Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Synth Biol ; 12(4): 1081-1093, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37011906

ABSTRACT

In recent years, targeted protein degradation (TPD) of plasma membrane proteins by hijacking the ubiquitin proteasome system (UPS) or the lysosomal pathway has emerged as a novel therapeutic avenue in drug development to address and inhibit canonically difficult targets. While TPD strategies have been successful in targeting cell surface receptors, these approaches are limited by the availability of suitable binders to generate heterobifunctional molecules. Here, we present the development of a nanobody (VHH)-based degradation toolbox termed REULR (Receptor Elimination by E3 Ubiquitin Ligase Recruitment). We generated human and mouse cross-reactive nanobodies against five transmembrane PA-TM-RING-type E3 ubiquitin ligases (RNF128, RNF130, RNF167, RNF43, and ZNRF3), covering a broad range and selectivity of tissue expression, with which we characterized the expression in human and mouse cell lines and immune cells (PBMCs). We demonstrate that heterobifunctional REULR molecules can enforce transmembrane E3 ligase interactions with a variety of disease-relevant target receptors (EGFR, EPOR, and PD-1) by induced proximity, resulting in effective membrane clearance of the target receptor at varying levels. In addition, we designed E3 ligase self-degrading molecules, "fratricide" REULRs (RNF128, RNF130, RENF167, RNF43, and ZNRF3), that allow downregulation of one or several E3 ligases from the cell surface and consequently modulate receptor signaling strength. REULR molecules represent a VHH-based modular and versatile "mix and match" targeting strategy for the facile modulation of cell surface proteins by induced proximity to transmembrane PA-TM-RING E3 ligases.


Subject(s)
Proteasome Endopeptidase Complex , Ubiquitin-Protein Ligases , Humans , Animals , Mice , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Proteolysis , Proteasome Endopeptidase Complex/metabolism , Carrier Proteins/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism
2.
Cell ; 185(24): 4560-4573.e19, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36368322

ABSTRACT

Binding of arrestin to phosphorylated G protein-coupled receptors (GPCRs) is crucial for modulating signaling. Once internalized, some GPCRs remain complexed with ß-arrestins, while others interact only transiently; this difference affects GPCR signaling and recycling. Cell-based and in vitro biophysical assays reveal the role of membrane phosphoinositides (PIPs) in ß-arrestin recruitment and GPCR-ß-arrestin complex dynamics. We find that GPCRs broadly stratify into two groups, one that requires PIP binding for ß-arrestin recruitment and one that does not. Plasma membrane PIPs potentiate an active conformation of ß-arrestin and stabilize GPCR-ß-arrestin complexes by promoting a fully engaged state of the complex. As allosteric modulators of GPCR-ß-arrestin complex dynamics, membrane PIPs allow for additional conformational diversity beyond that imposed by GPCR phosphorylation alone. For GPCRs that require membrane PIP binding for ß-arrestin recruitment, this provides a mechanism for ß-arrestin release upon translocation of the GPCR to endosomes, allowing for its rapid recycling.


Subject(s)
Arrestins , Phosphatidylinositols , beta-Arrestins/metabolism , Phosphatidylinositols/metabolism , Arrestins/metabolism , beta-Arrestin 1/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
Elife ; 112022 09 30.
Article in English | MEDLINE | ID: mdl-36178190

ABSTRACT

Secreted proteins, which include cytokines, hormones, and growth factors, are extracellular ligands that control key signaling pathways mediating cell-cell communication within and between tissues and organs. Many drugs target secreted ligands and their cell surface receptors. Still, there are hundreds of secreted human proteins that either have no identified receptors ('orphans') or are likely to act through cell surface receptors that have not yet been characterized. Discovery of secreted ligand-receptor interactions by high-throughput screening has been problematic, because the most commonly used high-throughput methods for protein-protein interaction (PPI) screening are not optimized for extracellular interactions. Cell-based screening is a promising technology for the deorphanization of ligand-receptor interactions, because multimerized ligands can enrich for cells expressing low affinity cell surface receptors, and such methods do not require purification of receptor extracellular domains. Here, we present a proteo-genomic cell-based CRISPR activation (CRISPRa) enrichment screening platform employing customized pooled cell surface receptor sgRNA libraries in combination with a magnetic bead selection-based enrichment workflow for rapid, parallel ligand-receptor deorphanization. We curated 80 potentially high-value orphan secreted proteins and ultimately screened 20 secreted ligands against two cell sgRNA libraries with targeted expression of all single-pass (TM1) or multi-pass transmembrane (TM2+) receptors by CRISPRa. We identified previously unknown interactions in 12 of these screens, and validated several of them using surface plasmon resonance and/or cell binding assays. The newly deorphanized ligands include three receptor protein tyrosine phosphatase (RPTP) ligands and a chemokine-like protein that binds to killer immunoglobulin-like receptors (KIRs). These new interactions provide a resource for future investigations of interactions between the human-secreted and membrane proteomes.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Proteome , Humans , Ligands , Proteome/metabolism , Receptors, Cell Surface/metabolism , Protein Binding/physiology , Cytokines/metabolism , Hormones , Immunoglobulins/metabolism
4.
Cell ; 182(4): 1027-1043.e17, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32822567

ABSTRACT

Cell-surface protein-protein interactions (PPIs) mediate cell-cell communication, recognition, and responses. We executed an interactome screen of 564 human cell-surface and secreted proteins, most of which are immunoglobulin superfamily (IgSF) proteins, using a high-throughput, automated ELISA-based screening platform employing a pooled-protein strategy to test all 318,096 PPI combinations. Screen results, augmented by phylogenetic homology analysis, revealed ∼380 previously unreported PPIs. We validated a subset using surface plasmon resonance and cell binding assays. Observed PPIs reveal a large and complex network of interactions both within and across biological systems. We identified new PPIs for receptors with well-characterized ligands and binding partners for "orphan" receptors. New PPIs include proteins expressed on multiple cell types and involved in diverse processes including immune and nervous system development and function, differentiation/proliferation, metabolism, vascularization, and reproduction. These PPIs provide a resource for further biological investigation into their functional relevance and may offer new therapeutic drug targets.


Subject(s)
Ligands , Protein Interaction Maps/physiology , Receptors, Cell Surface/metabolism , DCC Receptor/chemistry , DCC Receptor/metabolism , Humans , Phylogeny , Receptor-Like Protein Tyrosine Phosphatases, Class 2/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/classification , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/metabolism , Signaling Lymphocytic Activation Molecule Family/chemistry , Signaling Lymphocytic Activation Molecule Family/metabolism , Surface Plasmon Resonance
5.
Cell Rep ; 25(2): 339-349.e9, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30304675

ABSTRACT

WNT7A and WNT7B control CNS angiogenesis and blood-brain barrier formation by activating endothelial Wnt/ß-catenin signaling. The GPI-anchored protein RECK and adhesion G protein-coupled receptor GPR124 critically regulate WNT7-specific signaling in concert with FZD and LRP co-receptors. Here, we demonstrate that primarily the GPR124 ectodomain, but not its transmembrane and intracellular domains, mediates RECK/WNT7-induced canonical Wnt signaling. Moreover, RECK is the predominant binding partner of GPR124 in rat brain blood vessels in situ. WNT7A and WNT7B, but not WNT3A, directly bind to purified recombinant soluble RECK, full-length cell surface RECK, and the GPR124:RECK complex. Chemical cross-linking indicates that RECK and WNT7A associate with 1:1 stoichiometry, which stabilizes short-lived, active, monomeric, hydrophobic WNT7A. In contrast, free WNT7A rapidly converts into inactive, hydrophilic aggregates. Overall, RECK is a selective WNT7 receptor that mediates GPR124/FZD/LRP-dependent canonical Wnt/ß-catenin signaling by stabilizing active cell surface WNT7, suggesting isoform-specific regulation of Wnt bioavailability.


Subject(s)
Frizzled Receptors/metabolism , GPI-Linked Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/metabolism , Wnt3A Protein/metabolism , Animals , Biological Availability , Blood-Brain Barrier , Female , Frizzled Receptors/genetics , GPI-Linked Proteins/genetics , HEK293 Cells , Humans , Male , Protein Binding , Protein Interaction Domains and Motifs , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Wnt Proteins/genetics , Wnt3A Protein/genetics , beta Catenin/genetics , beta Catenin/metabolism
6.
Nature ; 545(7653): 234-237, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28467818

ABSTRACT

Wnt proteins modulate cell proliferation and differentiation and the self-renewal of stem cells by inducing ß-catenin-dependent signalling through the Wnt receptor frizzled (FZD) and the co-receptors LRP5 and LRP6 to regulate cell fate decisions and the growth and repair of several tissues. The 19 mammalian Wnt proteins are cross-reactive with the 10 FZD receptors, and this has complicated the attribution of distinct biological functions to specific FZD and Wnt subtype interactions. Furthermore, Wnt proteins are modified post-translationally by palmitoylation, which is essential for their secretion, function and interaction with FZD receptors. As a result of their acylation, Wnt proteins are very hydrophobic and require detergents for purification, which presents major obstacles to the preparation and application of recombinant Wnt proteins. This hydrophobicity has hindered the determination of the molecular mechanisms of Wnt signalling activation and the functional importance of FZD subtypes, and the use of Wnt proteins as therapeutic agents. Here we develop surrogate Wnt agonists, water-soluble FZD-LRP5/LRP6 heterodimerizers, with FZD5/FZD8-specific and broadly FZD-reactive binding domains. Similar to WNT3A, these Wnt agonists elicit a characteristic ß-catenin signalling response in a FZD-selective fashion, enhance the osteogenic lineage commitment of primary mouse and human mesenchymal stem cells, and support the growth of a broad range of primary human organoid cultures. In addition, the surrogates can be systemically expressed and exhibit Wnt activity in vivo in the mouse liver, regulating metabolic liver zonation and promoting hepatocyte proliferation, resulting in hepatomegaly. These surrogates demonstrate that canonical Wnt signalling can be activated by bi-specific ligands that induce receptor heterodimerization. Furthermore, these easily produced, non-lipidated Wnt surrogate agonists facilitate functional studies of Wnt signalling and the exploration of Wnt agonists for translational applications in regenerative medicine.


Subject(s)
Signal Transduction , Wnt Proteins/agonists , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Frizzled Receptors/metabolism , HEK293 Cells , Hepatocytes/cytology , Hepatomegaly/metabolism , Hepatomegaly/pathology , Humans , Hydrophobic and Hydrophilic Interactions , Intestines/cytology , Ligands , Liver/metabolism , Liver/pathology , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Models, Molecular , Organoids/cytology , Organoids/metabolism , Protein Multimerization , Solubility , Tissue Culture Techniques
8.
Nat Cell Biol ; 11(8): 967-72, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19597489

ABSTRACT

Pin1, a conserved eukaryotic peptidyl-prolyl cis/trans isomerase, has important roles in cellular regulation. Because of its activity to switch the conformation of peptidyl-proline bonds in polypeptide chains, Pin1 operates as a binary switch, often in fate-determining pathways. Pin1 activity is usually controlled by substrate phosphorylation, but how Pin1 switches protein fates has been unclear. Here we show that Pin1 controls the degree of substrate ubiquitylation and thereby protein functions. We found that yeast Pin1 (Ess1) is essential for viability because it controls the NF-kappaB-related Spt23 transcription factor involved in unsaturated fatty-acid synthesis. High Pin1 activity results in low ubiquitylation of Spt23, which triggers Spt23 precursor processing and hence transcription factor activation. By contrast, decreased Pin1 activity leads to robust Spt23 polyubiquitylation and subsequent proteasomal degradation. Inhibition of Pin1 in mammalian cells changes the ubiquitylation status of the tumour suppressor protein p53 from oligoubiquitylation, which is known to trigger nuclear export, to polyubiquitylation, which causes nuclear p53 degradation. This suggests that the Pin1 activity is often translated into a fate-determining ubiquitylation switch, and that Pin1 may affect the degree of substrate ubiquitylation in other pathways as well.


Subject(s)
Peptidylprolyl Isomerase/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Ubiquitination , Active Transport, Cell Nucleus/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Enzyme Inhibitors/pharmacology , Humans , Immunoblotting , Immunoprecipitation , Leupeptins/pharmacology , Membrane Proteins , Microscopy, Fluorescence , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Binding , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity , Trans-Activators/genetics , Transcription Factors , Tumor Suppressor Protein p53/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...