Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
J Biol Chem ; 299(2): 102804, 2023 02.
Article in English | MEDLINE | ID: mdl-36529290

ABSTRACT

Fibroblast growth factor (FGF) is a multifunctional protein that exhibits a wide range of biological effects. Most commonly, it acts as a mitogen, but it also has regulatory, morphological, and endocrine effects. The four receptor subtypes of FGF are activated by more than 20 different FGF ligands. FGF2, one of the FGF ligands, is an essential factor for cell culture in stem cells for regenerative medicine; however, recombinant FGF2 is extremely unstable. Here, we successfully generated homobivalent agonistic single-domain antibodies (variable domain of heavy chain of heavy chain antibodies referred to as VHHs) that bind to domain III and induce activation of the FGF receptor 1 and thus transduce intracellular signaling. This agonistic VHH has similar biological activity (EC50) as the natural FGF2 ligand. Furthermore, we determined that the agonistic VHH could support the proliferation of human-induced pluripotent stem cells (PSCs) and human mesenchymal stem cells, which are PSCs for regenerative medicine. In addition, the agonistic VHH could maintain the ability of mesenchymal stem cells to differentiate into adipocytes or osteocytes, indicating that it could maintain the properties of PSCs. These results suggest that the VHH agonist may function as an FGF2 mimetic in cell preparation of stem cells for regenerative medicine with better cost effectiveness.


Subject(s)
Fibroblast Growth Factor 2 , Protein Domains , Receptor, Fibroblast Growth Factor, Type 1 , Single-Domain Antibodies , Humans , Adipocytes/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Ligands , Mesoderm/cytology , Mesoderm/drug effects , Osteocytes/drug effects , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Regenerative Medicine , Signal Transduction/drug effects , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology
2.
Biomacromolecules ; 22(12): 5349-5362, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34855396

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) is an integral membrane protein that transmits prolife signals through the plasma membrane. Overexpression of FGFR1 has been reported in various tumor types, and therefore, this receptor constitutes an attractive molecular target for selective anticancer therapies. Here, we present a novel system for generation of intrinsically fluorescent, self-assembling, oligomeric cytotoxic conjugates with high affinity and efficient internalization targeting FGFR1. In our approach, we employed FGF1 as an FGFR1 recognizing molecule and genetically fused it to green fluorescent protein polygons (GFPp), a fluorescent oligomerization scaffold, resulting in a set of GFPp_FGF1 oligomers with largely improved receptor binding. To validate the applicability of using GFPp_FGF1 oligomers as cancer probes and drug carriers in targeted therapy of cancers with aberrant FGFR1, we selected a trimeric variant from generated GFPp_FGF1 oligomers and further engineered it by introducing FGF1-stabilizing mutations and by incorporating the cytotoxic drug monomethyl auristatin E (MMAE) in a site-specific manner. The resulting intrinsically fluorescent, trimeric cytotoxic conjugate 3xGFPp_FGF1E_LPET_MMAE exhibits nanomolar affinity for the receptor and very high stability. Notably, the intrinsic fluorescence of 3xGFPp_FGF1E_LPET_MMAE allows for tracking the cellular transport of the conjugate, demonstrating that 3xGFPp_FGF1E_LPET_MMAE is efficiently and selectively internalized into cells expressing FGFR1. Importantly, we show that 3xGFPp_FGF1E_LPET_MMAE displays very high cytotoxicity against a panel of different cancer cells overproducing FGFR1 while remaining neutral toward cells devoid of FGFR1 expression. Our data implicate that the engineered fluorescent conjugates can be used for imaging and targeted therapy of FGFR1-overproducing cancers.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
3.
Biomolecules ; 11(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34439755

ABSTRACT

FGFRs are cell surface receptors that, when activated by specific FGFs ligands, transmit signals through the plasma membrane, regulating key cellular processes such as differentiation, division, motility, metabolism and death. We have recently shown that the modulation of the spatial distribution of FGFR1 at the cell surface constitutes an additional mechanism for fine-tuning cellular signaling. Depending on the multivalent, engineered ligand used, the clustering of FGFR1 into diverse supramolecular complexes enhances the efficiency and modifies the mechanism of receptor endocytosis, alters FGFR1 lifetime and modifies receptor signaling, ultimately determining cell fate. Here, we present a novel approach to generate multivalent FGFR1 ligands. We functionalized FGF1 for controlled oligomerization by developing N- and C-terminal fusions of FGF1 with the Fc fragment of human IgG1 (FGF1-Fc and Fc-FGF1). As oligomerization scaffolds, we employed GFPpolygons, engineered GFP variants capable of well-ordered multivalent display, fused to protein G to ensure binding of Fc fragment. The presented strategy allows efficient assembly of oligomeric FGFR1 ligands with up to twelve receptor binding sites. We show that multivalent FGFR1 ligands are biologically active and trigger receptor clustering on the cell surface. Importantly, the approach described in this study can be easily adapted to oligomerize alternative growth factors to control the activity of other cell surface receptors.


Subject(s)
Fibroblast Growth Factor 1/chemistry , Immunoglobulin G/chemistry , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Animals , Binding Sites , Cell Line, Tumor , Cluster Analysis , Endocytosis , Endosomes/metabolism , Green Fluorescent Proteins/chemistry , Humans , Ligands , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Phosphorylation , Protein Binding , Protein Domains , Protein Engineering , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Proteins/chemistry , Signal Transduction
4.
Food Funct ; 12(11): 5038-5050, 2021 Jun 08.
Article in English | MEDLINE | ID: mdl-33960359

ABSTRACT

Rheumatoid Arthritis (RA) is an autoimmune disease that commences as inflammation and progressively destroys the articular joint. In this study, we assess the anti-rheumatic potential of the monoterpenoid class of thymol conjugated with Carbon Dots (CDs). Waste biomass in the form of dried rose petals was chosen as a precursor for the synthesis of CDs via a one-step hydrothermal bottom-up methodology. The prepared CDs exhibited absorption in the near-visible region, and unique excitation-dependent emission behaviour was confirmed from UV-Visible and fluorescence measurements. The surface morphology of CDs was confirmed by SEM and HR-TEM analysis to be quasi-spherical particles with an average size of ∼5-6 nm. The presence of various functional moieties (hydroxyl, carbonyl, and amino) was confirmed via FT-IR measurement. The graphitization of CDs was confirmed by the D and G bands for sp2 and sp3 hybridization, respectively, through Raman analysis. Esterification methodology was adopted to prepare the CDs-thymol conjugate and confirmed via FT-IR analysis. CDs play the role of a nanocarrier for thymol, an anti-arthritic agent. The bioactive compound of thymol showed potent anti-arthritic activity against RA targets through in silico docking studies. Further, the in vivo studies revealed that CDs-thymol conjugates (10 mg per kg body weight) showed a significant reduction in rat paw volume along with reduced levels of RF and CRP (2.23 ± 0.42 IU ml-1 and 16.96 ± 0.22 mg ml-1) when compared to the disease control rats. X-ray radiography and ultrasonic imaging revealed less bone destruction, joint derangement, and swelling in arthritis-induced Wistar rats. They could also potentially improve the Hb (14.14 ± 0.19), RBC (6.01 ± 0.11), PCV (6.01 ± 0.11) levels and elevate the status of antioxidant enzymes (GPx, SOD, MDA), and the activity was comparable to the standard drug, ibuprofen (10 mg kg-1), suggesting that the CDs-thymol conjugate at 10 mg kg-1 could act as a strong anti-arthritic agent. This work is evidence for the utilization of waste biomass as a value-added product such as a nanocarrier for biomedical applications.


Subject(s)
Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Carbon/chemistry , Quantum Dots , Thymol/chemistry , Animals , Antioxidants , Arthritis, Rheumatoid/drug therapy , Female , Interleukins/chemistry , Matrix Metalloproteinase 1/chemistry , Matrix Metalloproteinase 3/chemistry , Molecular Docking Simulation , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Spectroscopy, Fourier Transform Infrared , Transforming Growth Factor beta/chemistry
5.
Mol Oncol ; 14(9): 1998-2021, 2020 09.
Article in English | MEDLINE | ID: mdl-32511887

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) transmits signals through the plasma membrane regulating essential cellular processes like division, motility, metabolism, and death. Overexpression of FGFR1 is observed in numerous tumors and thus constitutes an attractive molecular target for selective cancer treatment. Targeted anti-cancer therapies aim for the precise delivery of drugs into cancer cells, sparing the healthy ones and thus limiting unwanted side effects. One of the key steps in targeted drug delivery is receptor-mediated endocytosis. Here, we show that the efficiency and the mechanism of FGFR1 internalization are governed by the spatial distribution of the receptor in the plasma membrane. Using engineered antibodies of different valency, we demonstrate that dimerization of FGFR1 with bivalent antibody triggers clathrin-mediated endocytosis (CME) of the receptor. Clustering of FGFR1 into larger oligomers with tetravalent antibody stimulates fast and highly efficient uptake of the receptor that occurs via two distinct mechanisms: CME and dynamin-dependent clathrin-independent endocytic routes. Furthermore, we show that all endocytic pathways engaged in FGFR1 internalization do not require receptor activation. Our data provide novel insights into the mechanisms of intracellular trafficking of FGFR1 and constitute guidelines for development of highly internalizing antibody-based drug carriers for targeted therapy of FGFR1-overproducing cancers.


Subject(s)
Antibodies/metabolism , Endocytosis , Protein Engineering , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Antibody Affinity/immunology , CHO Cells , Cell Line, Tumor , Clathrin/metabolism , Cluster Analysis , Cricetulus , Dynamins/metabolism , Humans , Mice , Models, Biological , NIH 3T3 Cells , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptors, Fc/metabolism
6.
J Mol Biol ; 432(13): 3749-3760, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32302608

ABSTRACT

Optically controlled receptor tyrosine kinases (opto-RTKs) allow regulation of RTK signaling using light. Until recently, the majority of opto-RTKs were activated with blue-green light. Fusing a photosensory core module of Deinococcus radiodurans bacterial phytochrome (DrBphP-PCM) to the kinase domains of neurotrophin receptors resulted in opto-RTKs controlled with light above 650 nm. To expand this engineering approach to RTKs of other families, here we combined the DrBpP-PCM with the cytoplasmic domains of EGFR and FGFR1. The resultant Dr-EGFR and Dr-FGFR1 opto-RTKs are rapidly activated with near-infrared and inactivated with far-red light. The opto-RTKs efficiently trigger ERK1/2, PI3K/Akt, and PLCγ signaling. Absence of spectral crosstalk between the opto-RTKs and green fluorescent protein-based biosensors enables simultaneous Dr-FGFR1 activation and detection of calcium transients. Action mechanism of the DrBphP-PCM-based opto-RTKs is considered using the available RTK structures. DrBphP-PCM represents a versatile scaffold for engineering of opto-RTKs that are reversibly regulated with far-red and near-infrared light.


Subject(s)
Phytochrome/ultrastructure , Receptor Protein-Tyrosine Kinases/ultrastructure , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biosensing Techniques , Deinococcus/chemistry , Deinococcus/genetics , Green Fluorescent Proteins/chemistry , HeLa Cells , Humans , Light , MAP Kinase Signaling System/radiation effects , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Phosphatidylinositol 3-Kinases/genetics , Phytochrome/chemistry , Phytochrome/genetics , Protein Conformation/radiation effects , Proto-Oncogene Proteins c-akt/genetics , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/genetics , Signal Transduction/radiation effects
7.
EBioMedicine ; 48: 462-477, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31631034

ABSTRACT

BACKGROUND: The development of a clinically useful fibroblast growth factor 21 (FGF21) hormone has been impeded by its inherent instability and weak FGF receptor (FGFR) binding affinity. There is an urgent need for innovative approaches to overcome these limitations. METHODS: We devised a structure-based chimerisation strategy in which we substituted the thermally labile and low receptor affinity core of FGF21 with an HS binding deficient endocrinised core derived from a stable and high receptor affinity paracrine FGF1 (FGF1ΔHBS). The thermal stability, receptor binding ability, heparan sulfate and ßKlotho coreceptor dependency of the chimera were measured using a thermal shift assay, SPR, SEC-MALS and cell-based studies. The half-life, tissue distribution, glucose lowering activity and adipose tissue remodeling were analyzed in normal and diabetic mice and monkeys. FINDINGS: The melting temperature of the engineered chimera (FGF1ΔHBS-FGF21C-tail) increased by ∼22 °C relative to wild-type FGF21 (FGF21WT), and resulted in a ∼5-fold increase in half-life in vivo. The chimera also acquired an ability to bind the FGFR1c isoform - the principal receptor that mediates the metabolic actions of FGF21 - and consequently was dramatically more effective than FGF21WT in correcting hyperglycemia and in ameliorating insulin resistance in db/db mice. Our chimeric FGF21 also exerted a significant beneficial effect on glycemic control in spontaneous diabetic cynomolgus monkeys. INTERPRETATION: Our study describes a structure-based chimerisation approach that effectively mitigates both the intrinsically weak receptor binding affinities and short half-lives of endocrine FGFs, and advance the development of the FGF21 hormone into a potentially useful drug for Type 2 diabetes.


