Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Br Dent J ; 224(6): 413-418, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29569608

ABSTRACT

Post crowns are restorations which utilise the root canal space to improve the retention and resistance form of teeth which lack coronal tooth structure. In recent years there have been significant developments in the materials, systems and evidence-base surrounding the provision of post crowns. This review aims to refresh the general dental practitioner's (GDPs) knowledge of the different factors that must be considered when placing a post crown, and how these factors can help guide the dentist in their decision to provide either a direct or indirect post and core.


Subject(s)
Post and Core Technique , Tooth, Nonvital/pathology , Dental Materials , Dental Prosthesis Design , Humans , Post and Core Technique/instrumentation , Tooth Root/pathology , Tooth, Nonvital/therapy
2.
Bone ; 42(5): 871-85, 2008 May.
Article in English | MEDLINE | ID: mdl-18314002

ABSTRACT

Connective tissue growth factor (CTGF/CCN2) is a cysteine rich, extracellular matrix protein that acts as an anabolic growth factor to regulate osteoblast differentiation and function. In osteoblasts, CTGF is induced by TGF-beta1 where it acts as a downstream mediator of TGF-beta1 induced matrix production. The molecular mechanisms that control CTGF induction by TGF-beta1 in osteoblasts are not known. To assess the role of individual Smads in mediating the induction of CTGF by TGF-beta1, we used specific Smad siRNAs to block Smad expression. These studies demonstrated that Smads 3 and 4, but not Smad 2, are required for TGF-beta1 induced CTGF promoter activity and expression in osteoblasts. Since the activation of MAPKs (Erk, Jnk and p38) by TGF-beta1 is cell type specific, we were interested in determining the role of individual MAPKs in TGF-beta1 induction of CTGF promoter activity and expression. Using dominant negative (DN) mutants for Erk, Jnk and p38, we demonstrated that the expression of DN-Erk caused a significant inhibition of TGF-beta1 induced CTGF promoter activity. In contrast, the expression of DN-p38 or DN-Jnk failed to inhibit activation of CTGF promoter activity. To confirm the vital role of Erk, we used the Erk inhibitor (PD98059) to block its activation, demonstrating that it prevented TGF-beta1 activation of the CTGF promoter and up-regulation of CTGF expression in osteoblasts. Since Src can also act as a downstream signaling effector for TGF-beta in some cell types, we determined its role in TGF-beta1 induction of CTGF in osteoblasts. Treatment of osteoblasts with a Src family kinase inhibitor, PP2, or the expression of two independent kinase-dead Src mutant constructs caused significant inhibition of TGF-beta1 induced CTGF promoter activity and expression. Additionally, blocking Src activation prevented Erk activation by TGF-beta1 demonstrating a role for Src as an upstream mediator of Erk in regulating CTGF expression in osteoblasts. To investigate the involvement of the TGF-beta1 response element (TRE) and the SMAD binding element (SBE) in CTGF induction, we cloned the rat CTGF proximal promoter (-787 to +1) containing the TRE and SBE motifs into a pGL3-Luciferase reporter construct. Using a combination of CTGF promoter deletion constructs and site-directed mutants, we demonstrated the unique requirement of both the TRE and SBE for CTGF induction by TGF-beta1 in osteoblasts. Electro-mobility shift assays using specific probes containing the TRE, SBE or both showed TGF-beta1 inducible complexes that can be ablated by mutation of the respective motif, confirming their requirement for TGF-beta1 induced CTGF promoter activity. In conclusion, these studies demonstrate that CTGF induction by TGF-beta1 in osteoblasts involves Smads 3 and 4, the Erk and Src signaling pathways, and requires both the TRE and SBE motifs in the CTGF proximal promoter.


Subject(s)
Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Osteoblasts/drug effects , Transforming Growth Factor beta1/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Connective Tissue Growth Factor , Electrophoretic Mobility Shift Assay , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Luciferases/genetics , Luciferases/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutagenesis, Site-Directed , Osteoblasts/cytology , Osteoblasts/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism
3.
Crit Rev Eukaryot Gene Expr ; 13(2-4): 205-20, 2003.
Article in English | MEDLINE | ID: mdl-14696968

ABSTRACT

Osteoactivin (OA) is more highly expressed in the bones of osteopetrotic mutant rats (op/op) than in those of their normal littermates and is the homologue of human nmb, a cDNA more highly expressed in melanoma-derived cell lines of low metastatic potential, and of mouse DC-HIL, which has been implicated in endothelial cell adhesion. The human OA gene is found on chromosome 7p15.1 and consists of 11 exons spanning 28.3 kb. Murine OA is encoded by a highly similar gene of 11 exons spanning 20.2 kb on mouse chromosome 6. Human OA uses the same transcriptional initiation site in both bone and kidney as was reported for melanoma cells. OA is expressed in primary human and mouse osteoblast cultures at all stages of differentiation, with increased levels observed concurrently with the expression of osteoblast phenotype markers. OA is also expressed in a wide variety of human and mouse tissues as determined by RT-PCR analysis. Immunohistochemical investigation of OA expression in late mouse embryonic development showed very high, cell-specific expression in the nervous system, basal layer of the skin, germinal cells of hair follicles, and in the forming nephrons of the kidney. Continuing investigation of the cell-specific expression of OA in bone as well as in other tissues will lead to a better understanding of its function in the development of these cell types.


