Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nature ; 600(7887): 153-157, 2021 12.
Article in English | MEDLINE | ID: mdl-34819673

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that regulates important functions in the central nervous system1,2. The ALK gene is a hotspot for chromosomal translocation events that result in several fusion proteins that cause a variety of human malignancies3. Somatic and germline gain-of-function mutations in ALK were identified in paediatric neuroblastoma4-7. ALK is composed of an extracellular region (ECR), a single transmembrane helix and an intracellular tyrosine kinase domain8,9. ALK is activated by the binding of ALKAL1 and ALKAL2 ligands10-14 to its ECR, but the lack of structural information for the ALK-ECR or for ALKAL ligands has limited our understanding of ALK activation. Here we used cryo-electron microscopy, nuclear magnetic resonance and X-ray crystallography to determine the atomic details of human ALK dimerization and activation by ALKAL1 and ALKAL2. Our data reveal a mechanism of RTK activation that allows dimerization by either dimeric (ALKAL2) or monomeric (ALKAL1) ligands. This mechanism is underpinned by an unusual architecture of the receptor-ligand complex. The ALK-ECR undergoes a pronounced ligand-induced rearrangement and adopts an orientation parallel to the membrane surface. This orientation is further stabilized by an interaction between the ligand and the membrane. Our findings highlight the diversity in RTK oligomerization and activation mechanisms.


Subject(s)
Anaplastic Lymphoma Kinase/chemistry , Anaplastic Lymphoma Kinase/metabolism , Anaplastic Lymphoma Kinase/ultrastructure , Binding Sites , Cell Membrane/chemistry , Cell Membrane/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Cytokines/chemistry , Cytokines/metabolism , Cytokines/ultrastructure , Enzyme Activation , Humans , Ligands , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Domains , Protein Multimerization
2.
Proc Natl Acad Sci U S A ; 120(7): e2219128120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36745784

ABSTRACT

While important insights were gained about how FGF21 and other endocrine fibroblast growth factors (FGFs) bind to Klotho proteins, the exact mechanism of Klotho/FGF receptor assembly that drives receptor dimerization and activation has not been elucidated. The prevailing dogma is that Klotho proteins substitute for the loss of heparan sulfate proteoglycan (HSPG) binding to endocrine FGFs by high-affinity binding of endocrine FGF molecules to Klotho receptors. To explore a potential role of HSPG in FGF21 signaling, we have analyzed the dynamic properties of FGF21-induced FGF21-ßKlotho-FGFR1c complexes on the surface of living wild-type (WT) or HSPG-deficient Chinese hamster ovary (CHO) cells by employing quantitative single-molecule fluorescence imaging analyses. Moreover, detailed analyses of FGF21 and FGF1 stimulation of cellular signaling pathways activated in WT or in HSPG-deficient CHO cells are also analyzed and compared. These experiments demonstrate that heparin is required for the formation of FGF21-ßKlotho-FGFR1c complexes on the cell membrane and that binding of heparin or HSPG to FGFR1c is essential for optimal FGF21 stimulation of FGFR1c activation, mitogen-activated protein kinase responses, and intracellular Ca2+ release. It is also shown that FGF1 binding stimulates assembly of ßKlotho and FGFR1c on cell membranes, resulting in endocytosis and degradation of ßKlotho. We conclude that heparin or HSPG is essential for FGF21 signaling and for regulation of ßKlotho cellular stability by acting as a coligand of FGFR1c.


Subject(s)
Heparan Sulfate Proteoglycans , Klotho Proteins , Cricetinae , Animals , CHO Cells , Cricetulus , Heparin , Fibroblast Growth Factor 1 , Fibroblast Growth Factors/metabolism , Signal Transduction/physiology
3.
Proc Natl Acad Sci U S A ; 120(13): e2300054120, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36943885

