Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters








Publication year range
1.
Fungal Syst Evol ; 2: 45-56, 2018 Dec.
Article in English | MEDLINE | ID: mdl-32467887

ABSTRACT

Caliciopsis pleomorpha sp. nov. is described from a severe stem canker disease of cultivated Eucalyptus cladocalyx 'Nana' (dwarf sugar gum) in Australia. The fungus is a pleomorphic ascomycete (Coryneliales), with pycnidial (pleurophoma-like) and hyphomycetous (phaeoacremonium-like) morphs, and differs in these respects and in ITS sequences from other Caliciopsis spp. The fungus was also found associated with cankers on other Eucalyptus species growing in native habitats, and was successfully inoculated under glasshouse conditions into a wide range of Eucalyptus species on which it caused cankers of varying severity.

2.
J Cell Biol ; 152(6): 1307-12, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11257130

ABSTRACT

Although growth factor receptors are generally thought to carry out their role in signal transduction at the cell surface, many of these transmembrane proteins translocate to the nucleus after ligand stimulation. Here, we show that the nuclear translocation of fibroblast growth factor receptor (FGFR)1 occurs via a mechanism distinct from classical nuclear import but dependent on importin beta, a component of multiple nuclear import pathways. Furthermore, we show that nuclear FGFR1 induces c-Jun and is involved in the regulation of cell proliferation. These data are the first description of a nuclear import pathway for transmembrane growth factor receptors and elucidate a novel signal transduction pathway from the cell surface to the nucleus.


Subject(s)
Active Transport, Cell Nucleus/physiology , Cell Division/physiology , Nuclear Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology , 3T3 Cells , Active Transport, Cell Nucleus/drug effects , Adenosine Triphosphate/metabolism , Animals , Antimetabolites/pharmacology , Cell Cycle/physiology , Cell Fractionation , Deoxyglucose/pharmacology , Enzyme Inhibitors/pharmacology , Immunoblotting , Karyopherins , Mice , Microscopy, Confocal , Oligomycins/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Recombinant Proteins/metabolism , Transfection
3.
Exp Cell Res ; 260(2): 189-98, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11035913

ABSTRACT

Treatment of confluent chicken embryo fibroblasts (CEFs) with trypsin results in a dose- and time-dependent increase in c-Src protein tyrosine kinase (PTK) activity. A similar, but less marked, increase in c-Src PTK activity occurs upon incubation of CEFs in calcium-free phosphate-buffered saline, which also causes a decrease in cell-substrate adhesion. The increase in c-Src PTK activity following disruption of cell-substrate adhesion correlates with a decrease in the phosphorylation of c-Src at the regulatory site, Tyr527. The phosphotyrosine phosphatase inhibitor phenylarsine oxide blocks the increase in c-Src PTK activity seen following treatment with trypsin and the morphological changes associated with the disruption of cell-substrate adhesion. In contrast, disruption of cell-substrate adhesion causes a decrease in FAK PTK activity that rapidly returns to control levels when the cells are plated on fibronection-coated dishes. Treatment of cells with cytochalasin D, which disrupts actin filaments but not cell-substrate adhesion, causes only a slight increase in c-Src PTK activity. Thus, these studies demonstrate a ligand-independent mechanism for the activation of c-Src that is consistent with its role in both cell adhesion and cell motility. Furthermore, these data suggest that similar to adhesion, loss of adhesion is not a passive process but can activate specific signaling pathways that may have significant effects on cellular function.


Subject(s)
Cell Adhesion/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Animals , Arsenicals/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Chick Embryo , Cytochalasin D/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , Fibronectins/metabolism , Focal Adhesion Protein-Tyrosine Kinases , Phosphorylation , Polylysine/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Trypsin , Tyrosine/metabolism
4.
J Biol Chem ; 275(11): 7771-8, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10713090

