgenic (Tg) mice displayed skeleto-hematopoietic defects in tissues derived by endochondral skeletogenesis.(1) Here we demonstrate that co-expression of the transgene product containing truncated chicken collagen X with full-length mouse collagen X in a cell-free translation system yielded chicken-mouse hybrid trimers and truncated chicken homotrimers; this indicated that the mutant could assemble with endogenous collagen X and thus had potential for dominant interference. Moreover, species-specific collagen X antibodies co-localized the transgene product with endogenous collagen X to hypertrophic cartilage in growth plates and ossification centers; proliferative chondrocytes also stained diffusely. Electron microscopy revealed a disrupted hexagonal lattice network in the hypertrophic chondrocyte pericellular matrix in Tg growth plates, as well as altered mineral deposition. Ruthenium hexamine trichloride-positive aggregates, likely glycosaminoglycans (GAGs)/proteoglycans (PGs), were also dispersed throughout the chondro-osseous junction. These defects likely resulted from transgene co-localization and dominant interference with endogenous collagen X. Moreover, altered GAG/PG distribution in growth plates of both collagen X Tg and null mice was confirmed by a paucity of staining for hyaluronan and heparan sulfate PG. A provocative hypothesis links the disruption of the collagen X pericellular network and GAG/PG decompartmentalization to the potential locus for hematopoietic failure in the collagen X mice.


Subject(s)
Chondrocytes/metabolism , Collagen Type X/physiology , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Proteoglycans/metabolism , Animals , Animals, Newborn , Antibodies, Monoclonal/immunology , Cartilage/metabolism , Cartilage/pathology , Cartilage/ultrastructure , Chickens , Collagen Type X/genetics , Collagen Type X/immunology , Gene Expression , Genotype , Growth Plate/metabolism , Growth Plate/pathology , Growth Plate/ultrastructure , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Mutation , Phenotype , Time Factors , Transgenes/genetics
15.
Osteoarthritis Cartilage ; 9 Suppl A: S160-73, 2001.
Article in English | MEDLINE | ID: mdl-11680681

ABSTRACT

OBJECTIVE: Chondrocytes in the growth plate at different stages of differentiation synthesize characteristic extracellular matrix (ECM) components. Mutations in some ECM genes result in chondrodysplasia in humans and mice. We aimed to evaluate the impact of loss- and gain-of-function mutations of ECM genes on matrix structure, gene expression and formation of the growth plate. DESIGN: We review information on the impact of deficiencies in proteoglycans, and types X and II collagens on skeletal development. Additionally, we compare the impact of a glycine904 to cysteine (G904C) mutation in the triple helical coding domain of mouse Col2a1 with two previously reported Col2a1 mutations (exon7 deletion (Del1) and G85C). The G904C Col2a1 gene was introduced as a transgene into mice. Transgenic newborn mice were examined for skeletal development. The histology of the epiphyseal cartilage and the growth plate, and the ultrastructure of chondrocytes and collagen fibrillar morphology in the ECM were studied in 18.5-day transgenic and wild-type fetuses. The distribution of the mRNAs for Col2a1, Col11a1, Col9a1, Matn1, Agc and Ihh in the growth plate of 18.5-day G904C/G904C and wild type fetuses were compared by in situ hybridization. RESULTS: Heterozygous transgenic mice harbouring five copies of the G904C Col2a1 transgene developed skeletal abnormalities and dwarfism. Homozygous G904C/G904C mice died at birth, showing cleft palate, disrupted zonation of chondrocytes and reduction of the zone of hypertrophic chondrocytes. Fewer collagen fibrils were found in ECM of the cartilage. Rough endoplasmic reticulum of the chondrocytes of G904C/+ and G904C/G904C mice was distended. In G904C/G904C mutant mice, Agc gene activity was extended to the hypertrophic zone. Expression of the other genes studied was unchanged. Calcified materials that were not found normally in the maturing and only at low abundance in the hypertrophic zones of the wild type growth plate, were present in these zones in G904C/G904C mice. Despite phenotypic similarities for the G904C and Del1 mice, reduced expression of types I, II, IX, X collagens and aggrecan were reported for the latter mutation. Changes in gene activity and matrix organization in the growth plate also accompanied deficiencies in aggrecan, perlecan and collagen II. CONCLUSIONS: The data suggest that a single amino acid alteration in collagen II could lead to skeletal abnormalities through multiple secondary effects on the synthesis and assembly of ECM components. The functional impact of mutations of ECM genes reveals that chondrodysplasia is caused not just by the formation of abnormal matrix molecules, but that the alteration of one ECM component may lead to a cascade of disruption of other gene activities in chondrocytes which collectively contribute to the pathological changes in the architecture of the growth plate.


Subject(s)
Cartilage, Articular/physiology , Extracellular Matrix Proteins/genetics , Extracellular Matrix/physiology , Mutation/genetics , Aggrecans , Animals , Chondrocytes/ultrastructure , Collagen Type I/physiology , Collagen Type I/ultrastructure , Collagen Type II/physiology , Collagen Type II/ultrastructure , Collagen Type IX/physiology , Collagen Type IX/ultrastructure , Collagen Type X/physiology , Collagen Type X/ultrastructure , Gene Expression , Growth Plate/anatomy & histology , In Situ Hybridization , Lectins, C-Type , Mice , Mice, Transgenic , Proteoglycans/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...