Subject(s)
Fibroblast Growth Factors/metabolism , Metabolic Diseases/metabolism , Paracrine Communication , Adipocytes/metabolism , Animals , Biomarkers , Chromatography, High Pressure Liquid , Disease Models, Animal , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Gene Expression , Humans , Insulin/metabolism , Male , Metabolic Diseases/drug therapy , Metabolic Diseases/etiology , Mice , Models, Molecular , Paracrine Communication/drug effects , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Proteins , Structure-Activity Relationship
8.
Gene ; 717: 144047, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31421190

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) signaling pathways play important roles in the formation of the blood vascular system and nervous system across animal phyla. We have earlier reported VEGF and FGF from Hydra vulgaris Ind-Pune, a cnidarian with a defined body axis, an organized nervous system and a remarkable ability of regeneration. We have now identified three more components of VEGF and FGF signaling pathways from hydra. These include FGF-1, FGF receptor 1 (FGFR-1) and VEGF receptor 2 (VEGFR-2) with a view to deciphering their possible roles in regeneration. METHODS: In silico analysis of proteins was performed using Clustal omega, Swiss model, MEGA 7.0, etc. Gene expression was studied by whole mount in situ hybridization. VEGF and FGF signaling was inhibited using specific pharmacological inhibitors and their effects on head regeneration were studied. RESULTS: Expression patterns of the genes indicate a possible interaction between FGF-1 and FGFR-1 and also VEGF and VEGFR-2. Upon treatment of decapitated hydra with pharmacological inhibitor of FGFR-1 or VEGFR-2 for 48 h, head regeneration was delayed in treated as compared to untreated, control regenerates. When we studied the expression of head specific genes HyBra1 and HyKs1 and tentacle specific gene HyAlx in control and treated regenerates using whole mount in situ hybridization, expression of all the three genes was found to be adversely affected in treated regenerates. CONCLUSIONS: The results suggest that VEGF and FGF signaling play important roles in regeneration of hypostome and tentacles in hydra.


Subject(s)
Fibroblast Growth Factors/metabolism , Head/physiology , Hydra/physiology , Regeneration/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Computer Simulation , Fibroblast Growth Factor 1/chemistry , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Gene Expression Regulation , Humans , Hydra/drug effects , Indoles/pharmacology , Protein Domains , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Regeneration/drug effects , Signal Transduction , Structural Homology, Protein , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Cells ; 8(7)2019 07 13.
Article in English | MEDLINE | ID: mdl-31337028

ABSTRACT

Glioblastoma is the most lethal brain cancer in adults, with no known cure. This cancer is characterized by a pronounced genetic heterogeneity, but aberrant activation of receptor tyrosine kinase signaling is among the most frequent molecular alterations in glioblastoma. Somatic mutations of fibroblast growth factor receptors (FGFRs) are rare in these cancers, but many studies have documented that signaling through FGFRs impacts glioblastoma progression and patient survival. Small-molecule inhibitors of FGFR tyrosine kinases are currently being trialed, underlining the therapeutic potential of blocking this signaling pathway. Nevertheless, a comprehensive overview of the state of the art of the literature on FGFRs in glioblastoma is lacking. Here, we review the evidence for the biological functions of FGFRs in glioblastoma, as well as pharmacological approaches to targeting these receptors.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Receptor, Fibroblast Growth Factor, Type 4/physiology , Disease Progression , Humans , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Receptor, Fibroblast Growth Factor, Type 3/chemistry , Receptor, Fibroblast Growth Factor, Type 4/chemistry
10.
Gene ; 702: 123-132, 2019 Jun 20.
Article in English | MEDLINE | ID: mdl-30926307

ABSTRACT

Fibroblast growth factor (FGF) 5 regulates the development and periodicity of hair follicles, which can affect hair traits. Loss-of-function mutations associated with long-hair phenotypes have been described in several mammalian species. Sheep is an important economic animal, however, the evolution characterizations and biological mechanism of oFGF5 (Ovis aries FGF5) gene are still poorly understood. In this study, oFGF5 gene was obtained by resequencing the whole genome of three Dorper sheep and RACE of two Kazakh sheep FGF5. We proposed FGF5 was phylogenetically related to FGF4 family and oFGF5 clearly orthologed to goat FGF5. Six loci were found from the positive selection results of FGF5 and half of them located on signal peptide. The basically similar rates of function-altering substitutions in sheep and goat lineage and the rest of the mammalian lineage of 365 SNPs indicated that the FGF5 gene was quite conservative during evolution. Homology modeling of the oFGF5 suggested that it has a highly conserved FGF superfamily domain containing 10 ß-strands. Furthermore, the protein-protein docking analysis revealed that oFGF5 have the potential to form heterodimers with oFGFR1, the predicted interaction interface of FGF5-FGFR1 heterodimer was formed mainly by residues from FGF superfamily domain. Our observations suggested the evolutionary and structural biology features of oFGF5 might be relevant to its function about hair follicle development and modulating hair growth, and we confirmed our speculation by using the FGF5 gene editing sheep produced by CRISPR/Cas9 technology.


Subject(s)
Fibroblast Growth Factor 5/chemistry , Fibroblast Growth Factor 5/genetics , Sheep/genetics , Animals , CRISPR-Cas Systems , Computational Biology , Evolution, Molecular , Fibroblast Growth Factor 4/classification , Fibroblast Growth Factor 5/classification , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Humans , Mice , Models, Molecular , Mutation , Phylogeny , Protein Conformation, beta-Strand , Protein Multimerization , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Sequence Alignment , Sheep/anatomy & histology , Wool/anatomy & histology
11.
J Cell Sci ; 132(1)2019 01 03.
Article in English | MEDLINE | ID: mdl-30478195

ABSTRACT

Both fibroblast growth factor-2 (FGF2) and neural cell adhesion molecule (NCAM) trigger FGF receptor 1 (FGFR1) signaling; however, they induce remarkably distinct receptor trafficking and cellular responses. The molecular basis of such a dichotomy and the role of distinct types of ligand-receptor interaction remain elusive. Number of molecules and brightness (N&B) analysis revealed that FGF2 and NCAM promote different FGFR1 assembly and dynamics at the plasma membrane. NCAM stimulation elicits long-lasting cycles of short-lived FGFR1 monomers and multimers, a behavior that might reflect a rapid FGFR1 internalization and recycling. FGF2, instead, induces stable dimerization at the dose that stimulates cell proliferation. Reducing the occupancy of FGFR1 in response to low FGF2 doses causes a switch towards cyclically exposed and unstable receptor dimers, consistently with previously reported biphasic response to FGF2 and with the divergent signaling elicited by different ligand concentrations. Similar instability was observed upon altering the endocytic pathway. Thus, FGF2 and NCAM induce differential FGFR1 clustering at the cell surface, which might account for the distinct intracellular fate of the receptor and, hence, for the different signaling cascades and cellular responses.


Subject(s)
Cell Membrane/metabolism , Cell Proliferation , Fibroblast Growth Factor 2/metabolism , Neural Cell Adhesion Molecules/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Cell Movement , Endocytosis , Fibroblast Growth Factor 2/genetics , HeLa Cells , Humans , Neural Cell Adhesion Molecules/genetics , Protein Binding , Protein Multimerization , Protein Transport , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics
12.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30217817

ABSTRACT

Fibroblast growth factor receptor-1 (FGFR1) activity at the plasma membrane is tightly controlled by the availability of co-receptors and competing receptor isoforms. We have previously shown that FGFR1 activity in pancreatic beta-cells modulates a wide range of processes, including lipid metabolism, insulin processing, and cell survival. More recently, we have revealed that co-expression of FGFR5, a receptor isoform that lacks a tyrosine-kinase domain, influences FGFR1 responses. We therefore hypothesized that FGFR5 is a co-receptor to FGFR1 that modulates responses to ligands by forming a receptor heterocomplex with FGFR1. We first show here increased FGFR5 expression in the pancreatic islets of nonobese diabetic (NOD) mice and also in mouse and human islets treated with proinflammatory cytokines. Using siRNA knockdown, we further report that FGFR5 and FGFR1 expression improves beta-cell survival. Co-immunoprecipitation and quantitative live-cell imaging to measure the molecular interaction between FGFR5 and FGFR1 revealed that FGFR5 forms a mixture of ligand-independent homodimers (∼25%) and homotrimers (∼75%) at the plasma membrane. Interestingly, co-expressed FGFR5 and FGFR1 formed heterocomplexes with a 2:1 ratio and subsequently responded to FGF2 by forming FGFR5/FGFR1 signaling complexes with a 4:2 ratio. Taken together, our findings identify FGFR5 as a co-receptor that is up-regulated by inflammation and promotes FGFR1-induced survival, insights that reveal a potential target for intervention during beta-cell pathogenesis.


Subject(s)
Cytokines/immunology , Diabetes Mellitus/genetics , Insulin-Secreting Cells/immunology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Animals , Diabetes Mellitus/immunology , Dimerization , Female , Fibroblast Growth Factor 2/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 5/chemistry , Receptor, Fibroblast Growth Factor, Type 5/immunology , Up-Regulation
13.
Cell Physiol Biochem ; 48(4): 1480-1491, 2018.
Article in English | MEDLINE | ID: mdl-30107380

ABSTRACT

BACKGROUND/AIMS: The up-regulation of hepatocyte growth factor/receptor, HGF/Met, signal transduction is observed in most of human cancers. Specific heparan sulfate structures enhance the HGF/Met signaling at both cell and animal-based model systems. Biochemical studies indicate that heparan sulfate interacts with HGF and a natural occurring splicing variant NK1 of HGF with similar affinity. However, it is currently unknown if cell surface heparan sulfate binds to Met at physiological conditions and if specific cell surface heparan sulfate structures are required for effective HGF/Met or NK1/Met signaling. METHODS: An established flow sorting strategy was used to isolate a soluble Met recombinant protein-binding positive or negative CHO cell clones different only in specific heparan sulfate structures. The cell surface bindings were imaged by confocal microscopy and flow cytometry analysis. Glucosamine vs. galactosamine contents from media-, cell surface-, and cell association glycosaminoglycans were quantified by HPLC. 35S-sulfate labeled glycosaminoglycans were characterized by anion exchange and size-exclusion HPLC. Heparan sulfate disaccharide compositions were determined by HPLC-MS analysis. Western blot analyses of MAPK-p42/44 were used to monitor HGF- and NK1-facillated Met signaling. RESULTS: CHO-Positive but not CHO-Negative cell surface heparan sulfate bound to Met recombinant protein and HGF/NK1 further promoted the binding. Overall glycosaminoglycan analysis results indicated that the CHO-Negative cells had reduced amount of heparan sulfate, shorter chain length, and less 6-O-sulfated disaccharides compared to that of CHO-Positive cells. Moreover, CHO-Negative cells were defective in NK1/Met but not HGF/Met signaling. CONCLUSIONS: This study demonstrated that soluble Met recombinant protein bound to cell surface HS at physiological conditions and a Met /HGF or NK1/HS ternary signaling complex might be involved in Met signaling. Shorter HS chains and reduced 6-O-sulfation might be responsible for reduced Met binding and the diminished NK1-initiated signaling in the CHO-Negative cells. The unique CHO-Positive and CHO-Negative cell clones established in current study should be effective tools for studying the role of specific glycosaminoglycan structures in regulating Met signaling. Such knowledge should be useful in developing glycosaminoglycan-based compounds that target HGF/Met signaling.


Subject(s)
Heparitin Sulfate/analysis , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction , Animals , CHO Cells , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Disaccharides/analysis , Glycosaminoglycans/analysis , Glycosaminoglycans/chemistry , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Hepatocyte Growth Factor/chemistry , Humans , Mitogen-Activated Protein Kinases/metabolism , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Neurokinin-1/genetics
14.
Curr Opin Nephrol Hypertens ; 27(4): 229-235, 2018 07.
Article in English | MEDLINE | ID: mdl-29851418

ABSTRACT

PURPOSE OF REVIEW: α-Klotho (Klotho) occurs in three isoforms, a membrane-bound form acting as a coreceptor for fibroblast growth factor-23 (FGF23) signalling, a shed soluble form consisting of Klotho's large ectodomain thought to act as an enzyme or a hormone, and a secreted truncated form generated by alternative splicing of the Klotho mRNA with unknown function. The purpose of this review is to highlight the recent advances in our understanding of Klotho's function in mineral homeostasis. RECENT FINDINGS: A number of seminal discoveries have recently been made in this area, shifting existing paradigms. The crystal structure of the ternary FGF receptor (FGFR)-1c/Klotho/FGF23 complex has been uncovered, revealing how the ligand FGF23 interacts with FGFR1c and the coreceptor Klotho at atomic resolution. Furthermore, it was shown that soluble Klotho lacks any glycosidase activity and serves as a bona fide coreceptor for FGF23 signalling. Experiments with a combination of Klotho and Fgf23-deficient mouse models demonstrated that all isoforms of Klotho lack any physiologically relevant, FGF23-independent functions in mineral homeostasis or ageing. Finally, it was demonstrated that the alternatively spliced Klotho mRNA is degraded and is not translated into a secreted Klotho protein isoform in humans. SUMMARY: Taken together, there is now overwhelming evidence that the main physiological function of transmembrane and soluble Klotho for mineral homeostasis is their role as coreceptors mediating FGF23 actions. In light of these findings, the main pathophysiological consequence of the downregulation of Klotho observed in acute and chronic renal failure may be the induction of renal FGF23 resistance.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucuronidase/genetics , Glucuronidase/metabolism , Minerals/metabolism , RNA, Messenger/metabolism , Acute Kidney Injury/metabolism , Animals , Fibroblast Growth Factor-23 , Glucuronidase/chemistry , Homeostasis , Humans , Kidney Failure, Chronic/metabolism , Klotho Proteins , Molecular Structure , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
15.
J Chem Theory Comput ; 14(6): 2867-2880, 2018 Jun 12.
Article in English | MEDLINE | ID: mdl-29678106

ABSTRACT

Alchemical free energy methods have gained much importance recently from several reports of improved ligand-protein binding affinity predictions based on their implementation using molecular dynamics simulations. A large number of variants of such methods implementing different accelerated sampling techniques and free energy estimators are available, each claimed to be better than the others in its own way. However, the key features of reproducibility and quantification of associated uncertainties in such methods have barely been discussed. Here, we apply a systematic protocol for uncertainty quantification to a number of popular alchemical free energy methods, covering both absolute and relative free energy predictions. We show that a reliable measure of error estimation is provided by ensemble simulation-an ensemble of independent MD simulations-which applies irrespective of the free energy method. The need to use ensemble methods is fundamental and holds regardless of the duration of time of the molecular dynamics simulations performed.


Subject(s)
Ligands , Molecular Dynamics Simulation , Proteins/metabolism , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Proteins/chemistry , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Thermodynamics , Thrombin/chemistry , Thrombin/metabolism
16.
J Pharmacol Toxicol Methods ; 94(Pt 1): 19-25, 2018.
Article in English | MEDLINE | ID: mdl-29660397

ABSTRACT

INTRODUCTION: bFKB1 is a humanized bispecific IgG1 antibody, created by conjoining an anti-Fibroblast Growth Factor Receptor 1 (FGFR1) half-antibody to an anti-Klothoß (KLB) half-antibody, using the knobs-into-holes strategy. bFKB1 acts as a highly selective agonist for the FGFR1/KLB receptor complex and is intended to ameliorate obesity-associated metabolic defects by mimicking the activity of the hormone FGF21. An important aspect of the biologics product manufacturing process is to establish meaningful product specifications regarding the tolerable levels of impurities that copurify with the drug product. The aim of the current study was to determine acceptable levels of product-related impurities for bFKB1. METHODS: To determine the tolerable levels of these impurities, we dosed obese mice with bFKB1 enriched with various levels of either HMW impurities or anti-FGFR1-related impurities, and measured biomarkers for KLB-independent FGFR1 signaling. RESULTS: Here, we show that product-related impurities of bFKB1, in particular, high molecular weight (HMW) impurities and anti-FGFR1-related impurities, when purposefully enriched, stimulate FGFR1 in a KLB-independent manner. By taking this approach, the tolerable levels of product-related impurities were successfully determined. DISCUSSION: Our study demonstrates a general pharmacology-guided approach to setting a product specification for a bispecific antibody whose homomultimer-related impurities could lead to undesired biological effects.


Subject(s)
Antibodies/chemistry , Antibodies/pharmacology , Animals , CHO Cells , Cell Line , Cricetulus , Disease Models, Animal , Drug Combinations , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Male , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Weight , Obesity/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Signal Transduction/drug effects
17.
Article in English | MEDLINE | ID: mdl-29610389

ABSTRACT

We report the case of a 27-yr-old male with visual field loss who had a 4.9-cm complex cystic mass in the right occipital lobe. Histologic examination showed pilocytic astrocytoma (PA) with anaplasia, and molecular characterization revealed FGFR1 duplication with additional variants of unknown significance in several genes (ARID1A, ARID1B, CHEK2, EPHA5, and MLL2). This is one of only a very few reported cases of anaplastic PA with characterization of molecular alterations.


Subject(s)
Astrocytoma/diagnosis , Astrocytoma/genetics , Gene Duplication , Protein Interaction Domains and Motifs/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adult , Anaplasia , Biopsy , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Neoplasm Grading , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Tomography, X-Ray Computed
18.
Molecules ; 23(4)2018 Mar 27.
Article in English | MEDLINE | ID: mdl-29584670

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) has become a potential target for the treatment of cancer. Designing FGFR1-selective inhibitors remains fundamental to the development of anti-cancer drugs because of highly sequential homology among FGFR subtypes. In present work, four inhibitors were examined with intermolecular interaction patterns with FGFR1 and FGFR4, respectively, for the exploration of binding mechanisms by applying a combined approach of computational techniques, including flexible docking, binding site analyses, electronic structure computations, molecular dynamic simulations, and binding free energy predictions. Molecular simulation-predicted binding conformations and pharmacophoric features of these molecules in the active pocket of either FGFR1 or FGFR4. MMPB(GB)SA-calculated binding free energies were accordant with the ordering of their tested potency values. Furthermore, in silico mutations of two residues (FGFR1: Tyr563 and Ser565) were also performed to check their impact on ligand binding by applying MD simulations and binding free energy calculations. The present studies may provide a structural understanding of the FGFR1-selective mechanism. The viewpoints from computational simulations would be valuable guidelines for the development of novel FGFR1-selective inhibitors.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Binding Sites , Catalytic Domain/drug effects , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Mutation , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Receptor, Fibroblast Growth Factor, Type 4/genetics
19.
Cancer Lett ; 423: 36-46, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29452146

ABSTRACT

The Hippo pathway plays a critical role in organ size control, tissue homeostasis and tumor genesis through its key transcription regulator Yes-associated protein1 (YAP1), but the mechanism underlying its role in lung cancer is unclear. We hypothesized that YAP1 influences FGFR1 signaling to maintain cancer stem-like cell (CSC) properties in FGFR1-amplified lung cancer. In support of this, our data confirms that expression levels of YAP1 are positively associated with those of FGFR1 in clinical lung carcinoma samples as measured by real-time PCR, western blot, and immunohistochemistry (IHC) staining. Mechanistically, YAP1 up-regulates FGFR1 expression at the level of promoter through the TEAD binding site while bFGF/FGFR1 induces YAP1 expression via large tumor suppressors 1(LATS1). In addition, the absence of YAP1 abolishes self-renewal ability in lung cancer. Furthermore, an orthotropic mouse model highlights the function of YAP1 in the initiation and metastasis of lung cancer. Verteporfin, a YAP1 inhibitor, effectively inhibits both YAP1 and FGFR1 expression in lung cancer. Thus, we conclude that YAP1 is a potential therapeutic target for lung cancer. Combined targeting of YAP1 and FGFR1 may provide benefits to patients with FGFR1-amplified lung cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Phosphoproteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Up-Regulation , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , Gene Amplification , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Neoplasm Transplantation , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Transcription Factors , YAP-Signaling Proteins
20.
Nature ; 553(7689): 461-466, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29342138

ABSTRACT

The ageing suppressor α-klotho binds to the fibroblast growth factor receptor (FGFR). This commits FGFR to respond to FGF23, a key hormone in the regulation of mineral ion and vitamin D homeostasis. The role and mechanism of this co-receptor are unknown. Here we present the atomic structure of a 1:1:1 ternary complex that consists of the shed extracellular domain of α-klotho, the FGFR1c ligand-binding domain, and FGF23. In this complex, α-klotho simultaneously tethers FGFR1c by its D3 domain and FGF23 by its C-terminal tail, thus implementing FGF23-FGFR1c proximity and conferring stability. Dimerization of the stabilized ternary complexes and receptor activation remain dependent on the binding of heparan sulfate, a mandatory cofactor of paracrine FGF signalling. The structure of α-klotho is incompatible with its purported glycosidase activity. Thus, shed α-klotho functions as an on-demand non-enzymatic scaffold protein that promotes FGF23 signalling.


Subject(s)
Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Glucuronidase/chemistry , Glucuronidase/metabolism , Paracrine Communication , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Animals , Binding Sites/genetics , Body Fluids/metabolism , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Glucuronidase/genetics , Heparitin Sulfate/metabolism , Humans , Klotho Proteins , Ligands , Male , Mice , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Protein Binding , Protein Domains , Protein Multimerization , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...