ABSTRACT

Using the cytoplasmic domain of fibroblast growth factor receptor 1 (FGFR1) as bait in a yeast two-hybrid screen, Grb14 was identified as a FGFR1 binding partner. A kinase-inactive mutant of FGFR1 failed to interact with Grb14, indicating that activation of FGFR1 is necessary for binding. Deletion of the C-tail or mutation of both C-tail tyrosine residues of FGFR1 to phenylalanine abolished binding, and deletion of the juxtamembrane domain of the receptor reduced binding, suggesting that Grb14 binds to FGFR1 at multiple sites. Co-immunoprecipitation and in vitro binding assays demonstrated that binding of Grb14 to FGFR1 in mammalian cells was dependent on receptor activation by fibroblast growth factor-2 (FGF-2). Deletion of the Src homology 2 (SH2) domain of Grb14 reduced but did not block binding to FGFR1 and eliminated dependence on receptor activation. The SH2 domain alone bound both FGFR1 and platelet-derived growth factor receptor, whereas full-length Grb14 bound only FGFR1, suggesting that regions upstream of the SH2 domain confer specificity for FGFR1. Grb14 was phosphorylated on serine and threonine residues in unstimulated cells, and treatment with FGF-2 enhanced this phosphorylation. Expression of exogenous Grb14 inhibited FGF-2-induced cell proliferation, whereas a point-mutated form of Grb14 incapable of binding to FGFR1 enhanced FGF-2-induced mitogenesis. These data demonstrate an interaction between activated FGFR1 and Grb14 and suggest a role for Grb14 in FGF signaling.


Subject(s)
Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Fibroblast Growth Factors/metabolism , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1 , Sequence Homology, Amino Acid , Signal Transduction , Two-Hybrid System Techniques , src Homology Domains
5.
J Cell Physiol ; 178(2): 144-53, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10048578

ABSTRACT

Basic fibroblast growth factor (FGF2) is synthesized as four isoforms with molecular weights of 24, 22.5, 22, and 18 kDa, with each of the three higher molecular weight forms (hmwFGF2) produced by the initiation of translation at one of three upstream CUG codons. We have shown that bovine arterial endothelial cells export the high molecular weight forms of FGF2 (hmwFGF2) in a 17beta-estradiol-dependent manner (Piotrowicz et al., 1997, J Biol Chem 272:7042-7047). To determine whether the hmwFGF2 forms affected cell behavior after release, we evaluated the effect of recombinant hmwFGF2 on the growth and migration of endothelial cells and mammary carcinoma cells (MCF-7). Treatment with the recombinant protein resulted in the inhibition of endothelial cell migration by 45% and MCF-7 cell migration by 70%. HmwFGF2-dependent inhibition was observed when endothelial cell migration was stimulated by 18 kDa FGF2 or vascular endothelial growth, and MCF cell migration was stimulated with insulin-like growth factor. In each case, inclusion of an antibody against the 55 amino acid amino terminal end of 24 kDa FGF2 abrogated the inhibition of migration, while antibodies to the 18 kDa FGF2 domain had no effect. When endothelial cells were cultured under conditions which promoted export of hmwFGF2, a 40% decrease in motility was observed which was reversed by the antibodies to the 24 kDa FGF2. Thus, both recombinant and endogenously produced hmw-FGF2 are capable of inhibiting migration. In contrast to the ubiquitous effect on migration, hmwFGF2 had no effect on endothelial cell growth but stimulated MCF-7 growth equally as well as the 18 kDa FGF2 (threefold). Antibodies to the 18 kDa domain of 24 kDa FGF2 blocked the growth-promoting activity of hmwFGF2, but those to the amino terminal end were ineffective. These data suggest that hmwFGF2 has dual activities, an inhibitory effect on cell migration and a growth-stimulating effect. The two activities can be localized to different parts of hmwFGF2: inhibitory activity to the amino terminal 55 amino acids (which are absent from the 18 kDa FGF2) and growth-promoting activity to the 18 kDa domain. Therefore, the ratio of hmwFGF2 and 18 kDa FGF2 in the extracellular space may provide a mechanism of control for angiogenesis and mammary tumor development.


Subject(s)
Cell Division/drug effects , Cell Movement/drug effects , Fibroblast Growth Factor 2/pharmacology , Animals , COS Cells , Cattle , Cell Division/physiology , Cell Line , Cell Movement/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/injuries , Female , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/physiology , Humans , Molecular Weight , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Wound Healing/drug effects , Wound Healing/physiology
6.
Neuroreport ; 9(11): 2501-3, 1998 Aug 03.
Article in English | MEDLINE | ID: mdl-9721922

ABSTRACT

TA20 is a cDNA clone which was previously reported to encode a novel neuronal differentiation factor. However, the majority of this clone has near perfect DNA sequence homology with cytochrome b mitochondrial sequence and an additional 5' region which lacks homology with any previously reported sequence. Furthermore, careful study of the library construction method indicates that the full length TA20 clone is likely an artifact arising from the head-to-head blunt-ended ligation of two unrelated cDNAs. Thus, the effects on growth and neurite outgrowth observed in correlation with the overexpression of TA20 in neuronal cells, as well as its mRNA distribution in the developing rat brain require further study.