Subject(s)
Protein Biosynthesis , Proteins/genetics , Amino Acid Sequence , Animals , Bone and Bones/metabolism , Cell Adhesion , Cell Differentiation , Cells, Cultured , Chromosome Mapping , Chromosomes, Human, Pair 6/genetics , DNA, Complementary/metabolism , Databases as Topic , Exons , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Introns , Kidney/metabolism , Membrane Glycoproteins , Mice , Models, Genetic , Molecular Sequence Data , Mutation , Osteoblasts/metabolism , Osteopetrosis , RNA, Messenger/metabolism , Radiation Hybrid Mapping , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution , Transcription, Genetic
4.
Proc Natl Acad Sci U S A ; 100(11): 6736-40, 2003 May 27.
Article in English | MEDLINE | ID: mdl-12748385

ABSTRACT

The morbidity and mortality associated with impaired/delayed fracture healing remain high. Our objective was to identify a small nonpeptidyl molecule with the ability to promote fracture healing and prevent malunions. Prostaglandin E2 (PGE2) causes significant increases in bone mass and bone strength when administered systemically or locally to the skeleton. However, due to side effects, PGE2 is an unacceptable therapeutic option for fracture healing. PGE2 mediates its tissue-specific pharmacological activity via four different G protein-coupled receptor subtypes, EP1, -2, -3, and -4. The anabolic action of PGE2 in bone has been linked to an elevated level of cAMP, thereby implicating the EP2 and/or EP4 receptor subtypes in bone formation. We identified an EP2 selective agonist, CP-533,536, which has the ability to heal canine long bone segmental and fracture model defects without the objectionable side effects of PGE2, suggesting that the EP2 receptor subtype is a major contributor to PGE2's local bone anabolic activity. The potent bone anabolic activity of CP-533,536 offers a therapeutic alternative for the treatment of fractures and bone defects in patients.


Subject(s)
Dinoprostone/agonists , Fracture Healing/drug effects , Pyridines/pharmacology , Receptors, Prostaglandin E/agonists , Animals , Bone Development , Cell Line , Dogs , Humans , Male , Pyridines/blood , Rats , Receptors, Prostaglandin E, EP2 Subtype
5.
J Cell Physiol ; 190(2): 207-17, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11807825

ABSTRACT

Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-beta (TGF-beta) gene superfamily of growth and differentiation factors. Members of the BMP family were originally cloned and characterized by their ability to induce ectopic bone formation. Of the various BMPs cloned, the bone inductive ability of BMP-7 (OP-1) and BMP-2 has been well characterized. Both BMP-7 and -2 have been shown to have clinical utility in the healing of non-union fractures. However, in spite of the various advances in BMP research, the physiological regulation of BMPs is not well understood. Here we studied the expression of BMP-7 by cloning a 4.6-kB fragment of the human BMP-7 promoter (hBMP-7p) and placing it upstream of a luciferase reporter. The promoter reporter construct was stably transfected into different cell backgrounds and its regulation by various factors was investigated. We show that retinoic acid (RA) treatment results in an upregulation of the hBMP-7p reporter activity. This regulation of the hBMP-7p was further confirmed by Northern blot, PCR, and Western blot analyses, which showed an increase in both BMP-7 mRNA and protein expression upon treatment with RA. We further show that RA specifically upregulates expression of osteocalcin via activation of BMP-7 mRNA and protein in vitro. Similarly, prostaglandin E(2) (PGE(2)) treatment increases BMP-7 mRNA and protein levels, but does not transcriptionally activate the hBMP-7p. Additionally, in vivo expression of BMP-7 in bone was increased upon PGE(2) treatment. In conclusion, RA and PGE(2) upregulate BMP-7 protein expression both in vitro and in vivo.


Subject(s)
Bone Morphogenetic Proteins/genetics , Dinoprostone/pharmacology , Gene Expression Regulation/drug effects , Transforming Growth Factor beta , Tretinoin/pharmacology , Base Sequence/genetics , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/metabolism , Cell Line , Cloning, Molecular , Humans , Molecular Sequence Data , Osteoblasts/drug effects , Osteoblasts/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Up-Regulation
6.
J Cell Biochem ; 84(1): 12-26, 2001.
Article in English | MEDLINE | ID: mdl-11746512

ABSTRACT

Osteoblast development is a complex process involving the expression of specific growth factors and regulatory proteins that control cell proliferation, differentiation, and maturation. In this study, we used the rat mutation, osteopetrosis (op), to examine differences in skeletal gene expression between mutant op and normal littermates. Total RNA isolated from long bone and calvaria was used as a template for mRNA differential display. One of many cDNAs that were selectively expressed in either normal or mutant bone was cloned and sequenced and found to share some homology to the human nmb and Pmel 17 genes. This novel cDNA was named osteoactivin. Osteoactivin has an open reading frame of 1716 bp that encodes a protein of 572 amino acids with a predicted molecular weight of 63.8 kD. Protein sequence analysis revealed the presence of a signal peptide and a cleavage site at position 23. The protein also has thirteen predicted N-linked glycosylation sites and a potential RGD integrin recognition site at position 556. Northern blot analysis confirmed that osteoactivin was 3- to 4-fold overexpressed in op versus normal bone. RT-PCR analysis showed that osteoactivin is most highly expressed in bone compared with any of the other non-osseous tissues examined. In situ hybridization analysis of osteoactivin in normal bone revealed that it is primarily expressed in osteoblasts actively engaged in bone matrix production and mineralization. In primary rat osteoblast cultures, osteoactivin showed a temporal pattern of expression being expressed at highest levels during the later stages of matrix maturation and mineralization and correlated with the expression of alkaline phosphatase and osteocalcin. Our findings show that osteoactivin expression in bone is osteoblast-specific and suggest that it may play an important role in osteoblast differentiation and matrix mineralization. Furthermore, osteoactivin overexpression in op mutant bone may be secondary to the uncoupling of bone resorption and formation resulting in abnormalities in osteoblast gene expression and function.


Subject(s)
Gene Expression , Osteoblasts/physiology , Proteins/chemistry , Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Bone and Bones/cytology , Bone and Bones/metabolism , Cloning, Molecular , DNA, Complementary/analysis , DNA, Complementary/metabolism , Humans , In Situ Hybridization , Membrane Glycoproteins , Mice , Models, Animal , Molecular Sequence Data , Rats , Sequence Homology
7.
Prostaglandins Other Lipid Mediat ; 65(4): 167-87, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11444589

ABSTRACT

Prostaglandin E2 (PGE2) is an important mediator of diverse biologic functions in many tissues and binds with high affinity to four cell surface, seven-transmembrane domain, G protein-coupled receptors (EP1-EP4). The EP4 receptor subtype has a long intracellular carboxy-terminal region and is functionally coupled to adenylate cyclase, resulting in elevated intracellular cyclic adenosine 5' monophosphate (cAMP) levels upon activation. To further study EP4 receptor subtype function, a canine kidney cDNA library was screened and three clones were isolated and sequenced. The longest clone was 3,103 bp and contained a single open reading frame of 1,476 bp, potentially encoding a protein of 492 amino acids with a predicted molecular weight of 53.4 kDa. Sequence analysis of this open reading frame reveals 89% identity to the human EP4 protein coding region at the nucleotide level and 90% identity when the putative canine and human protein sequences are compared. Northern blot analysis showed relatively high levels of canine EP4 expression in heart, lung and kidney, while Southern blot analysis of canine genomic DNA suggests the presence of a single copy gene. Following transfection of canine EP4 into CHO-KI cells, Scatchard analysis revealed a dissociation constant of 24 nM for PGE, while competition binding studies using 3H-PGE2 as ligand demonstrated specific displacement by PGE2 prostaglandin E, (PGE1), and prostaglandin A3 (PGA3). Treatment with PGE2 also resulted in increased levels of cAMP in transfected, but not in parental, CHO-KI cells. In contrast, butaprost, an EP2 selective ligand, and sulprostone, an EP1/EP3 selective ligand, did not bind to this receptor at the maximal concentration used (320 nM). To further investigate secondary signaling, the canine EP4 cDNA was truncated to produce an 1,117 bp fragment encoding a 356 amino acid protein lacking the intracellular carboxy-terminus. When transfected, this truncated cDNA produced a protein with a dissociation constant of 11 nM for PGE2 and a binding and cAMP accumulation profile similar to that of the full-length protein. Both full-length and truncated canine EP4 underwent short term PGE2-induced desensitization as shown by a lack of continuing cAMP accumulation after the initial PGE2 stimulation, suggesting no involvement of the C-terminal intracellular tail. This result is in contrast to that reported for the human EP4 receptor, where residues within the C-terminal intracellular tail were shown to mediate short term, ligand induced desensitization.


Subject(s)
Dinoprostone/metabolism , Receptors, Prostaglandin E/metabolism , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cloning, Molecular , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Dogs , Humans , Molecular Sequence Data , Prostaglandins A/metabolism , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Prostaglandin E/chemistry , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP4 Subtype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Distribution
8.
J Musculoskelet Neuronal Interact ; 2(1): 77-83, 2001 Sep.
Article in English | MEDLINE | ID: mdl-15758479

ABSTRACT

The current study was designed to compare the skeletal effects of comparable doses of rat parathyroid hormone 1-34 (rPTH) and bovine parathyroid hormone 1-34 (bPTH) in ovariectomized (OVX) rats. Female Sprague-Dawley rats were OVX or sham-operated at 6 months of age and maintained untreated for 28 days after surgery. Baseline control and OVX rats were sacrificed at the beginning of treatment. Beginning 28 days post-OVX, the remaining rats were subcutaneously injected daily with rPTH or bPTH at 0, 5, 25, or 50 microg/kg/d for 28 days. Bone area, bone mineral content (BMC), and bone mineral density (BMD) of the distal femoral metaphyses were determined ex vivo using dual energy X-ray absorptiometry. Quantitative bone histomorphometry was performed on undecalcified longitudinal sections of the proximal tibia from each rat. Baseline OVX rats exhibited osteopenia as demonstrated by their significantly reduced femoral BMD and proximal tibia cancellous bone volume compared with those of baseline sham controls. Both rPTH and bPTH restored bone in OVX rats by markedly stimulating bone formation in a dose-dependent manner. However, a difference in potency between the two forms of PTH was evident. The percentage increases of BMC, BMD, cancellous bone volume, trabecular thickness, mineralizing surface, and bone formation rate in the OVX rats treated with bPTH at 5 microg/kg/d were the same as or above those treated with rPTH at the 25 microg/kg/d dose level. A relative potency analysis showed that bPTH was approximately 4- to 6-fold relatively more potent than rPTH in increasing distal femoral BMD as well as cancellous bone volume, mineralizing surface, and bone formation rate of proximal tibial metaphyses at comparable dose levels and a given time. These results may serve as a reference for in vivo study design when rPTH or bPTH are to be the agents for studies on bone anabolism.

9.
Mol Biol Rep ; 28(4): 235-43, 2001.
Article in English | MEDLINE | ID: mdl-12153143

ABSTRACT

Parathyroid hormone (PTH) is a major mediator of calcium and phosphate metabolism through its interactions with receptors in kidney and bone. PTH binds with high affinity to PTH1 and PTH2, members of the superfamily of G protein-coupled receptors. In order to clone the canine PTH1 receptor, a canine kidney cDNA library was screened using the human PTH1 receptor cDNA and two clones were further characterized. The longest clone was 2177 bp and contained a single open reading frame of 1785 bp, potentially encoding a protein of 595 amino acids with a predicted molecular weight of 66.4 kD. This open reading frame exhibits >91% identity to the human PTH1 receptor cDNA and >95% identity when the putative canine and human protein sequences are compared. Competition binding following transfection of the canine PTH1 receptor into CHO cells demonstrated specific displacement of 125I-human PTH1-34 by canine PTH1-34, human PTH1-34, and canine/human parathyroid hormone related peptide (PTHrP) 1-34. Treatment of canine PTH1 receptor transfected cells, but not mock transfected cells, with these ligands also resulted in increased levels of intracellular cAMP. In contrast, the non-related aldosterone secretion inhibiting factor 1-35 neither bound nor activated the canine PTH1 receptor. Northern blot analysis revealed high levels of PTH1 receptor mRNA in the kidney, with much lower, but detectable, levels in aorta. heart, lung, prostate, testis, and skeletal muscle. Together, these data indicate that we have cloned the canine PTH1 receptor and that it is very similar, both in sequence and in functional characteristics, to the other known PTH1 receptors.


Subject(s)
Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive , CHO Cells , Cloning, Molecular , Cricetinae , Cyclic AMP/metabolism , Dogs , Gene Expression Profiling , Humans , Ligands , Molecular Sequence Data , Organ Specificity , Polymerase Chain Reaction , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/chemistry , Sequence Homology, Amino Acid , Transfection
10.
Mol Cell Biochem ; 226(1-2): 129-40, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11768233

ABSTRACT

Sex steroids, including testosterone, play a major role in determining peak bone mass in mammals and the subsequent loss of total bone mass with advancing age. Testosterone, and its active metabolite dihydrotestosterone (DHT), bind with high affinity to the androgen receptor (AR), a member of the nuclear hormone receptor superfamily. These receptors function as transcription factors, binding together with accessory proteins to specific DNA response elements in the promoters of androgen responsive genes. To further characterize AR function in a model species of relevance to bone and pharmaceutical research, we cloned a partial canine AR from a canine kidney cDNA library and then cloned the remaining 5' segment by PCR from canine ventral prostate cDNA. The complete sequence obtained was 3577 bp. This sequence contained a single open reading frame of 2721 bp, potentially encoding a protein of 907 amino acids with a predicted molecular weight of 98.7 kD. Sequence analysis of the protein encoded by this open reading frame reveals that the modular domains providing the DNA binding and ligand binding functions are identical to those reported for eight other mammalian ARs. Northern analysis of poly-A+ RNA from ventral prostate revealed three very low abundance transcripts of approximately 9 kb and RT-PCR analysis showed relatively high expression of AR in canine ventral prostate, testis, and kidney, with lower levels detectable in spleen, skeletal muscle, heart, and liver. Competition binding studies using 3H-DHT as ligand demonstrated specific displacement by DHT, testosterone, and the anabolic steroid stanozolol, with IC50 values of 1.3, 2.5 and 3.8 nM, respectively. Binding of DHT also resulted in the stimulation of an androgen responsive-luciferase reporter following cotransfection with the canine AR into 293 cells. Immunohistochemistry using an antibody directed to the C-terminal 19 amino acids of the human AR showed strong staining of the secretory epithelial cells in canine ventral prostate. Together, these data indicate that we have cloned the canine AR and that its functional DNA binding and ligand binding domains are absolutely conserved with those reported for eight other species.


Subject(s)
Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cell Line , Cloning, Molecular , DNA/metabolism , DNA, Complementary/metabolism , Dogs , Dose-Response Relationship, Drug , Gene Library , Humans , Immunohistochemistry , Inhibitory Concentration 50 , Kidney/metabolism , Ligands , Liver/metabolism , Luciferases/metabolism , Male , Molecular Sequence Data , Muscle, Skeletal/metabolism , Myocardium/metabolism , Prostate/metabolism , Protein Binding , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Spleen/metabolism , Testis/metabolism , Tissue Distribution , Transcriptional Activation , Transfection
11.
J Cell Biochem ; 77(1): 103-15, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10679821

ABSTRACT

The mammalian osteopetroses represent a pathogenetically diverse group of skeletal disorders characterized by excess bone mass resulting from reduced osteoclastic bone resorption. Abnormalities involving osteoblast function and skeletal development have also been reported in many forms of the disease. In this study, we used the rat mutation, osteopetrosis (op), to examine differences in skeletal gene expression between op mutants and their normal littermates. RNA isolated from calvaria and long bones was used as a template for mRNA-differential display. Sequence information for one of the many cDNA that were selectively expressed in either normal or mutant bone suggested that it is the rat homologue of connective tissue growth factor (CTGF) previously cloned in the human, mouse, and other species. A consensus sequence was assembled from overlapping 5'-RACE clones and used to confirm the rat CTGF cDNA protein coding region. Northern blot analysis confirmed that this message was highly (8- to 10-fold) over-expressed in op versus normal bone; it was also upregulated in op kidney but none of the other tissues (brain, liver, spleen, thymus) examined. In primary rat osteoblast cultures, the CTGF message exhibits a temporal pattern of expression dependent on their state of differentiation. Furthermore, CTGF expression is regulated by prostaglandin E(2), a factor known to modulate osteoblast differentiation. Since members of the CTGF family regulate the expression of specific genes, such as collagen and fibronectin, we propose that CTGF may play a previously unreported role in normal skeletal modeling/remodeling. Its dramatic over-expression in the op mutant skeleton may be secondary to the uncoupling of bone resorption and bone formation resulting in dysregulation of osteoblast gene expression and function.


Subject(s)
Bone and Bones/physiology , DNA, Complementary/genetics , Growth Substances/genetics , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins , Amino Acid Sequence , Animals , Base Sequence , Bone and Bones/embryology , Cloning, Molecular , Connective Tissue Growth Factor , DNA, Complementary/isolation & purification , Gene Expression Regulation, Developmental , Humans , Mice , Mitogens/genetics , Molecular Sequence Data , Rats , Sequence Alignment , Sequence Analysis
12.
Gene ; 237(2): 393-402, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10521663

ABSTRACT

Northern blot analysis of human placental RNA using a probe to the 5' end of the human prostaglandin E(2) (PGE(2)) EP2 receptor subtype coding region revealed the existence of a high abundance, low molecular weight transcript. To investigate the origin of this transcript, and its possible relationship to the human EP2 mRNA, we have cloned and characterized the gene encoding the human PGE(2) EP2 receptor subtype, identified transcriptional initiation and termination sites in two tissues (spleen and thymus), and determined its chromosomal localization. The human EP2 gene consists of two exons separated by a large intron, utilizes a common initiation site in both spleen and thymus at 1113 bp upstream of the translation initiation site, and has 3' transcript termini at 1140 bp and 1149 bp downstream of the translation stop site in spleen and thymus respectively. Southern and fluorescence in situ hybridization analysis demonstrated the human EP2 gene to be a single copy gene located in band 22 of the long arm of chromosome 14 (14q22). Though our initial interest in this gene was to investigate potential differential splicing of the human EP2 gene in placenta, this work demonstrates that the atypical transcript observed in placenta probably arises from a distinct, yet related, gene. Knowledge of the sequence, structure, and transcription events associated with the human EP2 gene will enable a broader understanding of its regulation and potential role in normal physiology and disease.


Subject(s)
Genes/genetics , Receptors, Prostaglandin E/genetics , Amino Acid Sequence , Base Sequence , Blotting, Northern , Blotting, Southern , Chromosome Banding , Chromosome Mapping , Chromosomes, Human, Pair 14/genetics , Cloning, Molecular , DNA/chemistry , DNA/genetics , Exons , Female , Genetic Variation , Humans , In Situ Hybridization, Fluorescence , Introns , Male , Molecular Sequence Data , Placenta/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Sequence Analysis, DNA , T-Lymphocytes/metabolism , Tissue Distribution
13.
Prostaglandins Other Lipid Mediat ; 57(2-3): 133-47, 1999 May.
Article in English | MEDLINE | ID: mdl-10410384

ABSTRACT

Prostaglandin E2 (PGE2) binds to four G-protein coupled cell surface receptors (EP1-EP4) and has been implicated as a local mediator of bone anabolism via a cyclic AMP mediated pathway following activation of the EP2 and/or EP4 receptor subtype. A canine kidney cDNA library was screened using a human EP2 probe, and a clone with an open reading frame of 1083 bp, potentially encoding a protein of 361 amino acids, was characterized. This open reading frame has 89% identity to the human EP2 cDNA at the nucleotide level and 87% identity at the predicted protein level. Scatchard analysis of a CHO cell line stably transfected with canine EP2 yielded a dissociation constant of 22 nM for PGE2. Competition binding studies, using 3H-PGE2 as ligand, demonstrated specific displacement by PGE2, Prostaglandin E1, Prostaglandin A3, and butaprost (an EP2 selective ligand), but not by ligands with selectivity for the related DP, FP, IP, or TP receptors. Specific ligand binding also resulted in increased levels of cAMP in EP2 transfected cells with no evidence of short-term, ligand-induced desensitization. Northern blot analysis revealed two transcripts of 3300 and 2400 bp in canine lung, and reverse-transcription polymerase chain reaction showed expression in all tissues examined. Southern blot analysis suggests the presence of a single-copy gene for EP2 in the dog.


Subject(s)
Receptors, Prostaglandin E/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive , Blotting, Southern , CHO Cells , Cloning, Molecular , Cricetinae , Cyclic AMP/metabolism , Dinoprostone/metabolism , Dogs , Gene Expression Regulation , Ligands , Molecular Sequence Data , RNA, Messenger/metabolism , Receptors, Prostaglandin E/biosynthesis , Receptors, Prostaglandin E/chemistry , Receptors, Prostaglandin E, EP2 Subtype , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Transfection
14.
J Biol Chem ; 273(22): 13760-7, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9593718

ABSTRACT

Members of the transforming growth factor-beta (TGF-beta) superfamily of growth and differentiation factors have been identified in a wide variety of organisms, ranging from invertebrates to mammals. Bone morphogenetic proteins (BMPs) constitute a subgroup of proteins belonging to the TGF-beta superfamily. BMPs were initially identified by their ability to induce endochondral bone formation at ectopic sites, suggesting a critical role for this family in development and regeneration of the skeleton. They are also expressed at a variety of nonskeletal sites during development, suggesting possible extraskeletal roles for these proteins. We cloned a novel member of the BMP family that is expressed at high levels in the placenta and the prostate and that we have designated as prostate-derived factor (PDF). Based on cDNA sequence analysis, the predicted PDF protein contains two cysteines in addition to the seven conserved cysteines that are the hallmark of the members of the TGF-beta superfamily. In addition, Northern blot hybridization to poly(A)+ RNA showed low levels of expression in the kidney and pancreas. We further characterized the expression of this member of the BMP family by in situ hybridization and immunohistochemistry. These results show high expression in the terminal villae of the placenta. The expression of the protein as visualized by immunohistochemistry shows an expression pattern identical to that of the message in the terminal villae of the placenta. In day 18 rat embryos, protein expression was also seen in the skin and in the cartilaginous tissue of developing skeleton. Orchidectomy and dihydrotestosterone treatment of rats revealed that PDF expression is regulated by androgens in the prostate. In addition, subcutaneous implantation of recombinant PDF induced cartilage formation and the early stages of endochondral bone formation. These data indicate that PDF has a functional relationship to the BMPs.


Subject(s)
Bone Morphogenetic Proteins/genetics , Membrane Proteins/genetics , Transforming Growth Factor beta/genetics , Amino Acid Sequence , Androgens/physiology , Animals , Base Sequence , Blotting, Northern , Cell Line , Cloning, Molecular , DNA, Complementary , Gene Expression Regulation/physiology , Growth Differentiation Factor 15 , Male , Membrane Proteins/metabolism , Mink , Molecular Sequence Data , Orchiectomy , Rats , Sequence Homology, Amino Acid , Signal Transduction
15.
Endocrinology ; 139(4): 2068-76, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9528995

ABSTRACT

We have discovered a new, nonsteroidal, potent estrogen agonist/antagonist, CP-336,156. CP-336,156 binds selectively and with high affinity to the human estrogen receptor-alpha with a half-inhibition concentration of 1.5 nM, which is similar to that seen with estradiol (4.8 nM). When given orally to immature (3-week-old) female Sprague-Dawley rats for 3 days at doses of 0.1, 1.0, 10, or 100 microg/kg x day, unlike 17alpha-ethynyl estradiol, CP-336,156 had no effect on uterine wet or dry weight. Similarly, no uterine hypertrophy was observed in aged (17-month-old) female rats treated (p.o.) with CP-336,156 at 10 or 100 microg/kg x day for 28 days. We also found that CP-336,156 decreased total serum cholesterol and fat body mass and had no effect on lean body mass in these aged female rats. In 5-month-old ovariectomized (OVX) Sprague-Dawley female rats, CP-336,156 completely prevented OVX-induced increases in body weight gain, total serum cholesterol, and serum osteocalcin at doses between 10 and 1000 microg/kg x day after 4 weeks. At these doses, CP-336,156 completely prevented OVX-induced bone loss and inhibited the increased bone turnover associated with estrogen deficiency in lumbar vertebrae, proximal tibiae, and distal femora. Similar to estrogen, CP-336,156 induced apoptosis and p53 expression with a concomitant decrease in the number of tartrate-resistant acid phosphatase-positive multinuclear cells in rat bone marrow cell cultures in vitro, suggesting that the induction of apoptosis may be a mechanism for the estrogenic activities of CP-336,156 in bone. In summary, CP-336,156 is a new, orally active, nonsteroidal, potent estrogen agonist/antagonist that has similar effects in bone as estradiol but without the uterine-stimulating effects associated with estradiol in rats.


Subject(s)
Body Composition/drug effects , Bone and Bones/drug effects , Cholesterol/blood , Estrogen Antagonists/pharmacology , Pyrrolidines/pharmacology , Tetrahydronaphthalenes/pharmacology , Uterus/drug effects , Adipose Tissue , Aging , Animals , Apoptosis , Bone Marrow Cells/drug effects , Cells, Cultured , Female , Gene Expression/drug effects , Genes, p53 , Organ Size/drug effects , Osteocalcin/blood , Ovariectomy , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Uterus/anatomy & histology , Weight Gain/drug effects
16.
J Cell Biochem ; 57(3): 557-73, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7768989

ABSTRACT

The development of implantable cardiac assist devices for prolonged circulatory support has been impeded by the problem of excessive thrombogenesis on the blood-prosthetic interface, with subsequent embolization. To overcome this obstacle, a ventricular assist device has been developed with textured blood-contacting surfaces to encourage the formation of a tightly adherent, hemocompatible, biological lining. In this study, we applied molecular biological techniques, in conjunction with conventional ultrastructural and biochemical techniques, to characterize the biological linings associated with the blood-contacting surfaces of 11 of these devices, which had been clinically implanted for durations ranging from 21 to 324 days. No clinical thromboembolic events or pump-related thromboembolism occurred. Biological linings developed on the textured surfaces composed of patches of cellular tissue intermingled with areas of compact fibrinous material. In addition, islands of collagenous tissue containing fibroblast-like cells appeared after 30 days of implantation. Many of these cells contained microfilaments with dense bodies indicative of myofibroblasts. RNA hybridization analyses demonstrated that the colonizing cells actively expressed genes encoding proteins for cell proliferation (histones), adhesion (fibronectin), cytoskeleton (actin, vimentin) and extracellular matrix (types I and III collagen). Linings, which never exceeded 150 microns in thickness, remained free of pathological calcification. Textured blood-contacting surfaces induced the formation of a thin, tightly adherent, viable lining which exhibited excellent long-term hemocompatibility.


Subject(s)
Biocompatible Materials , Blood , Gene Expression , Heart-Assist Devices , Adult , Case-Control Studies , Cell Adhesion , Evaluation Studies as Topic , Female , Humans , Male , Middle Aged , Surface Properties
17.
J Cell Biochem ; 55(2): 218-29, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8089197

ABSTRACT

Vitamin D responsive transcription of the bone-specific osteocalcin gene differs markedly in osteosarcoma cells and normal diploid osteoblasts. In osteoblasts the osteocalcin gene is transcribed, and upregulated by Vitamin D, only in post-proliferative cells, but in osteosarcoma cells expression is constitutive. This distinction in transcriptional regulation of the osteocalcin gene correlates with striking differences in the relative representation of two principal Vitamin D-dependent protein/DNA complexes designated V1 and V2 at the Vitamin D responsive element in the osteocalcin promoter. Formation of both complexes is Vitamin D dependent and they contain the Vitamin D receptor as well as an RXR related protein. Pore size exclusion and sedimentation velocity analyses suggest that the V1 and V2 complexes represent oligomeric protein assemblies (respectively, tetramers and trimers), and reflect primarily DNA-directed association of the monomeric protein components at the osteocalcin Vitamin D responsive element. UV crosslinking and methylation interference analyses of the V1 and V2 complexes at the osteocalcin Vitamin D responsive element indicate differences in protein/DNA recognition. For example, the V1 complex interacts with both steroid half-elements, whereas the V2 complex appears to recognize the proximal half-element. Our findings suggest variations in protein/protein and protein/DNA interactions of the VDR and RXR related complexes V1 and V2 at the osteocalcin Vitamin D responsive element that reflect unique properties of the osteosarcoma and normal diploid osteoblast phenotype.


Subject(s)
DNA/metabolism , Osteoblasts/metabolism , Osteocalcin/genetics , Osteosarcoma/genetics , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid , Transcription Factors , Vitamin D/pharmacology , Animals , Base Sequence , Gene Expression Regulation/drug effects , Molecular Sequence Data , Phosphorylation , Promoter Regions, Genetic , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoid X Receptors , Transcription, Genetic/drug effects , Tumor Cells, Cultured
18.
J Cell Biochem ; 54(2): 256-63, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8175900

ABSTRACT

Pre-adipocyte 3T3-L1 cells, after an appropriate induction stimulus, proceed through a defined change in morphology as differentiation progresses. Transforming growth factor beta 1 (TGF beta 1) is able to block the morphological and biochemical changes which occur with differentiation of these cells if given within 36-40 h of induction [Ignotz and Massague (1985): Proc Natl Acad Sci USA 82:8530-8534]. To begin to elucidate the role of the extracellular matrix in adipogenesis, as well as the mechanism whereby TGF beta 1 inhibits differentiation, we examined the expression of two extracellular matrix genes, type I (alpha 1) procollagen and fibronectin, as well as endogenous TGF beta 1. Confluent cells were induced to differentiate by treatment with insulin, dexamethasone, and isobutylmethylxanthine in the presence or absence of TGF beta 1. Following 6 days of treatment, the cells in the differentiated group acquired the rounded shape of mature adipocytes; the cytosol of these cells also contained numerous lipid-filled vesicles, as demonstrated by oil red O staining. Cells treated with the differentiation compounds in the presence of TGF beta 1 maintained the fibroblast-like appearance of control cells and did not stain with oil red O. At the level of gene expression, both procollagen and fibronectin mRNAs were down-regulated during differentiation of 3T3-L1 cells. When cells from the control or differentiation groups were treated with TGF beta 1, there was a 2-5-fold induction of procollagen and fibronectin mRNAs throughout the 6-day time course.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
3T3 Cells/drug effects , Adipocytes/drug effects , Fibronectins/genetics , Procollagen/genetics , Stem Cells/drug effects , Transforming Growth Factor beta/pharmacology , 3T3 Cells/cytology , Adipocytes/cytology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Gene Expression , Histocytochemistry , Mice , Stem Cells/cytology , Transforming Growth Factor beta/genetics
19.
J Cell Biochem ; 53(3): 240-50, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8263041

