Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 172: 113741, 2020 02.
Article in English | MEDLINE | ID: mdl-31812679

ABSTRACT

Na+/K+-ATPase α1 was reported to directly interact with and recruit FGF2 (fibroblast growth factor 2), a vital cell signaling protein implicated in angiogenesis, to the inner plasma membrane for subsequent secretion. Cardenolides, a class of cardiac glycosides, were reported to downregulate FGF2 secretion upon binding to Na+/K+-ATPase α1 in a cell system with ectopically expressed FGF2 and Na+/K+-ATPase α1. Herein, we disclose that the cardenolides ouabain and reevesioside A significantly enhance the secretion/release of FGF2 and the phosphorylation of FGFR1 (fibroblast growth factor receptor 1) in a time- and dose-dependent manner, in A549 carcinoma cells. A pharmacological approach was used to elucidate the pertinent upstream effectors. Only the ERK1/2 inhibitor U0126 but not the other inhibitors examined (including those inhibiting the unconventional secretion of FGF2) was able to reduce ouabain-induced FGF2 secretion and FGFR1 activation. ERK1/2 phosphorylation was increased upon ouabain treatment, a process found to be mediated through upstream effectors including ouabain-induced phosphorylated EGFR and a reduced MKP1 protein level. Therefore, at least two independent lines of upstream effectors are able to mediate ouabain-induced ERK1/2 phosphorylation and the subsequent FGF2 secretion and FGFR1 activation. These finding constitute unprecedent insights into the regulation of FGF2 secretion by cardenolides.


Subject(s)
Cardenolides/pharmacology , Fibroblast Growth Factor 2/agonists , Ouabain/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , A549 Cells , Cardenolides/chemistry , Cell Survival/drug effects , Drug Interactions , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , MAP Kinase Signaling System , Molecular Structure , Ouabain/chemistry , Pyrroles/administration & dosage , Pyrroles/pharmacology
2.
Sci Rep ; 6: 20643, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26865366

ABSTRACT

Tendon injury during limb motion is common. Damaged tendons heal poorly and frequently undergo unpredictable ruptures or impaired motion due to insufficient innate healing capacity. By basic fibroblast growth factor (bFGF) or vascular endothelial growth factor (VEGF) gene therapy via adeno-associated viral type-2 (AAV2) vector to produce supernormal amount of bFGF or VEGF intrinsically in the tendon, we effectively corrected the insufficiency of the tendon healing capacity. This therapeutic approach (1) resulted in substantial amelioration of the low growth factor activity with significant increases in bFGF or VEGF from weeks 4 to 6 in the treated tendons (p < 0.05 or p < 0.01), (2) significantly promoted production of type I collagen and other extracellular molecules (p < 0.01) and accelerated cellular proliferation, and (3) significantly increased tendon strength by 68-91% from week 2 after AAV2-bFGF treatment and by 82-210% from week 3 after AAV2-VEGF compared with that of the controls (p < 0.05 or p < 0.01). Moreover, the transgene expression dissipated after healing was complete. These findings show that the gene transfers provide an optimistic solution to the insufficiencies of the intrinsic healing capacity of the tendon and offers an effective therapeutic possibility for patients with tendon disunion.


Subject(s)
Dependovirus/genetics , Fibroblast Growth Factor 2/genetics , Tendon Injuries/therapy , Vascular Endothelial Growth Factor A/genetics , Wound Healing/genetics , Animals , Cell Proliferation , Chickens , Collagen Type I/agonists , Collagen Type I/genetics , Collagen Type I/metabolism , Dependovirus/metabolism , Fibroblast Growth Factor 2/agonists , Fibroblast Growth Factor 2/metabolism , Fibronectins/agonists , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Laminin/agonists , Laminin/genetics , Laminin/metabolism , Primary Cell Culture , Tendon Injuries/genetics , Tendon Injuries/metabolism , Tendon Injuries/pathology , Tendons/metabolism , Tendons/pathology , Tenocytes/cytology , Tenocytes/metabolism , Tensile Strength , Transgenes , Vascular Endothelial Growth Factor A/agonists , Vascular Endothelial Growth Factor A/metabolism
3.
Biomaterials ; 81: 157-168, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26731578