Subject(s)
Cytochrome b Group/physiology , Neurons/physiology , Transcription Factors/physiology , 5' Untranslated Regions/genetics , 5' Untranslated Regions/physiology , Animals , Base Sequence , Cell Differentiation/physiology , Cytochrome b Group/genetics , Mice , Molecular Sequence Data , Neurites/physiology , Neurons/enzymology , Protein Biosynthesis , RNA, Messenger/biosynthesis , Rats , Reading Frames , Transcription Factors/genetics
7.
Glia ; 22(2): 202-10, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9537840

ABSTRACT

Astrocytes play a critical role in the development of the CNS and its response to injury and disease. A key indicator of astrocyte activation is the increased accumulation of intermediate filaments composed of glial fibrillary acidic protein (GFAP). Treatment of astrocytes in vitro with transforming growth factor-beta1 (TGF-beta1) produced little morphological change, but resulted in a significant increase in GFAP mRNA and protein. Treatment with basic fibroblast growth factor (FGF-2) produced a dramatic change from a polygonal to a stellate morphology, and resulted in a significant decrease in GFAP mRNA and protein. FGF-2 also inhibited the TGF-beta1-mediated increase in GFAP mRNA and protein. Cycloheximide did not block the effects of TGF-beta1 or FGF-2 on GFAP mRNA levels, but blocked the inhibitory effects of FGF-2 on the TGF-beta1-mediated increase in GFAP expression. All effects of FGF-2 were blocked by co-incubation with 5'-methylthioadenosine, a specific inhibitor of FGF-2-induced tyrosine kinase activity and FGF receptor (FGFR) autophosphorylation. We also examined astrocyte expression of FGFR, and demonstrate the presence of FGFR 1 and 2, and lower levels of FGFR 3. Our results demonstrate that TGF-beta1 and FGF-2 cause differential effects on the astrocyte cytoskeleton and morphology, suggesting an uncoupling of process outgrowth from GFAP synthesis.


Subject(s)
Astrocytes/metabolism , Fibroblast Growth Factor 2/pharmacology , Glial Fibrillary Acidic Protein/biosynthesis , Transforming Growth Factor beta/pharmacology , Animals , Astrocytes/drug effects , Blotting, Northern , Blotting, Western , Cells, Cultured , Cross-Linking Reagents , Cycloheximide/pharmacology , Deoxyadenosines/pharmacology , Immunohistochemistry , Iodine Radioisotopes , Precipitin Tests , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Rats , Rats, Sprague-Dawley , Thionucleosides/pharmacology
8.
Clin Exp Metastasis ; 15(6): 620-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9344046

ABSTRACT

Mitotoxins targeted via high-affinity growth factor receptors on the cell surface are a potential means of anticancer therapy. We have evaluated the effect of a chemically conjugated (FGF2-SAP) and a fusion protein (rFGF2-SAP) mitotoxin containing FGF-2 and saporin on normal (FHs 738B1) and malignant bladder cell lines (HT1197, TCCSUP, EJ-6, and RT4). The FGF-saporins demonstrated potent cytotoxicity in malignant bladder cell lines with an ID50 range of 0.13-13.6 nM, whereas cells derived from normal fetal bladder (FHs 738B1) were less sensitive to FGF2-saporins (ID50 > 100 nM). Greater than a 100-fold difference in cytotoxicity between FGF-saporins and unconjugated saporin was observed. Assessment of cellular FGF-2 content and secretion showed that FHs 738B1 and TCCSUP contained and secreted significantly more FGF-2 compared to other cell lines tested. (125)I-FGF-2 receptor binding studies showed the presence of high-affinity (pM) FGF receptors on all bladder cell lines. Cross-linking studies revealed the presence of a major receptor-ligand complex of 90 kDa on FHs 738B1 and 160-170 kDa on the other bladder cell lines. All cell lines studied, except RT4, expressed solely FGFR-1. These studies demonstrate that FGF2-saporins have antiproliferative activity on human bladder cancer cell lines. However, the number of high-affinity FGF receptors, and FGF-2 cellular content and secretion are not absolute determinants of cellular sensitivity to FGF2-saporins.