ABSTRACT

The rat osteocalcin gene encodes a 6-kD osteoblast-specific protein that is expressed post-proliferatively. The developmental and steroid hormone responsive expression of the osteocalcin gene is transcriptionally regulated by a promoter with multiple basal and enhancer elements that exhibit activity controlled by a series of physiological mediators (e.g., 1.25(OH)2D3, glucocorticoids). In this study, we established the contribution of the rat osteocalcin (OC) box domain (-99 to -76), a proximal basal element with a CCAAT motif as a central core, to transcriptional activity of the rat osteocalcin gene with in vivo co-transfection assays. By this same assay, however, the highly homologous (22 of 24 nt) human OC box element was unable to compete for transcription factor binding with the rat OC promoter. In vitro protein/DNA interaction studies confirm the presence of two protein binding sites in the OC box region, one of which overlaps the CCAAT motif and, at least in part, accounts for species-specific expression. Competition analysis established that the single nucleotide substitution of adenine for thymine, which converts the core motif of the rat OC box (CCAAT) to the core motif of the human OC box (CCAAA), accounts for observed species differences in transcription factor interactions. The CCAAT-specific protein/DNA interactions are heat stable and insensitive to phosphatase treatment. At second protein/DNA interaction located upstream of the CCAAT motif includes two steroid-like half-elements. These interactions are heat labile and sensitive to phosphatase treatment in contrast to the CCAAT-specific interactions. The human OC promoter contains only a single steroid-like half-element, while two steroid half-elements with an 11 nucleotide spacer are present in the rat OC promoter. These observed variations in sequence organization and transactivation factor binding in analogous proximal basal regulatory regions of the OC gene promoter may provide a basis for species-restricted variations in responsiveness to physiological mediators of OC gene expression at the transcriptional level.


Subject(s)
Gene Expression Regulation , Osteocalcin/genetics , Promoter Regions, Genetic , Animals , Base Sequence , Binding Sites , Binding, Competitive , DNA/metabolism , Humans , Molecular Sequence Data , Osteosarcoma , Rats , Species Specificity , Temperature , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured
20.
J Cell Biochem ; 52(2): 171-82, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8366135

ABSTRACT

We have addressed questions raised by the observation in fetal rats of delayed ossification induced by caffeine at maternal doses above 80 mg/kg body weight per day. The effect of caffeine on endochondral bone development and mineralization has been studied in an experimental model system of bone formation which involves implantation of demineralized bone particles (DBP) in subcutaneous pockets of young growing rats. Caffeine's effects on cellular events associated with endochondral ossification were examined directly by quantitating cellular mRNA levels of chondrocyte and osteoblast growth and differentiation markers in DBP implants from caffeine-treated rats harvested at specific stages of development (day 7 through day 15). Oral caffeine administration to rats implanted with DBP resulted in a dose dependent inhibition of the formation of cartilage tissue in the implants. Histologic examination of the implants revealed a decrease in the number of cells which were transformed to chondrocytes compared to control implants. Those cartilaginous areas that did form, however, proceeded through the normal sequelae of calcified cartilage and bone formation. At the 100 mg/kg dose, cellular levels of mRNA for histone, collagen type II, and TGF beta were all reduced by greater than 40% of control implants consistent with the histological findings. Alkaline phosphatase activity in the implants and mRNA levels for proteins reflecting the hypertrophic chondrocyte and bone phenotype, collagen type I and osteocalcin were markedly decreased compared to controls. Lower doses of 50 and 12.5 mg/kg caffeine also resulted in decreased cellular proliferation and transformation to cartilage histologically and reflected by significant inhibition of type II collagen mRNA levels (day 7). The effects of caffeine on gene expression observed in vivo during the period of bone formation (day 11 to day 15) in the DBP model were similar to the inhibited expression of H4, alkaline phosphatase, osteocalcin, and osteopontin found in fetal rat calvarial derived osteoblast cultures following 24 hour exposure of the cultures to 0.4 mM caffeine. Thus the observed delayed mineralization in the fetal skeleton associated with caffeine appears to be related to an inhibition of endochondral bone formation at the early stages of proliferation of undifferentiated mesenchymal cells to cartilage specific cells as well as at later stages of bone formation.


Subject(s)
Bone Development/drug effects , Caffeine/pharmacology , Cartilage/drug effects , Administration, Oral , Animals , Bone Transplantation , Cartilage/physiology , Cells, Cultured , Gene Expression , Male , Osteoblasts/drug effects , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...