Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Gene Ther ; 12(11): 881-90, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15908994

ABSTRACT

Nonviral gene delivery is limited to a large extent by multiple extracellular and intracellular barriers. One of the major barriers, especially in nondividing cells, is the nuclear envelope. Once in the cytoplasm, plasmids must make their way into the nucleus in order to be expressed. Numerous studies have demonstrated that transfections work best in dividing populations of cells in which the nuclear envelope disassembles during mitosis, thus largely eliminating the barrier. However, since many of the cells that are targets for gene therapy do not actively undergo cell division during the gene transfer process, the mechanisms of nuclear transport of plasmids in nondividing cells are of critical importance. In this review, we summarize recent studies designed to elucidate the mechanisms of plasmid nuclear import in nondividing cells and discuss approaches to either exploit or circumvent these processes to increase the efficiency of gene transfer and therapy.


Subject(s)
Active Transport, Cell Nucleus , Gene Transfer Techniques , Genetic Vectors/pharmacokinetics , DNA/pharmacokinetics , Humans , Nuclear Envelope/metabolism , Plasmids/pharmacokinetics
2.
Genetica ; 114(1): 1-9, 2002.
Article in English | MEDLINE | ID: mdl-11990753

ABSTRACT

One QTL and genes and phenotypes have been localized in the region between 92 cM and 95cM of mouse chromosome 1. The QTL locus contributes to approximately 40% of the variation of the peak bone density between C57BL/6J (B6) and CAST/EiJ (CAST) strains. Other loci located in this chromosomal region include a neural tube defect mutant loop-tail (Lp), a lymphocyte-stimulating determinant (Lsd), and the Transgelin 2 (Tagln 2). The human chromosome region homologous to this region is 1q21-23, which also contains a QTL locus for high bone mineral density (BMD). Furthermore, it has been reported that this region may have duplicated several times in the mouse genome. Therefore, genomic sequencing of this region will provide important information for mouse genome structure, for positional cloning of mouse genes, and for the study of human homologous genes. In order to provide a suitable template for genomic sequencing by the NIH-sponsored genomic centers, we have constructed a BAC contig of this region using the RPCI-23 library. We have also identified the currently available mouse genomic sequences localized in our BAC contig. Further analysis of these sequences and BAC clones indicated a high frequency of repetitive sequences within this chromosomal area. This region also contains L1 retrotransposon sequences, providing a potential mechanism for the repetitive sequences described in the literature.


Subject(s)
Chromosomes, Artificial, Bacterial , Chromosomes , Mice/genetics , Animals , Chromosome Mapping , Chromosomes, Human, Pair 21 , Contig Mapping , Female , Genome , Humans , Lod Score , Mice, Inbred Strains , Microsatellite Repeats , Physical Chromosome Mapping , Quantitative Trait, Heritable , Retroelements
3.
Biochim Biophys Acta ; 1518(1-2): 145-51, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11267670

ABSTRACT

The 1.7 kb human insulin-like growth factor binding protein (IGFBP)-6 gene 5'-flanking region was sequenced and found to have promoter activity in human osteoblasts. The sequence contains four clustered transcription start sites and three retinoic acid response elements (RAREs) with widely spaced half-sites. Only the proximal DR15 RARE was functional. Retinoids increased IGFBP-6 promoter activity up to 3-fold.


Subject(s)
Insulin-Like Growth Factor Binding Protein 6/genetics , Promoter Regions, Genetic , Response Elements , Retinoids/metabolism , Animals , Base Sequence , DNA, Complementary , Humans , Mice , Molecular Sequence Data , Osteoblasts , Rats , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
4.
Growth Horm IGF Res ; 11(6): 368-77, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11914024

ABSTRACT

All-trans -retinoic acid (atRA) inhibits osteoblast marker gene expression and markedly increases expression of insulin-like growth factor binding protein-6 (IGFBP-6) in human osteoblasts. The possibility that IGFBP-6 inhibits the osteoblast phenotype and also mediates the inhibitory effect of atRA on osteoblast marker gene expression was explored using an antisense approach. Stable human osteoblast-like osteosarcoma SaOS-2 cells were prepared that expressed antisense IGFBP-6 RNA under basal and atRA-stimulated conditions. The functional expression of IGFBP-6 antisense RNA was confirmed by measuring IGFBP-6 mRNA by Northern analysis or by measuring IGFBP-6 protein in the conditioned media (CM) by radioimmunoassay. Antisense clones produced less mRNA and had less IGFBP-6 protein in the CM than controls. IGFBP-6 protein levels in the CM were inversely correlated with alkaline phosphatase (ALP) activity, whereas IGFBP-3 and IGFBP-4 protein levels were not. We reasoned that atRA would have little or no effect on ALP activity in IGFBP-6 antisense clones if atRA mediated its inhibitory effects by recruiting IGFBP-6. In the majority of IGFBP-6 antisense clones with the lowest IGFBP-6 mRNA and CM protein levels and only modest changes in other IGF system components, atRA did not significantly decrease ALP activity. These findings provide evidence that atRA recruits IGFBP-6 to inhibit the human osteoblast phenotype.


Subject(s)
Insulin-Like Growth Factor Binding Protein 6/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Tretinoin/pharmacology , Alkaline Phosphatase/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , DNA, Antisense/pharmacology , Gene Expression/drug effects , Genetic Markers , Humans , Insulin-Like Growth Factor Binding Protein 6/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 6/genetics , Osteoblasts/cytology , Phenotype , Transfection
5.
J Cell Biochem ; 75(4): 566-77, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10572240

ABSTRACT

Basic helix-loop-helix (bHLH) transcription factors have been shown to play an important role in controlling cell type determination and differentiation. TWIST, a member of the bHLH transcription factor family, is involved in the development of mesodermally derived tissue, including the skeleton. We examined the role of human TWIST in osteoblast metabolism using stable expression of sense and antisense TWIST in human osteoblast HSaOS-2 cells. Changes in morphology and osteogenic phenotype characterized these stable clones. Cells that overexpressed TWIST exhibited a spindle shaped morphology, reduced levels of alkaline phosphatase, a reduced proliferation rate, and failed to respond to basic fibroblast growth factor (bFGF). In contrast, those that underexpressed TWIST demonstrated a cuboidal epithelial-like morphology characteristic of differentiated osteoblasts. TWIST antisense cells exhibited increased levels of alkaline phosphatase and type I collagen mRNA, initiated osteopontin mRNA expression, and had a reduced proliferation rate. These results indicate that TWIST overexpressing cells may de-differentiate and remain in an osteoprogenitor-like state, and antisense TWIST cells progress to a more differentiated mature osteoblast-like state. Therefore, the level of TWIST can influence osteogenic gene expression and may act as a master switch in initiating bone cell differentiation by regulating the osteogenic cell lineage.


Subject(s)
Helix-Loop-Helix Motifs , Nuclear Proteins , Transcription Factors/biosynthesis , Transcription Factors/physiology , Alkaline Phosphatase/biosynthesis , Antigens, Differentiation/biosynthesis , Blotting, Northern , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/genetics , Cell Line , Cell Size/genetics , DNA, Antisense/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Middle Aged , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/pharmacology , Transfection , Twist-Related Protein 1
6.
J Biol Chem ; 274(37): 26431-8, 1999 Sep 10.
Article in English | MEDLINE | ID: mdl-10473602

ABSTRACT

Insulin-like growth factor-binding protein-5 (IGFBP-5) is produced by osteoblasts and potentiates insulin-like growth factor mitogenic stimulation in osteoblast cell cultures. Progesterone (PG) increased IGFBP-5 expression in normal human osteoblasts and increased IGFBP-5 transcription in U2 human osteosarcoma cells. We developed a chloramphenicol acetyltransferase reporter construct containing the human IGFBP-5 proximal promoter sequence, which includes TATA and CAAT boxes, and five putative PG response element half-sites. 10(-8) M PG increased promoter activity of this construct in U2 cells co-transfected with a PG receptor isoform A (PR(A)) expression vector. Analysis of 5' deletion constructs indicates that PG transactivation of IGFBP-5 promoter activity does not require the PG response element half-sites but does require the region -162 to -124 containing two tandem CACCC box sequences. Mutation of the proximal CACCC box at -139 eliminated PG transactivation. Gel shift assays using a -162 to -124 DNA fragment, U2 cell nuclear extracts, and purified PR(A) protein indicate that nuclear factors bind to a CACCC sequence at -139 and that PR(A) alters the pattern of transcription factor interaction with the CACCC sequence. Using a luciferase reporter construct containing base pairs -252 to +24 of the IGFBP-5 promoter, we found that both PR(A) and PR(B) isoforms mediated PG stimulation of promoter activity. These results suggest that PG may stimulate IGFBP-5 gene transcription via a novel mechanism involving PR and CACCC-binding factors.


Subject(s)
Insulin-Like Growth Factor Binding Protein 5/genetics , Osteoblasts/drug effects , Progesterone/pharmacology , Promoter Regions, Genetic , Transcription, Genetic/drug effects , Base Sequence , DNA Primers , Hormone Antagonists/pharmacology , Humans , Mifepristone/pharmacology , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Tumor Cells, Cultured
7.
Mol Med Today ; 5(3): 133-40, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10203737

ABSTRACT

Osteoporosis is a common disease that affects millions of patients throughout the world. We anticipate that both the diagnosis and the treatment of this disease will be revolutionized by the integration of genomics and informatics. It is predicted that a genetic algorithm will be developed to identify at-risk patients before they develop osteoporosis, so that preventive measures can be instituted. The sequencing of the human genome will lead to revolutionary advances in at least three areas of osteoporosis therapy: small molecule therapy, protein therapy and gene therapy. One area of focus for future therapeutics in osteoporosis will be on osteogenic agents, which should have a high likelihood of success because the skeleton has the innate capacity to regenerate itself.


Subject(s)
Osteoporosis/diagnosis , Osteoporosis/therapy , Animals , Bone Density , Genetic Therapy , Humans , Mice , Risk Assessment
8.
J Bone Miner Res ; 14(12): 2079-88, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10620067

ABSTRACT

To characterize the insulin-like growth factor binding protein-4 (IGFBP-4) protease produced by human osteoblasts (hOBs), we localized and determined the role of the proteolytic domains in human IGFBP-4 (hIGFBP-4) in modulating IGF-II actions. N-terminal amino acid sequence and mass spectrometric analyses of the 6xHis-tagged IGFBP-4 proteolytic fragments revealed that Met135-Lys136 was the only cleavage site recognized by the IGF-II-dependent IGFBP-4 protease produced by hOBs. This cleavage site was confirmed by the finding that deletion of His121 to Pro141 blocked proteolysis. However, unexpectedly, deletion of Pro94 to Gln119 containing no cleavage site had no effect on IGF-II binding activity but blocked proteolysis. Addition of the synthetic peptide corresponding to this region at concentrations of 250 or 1000 molar excess failed to block IGFBP-4 proteolysis. These data suggest that residues 94-119 may be involved in maintaining the IGFBP-4 conformation required to expose the cleavage site rather than being involved in direct protease-substrate binding. To determine the physiological significance of the IGF-II-dependent IGFBP-4 protease, we compared the effect of the wild-type IGFBP-4 and the protease-resistant IGFBP-4 analogs in blocking IGF-II-induced cell proliferation in normal hOBs, which produce IGFBP-4 protease, and MG63 cells, which do not produce IGFBP-4 protease. It was found that protease-resistant IGFBP-4 analogs were more potent than the wild-type protein in inhibiting IGF-II-induced cell proliferation in hOBs but not in MG63 cells. These data suggest that IGFBP-4 proteolytic fragments are not biologically active and that IGFBP-4 protease plays an important role in regulating IGFBP-4 bioavailability and consequently the mitogenic activity of IGFs in hOBs.


Subject(s)
Insulin-Like Growth Factor Binding Protein 4/chemistry , Insulin-Like Growth Factor II/pharmacology , Metalloendopeptidases/metabolism , Osteoblasts/drug effects , Amino Acid Sequence , Cell Division/drug effects , Cell Line , Humans , Insulin-Like Growth Factor Binding Protein 4/pharmacology , Mass Spectrometry , Molecular Sequence Data , Mutation , Osteoblasts/metabolism , Peptide Fragments/pharmacology , Pregnancy-Associated Plasma Protein-A , Protein Binding , Substrate Specificity
9.
J Biol Chem ; 273(36): 23509-16, 1998 Sep 04.
Article in English | MEDLINE | ID: mdl-9722589

ABSTRACT

To identify the molecular mechanism by which insulin-like growth factor binding protein-4 (IGFBP-4) exerts its inhibitory effects on insulin-like growth factor (IGF) actions, we localized and determined the role of the IGF binding domain in modulating IGF actions in human osteoblasts. Deletion analysis using IGFBP-4 expressed in bacteria revealed that the N-terminal sequence Leu72-Ser91 was essential for IGF binding. The C-terminal fragments (His121-Glu237 or Arg142-Glu237) did not bind to IGF but loss of these regions decreased IGF binding activity. Detailed deletion analysis identified the residues Cys205-Val214 as the motif to facilitate IGF binding. Mitogenic studies revealed that an IGFBP-4 mutant (His74 replaced by Pro74) and an N-terminal peptide (N terminus to Thr71) with little IGF binding activity failed to inhibit IGF-II-induced human osteoblast proliferation. An N-terminal peptide (N terminus to Asn182) with reduced IGF binding activity inhibited IGF action but with lower potency. In contrast, an IGFBP-4 mutant (His74 replaced with Ala74) exhibited similar IGF binding activity and potency in inhibiting the activity of IGF-II compared with the wild type. Therefore, the N-terminal sequence (Leu72-Ser91) and the C-terminal sequence (Cys205-Val214) are necessary to form the high affinity IGF binding domain, which is the major structural determinant of the IGFBP-4 function.


Subject(s)
Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Osteoblasts/metabolism , Amino Acid Sequence , Binding Sites , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Molecular Sequence Data , Mutagenesis , Osteoblasts/cytology , Peptide Fragments/metabolism , Protein Binding , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
10.
Endocrinology ; 138(10): 4240-7, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9322936

ABSTRACT

To begin delineating molecular mechanisms by which osteogenic protein-1 (OP-1) modulates its effect on the insulin-like growth factor (IGF) system in human skeletal cells, we evaluated time-course effects of OP-1 on the expression of IGFBP-3 messenger RNA (mRNA) in human SaOS-2 osteosarcoma cells and found that 100 ng/ml of OP-1 increased (maximum 10.7-fold at 24 h; P < 0.01) the level of IGFBP-3 mRNA in a time-dependent manner (from 3-36 h; treatment x time interaction, P < 0.001). The stimulatory effect of OP-1 on IGFBP-3 mRNA was not promoted by transcript stabilization; actually, OP-1 treatment selectively increased the decay of mRNA for IGFBP-3 (T1/2 = 5 h vs. 24 h for OP-1 and controls), but not for IGFBP-4 or beta-actin. Conversely, OP-1 acutely increased IGFBP-3 nuclear transcript abundance in total RNA samples ranging between 1-24 h of treatment. After 6 h of treatment, OP-1 produced an average 4-fold increase (P < 0.02; n = 4 experiments) in the level of IGFBP-3 nuclear transcripts vs. a 3-fold increase (P < 0.01; n = 2 experiments) in mRNA abundance. The OP-1 stimulated induction of IGFBP-3 nuclear transcript and mRNA expression was dependent on de novo protein synthesis. Transient transfection experiments were undertaken to isolate putative OP-1 stimulatory cis-elements within 1.8-kb of the IGFBP-3 5'-flanking region in SaOS-2 and TE-85 osteosarcoma cells. In these experiments, OP-1 did not stimulate IGFBP-3 proximal promoter activity in either cell line, thus suggesting that OP-1 reactive domains may be located either beyond the currently established 5'-flanking region, or within internal exon/intron regions of the IGFBP-3 gene. In conclusion, OP-1 treatment stimulates IGFBP-3 expression in human osteoblastic cells by a mechanism that largely promotes the production of IGFBP-3 nuclear transcripts, a process that requires de novo protein synthesis, and overrides an OP-1-induced targeted degradation of IGFBP-3 steady-state mRNA.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Bone Neoplasms/chemistry , DNA, Neoplasm/analysis , Insulin-Like Growth Factor Binding Protein 3/genetics , Osteosarcoma/chemistry , Transcription, Genetic , Transforming Growth Factor beta , Actins/analysis , Actins/genetics , Base Sequence , Blotting, Northern , Bone Morphogenetic Protein 7 , Bone Neoplasms/pathology , DNA Primers/analysis , DNA Primers/chemistry , DNA Primers/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Insulin-Like Growth Factor Binding Protein 3/analysis , Insulin-Like Growth Factor Binding Protein 4/analysis , Insulin-Like Growth Factor Binding Protein 4/genetics , Introns , Osteosarcoma/pathology , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Time Factors , Tumor Cells, Cultured
11.
Calcif Tissue Int ; 60(3): 297-301, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9069169

ABSTRACT

Bone morphogenetic proteins (BMPs) are novel growth and differentiation factors that act on mesenchymal stem cells to initiate new bone formation in vivo and promote the growth and differentiation of cells in the osteoblastic lineage. In the present study, we examined the effects of recombinant human osteogenic protein-1 (also known as BMP-7) on the expression of related members of the BMP family using SaOS-2 and U2-OS, two human osteosarcoma cell strains. Evaluation of BMP-2, -4, and -6 mRNA expression indicates that OP-1 stimulated the mRNA levels of BMP-6 in both SaOS-2 cells (threefold) and U2-OS cells (fivefold) after 24 hours of treatment, while decreasing the mRNA levels of BMP-4 in SaOS-2 cells (80%) and BMP-2 and BMP-4 in U2-OS cells by 50% and 72%, respectively. BMP-2 mRNA expression, as examined by Northern blot analysis, was below detectable limits in SaOS-2 cultures. These results demonstrate that OP-1 modulates the mRNA expression of related members of the BMP family, suggesting a possible mode of action of OP-1 on the growth and differentiation of cells in the osteoblastic lineage in vitro.


Subject(s)
Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Bone Neoplasms/physiopathology , Osteosarcoma/physiopathology , RNA, Messenger/biosynthesis , Transforming Growth Factor beta/physiology , Blotting, Northern , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 6 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/drug effects , Bone Morphogenetic Proteins/pharmacology , Humans , RNA, Messenger/drug effects , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
12.
Biochim Biophys Acta ; 1350(2): 136-40, 1997 Feb 07.
Article in English | MEDLINE | ID: mdl-9048882

ABSTRACT

More than 3 kb of the human (h)IGFBP-4 gene 5'-flanking region was sequenced and assessed for promoter activity. The hIGFBP-4 promoter resides within a CpG island and demonstrates strong basal activity in human osteoblast-like osteosarcoma and COS-7 monkey kidney cells. Transient transfection of cells with hIGFBP-4 promoter-linked deletion constructs demonstrated that multiple cooperating cis-acting elements within 836 bp of the 5'-flanking region contributed to overall promoter strength.


Subject(s)
Insulin-Like Growth Factor Binding Protein 4/genetics , Promoter Regions, Genetic , Animals , Base Sequence , COS Cells , Cell Line , CpG Islands , DNA/genetics , Humans , Molecular Sequence Data , Oligonucleotides, Antisense/genetics , Osteoblasts/metabolism , Plasmids/genetics , Transfection
13.
Eur J Endocrinol ; 134(5): 591-601, 1996 May.
Article in English | MEDLINE | ID: mdl-8664980

ABSTRACT

Glucocorticoids (GCs) inhibit bone formation in vivo and inhibit osteoblast proliferation and collagen synthesis in vitro. These effects may be mediated by alterations in the insulin-like growth factor (IGF) system. In the present study of normal human osteoblast-like (HOB) cells, we tested the hypothesis that dexamethasone (Dex) inhibits IGF anabolic activity in bone by altering expression of IGF binding proteins (IGFBPs), particularly by decreasing expression of IGFBP-5 and IGFBP-3 (which enhance IGF activity) and increasing expression of IGFBP-4 (which inhibits IGF actions). Dexamethasone treatment caused a dose-dependent inhibition of HOB cell proliferation (69 +/- 4% of control at 10(-8) mol/l Dex) in seven separate experiments. Dexamethasone decreased IGFBP-5 mRNA levels to 20-30% of control (10(-8) and 10(-7) mol/l for 24 h). In six of six HOB preparations, 10(-8) mol/l Dex decreased IGFBP-5 mRNA levels (35 +/- 7% of control) and this effect was time dependent. Dexamethasone also decreased IGFBP-3 mRNA levels (74 +/- 9% of control in three HOB preparations). Dexamethasone decreased secretion of 29-31-kD IGFBP-5 and 38-42-kD IGFBP-3 proteins, determined by Western ligand blot and IGFBP-5 immunoblot, and induced a dose-dependent decrease in IGFBP-3 and IGFBP-5 secretion determined by specific radioimmunoassays. The effects of Dex on IGFBP-4 mRNA and on secretion of 25-kD IGFBP-4 levels were inconsistent between different cell preparations. Results suggest that GC inhibition of IGFBP-5 and IGFBP-3 production could decrease IGF activities and contribute to GC inhibition of bone formation.


Subject(s)
Cell Division/drug effects , Glucocorticoids/pharmacology , Insulin-Like Growth Factor Binding Proteins/physiology , Osteoblasts/cytology , Base Sequence , Blotting, Northern , Blotting, Western , Culture Media, Conditioned , Dexamethasone/pharmacology , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Molecular Sequence Data , RNA, Messenger/metabolism
14.
J Clin Endocrinol Metab ; 81(4): 1389-96, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8636339

ABSTRACT

Insulin-like growth factor-binding protein-4 (IGFBP-4), like the five other IGFBPs present in human serum, acts as a transport protein for insulin-like growth factor I (IGF-I) and IGF-II and modulates their biological effects. To investigate the role of IGFBP-4 in the physiology of the IGF system, we developed a sensitive RIA for IGFBP-4 employing, as antigen, tracer, and standard, recombinant human IGFBP-4 (rhIGFBP-4) expressed in Escherichia coli as a fusion protein with glutathione S-transferase and affinity purified with glutathione-derivatized resin. Antibody against the rhIGFBP-4 fusion protein was raised in guinea pigs; tracer and standard were provided by the rhIGFBP-4 moiety that had been cleaved from the rhIGFBP-4 fusion protein and repurified by reverse phase high pressure liquid chromatography. We report that both IGFBP-4 purified from PC3 human prostate cell-conditioned medium and rhIGFBP-4 bound IGF and migrated in electrophoresis gels in an identical manner; that in gel permeation chromatography, rhIGFBP-4 coeluted with the IGFBP-4 present in human serum; and that both are equally immunoreactive with the IGFBP-4 antiserum. Employing this IGFBP-4 RIA, we determined that no IGFBP other than IGFBP-4 reacted with the IGFBP-4 antiserum, and that recovery of IGFBP-4 from serum samples exceeded 90% when exogenous IGFBP-4 was added and was unaffected by the addition of IGFs or by repeated freezing and thawing of the sample. We employed this IGFBP-4 RIA to demonstrate an increase in IGFBP-4 in TE85 human osteosarcoma cell-conditioned medium after treatment with dibutyryl cAMP, PTH, and 1,25-dihydroxyvitamin D3, agents known to increase the IGFBP-4 messenger ribonucleic acid level. Application of this RIA to the measurement of IGFBP-4 in human serum revealed that the circulating level of IGFBP-4 in 41 individuals in the 61-87 yr age group (546 +/- 135 microgram/L) was 35% higher than that in 24 individuals in the 23-40 yr age group (404 +/- 156 microgram/L). The mean circulating level of PTH was also 20% higher in the 61-87 yr group compared to that in the 23-40 yr group (P < 0.01). In addition, serum IGFBP-4 amounts showed a significant positive correlation with age (r = 0.54; P < 0.001) and serum PTH (r = 0.26; P < 0.01). These data validate this IGFBP-4 RIA and illustrate its utility in illuminating the physiological mechanisms that regulate IGFBP-4 in vivo and influence its effects on the IGFs in both normal and abnormal pathology and in aging.


Subject(s)
Insulin-Like Growth Factor Binding Protein 4/biosynthesis , Insulin-Like Growth Factor Binding Protein 4/blood , Adult , Aged , Aged, 80 and over , Animals , Antibodies , Biological Assay , Blotting, Western , Body Fluids/chemistry , Bone Neoplasms , Bone and Bones/cytology , Cells, Cultured , Chromatography, Gel , Escherichia coli , Female , Guinea Pigs , Humans , Insulin-Like Growth Factor Binding Protein 4/analysis , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Male , Middle Aged , Osteosarcoma , Radioimmunoassay/methods , Recombinant Fusion Proteins/isolation & purification , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Reference Values , Regression Analysis , Reproducibility of Results , Sensitivity and Specificity , Tumor Cells, Cultured
15.
Endocrinology ; 137(3): 975-83, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8603611

ABSTRACT

Retinoic acid (RA) regulates the growth and differentiation of numerous cells types and plays a key role in skeletal development. Previous studies have demonstrated that insulin-like growth factors (IGFs) are important local regulators of bone cell proliferation and differentiation and that IGF-binding proteins (IGFBPs) modulate their activities. In an attempt to test the hypothesis that RA mediates its effects on bone cells in part by regulating IGFBP expression, we first examined the effect of RA on IGFBP expression in human osteoblast model systems and then compared these responses to the effects of RA on IGFBP expression in human skin fibroblasts. The most dramatic effect of RA on IGFBPs++ in all cell types tested was to increase IGFBP-6 messenger RNA (mRNA) abundance more than 1000% of the control value. Significant effects on IGFBP-5 mRNA abundance were also found, with maximal reductions to 35% of control within 24 h of treatment. In addition, RNA maximally increased IGFBP-3 and -4 mRNA to 580% and 390% of the control value, respectively, in SaOS-2 cells, but had variable effects on IGFBP-3 and -4 mRNA levels in human bone cells, U2-OS, and human skin fibroblasts. The levels of the 24-, 29- to 32-, and 38- to 42 kDa IGFBPs in the conditioned medium of RA-treated cultures increased, as determined by ligand blot analysis, whereas the amount of IGFBP-5 was reduced, as determined by RIA. Cycloheximide abolished the RA-stimulated increase in IGFBP-6 mRNA and reduced baseline IGFBP-5 mRNA levels, but did not affect RA-modulated mRNA levels of the other IGFBPs. RA modestly increased the stabilities of all four IGFBP mRNAs, which could contribute to the observed increases in IGFBP-3 and IGFBP-4 mRNA levels; however, the 217% increase in the IGFBP-5 mRNA half- life in the presence of RA could not contribute to the reduction in mRNA levels. In addition, the small increase in the IGFBP-6 mRNA half-life could not account for the 1900% increase in the mRNA level. These data suggest that RA stimulated changes in IGFBP-5 and -6 mRNA levels may in part be mediated by alterations in transcription or other early posttranscription regulatory mechanisms. In conclusion, RA significantly regulates IGFBP expression in human osteoblast cells.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/biosynthesis , Keratolytic Agents/pharmacology , Osteoblasts/metabolism , Tretinoin/pharmacology , Cells, Cultured , Culture Media, Conditioned , Dose-Response Relationship, Drug , Humans , RNA, Messenger/analysis
16.
Endocrinology ; 136(3): 857-65, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7532581

ABSTRACT

Bone morphogenetic proteins (BMPs) have the unique ability to convert mesenchymal cells into matrix-producing osteoblasts. To understand the mechanism(s) by which a BMP produces a multitude of effects on bone cells, we examined the effects of recombinant human osteogenic protein (OP)-1 (referred to as BMP-7) on the insulin-like growth factor (IGF) regulatory system, an important growth factor system in bone. After 48 h of treatment, OP-1 increased the level of IGF-II (3- and 2-fold, respectively, at 100 ng/ml) in the conditioned medium (CM) of SaOS-2 and TE85 human osteosarcoma cells with osteoblastic characteristics, whereas IGF-I levels were low to undetectable in the CM of either cell type. OP-1 treatment had no significant effect on the messenger RNA (mRNA) level for type 1 and type 2 IGF receptors. In TE85 and SaOS-2 cells, 100 ng/ml OP-1 increased the level of IGF binding protein (BP)-3 more than 10-fold, decreased the IGFBP-4 level by 50%, and increased the level of the 29-32.5 kDa IGFBP-5 3-fold in the CM as determined by analysis with Western ligand blot, Western immunoblot, and RIA. The effect of OP-1 on IGFBP production was time and dose dependent. The OP-1 induced changes in the levels of IGFBPs were associated with decreased IGFBP-3 and -5 protease activity (29% and 71%, respectively) and proportional changes in IGFBP mRNA levels. OP-1 increased the level of IGFBP-3 mRNA (2- and 10-fold, respectively, after 4 and 24 h of treatment at 100 ng/ml) and of IGFBP-5 mRNA (more than 5-fold after 24 h of treatment) but decreased the level of IGFBP-4 mRNA (> 50% after 24 h at 100 ng/ml). OP-1 treatment had no effect on IGFBP-4 protease activity. These results collectively demonstrate that OP-1 can act locally by modulating the IGF regulatory system, suggesting that the mitogenic/differentiative effect of OP-1 on human bone cells may in part be mediated via IGF-II by increasing its secretion, and by regulating the balance between the stimulatory (e.g. IGFBP-5) and inhibitory (e.g. IGFBP-4) classes of IGFBPs both at the level of production (mRNA) and at the level of degradation but not by up-regulating the IGF receptor.


Subject(s)
Bone Morphogenetic Proteins , Bone and Bones/drug effects , Proteins/pharmacology , Somatomedins/physiology , Bone Morphogenetic Protein 7 , Bone and Bones/cytology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Culture Media/metabolism , Dose-Response Relationship, Drug , Endopeptidases/metabolism , Humans , Insulin-Like Growth Factor Binding Proteins , RNA, Messenger/metabolism , Time Factors , Transforming Growth Factor beta/pharmacology
17.
Calcif Tissue Int ; 56(2): 154-9, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7736325

ABSTRACT

Recent studies indicate that the amount of alkaline phosphatase (ALP) activity in human osteoblast-line cells is proportional to the concentration of phosphate in the culture medium. The current studies were intended to extend those observations and to determine whether the effects of phosphate (and phosphate esters and analogs) to alter the cellular level of ALP activity, in human osteosarcoma SaOS-2 cells, reflected regulation at the level of transcription. Consistent with previous findings, we found direct, time- and dose-dependent correlations between the concentration of phosphate and the amount of ALP activity/mg cell protein (P < 0.05). Surprisingly, we also found a negative correlation between the phosphate concentration in the medium and the level of skeletal ALP mRNA (e.g., r = -0.98, P < 0.01 at 24 hours). As the highest cellular levels of skeletal ALP activity were associated with the lowest levels of ALP mRNA, these data indicated that the phosphate-dependent increase in ALP activity was not mediated by an increase in transcription and, conversely, that the effect of phosphate withdrawal to decrease ALP activity was not mediated by a decrease in transcription.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Alkaline Phosphatase/metabolism , Osteosarcoma/enzymology , Phosphates/pharmacology , Alkaline Phosphatase/genetics , Base Sequence , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Glycerophosphates/pharmacology , Humans , Molecular Sequence Data , Osteosarcoma/genetics , Phosphates/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
18.
Growth Factors ; 12(4): 245-50, 1995.
Article in English | MEDLINE | ID: mdl-8930016

ABSTRACT

The IGFBPs bind to and modulate the function of the IGFs in various ways. Human IGFBP-4 inhibits IGF mediated cell proliferation. The IGFBP exon-encoded regions were aligned and secondary structure predictions for hIGFBP-4 were developed yielding predicted 3D co-ordinates for each such region of hIGFBP-4. The exon 1 encoded region is the most conserved among the IGFBPs. That of hIGFBP-4 is predicted as an array of beta-strands that include the glycine and cysteine rich IGFBP consensus pattern and that terminate with a helix. The exon 2 encoded region is the most variable among the IGFBPs. That of hIGFBP-4 is predicted as mostly an amphipathic helix. The remaining regions are also conserved among the IGFBPs. Those of hIGFBP-4 are also predicted to contain helices. The predicted structure of hIGFBP-4 comprises amino terminal beta-strands with four helices in the carboxy terminal two thirds of the molecule.


Subject(s)
Exons/genetics , Insulin-Like Growth Factor Binding Protein 4/chemistry , Amino Acid Sequence , Animals , Cattle , Consensus Sequence/genetics , Conserved Sequence/genetics , Databases, Factual , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Rats , Sequence Homology, Amino Acid
19.
Calcif Tissue Int ; 55(4): 311-5, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7820783

ABSTRACT

Previous studies have demonstrated that when cells of the mouse osteoblastic cell line MC3T3-E1 are exposed to IGF-I and IGF-II they exhibit rapid and transient induction of the transcript from the proto-oncogene c-fos [8]. To clarify the relationship between induction of cell proliferation and proto-oncogene expression in MC3T3-E1 cells, the acute affects of IGF-I and IGF-II, growth factors that stimulate cell proliferation, and of TGF-beta 1, which inhibits cell proliferation, northern analyses with cDNA-derived probes for the proto-oncogenes c-jun, jun-B, and jun-D were undertaken. Concurrent northern analyses with a probe for c-fos extended our previous results to include the effect of TGF-beta 1 on c-fos. IGF-I does not induce the c-jun, jun-B, or jun-D transcripts, the former and latter being produced at detectable levels constitutively. After 1 hour of exposure to IGF-II the c-jun transcript response ranges from onefold to 13-fold and the jun-D transcript response ranges around two-fold. After 1 hour of exposure to TGF-beta 1, the jun-B transcript response ranges from eightfold to 24-fold, the c-fos transcript response ranges between sixfold and sevenfold. The differences observed in the magnitude and kinetics of the induction provoked by these growth factors is consistent with the presence of a regulatory circuit acting through the Jun family members which may act to stimulate transcription differentially when bound to DNA either as homodimers or, with Fos family proteins, as heterodimers.


Subject(s)
Genes, jun/drug effects , Insulin-Like Growth Factor II/pharmacology , Insulin-Like Growth Factor I/pharmacology , Transforming Growth Factor beta/pharmacology , 3T3 Cells , Animals , Genes, fos , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
20.
Eur J Endocrinol ; 131(3): 293-301, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7522842

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is a cytokine produced by immune cells, which has multiple effects on bone cells and is therefore thought to mediate changes in bone metabolism occurring during inflammation. In the present study we have investigated the effect of TNF-alpha on the secretion of insulin-like growth factor I (IGF-I) and IGF binding protein 4 (IGFBP-4) by clonal mouse osteoblasts (MC3T3-E1 cells) using subconfluent in vitro cultures and serum-free conditions. The IGF-I was determined by radioimmunoassay under conditions eliminating the interference of IGFBPs. Treatment of MC3T3-E1 cultures with TNF-alpha for 24 h resulted in a dose-dependent decrease in IGF-I secretion (maximally to 34 +/- 9.7% of control with 60 pmol/l TNF-alpha; mean +/- SD). The TNF-alpha treatment also resulted in decreased messenger ribonucleic acid (mRNA) levels of IGF-I at 4 and 24 h, as detected by Northern analysis. Because basal secretion of IGFBPs is very low in MC3T3-E1 cells, effects of TNF-alpha on IGFBP secretion were studied in cultures in which IGFBP-4 expression was increased by calcitriol (1,25(OH)2D3) treatment. The presence of TNF-alpha (600 pmol/l) inhibited this calcitriol-induced stimulation of IGFBP-4 mRNA levels from 4 h onwards, with complete inhibition of the calcitriol effect occurring at 24 h. We also observed a dose-dependent inhibition of calcitriol-stimulated IGFBP-4 secretion into the culture medium (as detected by Western ligand blot), with the maximal inhibition occurring with 600 pmol/l TFN-alpha to 25 +/- 7% of control levels.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carrier Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Osteoblasts/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Division/drug effects , Cell Line , Indomethacin/pharmacology , Insulin-Like Growth Factor Binding Protein 4 , Mice , Protein Biosynthesis , Somatomedins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...