ABSTRACT

Site-specific chemical dimerization of fibroblast growth factor 2 (FGF2) with the optimal linker length resulted in a FGF2 homodimer with improved granulation tissue formation and blood vessel formation at exceptionally low concentrations. Homodimers of FGF2 were synthesized through site-specific linkages to both ends of different molecular weight poly(ethylene glycols) (PEGs). The optimal linker length was determined by screening dimer-induced metabolic activity of human dermal fibroblasts and found to be that closest to the inter-cysteine distance, 70 Å, corresponding to 2 kDa PEG. A straightforward analysis of the kinetics of second ligand binding as a function of tether length showed that, as the polymerization index (the number of monomer repeat units in the polymer, N) of the tether decreases, the mean time for second ligand capture decreases as ∼N(3/2), leading to an enhancement of the number of doubly bound ligands in steady-state for a given (tethered) ligand concentration. FGF2-PEG2k-FGF2 induced greater fibroblast metabolic activity than FGF2 alone, all other dimers, and all monoconjugates, at each concentration tested, with the greatest difference observed at low (0.1 ng/mL) concentration. FGF2-PEG2k-FGF2 further exhibited superior activity compared to FGF2 for both metabolic activity and migration in human umbilical vein endothelial cells, as well as improved angiogenesis in a coculture model in vitro. Efficacy in an in vivo wound healing model was assessed in diabetic mice. FGF2-PEG2k-FGF2 increased granulation tissue and blood vessel density in the wound bed compared to FGF2. The results suggest that this rationally designed construct may be useful for improving the fibroblast matrix formation and angiogenesis in chronic wound healing.


Subject(s)
Fibroblast Growth Factor 2/agonists , Fibroblast Growth Factor 2/pharmacology , Granulation Tissue/pathology , Protein Multimerization , Wound Healing/drug effects , Animals , Blood Vessels/drug effects , Blood Vessels/pathology , Blotting, Western , Cell Movement/drug effects , Cross-Linking Reagents/chemistry , Dermis/cytology , Diabetes Mellitus, Experimental/pathology , Fibroblast Growth Factor 2/chemistry , Fibroblasts/cytology , Fibroblasts/drug effects , Granulation Tissue/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Male , Mice , Neovascularization, Physiologic/drug effects , Polyethylene Glycols/chemistry
4.
PLoS One ; 9(11): e112996, 2014.
Article in English | MEDLINE | ID: mdl-25426952

ABSTRACT

Neuroprotective proteins expressed in the fetus play a critical role during early embryonic neurodevelopment, especially during maternal exposure to alcohol and drugs that cause stress, glutamate neuroexcitotoxicity, and damage to the fetal brain, if prolonged. We have identified a novel protein, carboxypeptidase E-ΔN (CPE-ΔN), which is a splice variant of CPE that has neuroprotective effects on embryonic neurons. CPE-ΔN is transiently expressed in mouse embryos from embryonic day 5.5 to postnatal day 1. It is expressed in embryonic neurons, but not in 3 week or older mouse brains, suggesting a function primarily in utero. CPE-ΔN expression was up-regulated in embryonic hippocampal neurons in response to dexamethasone treatment. CPE-ΔN transduced into rat embryonic cortical and hippocampal neurons protected them from glutamate- and H2O2-induced cell death. When transduced into embryonic cortical neurons, CPE-ΔN was found in the nucleus and enhanced the transcription of FGF2 mRNA. Embryonic cortical neurons challenged with glutamate resulted in attenuated FGF2 levels and cell death, but CPE-ΔN transduced neurons treated in the same manner showed increased FGF2 expression and normal viability. This neuroprotective effect of CPE-ΔN was mediated by secreted FGF2. Through receptor signaling, FGF2 activated the AKT and ERK signaling pathways, which in turn increased BCL-2 expression. This led to inhibition of caspase-3 activity and cell survival.


Subject(s)
Carboxypeptidase H/genetics , Fibroblast Growth Factor 2/agonists , Glutamic Acid/pharmacology , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neuroprotective Agents/metabolism , Animals , Carboxypeptidase H/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Embryo, Mammalian , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/enzymology , Hydrogen Peroxide/pharmacology , Mice , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/enzymology , Primary Cell Culture , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/agonists , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction , Transduction, Genetic
5.
J Bone Miner Metab ; 32(6): 627-35, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24306516

ABSTRACT

In the present study, the effects of bone morphogenetic protein-2 (BMP-2), vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) on regulation of rat osteoblast (ROB) maturation in vitro were investigated. It was found that the proliferation, differentiation and mineralization of ROBs were all dose-dependently increased at particular times in the case of treatment with only one growth factor. To investigate the effects of combined treatment, ROBs were treated with either a single application of a relatively high dose of each growth factor, or binary/triple combined applications of relatively low doses of the growth factors. Osteogenic differentiation was significantly promoted in the triple combination treatment of BMP-2, VEGF and bFGF compared with the single or binary combination treatments. The optimal timing of the triple combination to enhance osteogenesis was also tested. When bFGF and VEGF were added in the early stage, and BMP-2 and VEGF were added in the late stage, osteogenic differentiation of ROBs could be enhanced more effectively. These results could be used to construct bone tissue engineering scaffolds that release growth factors sequentially.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Fibroblast Growth Factor 2/pharmacology , Osteoblasts/metabolism , Osteogenesis/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Bone Morphogenetic Protein 2/agonists , Cells, Cultured , Drug Synergism , Fibroblast Growth Factor 2/agonists , Osteoblasts/cytology , Rats , Vascular Endothelial Growth Factor A/agonists
6.
Am J Pathol ; 170(2): 764-73, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255342

ABSTRACT

Corneal neovascularization is a vision-threatening condition caused by various ocular pathological conditions. The aim of this study was to evaluate the function of the ephrin ligands and Eph receptors in vitro and in vivo in corneal angiogenesis in a mouse model. The Eph tyrosine kinase receptors and their ligands, ephrins, are expressed on the cell surface. The functions of Eph and ephrins have been shown to regulate axonal guidance, segmentation, cell migration, and angiogenesis. Understanding the roles of Eph and ephrin in corneal angiogenesis may provide a therapeutic intervention for the treatment of angiogenesis-related disorders. Immunohistochemical studies demonstrated that ephrinB1 and EphB1 were expressed in basic fibroblast growth factor (bFGF)-induced vascularized corneas. EphB1 was specifically colocalized with vascular endothelial marker CD31 surrounded by type IV collagen. EphrinB1 was expressed in corneal-resident keratocytes and neutrophils. Recombinant ephrinB1-Fc, which induces EphB receptor activation, enhanced bFGF-induced tube formation in an in vitro aortic ring assay and promoted bFGF-induced corneal angiogenesis in vivo in a corneal pocket assay. Synergistically enhanced and sustained activation of extracellular signal-regulated kinase was noted in vascular endothelial cell lines after stimulation with ephrin B1 and bFGF combinations. These results suggest that ephrinB1 plays a synergistic role in corneal neovascularization.


Subject(s)
Corneal Neovascularization/metabolism , Ephrin-B1/biosynthesis , Fibroblast Growth Factor 2/toxicity , Gene Expression Regulation/drug effects , Receptors, Eph Family/biosynthesis , Animals , Aorta/metabolism , Cattle , Collagen Type IV/biosynthesis , Corneal Neovascularization/chemically induced , Corneal Neovascularization/drug therapy , Corneal Neovascularization/pathology , Enzyme Activation/drug effects , Ephrin-B1/agonists , Ephrin-B1/pharmacology , Fibroblast Growth Factor 2/agonists , Humans , Mice , Organ Culture Techniques , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis
7.
Neurobiol Aging ; 25(10): 1333-42, 2004.
Article in English | MEDLINE | ID: mdl-15465631

ABSTRACT

Indirect trophic actions of nicotine on brain during aging are suggested from observations describing nicotine as a cognitive enhancer, increasing vigilance and improving learning and memory, and both in vitro and in vivo models have demonstrated neuroprotective effects of nAChR agonists. Previously, we have reported that an acute intermittent (-)nicotine treatment significantly increases fibroblast growth factor-2 (FGF-2) mRNA and protein in several brain regions of rat brain. The present study was designed to analyse if nicotine-induced FGF-2 expression in the rat brain was preserved during aging. Using in situ hybridization and quantitative RNase protection assay the present paper reports that during aging (12- and 24-month-old rats) the response of FGF-2 gene expression in the rat brain to nAChR stimulation by (-)nicotine is fully effective and involves both neurons and glial cells. The investigation was extended to other members of the FGF family, such as FGF-5 and -20, but this expression was not influenced by the (-)nicotine treatment at any age studied. Similarly following (-)nicotine treatment no changes were observed in FGF receptors (FGFR 1-3) mRNA levels in adult and aged rats. Taken together, the present and previous data support the hypothesis that neuroprotective effects of (-)nicotine and the potential beneficial effects of (-)nicotine agonists in the treatment of Alzheimer's and Parkinson's diseases, may at least in part involve an activation of the neuronal and glial FGF-2 signalling. Work is in progress to analyse the mechanism(s) linking nAChR activation to the up-regulation of FGF-2.


Subject(s)
Aging/drug effects , Aging/metabolism , Brain/metabolism , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation/physiology , Nicotine/pharmacology , RNA, Messenger/biosynthesis , Animals , Brain/drug effects , Fibroblast Growth Factor 2/agonists , Gene Expression Regulation/drug effects , RNA, Messenger/agonists , Rats , Tissue Distribution
8.
J Cell Sci ; 115(Pt 2): 267-74, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11839778

ABSTRACT

Angiogenic regulators modulate endothelial cell functions, including proliferation, migration, secretion, and adhesion, through their action on endothelial cells or other cell types. TWEAK, a novel member of the tumor necrosis factor family, appears to be a pro-angiogenic agent on the basis of previous studies demonstrating its ability to induce interleukin-8 production by epithelial tumor lines, stimulate proliferation of human vascular cell types and neovascularization in rat corneas. Here, we further characterized the angiogenic potential of TWEAK, revealing a dual role for TWEAK as an angiogenic regulator. We demonstrate that TWEAK is a potent inducer of endothelial cell survival and cooperates with basic fibroblast growth factor to induce the proliferation and migration of human endothelial cells and morphogenesis of capillary lumens. In contrast, TWEAK antagonizes the morphogenic response of endothelial cells to vascular endothelial growth factor (VEGF) without inhibiting VEGF-induced survival or proliferation. Thus, our observations suggest that TWEAK may differentially regulate microvascular growth, remodeling and/or maintenance in vivo, depending upon the angiogenic context.


Subject(s)
Angiogenesis Inhibitors/metabolism , Capillaries/metabolism , Carrier Proteins/metabolism , Endothelium, Vascular/metabolism , Neovascularization, Physiologic/physiology , Apoptosis Regulatory Proteins , Binding Sites/drug effects , Binding Sites/physiology , Capillaries/cytology , Capillaries/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cytokine TWEAK , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/agonists , Fibroblast Growth Factor 2/metabolism , Humans , Infant, Newborn , Lymphokines/antagonists & inhibitors , Lymphokines/metabolism , Neovascularization, Physiologic/drug effects , Tumor Necrosis Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Atherosclerosis ; 122(2): 255-63, 1996 May.
Article in English | MEDLINE | ID: mdl-8769688

ABSTRACT

Insulin-like growth factor I and II (IGF-I and -II)--two 7.65- and 7.47-kDA polypeptides belonging to the somatomedine family--are regular constituents of human blood plasma. Both factors exert mitogenic activity on a variety of cell types including arterial smooth muscle cells. In the present study, the effect of IGF-I and -II on cultured bovine coronary smooth muscle cells (cSMC) was assessed. Human recombinant IGF-I and IGF-II added to cSMC cultured in a medium containing 10% fetal bovine serum (FBS) decreased the cell number and [3H]thymidine incorporation in a dose dependent fashion up to 40% and 43% compared to control cells (100%). At the same time, the expression of basic fibroblast growth factor (bFGF) increased from 60 pg/5 x 10(4) cells (control) to 75 (IGF-I) and 113 pg/5 x 10(4) cells (IGF-II). In parallel with enhanced bFGF expression, the bFGF receptor content per cell and the [35S]sulfate incorporation into extracellular and cell-associated proteoglycans also increased under the influence of IGF-I and -II. In contrast, with low serum concentration (0.1% FBS) the addition of IGF-I and -II to bovine cSMC cultures resulted in a slight increase in cell number, protein content and [3H]thymidine incorporation as described in previous studies. These results suggest that the mitogenic activity of IGF-I and -II towards coronary smooth muscle cells depends on culture conditions. In the presence of 10% fetal bovine serum that mimics in vivo conditions, IGF-I and -II did not necessarily act as mitogenic factors but inhibited the proliferation of cSMC in vitro possibly by modulating antagonizing the action of other growth factors. Irrespective of the inhibition of cell division, the cellular bFGF, the bFGF receptor and the bFGF activity-related proteoheparan sulfate were overexpressed under the influence of IGF.


Subject(s)
Coronary Vessels/metabolism , Fibroblast Growth Factor 2/biosynthesis , Insulin-Like Growth Factor II/pharmacology , Insulin-Like Growth Factor I/pharmacology , Muscle, Smooth, Vascular/metabolism , Animals , Cattle , Cell Division/drug effects , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , DNA/biosynthesis , Fibroblast Growth Factor 2/agonists , Flow Cytometry , Heparan Sulfate Proteoglycans , Heparitin Sulfate/metabolism , Immunoenzyme Techniques , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Proteoglycans/metabolism , Receptors, Fibroblast Growth Factor/biosynthesis , Receptors, Fibroblast Growth Factor/drug effects , Recombinant Proteins/pharmacology
10.
Mol Biol Cell ; 7(3): 369-81, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8868466

ABSTRACT

Basic fibroblast growth factor (FGF-2) induces cell proliferation and urokinase-type plasminogen activator (uPA) production in fetal bovine aortic endothelial GM 7373 cells. In the present paper we investigated the role of the interaction of FGF-2 with tyrosine-kinase (TK) FGF receptors (FGFRs) in mediating uPA up-regulation in these cells. The results show that FGF-2 antagonists suramin, protamine, heparin, the synthetic peptide FGF-2(112-155), and a soluble form of FGFR-1 do not inhibit FGF-2-mediated uPA up-regulation at concentrations that affect growth factor binding to cell surface receptors and mitogenic activity. In contrast, tyrosine phosphorylation inhibitors and overexpression of a dominant negative TK- mutant of FGFR-1 abolish the uPA-inducing activity of FGF-2, indicating that FGFR and its TK activity are essential in mediating uPA induction. Accordingly, FGF-2 induces uPA up-regulation in Chinese hamster ovary cells transfected with wild-type FGFR-1, -2, -3, or -4 but not with TK- FGFR-1 mutant. Small unilamellar phosphatidyl choline:cholesterol vesicles loaded with FGF-2 increased uPA production in GM 7373 cells in the absence of a mitogenic response. Liposome-encapsulated FGF-2 showed a limited but significant capacity, relative to free FGF-2, to interact with FGFR both at 4 degrees C and 37 degrees C and to be internalized within the cell. uPA up-regulation by liposome-encapsulated FGF-2 was quenched by neutralizing anti-FGF-2 antibodies, indicating that the activity of liposome-delivered FGF-2 is mediated by an extracellular action of the growth factor. Taken together, the data indicate that a distinct interaction of FGF-2 with FGFR, quantitatively and/or qualitatively different from the one that leads to mitogenicity, is responsible for the uPA-inducing activity of the growth factor.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Up-Regulation , Urokinase-Type Plasminogen Activator/metabolism , Animals , Aorta/cytology , Aorta/embryology , CHO Cells , Cattle , Cell Line , Cricetinae , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/agonists , Humans , Liposomes/metabolism , Mitogens , Protein-Tyrosine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...