Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Oncogene ; 35(14): 1750-9, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26096936

ABSTRACT

Metastasis of tumors requires angiogenesis, which is comprised of multiple biological processes that are regulated by angiogenic factors. The fibroblast growth factor (FGF) is a potent angiogenic factor and aberrant FGF signaling is a common property of tumors. Yet, how the aberration in cancer cells contributes to angiogenesis in the tumor is not well understood. Most studies of its angiogenic signaling mechanisms have been in endothelial cells. FGF receptor substrate 2α (FRS2α) is an FGF receptor-associated protein required for activation of downstream signaling molecules that include those in the mitogen-activated protein and AKT kinase pathways. Herein, we demonstrated that overactivation and hyperactivity of FRS2α, as well as overexpression of cJUN and HIF1α, were positively correlated with vessel density and progression of human prostate cancer (PCa) toward malignancy. We also demonstrate that FGF upregulated the production of vascular endothelial growth factor A mainly by increasing expression of cJUN and HIF1α. This then promoted recruitment of endothelial cells and vessel formation for the tumor. Tumor angiogenesis in mouse PCa tissues was compromised by tissue-specific ablation of Frs2α in prostate epithelial cells. Depletion of Frs2α expression in human PCa cells and in a preclinical xenograft model, MDA PCa 118b, also significantly suppressed tumor angiogenesis accompanied with decreased tumor growth in the bone. The results underscore the angiogenic role of FRS2α-mediated signaling in tumor epithelial cells in angiogenesis. They provide a rationale for treating PCa with inhibitors of FGF signaling. They also demonstrate the potential of overexpressed FRS2α as a biomarker for PCa diagnosis, prognosis and response to therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Biomarkers, Tumor/biosynthesis , Membrane Proteins/biosynthesis , Neovascularization, Pathologic/genetics , Prostatic Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Biomarkers, Tumor/genetics , Blood Vessels/growth & development , Blood Vessels/pathology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/genetics , Mice , Neoplasm Metastasis , Neovascularization, Pathologic/pathology , Phosphorylation , Prostatic Neoplasms/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays
2.
Oncogene ; 32(35): 4181-8, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23045271

ABSTRACT

Cerebellar granule neurons are the most abundant neurons in the brain, and a critical element of the circuitry that controls motor coordination and learning. In addition, granule neuron precursors (GNPs) are thought to represent cells of origin for medulloblastoma, the most common malignant brain tumor in children. Thus, understanding the signals that control the growth and differentiation of these cells has important implications for neurobiology and neurooncology. Our previous studies have shown that proliferation of GNPs is regulated by Sonic hedgehog (Shh), and that aberrant activation of the Shh pathway can lead to medulloblastoma. Moreover, we have demonstrated that Shh-dependent proliferation of GNPs and medulloblastoma cells can be blocked by basic fibroblast growth factor (bFGF). But while the mitogenic effects of Shh signaling have been confirmed in vivo, the inhibitory effects of bFGF have primarily been studied in culture. Here, we demonstrate that mice lacking FGF signaling in GNPs exhibit no discernable changes in GNP proliferation or differentiation. In contrast, activation of FGF signaling has a potent effect on tumor growth: treatment of medulloblastoma cells with bFGF prevents them from forming tumors following transplantation, and inoculation of tumor-bearing mice with bFGF markedly inhibits tumor growth in vivo. These results suggest that activators of FGF signaling may be useful for targeting medulloblastoma and other Shh-dependent tumors.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellum/growth & development , Fibroblast Growth Factor 2/physiology , Medulloblastoma/pathology , Signal Transduction/physiology , Animals , Cell Cycle , Cell Differentiation , Cerebellar Neoplasms/etiology , Hedgehog Proteins/physiology , Medulloblastoma/etiology , Mice , Mice, Inbred C57BL , Neurons/cytology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Stem Cells/cytology
3.
Biochemistry ; 40(48): 14429-39, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11724555

ABSTRACT

Stromal cell-derived FGF-7 binds and activates only the resident FGFR2IIIb in epithelial cells while FGF-1 and FGF-2 exhibit a broader interaction with multiple isoforms of FGFR. Here we report the structure of FGF-7 that has been solved to 3.1 A resolution by molecular replacement with the structure of a dual function chimera of FGF-7 and FGF-1 (FGF-7/1) which was resolved to 2.3 A. Comparison of the FGF-7 structure to that of FGF-1 and FGF-2 revealed the strongly conserved Calpha backbone among the three FGF polypeptides and the surface hydrophobic patch that forms the primary receptor-binding domain. In contrast, a decrease and dispersion of the positive surface charge density characterized the heparin-binding domain of FGF-7 defined by homology to that of FGF-1 and FGF-2 in complexes with heparin. A simple heparin hexasaccharide that cocrystallized with FGF-1 and FGF-2 and protected both against protease in solution failed to exhibit the same properties with FGF-7. In contrast to FGF-1 and FGF-2, protection of FGF-7 was enhanced by heparin oligosaccharides of increased length with those exhibiting a 3-O-sulfate being the most effective. Protection of FGF-7 required interaction with specifically the fraction of crude heparin retained on antithrombin affinity columns. Conversely, heparin enriched by affinity for immobilized FGF-7 exhibited anti-factor Xa activity similar to that purified on an antithrombin affinity matrix. In contrast, an FGF-1 affinity matrix enriched the fraction of crude heparin with low anti-factor Xa activity. The results provide a structural basis to suggest that the unique FGF-7 heparin-binding (HB) domain underlies a specific restriction in respect to composition and length of the heparan sulfate motif that may impact specificity of localization, stability, and trafficking of FGF-7 in the microenvironment, and formation and activation of the FGFR2IIIb kinase signaling complex in epithelial cells.


Subject(s)
Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factors/metabolism , Heparitin Sulfate/metabolism , Amino Acid Sequence , Crystallization , Factor Xa/immunology , Factor Xa/metabolism , Fibroblast Growth Factor 1/chemistry , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/chemistry , Heparin/pharmacology , Molecular Sequence Data , Protein Structure, Secondary , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship
4.
Proc Natl Acad Sci U S A ; 98(20): 11336-40, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562460

ABSTRACT

We have reported that normal human salivary gland-derived epithelial cells exclusively express keratinocyte growth factor receptor (KGFR). In the process of malignant transformation of human salivary gland tumors, KGFR gene expression disappeared concomitantly with the de novo expression of the fibroblast growth factor receptor 1 (FGFR1) and FGFR4 genes. In the present study, we introduced wild-type KGFR cDNA or chimeric KGFR/FGFR1 cDNA, which encoded the extracellular domain of KGFR and the intracellular domain of FGFR1, into the HSY human salivary adenocarcinoma cell line. The KGFR tyrosine kinase suppressed the activity of FGF receptor substrate 2 (FRS2) and inhibited the growth of HSY by inducing differentiation and apoptosis in vitro and in vivo. Our results provided significant insight into the mechanism of KGFR tumor suppression and suggest that KGFR gene therapy might be a viable method of inhibiting human salivary adenocarcinoma growth.


Subject(s)
Adenocarcinoma/pathology , Epithelial Cells/cytology , Receptors, Fibroblast Growth Factor/physiology , Salivary Gland Neoplasms/pathology , Submandibular Gland/cytology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis , Cell Differentiation , Cell Division , Cells, Cultured , Female , Humans , Kinetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Nude , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Phosphorylation , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/physiology , Receptor, Fibroblast Growth Factor, Type 1 , Receptor, Fibroblast Growth Factor, Type 2 , Receptor, Fibroblast Growth Factor, Type 4 , Receptors, Fibroblast Growth Factor/genetics , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
5.
Nucleic Acids Res ; 29(17): 3557-65, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522825

ABSTRACT

The cell type-specific, mutually-exclusive alternative splicing of the fibroblast growth factor receptor 2 (FGFR2) pre-mRNA is tightly regulated. A sequence termed ISAR (intronic splicing activator and repressor) has been implicated as an important cis regulatory element in both activation of exon IIIb and repression of exon IIIc splicing in epithelial cells. In order to better understand how this single sequence could have dual roles, we transfected minigenes containing a series of 2-bp mutations in the 18 3'-most nucleotides of ISAR that we refer to as the ISAR core. Transfection of cells with dual-exon (IIIb and IIIc) minigenes revealed that mutation of terminal sequences of the core led to decreased exon IIIb inclusion and increased exon IIIc inclusion. Transfection of cells with single-exon IIIb minigenes and single-exon IIIc minigenes revealed that mutation of terminal sequences of the ISAR core led to decreased exon IIIb inclusion and increased exon IIIc inclusion, respectively. Nucleotides of the ISAR core responsible for exon IIIb activation appear to overlap very closely with those required for exon IIIc repression. We describe a model in which ISAR and a 5' intronic sequence known as IAS2 form a stem structure required for simultaneous exon IIIb activation and exon IIIc repression.


Subject(s)
Alternative Splicing , Exons/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Base Sequence , Gene Expression Regulation , Mutation , Plasmids/genetics , RNA/genetics , RNA/metabolism , Receptor, Fibroblast Growth Factor, Type 2 , Regulatory Sequences, Nucleic Acid/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
6.
Cancer Res ; 61(13): 5295-302, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431373

ABSTRACT

The abnormal appearance and age-dependent loss of resident fibroblast growth factor receptor-2 (FGFR2) and gain of activity of FGFR1 in epithelial cells is a hallmark of the slow progression to malignancy in some models of prostate cancer. Pericellular matrix heparan sulfate (HS) is an integral subunit of the FGFR tyrosine kinase complex that restricts activity in absence of FGF, facilitates binding of an activating FGF, and confers specificity for FGF isoforms. In this report, we isolated and purified HS proteoglycan (HSPG) from premalignant prostate tumor epithelial cells based on the ability of the HS chains to form a binary complex with immunoglobulin module II of the ectopic and progression-promoting FGFR1 that was competent to bind FGF. The FGFR1 affinity-purified product exhibited a specific activity of over 600 times that of crude cellular HSPG enriched from cell lysates by ion exchange chromatography. The purified preparation exhibited a single NH(2)-terminal sequence with 11 of 13 residues identical to syndecan-1. The activity of purified recombinant glutathione S-transferase-tagged syndecan-1 expressed in premalignant epithelial cells confirmed that syndecan-1 bears HS chains that exhibit the rare motif that forms the FGF-binding complex with ectopic FGFR1. These results are the first to identify by affinity purification a specific HSPG core protein, the HS chains of which act as an integral subunit of the FGFR complex. The results suggest that syndecan-1 provides HS chains in premalignant epithelial cells to both the FGFR2- and FGFR1-signaling complexes that are integral to their dual roles in progression to malignancy.


Subject(s)
Membrane Glycoproteins/metabolism , Proteoglycans/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromatography, Affinity , DNA, Complementary/genetics , Fibroblast Growth Factors/metabolism , Heparin/analogs & derivatives , Heparin/genetics , Heparin/isolation & purification , Heparin/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/isolation & purification , Molecular Sequence Data , Precancerous Conditions/chemistry , Precancerous Conditions/metabolism , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/metabolism , Proteoglycans/genetics , Proteoglycans/isolation & purification , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Fibroblast Growth Factor, Type 1 , Receptor, Fibroblast Growth Factor, Type 2 , Recombinant Proteins/metabolism , Syndecan-1 , Syndecans
7.
J Agric Food Chem ; 49(6): 3051-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11410008

ABSTRACT

This study was undertaken to characterize the pectin from four citrus species and to determine their in vitro inhibitory activities on the binding of fibroblast growth factor (FGF) to the FGF receptor (FGFR). Pectin from various parts of lemon, grapefruit, tangerine, and orange were isolated and characterized. Tangerine had the highest pectin content among the four citrus species. Segment membrane contained as much as or more pectin than flavedo/albedo. Anhydrogalacturonic content was highest in pectin from segment membrane of tangerine and flavedo/albedo of grapefruit. Lemon pectin contained the highest methoxyl content (MC), and grapefruit contained the largest proportion of lower molecular weight (<10000 Da) pectin. Tangerine contained the highest neutral sugar in both flavedo/albedo and segment membrane. The interdependency of heparin on factor-receptor interaction provides a means for identifying new antagonists of growth factor activity and thus for treatment of various diseases. These results showed that pectin significantly inhibited the binding of FGF-1 to FGFR1 in the presence of 0.1 microg/mL heparin. The pectin from the segment membrane of lemon was the most potent inhibitor. The inhibition activity was significantly correlated with sugar content, MC, and size of pectin. Kinetic studies revealed a competitive nature of pectin inhibition with the heparin, a crucial component of the FGF signal transduction process. The observation that the heparin-dependent biological activity of FGF signal transduction is antagonized by citrus pectin should be further investigated for the use of these pectins as anti-growth factor agents for potential health benefits.


Subject(s)
Citrus/chemistry , Fibroblast Growth Factors/metabolism , Pectins/analysis , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Fibroblast Growth Factors/chemistry , Heparin/chemistry , Heparin/metabolism , Kinetics , Molecular Weight , Pectins/classification , Signal Transduction
8.
Biochem Biophys Res Commun ; 283(4): 791-7, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11350054

ABSTRACT

Models of the oligomeric FGF signaling complex, including those derived from crystal structures, vary in stoichiometry and arrangement of the three subunits comprised of heparin/heparan sulfate chains, FGFR tyrosine kinase and activating FGF. Here, using covalent affinity crosslinking of radiolabeled FGF7 to binary complexes of FGFR2IIIb and heparin, we show that two molecules of FGF7 contact each FGFR2IIIb. This supports models that propose a dimeric complex of two units with stoichiometry 1 FGF:1 FGFR in which each FGF contacts both FGFR. The bivalent FGF7 contact was dependent on the full-length amino terminus of FGF7alpha and the intracellular domain of FGFR2IIIb extending through the juxtamembrane domain and the beta1 and beta2 strands of the kinase which is required for ATP binding. We propose that the differences in crosslinking report differences in relationships among subunits in the ectodomain of the complex that are affected by the amino terminus of FGF and the FGFR intracellular domain. From this, we suggest the corollary that conformational relationships among subunits in the ectodomain are transmitted to the intracellular and ATP binding domains during activation of the complex.


Subject(s)
Fibroblast Growth Factors/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Sequence , Base Sequence , DNA Primers , Fibroblast Growth Factor 7 , Ligands , Molecular Sequence Data , Protein Binding , Receptors, Fibroblast Growth Factor/chemistry
9.
J Biol Chem ; 276(6): 4158-67, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11042206

ABSTRACT

Exons IIIb and IIIc of the FGFR2 gene are alternatively spliced in a mutually exclusive manner in different cell types. A switch from expression of FGFR2IIIb to FGFR2IIIc accompanies the transition of nonmalignant rat prostate tumor epithelial cells (DTE) to cells comprising malignant AT3 tumors. Here we used transfection of minigenes with and without alterations in reading frame and with and without introns to examine how translation affects observed FGFR2 splice products. We observed that nonsense mutations in other than the last exon led to a dramatic reduction in mRNA that is abrogated by removal of downstream introns in both DTE and AT3 cells. The mRNA, devoid of both IIIb and IIIc exons (C1-C2), is a major splice product from minigenes lacking an intron downstream of the second common exon C2. From these observations, we suggest that repression of exon IIIc and activation of exon IIIb inclusion in DTE cells lead to the generation of both C1-IIIb-C2 and C1-C2 products. However, the C1-C2 product from the native gene is degraded due to a frameshift and a premature termination codon caused by splicing C1 and C2 together. Derepression of exon IIIc and repression of exon IIIb lead to the generation of both C1-IIIc-C2 and C1-C2 products in AT3 cells, but the C1-C2 product is degraded. The C1-IIIb-IIIc-C2 mRNA containing a premature termination codon in exon IIIc was present, but at apparently trace levels in both cell types. The nonsense-mediated mRNA decay pathway and cell type-dependent rates of inclusion of exons IIIb and IIIc result in the mutually exclusive expression of FGFR2IIIb and IIIc.


Subject(s)
Alternative Splicing , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Animals , Exons , Male , Molecular Sequence Data , Open Reading Frames , Protein Biosynthesis , Rats , Rats, Inbred F344 , Receptor, Fibroblast Growth Factor, Type 2 , Reverse Transcriptase Polymerase Chain Reaction , Transfection
10.
Cancer Res ; 60(15): 4049-52, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10945607

ABSTRACT

A nested reverse transcription-PCR analysis of FGFR3 from human colorectal carcinomas revealed novel mutant transcripts caused by aberrant splicing and activation of cryptic splice sequences. Two aberrantly spliced transcripts were detected with high frequency in 50% of 36 primary tumors and in 60% of 10 human colorectal cancer cell lines. Most transcripts used normal splice sites but skipped or included exons 8 and 9. Two mutant transcripts arose from cryptic splice donor sites in exon 7 that spliced to exon 10. The predicted translation products would exhibit frameshifts and a premature termination codon in exon 10. We propose that dysregulation of mRNA splicing frequently generates an aberrant FGFR3 transcript that may confer a selectable advantage on clones of cells in colorectal tumorigenesis.


Subject(s)
Alternative Splicing/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Protein-Tyrosine Kinases , RNA, Messenger/genetics , Receptors, Fibroblast Growth Factor/genetics , Amino Acid Sequence , Codon, Nonsense , Colorectal Neoplasms/metabolism , Down-Regulation , Exons/genetics , Frameshift Mutation , Humans , Molecular Sequence Data , Peptide Chain Termination, Translational , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/biosynthesis , Tumor Cells, Cultured
11.
Biochem Biophys Res Commun ; 272(3): 830-6, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10860838

ABSTRACT

Epithelial cells, which express FGFR2IIIb, bind and respond to FGF-1, FGF-7 and FGF-10, but not FGF-2. Stromal cells, which bind and respond to FGF-1 and FGF-2, but not FGF-7 and FGF-10, express FGFR2IIIc or FGFR1IIIc. Here we show that when both isolated FGFR2betaIIIb and FGFR2betaIIIc or their common Ig module II are allowed to affinity select heparin from a mixture, the resultant binary complexes bound FGF-1, FGF-2, and FGF-7 with nearly equal affinity. In addition, FGF-2 and FGF-7 bound to both heparin-Ig module IIIb and IIIc complexes, but FGF-1 bound to neither Ig module III. The results show that in isolation both Ig modules II and III of FGFR2 can interact with heparin and that each exhibits a binding site for FGF. We suggest that the specificity of FGFR2IIIb and FGFR2IIIc is dependent on the cell membrane environment and heparin/heparan sulfate. Ig modules II and III cooperate both within monomers and across dimers with cellular heparan sulfates to confer cell type-dependent specificity of the FGFR complex for FGF.


Subject(s)
Heparin/metabolism , Immunoglobulins/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Sequence , Animals , Binding Sites/drug effects , Cell Membrane/metabolism , Fibroblast Growth Factors/metabolism , Heparin/pharmacology , Ligands , Models, Biological , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Solubility , Substrate Specificity/drug effects , Thermodynamics
12.
J Biol Chem ; 275(20): 15482-9, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10809780

ABSTRACT

Heparan sulfate-regulated transmembrane tyrosine kinase receptor FGFR4 is the major FGFR isotype in mature hepatocytes. Fibroblast growth factor has been implicated in the definition of liver from foregut endoderm where FGFR4 is expressed and stimulation of hepatocyte DNA synthesis in vitro. Here we show that livers of mice lacking FGFR4 exhibited normal morphology and regenerated normally in response to partial hepatectomy. However, the FGFR4 (-/-) mice exhibited depleted gallbladders, an elevated bile acid pool and elevated excretion of bile acids. Cholesterol- and bile acid-controlled liver cholesterol 7alpha-hydroxylase, the limiting enzyme for bile acid synthesis, was elevated, unresponsive to dietary cholesterol, but repressed normally by dietary cholate. Expression pattern and cholate-dependent, cholesterol-induced hepatomegaly in the FGFR4 (-/-) mice suggested that activation of receptor interacting protein 140, a co-repressor of feed-forward activator liver X receptor alpha, may mediate the negative regulation of cholesterol- and bile acid-controlled liver cholesterol 7alpha-hydroxylase transcription by FGFR4 and cholate. The results demonstrate that transmembrane sensors interface with metabolite-controlled transcription networks and suggest that pericellular matrix-controlled liver FGFR4 in particular may ensure adequate cholesterol for cell structures and signal transduction.


Subject(s)
Bile Acids and Salts/biosynthesis , Cholesterol/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/physiology , Animals , Cell Membrane/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Gallbladder/metabolism , Gallbladder/pathology , Gene Expression Regulation, Enzymologic , Hepatectomy , Hydroxymethylglutaryl CoA Reductases/genetics , Liver/cytology , Liver/physiology , Liver Regeneration , Mice , Mice, Knockout , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 4 , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics
13.
Proc Natl Acad Sci U S A ; 97(3): 1113-8, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10655493

ABSTRACT

Recent experiments have established that Sox9 is required for chondrocyte differentiation. Here, we show that fibroblast growth factors (FGFs) markedly enhance Sox9 expression in mouse primary chondrocytes as well as in C3H10T1/2 cells that express low levels of Sox9. FGFs also strongly increase the activity of a Sox9-dependent chondrocyte-specific enhancer in the gene for collagen type II. Transient transfection experiments using constructs encoding FGF receptors strongly suggested that all FGF receptors, FGFR1-R4, can transduce signals that lead to the increase in Sox9 expression. The increase in Sox9 levels induced by FGF2 was inhibited by a specific mitogen-activated protein kinase kinase (MAPKK)/mitogen-activated protein kinase/ERK kinase (MEK) inhibitor U0126 in primary chondrocytes. In addition, coexpression of a dual-specificity phosphatase, CL100/MKP-1, that is able to dephosphorylate and inactivate mitogen-activated protein kinases (MAPKs) inhibited the FGF2-induced increase in activity of the Sox9-dependent enhancer. Furthermore, coexpression of a constitutively active mutant of MEK1 increased the activity of the Sox9-dependent enhancer in primary chondrocytes and C3H10T1/2 cells, mimicking the effects of FGFs. These results indicate that expression of the gene for the master chondrogenic factor Sox9 is stimulated by FGFs in chondrocytes as well as in undifferentiated mesenchymal cells and strongly suggest that this regulation is mediated by the MAPK pathway. Because Sox9 is essential for chondrocyte differentiation, we propose that FGFs and the MAPK pathway play an important role in chondrogenesis.


Subject(s)
Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System , Animals , Base Sequence , Butadienes/pharmacology , Cartilage/cytology , Cell Differentiation , Gene Expression Regulation/physiology , Mesoderm/cytology , Mice , Mice, Inbred C3H , Molecular Sequence Data , Nitriles/pharmacology , Recombinant Fusion Proteins/pharmacology , Transfection
14.
J Med Genet ; 37(1): 26-32, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10633130

ABSTRACT

The Antley-Bixler syndrome has been thought to be caused by an autosomal recessive gene. However, patients with this phenotype have been reported with a new dominant mutation at the FGFR2 locus as well as in the offspring of mothers taking the antifungal agent fluconazole during early pregnancy. In addition to the craniosynostosis and joint ankylosis which are the clinical hallmarks of the condition, many patients, especially females, have genital abnormalities. We now report abnormalities of steroid biogenesis in seven of 16 patients with an Antley-Bixler phenotype. Additionally, we identify FGFR2 mutations in seven of these 16 patients, including one patient with abnormal steroidogenesis. These findings, suggesting that some cases of Antley-Bixler syndrome are the outcome of two distinct genetic events, allow a hypothesis to be formulated under which we may explain all the differing and seemingly contradictory circumstances in which the Antley-Bixler phenotype has been recognised.


Subject(s)
Abnormalities, Multiple/genetics , Craniosynostoses/genetics , Genitalia, Female/abnormalities , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Abnormalities, Drug-Induced/genetics , Ankle Joint/abnormalities , Ankylosis/genetics , Eye Abnormalities/genetics , Female , Fluconazole/adverse effects , Humans , Infant , Karyotyping , Male , Pregnancy , Prenatal Exposure Delayed Effects , Receptor, Fibroblast Growth Factor, Type 2 , Sex Characteristics , Syndrome
15.
Endocr J ; 46(3): 405-13, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10503993

ABSTRACT

It has been suggested that prostate homeostasis is regulated indirectly by androgens through stromal-epithelial interactions in part by factors from the stromal cells acting on receptors in epithelial cells. In this report, the role of fibroblast growth factor (FGF)-10 in prostatic epithelial proliferation was investigated. The expression of FGF-10 mRNA was apparent in primary-cultured stromal cells, but not in epithelial cells derived from human tissue from patients with benign prostatic hyperplasia (BPH). The mitogenic activity of human recombinant FGF-10 assessed by 5-bromo-2'-deoxyuridine (BrdU) incorporation was demonstrated in isolated epithelial cells, but not in cultured stromal cells. No mitogenic activity of dihydrotestosterone (DHT) for either epithelial or stromal cells could be demonstrated, but quantitative PCR (real-time PCR) with a double-labeled fluorogenic probe demonstrated that expression of FGF-10 in stromal cells was enhanced 5.3-fold at a DHT concentration of 100 pM. Androgen receptor mRNA levels showed no significant change with DHT at concentrations less than 100 pM, but were reduced to 50% of control levels at a DHT concentration of 10 nM. These results suggest that stromal-derived FGF-10 stimulates human prostatic epithelial growth and its mRNA expression is induced by androgens, without an increase in the androgen receptor mRNA. Moreover, FGF-10 may be involved in the development or support of human BPH.


Subject(s)
Androgens/pharmacology , Cell Division/drug effects , Fibroblast Growth Factors/pharmacology , Prostate/pathology , Prostatic Hyperplasia/pathology , Stromal Cells/metabolism , Cells, Cultured , Dihydrotestosterone/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblast Growth Factor 10 , Fibroblast Growth Factors/genetics , Fluorescent Dyes , Gene Expression/drug effects , Humans , Male , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Recombinant Proteins/pharmacology
17.
J Biol Chem ; 274(31): 21511-4, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419453

ABSTRACT

A divalent cation-dependent association between heparin or heparan sulfate and the ectodomain of the fibroblast growth factor (FGF) receptor kinase (FGFR) restricts FGF-independent trans-phosphorylation between self-associated FGFR and determines specificity for and mediates binding of activating FGF. Here we show that only the fraction of commercial heparin or rat liver heparan sulfate which binds to immobilized antithrombin formed an FGF-binding binary complex with the ectodomain of the FGFR kinase. Conversely, only the fraction of heparin that binds to immobilized FGFR inhibited Factor Xa in the presence of antithrombin. Only the antithrombin-bound fraction of heparin competed with (3)H-heparin bound to FGFR in absence of FGF, whereas both antithrombin-bound and unretained fractions competed with radiolabeled heparin bound independently to FGF-1 and FGF-2. The antithrombin-bound fraction of heparin was required to support the heparin-dependent stimulation of DNA synthesis of endothelial cells by FGF-1. The requirement for divalent cations and the antithrombin-binding motif distinguish the role of heparan sulfate as an integral subunit of the FGFR complex from the wider range of effects of heparan sulfates and homologues on FGF signaling through FGFR-independent interactions with FGF.


Subject(s)
Fibroblast Growth Factors/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Liver/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Anticoagulants/isolation & purification , Anticoagulants/metabolism , Antithrombins/metabolism , Binding, Competitive , Chromatography, Affinity , Fibroblast Growth Factor 1 , Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/isolation & purification , Heparin/metabolism , Heparitin Sulfate/isolation & purification , Intestinal Mucosa/metabolism , Kinetics , Phosphorylation , Rats , Receptors, Fibroblast Growth Factor/isolation & purification , Swine
18.
J Biol Chem ; 274(22): 15947-52, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10336501

ABSTRACT

A divalent cation-dependent association between heparin or heparan sulfate and the ectodomain of the FGF receptor kinase (FGFR) restricts FGF-independent trans-phosphorylation and supports the binding of activating FGF to self-associated FGFR. Here we show that in contrast to heparin, cellular heparan sulfate forms a binary complex with FGFR that discriminates between FGF-1 and FGF-2. FGFR type 4 (FGFR4) in liver parenchymal cells binds only FGF-1, whereas FGFR1 binds FGF-1 and FGF-2 equally. Cell-free complexes of heparin and recombinant FGFR4 bound FGF-1 and FGF-2 equally. However, in contrast to FGFR1, when recombinant FGFR4 was expressed back in epithelial cells by transfection, it failed to bind FGF-2 unless heparan sulfate was depressed by chlorate or heparinase treatment. Isolated heparan sulfate proteoglycan (HSPG) from liver cells in cell-free complexes with FGFR4 restored the specificity for FGF-1 and supported the binding of both FGF-1 and FGF-2 when complexed with FGFR1. In contrast, FGF-2 bound equally well to complexes of both FGFR1 and FGFR4 formed with endothelial cell-derived HSPG, but the endothelial HSPG was deficient for the binding of FGF-1 to both FGFR complexes. These data suggest that a heparan sulfate subunit is a cell type- and FGFR-specific determinant of the selectivity of the FGFR signaling complex for FGF. In a physiological context, the heparan sulfate subunit may limit the redundancy among the current 18 FGF polypeptides for the 4 known FGFR.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Cells, Cultured , Fibroblast Growth Factor 1 , Heparan Sulfate Proteoglycans/metabolism , Liver/metabolism , Rats , Recombinant Proteins/metabolism , Signal Transduction
19.
J Biol Chem ; 274(18): 12827-34, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10212269

ABSTRACT

Fibroblast growth factor (FGF)-10, a homologue of FGF-7, is expressed significantly in normal rat prostate tissue, well differentiated rat prostate tumors with an epithelial and stromal compartment and only in derived prostate stromal cells in culture. Similar to FGF-7, recombinant rat FGF-10 was a specific mitogen for prostate epithelial cells. In contrast to FGF-7 which is widely expressed among stromal cells in tissues, the expression of FGF-10 correlated with the presence of stromal cells of muscle origin. Radioreceptor binding assays and covalent cross-linking analysis revealed that FGF-10 binds with an affinity equal to FGF-7 to resident epithelial cell receptor, FGFR2IIIb, but unlike FGF-7 also binds the IIIb splice variant of FGFR1. Analysis of mRNA expression by RNase protection revealed that, similar to FGF-7, the expression of FGF-10 was responsive to androgen in stromal cells from normal prostate and non-malignant differentiated tumors. Although FGF-10 cDNA exhibits a signal sequence for secretion, cultured stromal cells exhibit strictly a cell-associated FGF-10 antigen that correlates with an alternately translated intracellular isoform. FGF-10 requires 1.4 times higher NaCl for elution from immobilized heparin than does FGF-7 and binds to four times the number of sites on the pericellular matrix of epithelial cells. The results show that prostate stromal cell-derived FGF-10, like FGF-7, exhibits the properties of an andromedin which may indirectly mediate control of epithelial cell growth and function by androgen. Although FGF-10 and FGF-7 bind and activate the same resident epithelial cell receptor (FGFR2IIIb), differences in cell type of origin, compartmentation by alternate translation, the affinity for FGFR1IIIb, and access to FGFR by differential interaction with pericellular matrix heparan sulfate suggest they may play both independent and compensatory roles in prostate homeostasis.


Subject(s)
Fibroblast Growth Factors/metabolism , Prostate/metabolism , Animals , Base Sequence , DNA Primers , Epithelial Cells/metabolism , Fibroblast Growth Factor 10 , Heparin/metabolism , Male , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/metabolism , Tumor Cells, Cultured
20.
Biochemistry ; 38(1): 160-71, 1999 Jan 05.
Article in English | MEDLINE | ID: mdl-9890894

ABSTRACT

The assembly and activation of oligomeric complexes of FGF, the transmembrane receptor kinase (FGFR), and heparan sulfate transmit intracellular signals regulating growth and function of cells. An understanding of the structural relationships between the three subunits and their redundancy and specificity is essential for understanding the ubiquitous FGF signaling system in health and disease. Previously, we reported that a primary heparin or heparan sulfate binding site resides in a distinct sequence in immunoglobulin (Ig)-like module II of the three modules of FGFR. Here we report that in the absence of flanking sequences, isolated Ig module II of FGFR1 supports the binding of FGF-1, FGF-2, and FGF-7 in respective order of affinity. None of the three FGFs detectably bind Ig module I or the IIIb and IIIc splice variants of Ig module III in the absence of flanking sequences. Ig module I and the C-terminus of Ig module III are dispensable for high-affinity binding of FGF-1, FGF-2, and FGF-7. Alterations in highly conserved Ig module II in the heparin binding domain and substitution of individual sequence domains spanning the entire sequence of Ig module II with those from Ig module I obliterated FGF binding. Addition of a specific number of FGFR sequences to the C-terminus of Ig module II resulted in a gain in affinity for FGF-7. Several site-specific alterations in the C-terminus of full-length FGFR1IIIc, an isoform that otherwise absolutely rejects FGF-7, resulted in gain of FGF-7 binding. These results suggest that a complex of Ig module II and heparan sulfate is the base common active core of the FGFR ectodomain and that flanking structural domains modify FGF affinity and determine specificity.


Subject(s)
Fibroblast Growth Factors/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Receptors, Fibroblast Growth Factor/chemistry , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Conserved Sequence , Fibroblast Growth Factor 1 , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Growth Substances/metabolism , Heparin/metabolism , Heparin/physiology , Immunoglobulins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...