Subject(s)
Fibroblast Growth Factors/pharmacology , Immunotoxins , N-Glycosyl Hydrolases , Plant Proteins/pharmacology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder/drug effects , Cell Line , Humans , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/pharmacology , Ribosome Inactivating Proteins, Type 1 , Saporins , Tumor Cells, Cultured , Urinary Bladder/metabolism
9.
Oncogene ; 14(18): 2201-11, 1997 May 08.
Article in English | MEDLINE | ID: mdl-9174056

ABSTRACT

In this study we describe the presence of high affinity FGF-2 binding sites in the nuclei of U251MG glioma cells (K(d)=7 pM). Immunoprecipitation of total cell extracts with FGF receptor (FGFR) 1-4 antibodies showed that U251MG glioma cells express only FGFR1. [125I]FGF-2 cross linking to nuclear extracts followed by FGFR1 immunoprecipitation showed that FGFR1 may account for the nuclear FGF-2 binding sites. Western blot analysis demonstrated the presence of 103, 118 kDa and small amounts of 145 kDa FGFR1 isoforms in the nuclei of glioma cells. All isoforms contain both the C- and N-terminal domains. Nuclear FGFR1 retains kinase activity. Immunocytochemistry using confocal microscopy showed specific FGFR1 immunoreactivity within the nuclear interior. In continuously proliferating glioma cells, nuclear FGFR1 is constitutively expressed, independent of cell density. In contrast, in nontransformed human astrocytes, nuclear FGFR1 levels fluctuate with the proliferative state of the cell. In quiescent, confluent astrocytes nuclear FGFR1 protein was depleted. An accumulation of nuclear FGFR1 was observed following the transition to a subconfluent, proliferating state. Transfection of a pcDNA3.1-FGFR1 expression vector into glioma cells that do not express FGFR1 resulted in the nuclear accumulation of FGFR1, increased cell proliferation, and stimulated transition from the G0/G1 to the S-phase of the cell cycle. The increased proliferative rate was resistant to inhibition by the cell-impermeable FGF binding antagonist, myoinositol hexakis [dihydrogen phosphate]. Our results suggest that the constitutive nuclear presence of FGFR1 contributes to the increased proliferation of glioma cells while the transient nuclear accumulation of FGFR1 in normal astrocytes may play a role in the transition to a reactive state.


Subject(s)
Cell Nucleus/metabolism , Glioma/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/metabolism , Astrocytes/chemistry , Astrocytes/metabolism , Blotting, Western , Cell Division/physiology , Cell Nucleus/chemistry , Cells, Cultured , Fibroblast Growth Factor 2/metabolism , Glioma/chemistry , Humans , Immunohistochemistry , Iodine Radioisotopes , Mitogens/physiology , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Subcellular Fractions , Transfection
10.
J Cell Physiol ; 169(2): 380-90, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8908206

ABSTRACT

A novel, low molecular weight, intracellular isoform of FGF receptor-1 (FGFR-1) was identified in embryonic chicken tissues using several antibodies specific for different domains of FGF receptors. This low molecular weight isoform differs from the previously characterized isoforms of FGFR-1 in that it contains little or no carbohydrate. Furthermore, in contrast to the other isoforms of FGFR-1, this novel isoform is located exclusively intracellularly. However, it is capable of binding 125I-FGF-2 and it possesses intrinsic kinase activity. Pulse-chase experiments indicate that this isoform of FGFR-1 is not simply a precursor to glycosylated FGFR-1 since it can be detected long after the appearance of glycosylated FGFR-1 in the cells. These results suggest that the novel FGFR-1 isoform plays a role in regulating FGF activity distinct from cell surface, glycosylated FGFR-1. The possible roles of this FGFR-1 variant in FGF signaling are discussed.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Receptors, Fibroblast Growth Factor/chemistry , Animals , Antibodies/immunology , Antibodies/metabolism , Blotting, Northern , Cells, Cultured , Chick Embryo , Electrophoresis, Polyacrylamide Gel , Fibroblast Growth Factor 2/metabolism , Glycosylation , Immunoblotting , Molecular Weight , Polymerase Chain Reaction , Precipitin Tests , Protein Binding , Receptors, Fibroblast Growth Factor/immunology , Receptors, Fibroblast Growth Factor/metabolism
11.
Mol Biol Cell ; 7(8): 1299-317, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8856671

ABSTRACT

In an effort to determine the localization of fibroblast growth factor (FGF) receptors (FGFR) that could mediate the intracellular action of FGF-2, we discovered the presence of high-affinity. FGF-2 binding sites in the nuclei of bovine adrenal medullary cells (BAMC). Western blot analysis demonstrated the presence of 103-, 118-, and 145-kDa forms of FGFR1 in nuclei isolated from BAMC. 125I-FGF-2 cross-linking to nuclear extracts followed by FGFR1 immunoprecipitation showed that FGFR1 can account for the nuclear FGF-2 binding sites. Nuclear FGFR1 has kinase activity and undergoes autophosphorylation. Immunocytochemistry with the use of confocal and electron microscopes demonstrated the presence of FGFR1 within the nuclear interior. Nuclear subfractionation followed by Western blot or immunoelectron microscopic analysis showed that the nuclear FGFR1 is contained in the nuclear matrix and the nucleoplasm. Agents that induce translocation of endogenous FGF-2 to the nucleus (forskolin, carbachol, or angiotensin II) increased the intranuclear accumulation of FGFR1. This accumulation was accompanied by an overall increase in FGF-2-inducible tyrosine kinase activity. Our findings suggest a novel mode for growth factor action whereby growth factor receptors translocate to the nucleus in parallel with their ligand and act as direct mediators of nuclear responses to cell stimulation.


Subject(s)
Adrenal Medulla/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Adrenal Medulla/cytology , Animals , Cattle , Cell Nucleus/metabolism , Cells, Cultured , Kinetics , Microscopy, Immunoelectron , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Subcellular Fractions/metabolism
12.
J Cell Biol ; 134(2): 529-36, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8707835

ABSTRACT

Members of the FGF family of growth factors localize to the nuclei in a variety of different cell types. To determine whether FGF receptors are also present within nuclei and if this localization is regulated by FGFs, nuclei were prepared from quiescent and FGF-2-treated Swiss 3T3 fibroblasts and examined for the presence of FGF receptors by immunoblotting with an antibody produced against the extracellular domain of FGF receptor-1 (FGFR-1). Little or no FGFR-1 is detected in nuclei prepared from quiescent cells. When cells are treated with FGF-2, however, there is a time- and dose-dependent increase in the association of FGFR-1 immunoreactivity with the nucleus. In contrast, treatment with either EGF or 10% serum does not increase the association of FGFR-1 with the nucleus. When cell surface proteins are labeled with biotin, a biotinylated FGFR-1 is detected in the nuclear fraction prepared from FGF-2-treated, but not untreated, cells indicating that the nuclear-associated FGFR-1 immunoreactivity derives from the cell surface. The presence of FGFR-1 in the nuclei of FGF-2-treated cells was confirmed by immunostaining with a panel of different FGFR-1 antibodies, including one directed against the COOH-terminal domain of the protein. Fractionation of nuclei from FGF-2-treated cells indicates that nuclear FGFR-1 is localized to the nuclear matrix, suggesting that the receptor may play a role in regulating gene activity.


Subject(s)
Cell Nucleus/metabolism , Fibroblast Growth Factor 2/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/metabolism , 3T3 Cells , Animals , Biological Transport , Cell Nucleus/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Mice , Protein-Tyrosine Kinases/metabolism , Rabbits , Receptor, Fibroblast Growth Factor, Type 1 , Time Factors
13.
Brain Res Mol Brain Res ; 38(1): 161-5, 1996 May.
Article in English | MEDLINE | ID: mdl-8737680

ABSTRACT

Fractionation of human astrocytes revealed the presence of 103, 118, and 145 kDa forms of FGF receptor 1 (FGFR1) in isolated nuclei. Only trace amounts of FGFR1 proteins were detected in the cell membrane or cytoplasmic fractions. Nuclear FGFR1 is found in the nucleoplasm and nuclear matrix but not in chromatin. Immuno-confocal microscopy further demonstrates the intranuclear presence of FGFR1 and its colocalization with FGF-2. Nuclear FGFR1 binds to FGF-2 and has tyrosine kinase activity. Translocation of functional growth factor receptors into the cell nucleus offers a novel mechanism for growth factor action.


Subject(s)
Astrocytes/chemistry , Cell Nucleus/chemistry , Fibroblast Growth Factor 1 , Receptors, Fibroblast Growth Factor/analysis , Cell Membrane/chemistry , Cells, Cultured , Cytoplasm/chemistry , Humans , Molecular Weight
14.
Pediatr Res ; 39(3): 375-85, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8929854

ABSTRACT

Fibroblast growth factors (FGF) are known to have key roles in embryonic growth and morphogenesis, but their presence and contributions to fetal development are unclear. In particular, little information exists as to the relevance of FGF and their specific receptors to human fetal development. We studied the anatomical distribution of messenger RNA encoding FGF-2 and one of its high affinity receptors, FGFR1, using in situ hybridization in a variety of human fetal tissues in early second trimester. Corresponding protein distributions were determined by immunohistochemistry. Both FGF-2 and FGFR1 mRNA and proteins were found to be present in every organ and tissue examined, but with defined cellular localizations. In skeletal muscle, both FGF-2 and FGFR1 mRNA and peptides were present in differentiated fibers, and both co-localized to proliferating chondrocytes of the epiphyseal growth plate. FGF-2 and FGFR1 mRNA and peptides were also present within cardiac or gastrointestinal smooth muscle. Within the gastrointestinal tract FGF-2 mRNA and peptide were located in the submucosal tissue, whereas FGFR1 was expressed within the overlying mucosa. Similarly, in skin, FGF-2 was expressed within the dermis whereas FGFR1 mRNA and peptide were most apparent in the stratum germinativum of the epidermis. In kidney and lung, FGFR1 mRNA was located in the tubular and alveolar epithelia respectively, whereas FGF-2 was expressed in both epithelial and mesenchymal cell populations. Both growth factor and receptor were widespread in both neuroblasts and glioblasts in the cerebral cortex of the brain. Immunoreactivity for FGF-2 and FGFR1 was seen in all vascular endothelial cells of major vessels and capillaries. Within the skin, kidney, lung, and intestine FGF-2 immunoreactivity was found in basement membranes underlying epithelia, and was associated with the extracellular matrix and plasma membranes of many cell types. The results show that FGF-2 and one of its receptors are widely expressed anatomically in the mid-trimester human fetus.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/metabolism , Blood Vessels/metabolism , Brain/metabolism , Digestive System , Female , Fetus/metabolism , Fibroblast Growth Factor 2/genetics , Gene Expression , Humans , Lung/metabolism , Male , Musculoskeletal System/metabolism , Myocardium/metabolism , Pregnancy , Pregnancy Trimester, Second , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/genetics , Tissue Distribution
15.
Brain Res ; 701(1-2): 201-26, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-8925285

ABSTRACT

We have examined the cellular distribution of both FGF-2 and FGFR1 immunoreactivity and their mRNAs throughout the normal adult rat brain in order to reconcile numerous disparate findings in the published literature. The results confirm a widespread distribution of FGF-2 and FGFR1 in the rat brain, and different regions express distinct patterns of FGF-2 and FGFR1 mRNA and protein: neuronal and non-neuronal cells show different subcellular distributions that vary according to the area where they are located. The intensity of the staining and hybridization also varies according to the loci examined and the cell type involved. Astrocytes contain the highest levels of FGF-2 and FGFR1 mRNAs, and characteristically, possess high levels of immunoreactive FGF-2 within the nucleus. Amongst non-neuronal cells, oligodendrocytes do not synthesize or contain significant levels of FGF-2 immunoreactivity however, they do express FGFR1 mRNA. In these cells, immunoreactive FGFR1 is mainly associated with the myelin sheaths of neuronal fibers. In ventricular systems, ependymal cells synthesize and contain immunoreactive FGFR1. In contrast, only cells lining the lateral wall of the IIIrd ventricle express FGF-2 mRNA. Subependymal cells contain high levels of both FGF-2 and FGFR1 immunoreactivity. Neurons express low levels of FGF-2 mRNA and immunoreactive FGF-2 is localized predominantly to the perikaryon. However, selected populations of neurons, such as CA2 field of the hippocampus, show high levels of FGF-2 mRNA, in which the nucleus is strongly immunopositive. Similarly, high levels of FGFR1 mRNA are localized to select populations of neurons (e.g. amygdala). FGFR1 immunoreactivity is mainly associated with myelinated fiber tracts (e.g. striatum), and some neurons show immunoreactivity in the perikaryon (e.g. hippocampus), the nucleus (e.g. mesencephalic trigeminal nucleus), or in axonal projections (e.g. hypothalamus). Remarkably, in many of the areas studied, FGF-2 and FGFR1 mRNA and/or their translated protein do not co-localize in neurons (e.g. neo-cortices) or even in the same regions of the brain (e.g. substantia nigra). In other instances, mRNAs for both FGF-2 and FGFR1 colocalize (e.g. supraoptic nucleus). The brain, in contrast to peripheral tissues, contains high levels of FGF-2 and actively expresses its gene under normal physiological conditions. The highly specific anatomical distribution of immunoreactive FGF-2 in neuronal and non-neuronal brain cells, supports the notion that it plays a multifunctional role in the CNS under normal physiology. By correlating the localization and the synthesis of FGF-2 and one of its high affinity receptors, FGFR1, in the CNS, it should be possible to obtain a better understanding of the roles of FGF-2 in normal and pathological conditions.


Subject(s)
Brain Chemistry/physiology , Fibroblast Growth Factor 2/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/metabolism , Animals , Blotting, Western , Brain/anatomy & histology , Brain/cytology , Immunohistochemistry , In Situ Hybridization , Male , Neurons/metabolism , RNA Probes , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1 , Ribonucleases/metabolism
16.
J Neurochem ; 65(4): 1895-8, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7561890

ABSTRACT

Schwannoma-derived growth factor (SDGF) is a potent mitogen and neuronal differentiation factor. Because of its relationship to epidermal growth factor (EGF) and the heregulins, it was asked if SDGF interacts with the EGF receptor or HER2/neu. SDGF binds to and causes the phosphorylation on tyrosine of the EGF receptor but not HER2/neu.


Subject(s)
ErbB Receptors/metabolism , Glycoproteins , Growth Substances/metabolism , Intercellular Signaling Peptides and Proteins , Receptor, ErbB-2/metabolism , 3T3 Cells , Amphiregulin , Animals , Binding, Competitive , Cell Line , EGF Family of Proteins , Mice , PC12 Cells , Phosphorylation , Rats , Tyrosine/metabolism
17.
J Cell Biol ; 128(6): 1221-8, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7896883

ABSTRACT

We recently characterized three FGF-binding proteins (FGF-BPs) which are soluble forms of the extracellular domains of the high affinity FGF receptors (Hanneken, A. M., W. Ying, N. Ling, and A. Baird. Proc. Natl. Acad. Sci. USA. 1994. 91:9170-9174). These proteins circulate in blood and have been proposed to modulate the biological activity of the FGF family of proteins. Immunohistochemical studies now demonstrate that these soluble, truncated FGF receptors are also present in the basement membranes of retinal vascular endothelial cells. These immunoreactive proteins can be detected with antibodies raised to the extracellular domain of FGFR-1 but not with antibodies raised to either the juxtamembrane domain or the cytoplasmic domain of FGFR-1. Western blotting of human retinal extracts with the antibody raised to the extracellular domain of FGFR-1 detects specific, low molecular mass proteins at 85 kD and 55 kD, corresponding in size to the FGF-BPs, which are not detected with antibodies against the cytoplasmic domain of the receptor. The interaction of this receptor with the extracellular matrix is not dependent on the presence of FGF-2. Immunoreactive receptors are still detected in vascular basement membranes after the removal of FGF-2 with heparitinase. In addition, the recombinant extracellular domain of FGFR-1 continues to bind to corneal endothelial cell matrix after endogenous FGF-2 has been removed with 2 M NaCl. Acid treatment, which has been shown to disrupt protein interactions with the extracellular matrix, leads to a significant reduction in the presence of the matrix form of the FGF receptor. This loss can be restored with exogenous incubations of the recombinant extracellular domain of FGFR-1. This report is the first demonstration that a truncated form of a high affinity growth factor receptor can be localized to the extracellular matrix. These findings add to the list of binding proteins associated with the extracellular matrix (IGFBP-5) and suggest a potentially new regulatory mechanism for controlling the biological availability of FGF, and other peptide growth factors, in the extracellular matrix.


Subject(s)
Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Sequence , Animals , Basement Membrane/metabolism , Cattle , Humans , Molecular Sequence Data , Radioligand Assay , Receptors, Fibroblast Growth Factor/chemistry , Retina/metabolism
18.
Proc Natl Acad Sci U S A ; 90(23): 11177-81, 1993 Dec 01.
Article in English | MEDLINE | ID: mdl-7504298

ABSTRACT

Treatment of chicken embryo fibroblasts (CEFs) with trypsin results in a dose- and time-dependent loss of phosphotyrosine from cellular proteins. A similar, but less marked, reduction in protein tyrosine phosphorylation occurs upon incubation of CEFs in phosphate-buffered saline (PBS). The decrease in the phosphotyrosine content of proteins following treatment with trypsin or PBS, as determined by immunoblotting of cell extracts with anti-phosphotyrosine antibodies, corresponds with a loss of phosphotyrosine antibody immunoreactivity at focal contacts, as detected by immunofluorescence microscopy. The recovery of phosphotyrosine in cellular proteins occurs within 30 min following removal of trypsin, even in the presence of the protein synthesis inhibitor cycloheximide, indicating that the loss of phosphotyrosine-containing proteins is not due to their degradation by trypsin. Pretreatment of CEFs with inhibitors of protein-tyrosine-phosphatases greatly reduces the loss of phosphotyrosine from proteins brought about by trypsin. In addition, phosphotyrosine phosphatase activity is increased in extracts prepared from trypsin-treated CEFs. The loss of phosphotyrosine from proteins following treatment with trypsin or PBS is not specific to CEFs but is also observed in established fibroblast lines. Taken together these results suggest that the activity of one or more phosphotyrosine phosphatases is regulated by cell-substrate adhesion.


Subject(s)
Cell Adhesion , Phosphoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Tyrosine/analogs & derivatives , Actin Cytoskeleton/ultrastructure , Actins/ultrastructure , Animals , Blotting, Western , Cell Adhesion/drug effects , Cells, Cultured , Chick Embryo , Enzyme Activation , Fluorescent Antibody Technique , In Vitro Techniques , Phosphotyrosine , Trypsin/pharmacology , Tyrosine/metabolism
19.
J Cell Physiol ; 155(2): 368-75, 1993 May.
Article in English | MEDLINE | ID: mdl-8387074

ABSTRACT

Basic fibroblast growth factor (bFGF) is a multifunctional growth factor that can stimulate cell proliferation, production of proteases, and angiogenesis. Loss of mechanisms that regulate bFGF activity could result in tumor development. To test this idea, cells derived from an invasive bladder carcinoma (EJ) were compared with cells derived from a noninvasive bladder carcinoma (RT4) for the expression of bFGF and high and low affinity FGF receptors. bFGF was produced by the invasive EJ cells but not by the noninvasive RT4 cells, suggesting that bFGF could act in an autocrine fashion in the EJ cells to promote their invasion and growth in the surrounding tissue. The two cells lines also showed differences in FGF receptor expression. The EJ cells expressed both high and low affinity FGF receptors as determined by Scatchard analysis, whereas the RT4 cells expressed only high affinity receptors. The high affinity receptors on the RT4 cells were not recognized by an antibody to known FGF receptors. Furthermore, in contrast to the EJ cells, bFGF did not induce protein tyrosine phosphorylation in the RT4 cells. Thus these data suggest that the invasive potential of bladder carcinoma cells may be regulated by the expression of both bFGF and its receptors.


Subject(s)
Carcinoma/metabolism , Fibroblast Growth Factor 2/metabolism , Receptors, Cell Surface/metabolism , Urinary Bladder Neoplasms/metabolism , Binding, Competitive , Blotting, Northern , Carcinoma/pathology , Humans , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology
20.
J Biol Chem ; 268(6): 4244-9, 1993 Feb 25.
Article in English | MEDLINE | ID: mdl-8440708

ABSTRACT

Stimulation of fibroblasts with basic fibroblast growth factor (bFGF) led to the rapid tyrosine phosphorylation of a number of cellular proteins, including a major substrate of 90 kDa. The methyltransferase inhibitor 5'-methylthioadenosine (MTA) was found to be a specific inhibitor of bFGF-stimulated protein tyrosine phosphorylation in fibroblasts, blocking both receptor autophosphorylation and substrate phosphorylation. MTA had no effect on either epidermal growth factor- or platelet-derived growth factor-stimulated protein tyrosine phosphorylation in fibroblasts. MTA also inhibited both bFGF-stimulated protein tyrosine phosphorylation and neurite outgrowth in PC12 cells. MTA was a specific inhibitor of bFGF-stimulated protein tyrosine phosphorylation only in intact cells. MTA delayed and reduced, but did not inhibit, bFGF internalization and processing. The effects of MTA on bFGF-stimulated tyrosine phosphorylation required only a brief pretreatment with the agent and were readily reversible.


Subject(s)
Deoxyadenosines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Thionucleosides/pharmacology , 3T3 Cells , Animals , Fibroblast Growth Factor 2/pharmacology , Methyltransferases/antagonists & inhibitors , Mice , Mice, Inbred BALB C , PC12 Cells , Phosphorylation , Receptors, Fibroblast Growth Factor/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL