Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
iScience ; 25(3): 103870, 2022 Mar 18.
Article in English | MEDLINE | ID: mdl-35243233

ABSTRACT

The EphA2 receptor tyrosine kinase activates signaling pathways with different, and sometimes opposite, effects in cancer and other pathologies. Thus, highly specific and potent biased ligands that differentially control EphA2 signaling responses could be therapeutically valuable. Here, we use EphA2-specific monomeric peptides to engineer dimeric ligands with three different geometric configurations to combine a potential ability to differentially modulate EphA2 signaling responses with the high potency and prolonged receptor residence time characteristic of dimeric ligands. The different dimeric peptides readily induce EphA2 clustering, autophosphorylation and signaling, the best with sub-nanomolar potency. Yet, there are differences in two EphA2 signaling responses induced by peptides with different configurations, which exhibit distinct potency and efficacy. The peptides bias signaling when compared with the ephrinA1-Fc ligand and do so via different mechanisms. These findings provide insights into Eph receptor signaling, and proof-of-principle that different Eph signaling responses can be distinctly modulated.

2.
Nat Commun ; 12(1): 7047, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857764

ABSTRACT

Eph receptor tyrosine kinases play a key role in cell-cell communication. Lack of structural information on the entire multi-domain intracellular region of any Eph receptor has hindered understanding of their signaling mechanisms. Here, we use integrative structural biology to investigate the structure and dynamics of the EphA2 intracellular region. EphA2 promotes cancer malignancy through a poorly understood non-canonical form of signaling involving serine/threonine phosphorylation of the linker connecting its kinase and SAM domains. We show that accumulation of multiple linker negative charges, mimicking phosphorylation, induces cooperative changes in the EphA2 intracellular region from more closed to more extended conformations and perturbs the EphA2 juxtamembrane segment and kinase domain. In cells, linker negative charges promote EphA2 oligomerization. We also identify multiple kinases catalyzing linker phosphorylation. Our findings suggest multiple effects of linker phosphorylation on EphA2 signaling and imply that coordination of different kinases is necessary to promote EphA2 non-canonical signaling.


Subject(s)
Receptor, EphA2/chemistry , Serine/chemistry , Sterile Alpha Motif/genetics , Threonine/chemistry , A549 Cells , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Gene Expression , HEK293 Cells , Humans , Models, Molecular , Molecular Mimicry , PC-3 Cells , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine/genetics , Serine/metabolism , Static Electricity , Substrate Specificity , Threonine/genetics , Threonine/metabolism
3.
Dev Cell ; 54(3): 302-316.e7, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32574556

ABSTRACT

Mechanical cues from the extracellular matrix (ECM) regulate various cellular processes via distinct mechanotransduction pathways. In breast cancer, increased ECM stiffness promotes epithelial-to-mesenchymal transition (EMT), cell invasion, and metastasis. Here, we identify a mechanosensitive EPHA2/LYN protein complex regulating EMT and metastasis in response to increasing ECM stiffness during tumor progression. High ECM stiffness leads to ligand-independent phosphorylation of ephrin receptor EPHA2, which recruits and activates the LYN kinase. LYN phosphorylates the EMT transcription factor TWIST1 to release TWIST1 from its cytoplasmic anchor G3BP2 to enter the nucleus, thus triggering EMT and invasion. Genetic and pharmacological inhibition of this pathway prevents breast tumor invasion and metastasis in vivo. In human breast cancer samples, activation of this pathway correlates with collagen fiber alignment, a marker of increasing ECM stiffness. Our findings reveal an EPHA2/LYN/TWIST1 mechanotransduction pathway that responds to mechanical signals from the tumor microenvironment to drive EMT, invasion, and metastasis.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix/metabolism , Nuclear Proteins/metabolism , Receptor, EphA2/metabolism , Twist-Related Protein 1/metabolism , Animals , Breast Neoplasms/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Epithelial-Mesenchymal Transition/genetics , Humans , Mammary Neoplasms, Animal/metabolism , Mechanotransduction, Cellular/genetics , Mice , Receptor, EphA2/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...