ABSTRACT

The receptor tyrosine kinase KIT and its ligand stem cell factor (SCF) are required for the development of hematopoietic stem cells, germ cells, and other cells. A variety of human cancers, such as acute myeloid leukemia, gastrointestinal stromal tumor, and mast cell leukemia, are driven by somatic gain-of-function KIT mutations. Here, we report cryo electron microscopy (cryo-EM) structural analyses of full-length wild-type and two oncogenic KIT mutants, which show that the overall symmetric arrangement of the extracellular domain of ligand-occupied KIT dimers contains asymmetric D5 homotypic contacts juxtaposing the plasma membrane. Mutational analysis of KIT reveals in D5 region an "Achilles heel" for therapeutic intervention. A ligand-sensitized oncogenic KIT mutant exhibits a more comprehensive and stable D5 asymmetric conformation. A constitutively active ligand-independent oncogenic KIT mutant adopts a V-shaped conformation solely held by D5-mediated contacts. Binding of SCF to this mutant fully restores the conformation of wild-type KIT dimers, including the formation of salt bridges responsible for D4 homotypic contacts and other hallmarks of SCF-induced KIT dimerization. These experiments reveal an unexpected structural plasticity of oncogenic KIT mutants and a therapeutic target in D5.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-kit , Humans , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Ligands , Cryoelectron Microscopy , Receptor Protein-Tyrosine Kinases/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Phosphorylation
4.
Nature ; 553(7689): 501-505, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29342135

ABSTRACT

Canonical fibroblast growth factors (FGFs) activate FGF receptors (FGFRs) through paracrine or autocrine mechanisms in a process that requires cooperation with heparan sulfate proteoglycans, which function as co-receptors for FGFR activation. By contrast, endocrine FGFs (FGF19, FGF21 and FGF23) are circulating hormones that regulate critical metabolic processes in a variety of tissues. FGF19 regulates bile acid synthesis and lipogenesis, whereas FGF21 stimulates insulin sensitivity, energy expenditure and weight loss. Endocrine FGFs signal through FGFRs in a manner that requires klothos, which are cell-surface proteins that possess tandem glycosidase domains. Here we describe the crystal structures of free and ligand-bound ß-klotho extracellular regions that reveal the molecular mechanism that underlies the specificity of FGF21 towards ß-klotho and demonstrate how the FGFR is activated in a klotho-dependent manner. ß-Klotho serves as a primary 'zip code'-like receptor that acts as a targeting signal for FGF21, and FGFR functions as a catalytic subunit that mediates intracellular signalling. Our structures also show how the sugar-cutting enzyme glycosidase has evolved to become a specific receptor for hormones that regulate metabolic processes, including the lowering of blood sugar levels. Finally, we describe an agonistic variant of FGF21 with enhanced biological activity and present structural insights into the potential development of therapeutic agents for diseases linked to endocrine FGFs.


Subject(s)
Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Signal Transduction , Binding Sites , Crystallography, X-Ray , Extracellular Space/metabolism , Fibroblast Growth Factor-23 , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , HEK293 Cells , Humans , Klotho Proteins , Ligands , Models, Molecular , Protein Binding , Protein Domains , Receptors, Fibroblast Growth Factor/metabolism , Substrate Specificity
5.
Proc Natl Acad Sci U S A ; 117(50): 31800-31807, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257569

ABSTRACT

The three members of the endocrine-fibroblast growth factor (FGF) family, FGF19, 21, and 23 are circulating hormones that regulate critical metabolic processes. FGF23 stimulates the assembly of a signaling complex composed of α-Klotho (KLA) and FGF receptor (FGFR) resulting in kinase activation, regulation of phosphate homeostasis, and vitamin D levels. Here we report that the C-terminal tail of FGF23, a region responsible for KLA binding, contains two tandem repeats, repeat 1 (R1) and repeat 2 (R2) that function as two distinct ligands for KLA. FGF23 variants with a single KLA binding site, FGF23-R1, FGF23-R2, or FGF23-wild type (WT) with both R1 and R2, bind to KLA with similar binding affinity and stimulate FGFR1 activation and MAPK response. R2 is flanked by two cysteines that form a disulfide bridge in FGF23-WT; disulfide bridge formation in FGF23-WT is dispensable for KLA binding and for cell signaling via FGFRs. We show that FGF23-WT stimulates dimerization and activation of a chimeric receptor molecule composed of the extracellular domain of KLA fused to the cytoplasmic domain of FGFR and employ total internal reflection fluorescence microscopy to visualize individual KLA molecules on the cell surface. These experiments demonstrate that FGF23-WT can act as a bivalent ligand of KLA in the cell membrane. Finally, an engineered Fc-R2 protein acts as an FGF23 antagonist offering new pharmacological intervention for treating diseases caused by excessive FGF23 abundance or activity.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Protein Multimerization/physiology , Binding Sites , Calcinosis/drug therapy , Calcinosis/genetics , Cell Membrane/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/therapeutic use , HEK293 Cells , Humans , Hyperostosis, Cortical, Congenital/drug therapy , Hyperostosis, Cortical, Congenital/genetics , Hyperphosphatemia/drug therapy , Hyperphosphatemia/genetics , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/therapeutic use , Klotho Proteins , Mutation , Osteomalacia/drug therapy , Osteomalacia/genetics , Protein Binding/drug effects , Protein Binding/physiology , Protein Domains , Protein Multimerization/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Rickets, Hypophosphatemic/drug therapy , Rickets, Hypophosphatemic/genetics
6.
Proc Natl Acad Sci U S A ; 116(16): 7819-7824, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30944224

ABSTRACT

The three members of the endocrine fibroblast growth factor (FGF) family designated FGF19, FGF21, and FGF23 mediate their pleiotropic cellular effects by binding to and activating binary complexes composed of an FGF receptor (FGFR) bound to either α-Klotho or ß-Klotho receptors. Structural analyses of ligand-occupied Klotho extracellular domains have provided important insights concerning mechanisms underlying the binding specificities of FGF21 and FGF23 to ß-Klotho or α-Klotho, respectively. They have also demonstrated that Klotho proteins function as primary high-affinity receptors while FGFRs function as the catalytic subunits that mediate intracellular signaling. Here we describe the crystal structure the C-terminal tail of FGF19 (FGF19CT) bound to sKLB and demonstrate that FGF19CT and FGF21CT bind to the same binding site on sKLB, via a multiturn D-P motif to site 1 and via a S-P-S motif to the pseudoglycoside hydrolase region (site 2). Binding affinities to sKLB and cellular stimulatory activities of FGF19CT, FGF21CT, and a variety of chimeric mutants to cells expressing ß-Klotho together with FGFR1c or FGFR4 were also analyzed. These experiments as well as detailed comparison of the structures of free and ligand-occupied sKLB to the structure of ligand-occupied sKLA reveal a general mechanism for recognition of endocrine FGFs by Klotho proteins and regulatory interactions with FGFRs that control their pleiotropic cellular responses.


Subject(s)
Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Animals , Cell Line , Fibroblast Growth Factor-23 , Humans , Klotho Proteins , Membrane Proteins/genetics , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation , Rats , Signal Transduction/physiology , Substrate Specificity
7.
Proc Natl Acad Sci U S A ; 115(33): 8340-8345, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061385

ABSTRACT

Elucidating the physiological roles and modes of action of the recently discovered ligands (designated ALKAL1,2 or AUG-α,ß) of the receptor tyrosine kinases Anaplastic Lymphoma Kinase (ALK) and Leukocyte Tyrosine Kinase (LTK) has been limited by difficulties in producing sufficient amounts of the two ligands and their poor stability. Here we describe procedures for expression and purification of AUG-α and a deletion mutant lacking the N-terminal variable region. Detailed biochemical characterization of AUG-α by mass spectrometry shows that the four conserved cysteines located in the augmentor domain (AD) form two intramolecular disulfide bridges while a fifth, primate-specific cysteine located in the N-terminal variable region mediates dimerization through formation of a disulfide bridge between two AUG-α molecules. In contrast to AUG-α, the capacity of AUG-α AD to undergo dimerization is strongly compromised. However, full-length AUG-α and the AUG-α AD deletion mutant stimulate similar tyrosine phosphorylation of cells expressing either ALK or LTK. Both AUG-α and AUG-α AD also stimulate a similar profile of MAP kinase response in L6 cells and colony formation in soft agar by autocrine stimulation of NIH 3T3 cells expressing ALK. Moreover, both AUG-α and AUG-α AD stimulate neuronal differentiation of human neuroblastoma NB1 and PC12 cells in a similar dose-dependent manner. Taken together, these experiments show that deletion of the N-terminal variable region minimally affects the activity of AUG-α toward LTK or ALK stimulation in cultured cells. Reduced dimerization might be compensated by high local concentration of AUG-α AD bound to ALK at the cell membrane and by potential ligand-induced receptor-receptor interactions.


Subject(s)
Cytokines/isolation & purification , Receptor Protein-Tyrosine Kinases/isolation & purification , Amino Acid Motifs , Anaplastic Lymphoma Kinase , Animals , Cytokines/chemistry , Cytokines/physiology , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , PC12 Cells , Protein Multimerization , Rats , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism
8.
Proc Natl Acad Sci U S A ; 112(52): 15862-7, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26630010

ABSTRACT

Receptor tyrosine kinases (RTKs) are a class of cell surface receptors that, upon ligand binding, stimulate a variety of critical cellular functions. The orphan receptor anaplastic lymphoma kinase (ALK) is one of very few RTKs that remain without a firmly established protein ligand. Here we present a novel cytokine, FAM150B, which we propose naming augmentor-α (AUG-α), as a ligand for ALK. AUG-α binds ALK with high affinity and activates ALK in cells with subnanomolar potency. Detailed binding experiments using cells expressing ALK or the related receptor leukocyte tyrosine kinase (LTK) demonstrate that AUG-α binds and robustly activates both ALK and LTK. We show that the previously established LTK ligand FAM150A (AUG-ß) is specific for LTK and only weakly binds to ALK. Furthermore, expression of AUG-α stimulates transformation of NIH/3T3 cells expressing ALK, induces IL-3 independent growth of Ba/F3 cells expressing ALK, and is expressed in neuroblastoma, a cancer partly driven by ALK. These experiments reveal the hierarchy and specificity of two cytokines as ligands for ALK and LTK and set the stage for elucidating their roles in development and disease states.


Subject(s)
Cytokines/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cytokines/genetics , Doxycycline/pharmacology , Enzyme Activation/drug effects , HEK293 Cells , Heparin/pharmacology , Humans , Immunoblotting , Ligands , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Binding , Receptor Protein-Tyrosine Kinases/genetics , Sequence Homology, Amino Acid
9.
Cell Rep ; 41(4): 111545, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36288716

ABSTRACT

Cellular signaling by fibroblast growth factor receptors (FGFRs) is a highly regulated process mediated by specific interactions between distinct subsets of fibroblast growth factor (FGF) ligands and two FGFR isoforms generated by alternative splicing: an epithelial b- and mesenchymal c-isoforms. Here, we investigate the properties of a mini-protein, mb7, developed by an in silico design strategy to bind to the ligand-binding region of FGFR2. We describe structural, biophysical, and cellular analyses demonstrating that mb7 binds with high affinity to the c-isoforms of FGFR, resulting in inhibition of cellular signaling induced by a subset of FGFs that preferentially activate c-isoforms of FGFR. Notably, as mb7 blocks interaction between FGFR with Klotho proteins, it functions as an antagonist of the metabolic hormones FGF19 and FGF21, providing mechanistic insights and strategies for the development of therapeutics for diseases driven by aberrantly activated FGFRs.


Subject(s)
Fibroblast Growth Factors , Receptors, Fibroblast Growth Factor , Ligands , Receptors, Fibroblast Growth Factor/metabolism , Protein Isoforms/metabolism , Fibroblast Growth Factors/metabolism , Signal Transduction , Hormones
10.
J Med Chem ; 65(12): 8380-8400, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35653642

ABSTRACT

JAK2 is a non-receptor tyrosine kinase that regulates hematopoiesis through the JAK-STAT pathway. The pseudokinase domain (JH2) is an important regulator of the activity of the kinase domain (JH1). V617F mutation in JH2 has been associated with the pathogenesis of various myeloproliferative neoplasms, but JAK2 JH2 has been poorly explored as a pharmacological target. In light of this, we aimed to develop JAK2 JH2 binders that could selectively target JH2 over JH1 and test their capacity to modulate JAK2 activity in cells. Toward this goal, we optimized a diaminotriazole lead compound into potent, selective, and cell-permeable JH2 binders leveraging computational design, synthesis, binding affinity measurements for the JH1, JH2 WT, and JH2 V617F domains, permeability measurements, crystallography, and cell assays. Optimized diaminotriazoles are capable of inhibiting STAT5 phosphorylation in both WT and V617F JAK2 in cells.


Subject(s)
Janus Kinases , Myeloproliferative Disorders , Humans , Janus Kinase 2/metabolism , Janus Kinases/metabolism , Ligands , Mutation , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL