Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 254
Filtrer
1.
Arch Biochem Biophys ; 758: 110070, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38909834

RÉSUMÉ

Fibroblast Growth Factor Receptor (FGFR) is connected to numerous downstream signalling cascades regulating cellular behavior. Any dysregulation leads to a plethora of illnesses, including cancer. Therapeutics are available, but drug resistance driven by gatekeeper mutation impedes the treatment. Ponatinib is an FDA-approved drug against BCR-ABL kinase and has shown effective results against FGFR-mediated carcinogenesis. Herein, we undertake molecular dynamics simulation-based analysis on ponatinib against all the FGFR isoforms having Val to Met gatekeeper mutations. The results suggest that ponatinib is a potent and selective inhibitor for FGFR1, FGFR2, and FGFR4 gatekeeper mutations. The extensive electrostatic and van der Waals interaction network accounts for its high potency. The FGFR3_VM mutation has shown resistance towards ponatinib, which is supported by their lesser binding affinity than wild-type complexes. The disengaged molecular brake and engaged hydrophobic spine were believed to be the driving factors for weak protein-ligand interaction. Taken together, the inhibitory and structural characteristics exhibited by ponatinib may aid in thwarting resistance based on Val-to-Met gatekeeper mutations at an earlier stage of treatment and advance the design and development of other inhibitors targeted at FGFRs harboring gatekeeper mutations.


Sujet(s)
Imidazoles , Simulation de dynamique moléculaire , Mutation , Liaison aux protéines , Pyridazines , Pyridazines/composition chimique , Pyridazines/pharmacologie , Pyridazines/métabolisme , Imidazoles/composition chimique , Imidazoles/pharmacologie , Imidazoles/métabolisme , Humains , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/génétique
2.
Nature ; 618(7966): 862-870, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37286607

RÉSUMÉ

α/ßKlotho coreceptors simultaneously engage fibroblast growth factor (FGF) hormones (FGF19, FGF21 and FGF23)1,2 and their cognate cell-surface FGF receptors (FGFR1-4) thereby stabilizing the endocrine FGF-FGFR complex3-6. However, these hormones still require heparan sulfate (HS) proteoglycan as an additional coreceptor to induce FGFR dimerization/activation and hence elicit their essential metabolic activities6. To reveal the molecular mechanism underpinning the coreceptor role of HS, we solved cryo-electron microscopy structures of three distinct 1:2:1:1 FGF23-FGFR-αKlotho-HS quaternary complexes featuring the 'c' splice isoforms of FGFR1 (FGFR1c), FGFR3 (FGFR3c) or FGFR4 as the receptor component. These structures, supported by cell-based receptor complementation and heterodimerization experiments, reveal that a single HS chain enables FGF23 and its primary FGFR within a 1:1:1 FGF23-FGFR-αKlotho ternary complex to jointly recruit a lone secondary FGFR molecule leading to asymmetric receptor dimerization and activation. However, αKlotho does not directly participate in recruiting the secondary receptor/dimerization. We also show that the asymmetric mode of receptor dimerization is applicable to paracrine FGFs that signal solely in an HS-dependent fashion. Our structural and biochemical data overturn the current symmetric FGFR dimerization paradigm and provide blueprints for rational discovery of modulators of FGF signalling2 as therapeutics for human metabolic diseases and cancer.


Sujet(s)
Facteur-23 de croissance des fibroblastes , Protéoglycanes à sulfate d'héparane , Hormones , Récepteur facteur croissance fibroblaste , Transduction du signal , Humains , Cryomicroscopie électronique , Facteur-23 de croissance des fibroblastes/composition chimique , Facteur-23 de croissance des fibroblastes/métabolisme , Facteur-23 de croissance des fibroblastes/ultrastructure , Protéoglycanes à sulfate d'héparane/composition chimique , Protéoglycanes à sulfate d'héparane/métabolisme , Hormones/composition chimique , Hormones/métabolisme , Protéines Klotho/composition chimique , Protéines Klotho/métabolisme , Protéines Klotho/ultrastructure , Multimérisation de protéines , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/ultrastructure , Complexes multiprotéiques/composition chimique , Complexes multiprotéiques/métabolisme , Complexes multiprotéiques/ultrastructure
3.
J Med Chem ; 65(22): 15433-15442, 2022 11 24.
Article de Anglais | MEDLINE | ID: mdl-36356320

RÉSUMÉ

Upregulation of the fibroblast growth factor receptor (FGFR) signaling pathway has been implicated in multiple cancer types, including cholangiocarcinoma and bladder cancer. Consequently, small molecule inhibition of FGFR has emerged as a promising therapy for patients suffering from these diseases. First-generation pan-FGFR inhibitors, while highly effective, suffer from several drawbacks. These include treatment-related hyperphosphatemia and significant loss of potency for the mutant kinases. Herein, we present the discovery and optimization of novel FGFR2/3 inhibitors that largely maintain potency for the common gatekeeper mutants and have excellent selectivity over FGFR1. A combination of meticulous structure-activity relationship (SAR) analysis, structure-based drug design, and medicinal chemistry rationale ultimately led to compound 29, a potent and selective FGFR2/3 inhibitor with excellent in vitro absorption, distribution, metabolism, excretion (ADME), and pharmacokinetics in rat. A pharmacodynamic study of a closely related compound established that maximum inhibition of downstream ERK phosphorylation could be achieved with no significant effect on serum phosphate levels relative to vehicle.


Sujet(s)
Tumeurs , Inhibiteurs de protéines kinases , Récepteur facteur croissance fibroblaste , Animaux , Rats , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/composition chimique , Transduction du signal , Relation structure-activité , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/effets des médicaments et des substances chimiques
4.
Molecules ; 26(23)2021 Dec 03.
Article de Anglais | MEDLINE | ID: mdl-34885928

RÉSUMÉ

Considering our interest in the use of peptides as potential target-specific drugs or as delivery vectors of metallodrugs for various biomedical applications, it is crucial to explore improved synthetic methodologies to accomplish the highest peptide crude purity in the shortest time possible. Therefore, we compared "classical" fluorenylmethoxycarbonyl (Fmoc)-solid phase peptide synthesis (SPPS) with ultrasound(US)-assisted SPPS based on the preparation of three peptides, namely the fibroblast growth factor receptor 3(FGFR3)-specific peptide Pep1 (VSPPLTLGQLLS-NH2) and the novel peptides Pep2 (RQMATADEA-NH2) and Pep3 (AAVALLPAVLLALLAPRQMATADEA-NH2), which are being developed aimed at interfering with the intracellular protein-protein interaction(PPI) RANK-TRAF6. Our results demonstrated that US-assisted SPPS led to a 14-fold (Pep1) and 4-fold time reduction (Pep2) in peptide assembly compared to the "classical" method. Interestingly, US-assisted SPPS yielded Pep1 in higher purity (82%) than the "classical" SPPS (73%). The significant time reduction combined with high crude peptide purity attained prompted use to apply US-assisted SPPS to the large peptide Pep3, which displays a high number of hydrophobic amino acids and homooligo-sequences. Remarkably, the synthesis of this 25-mer peptide was attained during a "working day" (347 min) in moderate purity (approx. 49%). In conclusion, we have reinforced the importance of using US-SPPS towards facilitating the production of peptides in shorter time with increased efficacy in moderate to high crude purity. This is of special importance for long peptides such as the case of Pep3.


Sujet(s)
Peptides/synthèse chimique , Techniques de synthèse en phase solide/méthodes , Humains , Peptides/composition chimique , Récepteur activateur du facteur nucléaire Kappa B/composition chimique , Récepteur facteur croissance fibroblaste/composition chimique , Sonication/méthodes , Facteur-6 associé aux récepteurs de TNF/composition chimique
5.
Sci Rep ; 11(1): 7832, 2021 04 09.
Article de Anglais | MEDLINE | ID: mdl-33837264

RÉSUMÉ

Clostridium botulinum neurotoxin serotype A (BoNT/A) is a potent neurotoxin that serves as an effective therapeutic for several neuromuscular disorders via induction of temporary muscular paralysis. Specific binding and internalization of BoNT/A into neuronal cells is mediated by its binding domain (HC/A), which binds to gangliosides, including GT1b, and protein cell surface receptors, including SV2. Previously, recombinant HC/A was also shown to bind to FGFR3. As FGFR dimerization is an indirect measure of ligand-receptor binding, an FCS & TIRF receptor dimerization assay was developed to measure rHC/A-induced dimerization of fluorescently tagged FGFR subtypes (FGFR1-3) in cells. rHC/A dimerized FGFR subtypes in the rank order FGFR3c (EC50 ≈ 27 nM) > FGFR2b (EC50 ≈ 70 nM) > FGFR1c (EC50 ≈ 163 nM); rHC/A dimerized FGFR3c with similar potency as the native FGFR3c ligand, FGF9 (EC50 ≈ 18 nM). Mutating the ganglioside binding site in HC/A, or removal of GT1b from the media, resulted in decreased dimerization. Interestingly, reduced dimerization was also observed with an SV2 mutant variant of HC/A. Overall, the results suggest that the FCS & TIRF receptor dimerization assay can assess FGFR dimerization with known and novel ligands and support a model wherein HC/A, either directly or indirectly, interacts with FGFRs and induces receptor dimerization.


Sujet(s)
Toxines botuliniques de type A/métabolisme , Clostridium botulinum/enzymologie , Neurotoxines/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Sérogroupe , Transduction du signal/génétique , Animaux , Sites de fixation , Toxines botuliniques de type A/composition chimique , Membrane cellulaire/métabolisme , Dimérisation , Récepteurs ErbB/composition chimique , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Gangliosides/métabolisme , Glycoprotéines membranaires/métabolisme , Protéines de tissu nerveux/métabolisme , Neurones/métabolisme , Neurotoxines/composition chimique , Cellules PC12 , Liaison aux protéines , Domaines protéiques , Rats , Récepteurs de surface cellulaire/métabolisme , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/génétique , Transfection
6.
Int J Mol Sci ; 22(6)2021 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-33804608

RÉSUMÉ

Fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases that have been associated not only with various cellular processes, such as embryonic development and adult wound healing but also enhanced tumor survival, angiogenesis, and metastatic spread. Proteolytic cleavage of these single-pass transmembrane receptors has been suggested to regulate biological activities of their ligands during growth and development, yet little is known about the proteases responsible for this process. In this study, we monitored the release of membrane-anchored FGFRs 1, 2, 3, and 4 in cell-based assays. We demonstrate here that metalloprotease-dependent metalloprotease family, ADAM10 and ADAM17. Loss- and gain-of-function studies in murine embryonic fibroblasts showed that constitutive shedding as well as phorbol-ester-induced processing of FGFRs 1, 3, and 4 is mediated by ADAM17. In contrast, treatment with the calcium ionophore ionomycin stimulated ADAM10-mediated FGFR2 shedding. Cell migration assays with keratinocytes in the presence or absence of soluble FGFRs suggest that ectodomain shedding can modulate the function of ligand-induced FGFR signaling during cell movement. Our data identify ADAM10 and ADAM17 as differentially regulated FGFR membrane sheddases and may therefore provide new insight into the regulation of FGFR functions.


Sujet(s)
Metalloproteases/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Protéine ADAM10/métabolisme , Protéine ADAM17/métabolisme , Animaux , Lignée cellulaire , Mouvement cellulaire , Activation enzymatique , Cellules épithéliales/métabolisme , Protéines membranaires/composition chimique , Protéines membranaires/métabolisme , Famille multigénique , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Isoformes de protéines , Protéine kinase C/métabolisme , Protéolyse , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/génétique
7.
Chembiochem ; 22(1): 160-169, 2021 01 05.
Article de Anglais | MEDLINE | ID: mdl-32975328

RÉSUMÉ

Fibroblast growth factor (FGF2)/fibroblast growth factor receptor (FGFR) signalling plays a major role both in physiology and in several pathologies, including cancer development, metastasis formation and resistance to therapy. The development of small molecules, acting extracellularly to target FGF2/FGFR interactions, has the advantage of limiting the adverse effects associated with current intracellular FGFR inhibitors. Herein, we discuss the ability of the natural compound rosmarinic acid (RA) to induce FGF2/FGFR complex dissociation. The molecular-level description of the FGF2/FGFR/RA system, by NMR spectroscopy and docking, clearly demonstrates that RA binds to the FGFR-D2 domain and directly competes with FGF2 for the same binding site. Direct and allosteric perturbations combine to destabilise the complex. The proposed molecular mechanism is validated by cellular studies showing that RA inhibits FGF2-induced endothelial cell proliferation and FGFR activation. Our results can serve as the basis for the development of new extracellular inhibitors of the FGF/FGFR pathways.


Sujet(s)
Inhibiteurs de l'angiogenèse/pharmacologie , Produits biologiques/pharmacologie , Cinnamates/pharmacologie , Depsides/pharmacologie , Facteur de croissance fibroblastique de type 2/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/métabolisme , Inhibiteurs de l'angiogenèse/composition chimique , Animaux , Produits biologiques/composition chimique , Bovins , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cinnamates/composition chimique , Depsides/composition chimique , Facteur de croissance fibroblastique de type 2/composition chimique , Facteur de croissance fibroblastique de type 2/métabolisme , Simulation de docking moléculaire , Phosphorylation/effets des médicaments et des substances chimiques , Récepteur facteur croissance fibroblaste/composition chimique , Rosmarinic Acid
8.
Int J Biol Macromol ; 164: 3005-3011, 2020 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-32810535

RÉSUMÉ

The cartilages of marine fish, such as sharks and sturgeon, are important resources of the bioactive chondroitin sulfate (CS). To explore glycosaminoglycans from marine fish, polysaccharides from the cartilage of the sturgeon, Acipenser schrenckii, were extracted. Using enzyme-assisted extraction and anion-exchange chromatography, an uronic acid-containing polysaccharide, YG-1, was isolated. YG-1 is composed of GlcN, GlcUA, GalN, and Gal, in the ratio of 1.4: 3.4: 3.7: 1.0, and its molecular weight was determined to be 3.0 × 105 Da. YG-1 was confirmed to be chondroitin 4-sulfate (CS) composed of →4GlcAß1→3GalNAc4Sß1→ and minor →4GlcAß1→3GalNAcß1→, which was confirmed using IR spectroscopy, disaccharide composition analysis, and NMR. Bioactivity studies, including MTT assay and scratch-wound assays revealed that CS from Acipenser schrenckii had significant proliferation activity. The proliferation activity of the polysaccharide, YG-1, was related to Fibroblast growth factor 2 (FGF2). GalNAc 4S of YG-1 could be the binding sites of FGF2 and FGFR.


Sujet(s)
Chondroïtines sulfate/composition chimique , Chondroïtines sulfate/pharmacologie , Poissons/métabolisme , Animaux , Sites de fixation , Séquence glucidique , Prolifération cellulaire , Chromatographie d'échange d'ions , Facteur de croissance fibroblastique de type 2/composition chimique , Facteur de croissance fibroblastique de type 2/métabolisme , Cellules endothéliales de la veine ombilicale humaine , Humains , Spectroscopie par résonance magnétique , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/métabolisme
9.
Cells ; 8(8)2019 08 15.
Article de Anglais | MEDLINE | ID: mdl-31443196

RÉSUMÉ

Fibroblast growth factor 1 (FGF1) has been shown to interact with integrin αvß3 through a specific binding site, involving Arg35 residue. The FGF1 mutant (R35E) with impaired integrin binding was found to be defective in its proliferative response, although it was still able to interact with FGF receptors (FGFR) and heparin and induce the activation of downstream signaling pathways. Here, we demonstrate that the lack of mitogenic potential of R35E mutant is directly caused by its decreased thermodynamic stability and susceptibility to proteolytic degradation. Introduction of three stabilizing mutations into R35E variant compensated the effect of destabilizing R35E mutation and restored the proliferation potential of FGF1. Moreover, the stabilized R35E variant regained both anti-apoptotic and wound healing activities, while remaining defective in binding to integrin αvß3. Our results suggest that the thermodynamic stability and resistance to degradation, rather than the interaction with integrin are required for mitogenic response of FGF1.


Sujet(s)
Facteur de croissance fibroblastique de type 1/composition chimique , Intégrine alphaVbêta3/métabolisme , Stabilité protéique , Protéolyse , Animaux , Sites de fixation , Facteur de croissance fibroblastique de type 1/génétique , Héparine/composition chimique , Humains , Intégrine alphaVbêta3/composition chimique , Cinétique , Souris , Mutation , Cellules NIH 3T3 , Liaison aux protéines , Récepteur facteur croissance fibroblaste/composition chimique
10.
FASEB J ; 33(8): 9182-9193, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31063704

RÉSUMÉ

Soluble klotho (sKlotho), the shed ectodomain of α-klotho, protects the heart by down-regulating transient receptor potential canonical isoform 6 (TRPC6)-mediated calcium signaling. Binding to α2-3-sialyllactose moiety of gangliosides in lipid rafts and inhibition of raft-dependent signaling underlies the mechanism. A recent 3-Å X-ray structure of sKlotho in complex with fibroblast growth factor receptor (FGFR) and fibroblast growth factor 23 (FGF23) indicates that its ß6α6 loop might block access to the proposed binding site for α2-3-sialyllactose. It was concluded that sKlotho only functions in complex with FGFR and FGF23 and that sKlotho's pleiotropic effects all depend on FGF23. Here, we report that sKlotho can inhibit TRPC6 channels expressed in cells lacking endogenous FGFRs. Structural modeling and molecular docking show that a repositioned ß6α6 loop allows sKlotho to bind α2-3-sialyllactose. Molecular dynamic simulations further show the α2-3-sialyllactose-bound sKlotho complex to be stable. Domains mimicking sKlotho's sialic acid-recognizing activity inhibit TRPC6. The results strongly support the hypothesis that sKlotho can exert effects independent of FGF23 and FGFR.-Wright, J. D., An, S.-W., Xie, J., Lim, C., Huang, C.-L. Soluble klotho regulates TRPC6 calcium signaling via lipid rafts, independent of the FGFR-FGF23 pathway.


Sujet(s)
Signalisation calcique , Glucuronidase/métabolisme , Microdomaines membranaires/métabolisme , Membre-6 de la sous-famille C de canaux cationiques à potentiel de récepteur transitoire/métabolisme , Animaux , Sites de fixation , Fixation compétitive , Lignée cellulaire , Facteur-23 de croissance des fibroblastes , Facteurs de croissance fibroblastique/composition chimique , Facteurs de croissance fibroblastique/métabolisme , Glucuronidase/composition chimique , Cellules HEK293 , Humains , Protéines Klotho , Lactose/analogues et dérivés , Lactose/composition chimique , Lactose/métabolisme , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Motifs et domaines d'intéraction protéique , Rats , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/métabolisme , Protéines recombinantes/composition chimique , Protéines recombinantes/métabolisme , Acides sialiques/composition chimique , Acides sialiques/métabolisme , Solubilité , Membre-6 de la sous-famille C de canaux cationiques à potentiel de récepteur transitoire/antagonistes et inhibiteurs , Membre-6 de la sous-famille C de canaux cationiques à potentiel de récepteur transitoire/composition chimique
11.
Trends Pharmacol Sci ; 40(2): 142-153, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30616873

RÉSUMÉ

Endocrine fibroblast growth factors (eFGFs) control pathways that are crucial for maintaining metabolic homeostasis of lipids, glucose, energy, bile acids, and minerals. Unlike the heparin-binding paracrine FGFs, eFGFs require a unique Klotho family protein to form a productive triad complex, but the structural and mechanistical details of this complex have remained obscure since the beginning of the eFGF field. However, recent breakthroughs in resolving the 3D structures of eFGF signaling complexes have now unveiled the atomic details of multivalent interactions among eFGF, FGFR, and Klotho. We provide here a timely review on the architecture and the structure-function relationships of these complexes, and highlight how the structural knowledge opens a new door to structure-based drug design against a repertoire of eFGF-associated metabolic diseases.


Sujet(s)
Facteurs de croissance fibroblastique/composition chimique , Facteurs de croissance fibroblastique/métabolisme , Maladies métaboliques/traitement médicamenteux , Maladies métaboliques/métabolisme , Animaux , Conception de médicament , Système endocrine/métabolisme , Facteur-23 de croissance des fibroblastes , Glucuronidase/composition chimique , Glucuronidase/métabolisme , Humains , Protéines Klotho , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/métabolisme , Relation structure-activité
12.
Biochem Soc Trans ; 46(6): 1753-1770, 2018 12 17.
Article de Anglais | MEDLINE | ID: mdl-30545934

RÉSUMÉ

The receptor tyrosine kinase family of fibroblast growth factor receptors (FGFRs) play crucial roles in embryonic development, metabolism, tissue homeostasis and wound repair via stimulation of intracellular signalling cascades. As a consequence of FGFRs' influence on cell growth, proliferation and differentiation, FGFR signalling is frequently dysregulated in a host of human cancers, variously by means of overexpression, somatic point mutations and gene fusion events. Dysregulation of FGFRs is also the underlying cause of many developmental dysplasias such as hypochondroplasia and achondroplasia. Accordingly, FGFRs are attractive pharmaceutical targets, and multiple clinical trials are in progress for the treatment of various FGFR aberrations. To effectively target dysregulated receptors, a structural and mechanistic understanding of FGFR activation and regulation is required. Here, we review some of the key research findings from the last couple of decades and summarise the strategies being explored for therapeutic intervention.


Sujet(s)
Récepteurs à activité tyrosine kinase/composition chimique , Récepteurs à activité tyrosine kinase/métabolisme , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/métabolisme , Animaux , Humains , Transduction du signal/physiologie
14.
Biochemistry (Mosc) ; 83(8): 930-943, 2018 Aug.
Article de Anglais | MEDLINE | ID: mdl-30208830

RÉSUMÉ

Fibroblast growth factor (FGF) plays an important role in human embryogenesis, angiogenesis, cell proliferation, and differentiation. Carcinogenesis is accompanied by aberrant constitutive activation of FGF receptors (FGFRs) resulting from missense mutation in the FGFR1-4 genes, generation of chimeric oncogenes, FGFR1-4 gene amplification, alternative splicing shift toward formation of mesenchymal FGFR isoforms, and FGFR overexpression. Altogether, these alterations contribute to auto- and paracrine stimulation of cancer cells and neoangiogenesis. Certain missense mutations are found at a high rate in urinary bladder cancer and can be used for non-invasive cancer recurrence diagnostics by analyzing urine cell pellet DNA. Chimeric FGFR1/3 and amplified FGFR1/2 genes can predict cell response to the targeted therapy in various oncological diseases. In recent years, high-throughput sequencing has been used to analyze exomes of virtually all human tumors, which allowed to construct phylogenetic trees of clonal cancer evolution with special emphasis on driver mutations in FGFR1-4 genes. At present, FGFR blockers, such as multi-kinase inhibitors, specific FGFR inhibitors, and FGF ligand traps are being tested in clinical trials. In this review, we discuss current data on the functioning of the FGFR family proteins in both normal and cancer cells, mutations in the FGFR1-4 genes, and mechanisms underlying their oncogenic potential, which might be interesting to a broad range of scientists searching for specific tumor markers and targeted anti-cancer drugs.


Sujet(s)
Carcinogenèse , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/métabolisme , Animaux , Marqueurs biologiques tumoraux/antagonistes et inhibiteurs , Marqueurs biologiques tumoraux/composition chimique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Carcinogenèse/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Humains , Mutation , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/génétique , Transduction du signal/effets des médicaments et des substances chimiques
15.
Chemistry ; 24(31): 7861-7865, 2018 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-29656465

RÉSUMÉ

The interaction of fibroblast growth factors (FGFs) with their fibroblast growth factor receptors (FGFRs) are important in the signaling network of cell growth and development. SSR128129E (SSR), a ligand of small molecular weight with potential anti-cancer properties, acts allosterically on the extracellular domains of FGFRs. Up to now, the structural basis of SSR binding to the D3 domain of FGFR remained elusive. This work reports the structural characterization of the interaction of SSR with one specific receptor, FGFR3, by NMR spectroscopy. This information provides a basis for rational drug design for allosteric FGFR inhibitors.


Sujet(s)
Antinéoplasiques/composition chimique , Indolizine/composition chimique , Inhibiteurs de protéines kinases/composition chimique , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , ortho-Aminobenzoates/composition chimique , Régulation allostérique , Conception de médicament , Simulation de docking moléculaire , Liaison aux protéines , Récepteur facteur croissance fibroblaste/composition chimique , Relation structure-activité , Thermodynamique
16.
J Med Chem ; 61(20): 9085-9104, 2018 10 25.
Article de Anglais | MEDLINE | ID: mdl-29522671

RÉSUMÉ

Fibroblast growth factor receptors (FGFR1-4) are promising therapeutic targets in many cancers. With the resurgence of interest in irreversible inhibitors, efforts have been directed to the discovery of irreversible FGFR inhibitors. Currently, several selective irreversible inhibitors are being evaluated in clinical trials that could covalently target a conserved cysteine in the P-loop of FGFR. In this article, we used a structure-guided approach that is rationalized by a computer-aided simulation to discover the novel and irreversible pan-FGFR inhibitor, 9g, which provided superior FGFR in vitro activities and decent selectivity over VEGFR2 (vascular endothelia growth factor receptor 2). In in vivo studies, 9g displayed clear antitumor activities in NCI-H1581 and SNU-16 xenograft mice models. Additionally, the diluting method confirmed the irreversible binding of 9g to FGFR.


Sujet(s)
Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Conception de médicament , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/métabolisme , Antinéoplasiques/pharmacocinétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Concentration inhibitrice 50 , Mâle , Simulation de docking moléculaire , Conformation des protéines , Pyrimidines/métabolisme , Pyrimidines/pharmacocinétique , Rats , Rat Sprague-Dawley , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/métabolisme , Relation structure-activité , Distribution tissulaire
17.
Cell Rep ; 20(7): 1717-1728, 2017 08 15.
Article de Anglais | MEDLINE | ID: mdl-28813681

RÉSUMÉ

The recent discovery of metabolic roles for fibroblast growth factor 1 (FGF1) in glucose homeostasis has expanded the functions of this classically known mitogen. To dissect the molecular basis for this functional pleiotropy, we engineered an FGF1 partial agonist carrying triple mutations (FGF1ΔHBS) that diminished its ability to induce heparan sulfate (HS)-assisted FGF receptor (FGFR) dimerization and activation. FGF1ΔHBS exhibited a severely reduced proliferative potential, while preserving the full metabolic activity of wild-type FGF1 in vitro and in vivo. Hence, suboptimal FGFR activation by a weak FGF1-FGFR dimer is sufficient to evoke a metabolic response, whereas full FGFR activation by stable and sustained dimerization is required to elicit a mitogenic response. In addition to providing a physical basis for the diverse activities of FGF1, our findings will impact ongoing drug discoveries targeting FGF1 and related FGFs for the treatment of a variety of human diseases.


Sujet(s)
Facteur de croissance fibroblastique de type 1/composition chimique , Hépatocytes/effets des médicaments et des substances chimiques , Mitogènes/composition chimique , Récepteur facteur croissance fibroblaste/composition chimique , Cellules 3T3-L1 , Animaux , Sites de fixation , Lignée cellulaire tumorale , Clonage moléculaire , Escherichia coli/génétique , Escherichia coli/métabolisme , Facteur de croissance fibroblastique de type 1/génétique , Facteur de croissance fibroblastique de type 1/métabolisme , Facteur de croissance fibroblastique de type 1/pharmacologie , Expression des gènes , Hépatocytes/cytologie , Hépatocytes/métabolisme , Humains , Souris , Souris de lignée C57BL , Mitogènes/génétique , Mitogènes/métabolisme , Mitogènes/pharmacologie , Modèles moléculaires , Cellules NIH 3T3 , Liaison aux protéines , Structure en hélice alpha , Structure en brin bêta , Motifs et domaines d'intéraction protéique , Multimérisation de protéines , Stabilité protéique , Rats , Récepteur facteur croissance fibroblaste/génétique , Récepteur facteur croissance fibroblaste/métabolisme , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme
18.
Molecules ; 22(4)2017 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-28379191

RÉSUMÉ

Abnormality of fibroblast growth factor receptor (FGFR)-mediated signaling pathways were frequently found in various human malignancies, making FGFRs hot targets for cancer treatment. To address the consistent need for a new chemotype of FGFR inhibitors, here, we started with a hit structure identified from our internal hepatocyte growth factor receptor (also called c-Met) inhibitor project, and conducted a chemical optimization. After exploring three parts of the hit compound, we finally discovered a new series of pyrrolo[2,3-b]pyrazine FGFR inhibitors, which contain a novel scaffold and unique molecular shape. We believe that our findings can help others to further develop selective FGFR inhibitors.


Sujet(s)
Pyrazines/composition chimique , Pyrazines/pharmacologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/composition chimique , Sites de fixation , Domaine catalytique , Activation enzymatique , Concentration inhibitrice 50 , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Structure moléculaire , Liaison aux protéines , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Relation structure-activité
19.
Molecules ; 21(10)2016 Oct 23.
Article de Anglais | MEDLINE | ID: mdl-27782099

RÉSUMÉ

Tyrosine kinase fibroblast growth factor receptor (FGFR), which is aberrant in various cancer types, is a promising target for cancer therapy. Here we reported the design, synthesis, and biological evaluation of a new series of 6-(2,6-dichloro-3,5-dimethoxyphenyl)-4-substituted-1H-indazole derivatives as potent FGFR inhibitors. The compound 6-(2,6-dichloro-3,5-dimethoxyphenyl)-N-phenyl-1H-indazole-4-carboxamide (10a) was identified as a potent FGFR1 inhibitor, with good enzymatic inhibition. Further structure-based optimization revealed that 6-(2,6-dichloro-3,5-dimethoxyphenyl)-N-(3-(4-methylpiperazin-1-yl)phenyl)-1H-indazole-4-carboxamide (13a) is the most potent FGFR1 inhibitor in this series, with an enzyme inhibitory activity IC50 value of about 30.2 nM.


Sujet(s)
Indazoles/synthèse chimique , Indazoles/pharmacologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Conception de médicament , Humains , Indazoles/composition chimique , Simulation de docking moléculaire , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Récepteur FGFR1/antagonistes et inhibiteurs , Récepteur FGFR1/composition chimique , Récepteur facteur croissance fibroblaste/composition chimique , Relation structure-activité
20.
Mol Biosyst ; 12(12): 3651-3665, 2016 11 15.
Article de Anglais | MEDLINE | ID: mdl-27731453

RÉSUMÉ

Multiple sequence alignments (MSAs) are a fundamental analysis tool used throughout biology to investigate relationships between protein sequence, structure, function, evolutionary history, and patterns of disease-associated variants. However, their widespread application in systems biology research is currently hindered by the lack of user-friendly tools to simultaneously visualize, manipulate and query the information conceptualized in large sequence alignments, and the challenges in integrating MSAs with multiple orthogonal data such as cancer variants and post-translational modifications, which are often stored in heterogeneous data sources and formats. Here, we present the Multiple Sequence Alignment Ontology (MSAOnt), which represents a profile or consensus alignment in an ontological format. Subsets of the alignment are easily selected through the SPARQL Protocol and RDF Query Language for downstream statistical analysis or visualization. We have also created the Kinome Viewer (KinView), an interactive integrative visualization that places eukaryotic protein kinase cancer variants in the context of natural sequence variation and experimentally determined post-translational modifications, which play central roles in the regulation of cellular signaling pathways. Using KinView, we identified differential phosphorylation patterns between tyrosine and serine/threonine kinases in the activation segment, a major kinase regulatory region that is often mutated in proliferative diseases. We discuss cancer variants that disrupt phosphorylation sites in the activation segment, and show how KinView can be used as a comparative tool to identify differences and similarities in natural variation, cancer variants and post-translational modifications between kinase groups, families and subfamilies. Based on KinView comparisons, we identify and experimentally characterize a regulatory tyrosine (Y177PLK4) in the PLK4 C-terminal activation segment region termed the P+1 loop. To further demonstrate the application of KinView in hypothesis generation and testing, we formulate and validate a hypothesis explaining a novel predicted loss-of-function variant (D523NPKCß) in the regulatory spine of PKCß, a recently identified tumor suppressor kinase. KinView provides a novel, extensible interface for performing comparative analyses between subsets of kinases and for integrating multiple types of residue specific annotations in user friendly formats.


Sujet(s)
Biologie informatique/méthodes , Protein kinases/composition chimique , Protein kinases/génétique , Analyse de séquence/méthodes , Logiciel , Séquence d'acides aminés , Mutation , Phosphorylation , Matrices de scores , Motifs et domaines d'intéraction protéique , Protein kinase C beta/génétique , Protein kinases/métabolisme , Maturation post-traductionnelle des protéines , Récepteur facteur croissance fibroblaste/composition chimique , Récepteur facteur croissance fibroblaste/génétique , Récepteurs aux facteurs de croissance dérivés des plaquettes/composition chimique , Récepteurs aux facteurs de croissance dérivés des plaquettes/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE