Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell ; 168(6): 1041-1052.e18, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283060

ABSTRACT

Most secreted growth factors and cytokines are functionally pleiotropic because their receptors are expressed on diverse cell types. While important for normal mammalian physiology, pleiotropy limits the efficacy of cytokines and growth factors as therapeutics. Stem cell factor (SCF) is a growth factor that acts through the c-Kit receptor tyrosine kinase to elicit hematopoietic progenitor expansion but can be toxic when administered in vivo because it concurrently activates mast cells. We engineered a mechanism-based SCF partial agonist that impaired c-Kit dimerization, truncating downstream signaling amplitude. This SCF variant elicited biased activation of hematopoietic progenitors over mast cells in vitro and in vivo. Mouse models of SCF-mediated anaphylaxis, radioprotection, and hematopoietic expansion revealed that this SCF partial agonist retained therapeutic efficacy while exhibiting virtually no anaphylactic off-target effects. The approach of biasing cell activation by tuning signaling thresholds and outputs has applications to many dimeric receptor-ligand systems.


Subject(s)
Anaphylaxis/metabolism , Hematopoietic Stem Cells/immunology , Mast Cells/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Stem Cell Factor/metabolism , Anaphylaxis/immunology , Animals , Dimerization , Humans , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Engineering , Proto-Oncogene Proteins c-kit/agonists , Proto-Oncogene Proteins c-kit/chemistry , Stem Cell Factor/chemistry , Stem Cell Factor/genetics
2.
Cell ; 168(6): 1053-1064.e15, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283061

ABSTRACT

Cytokines are classically thought to stimulate downstream signaling pathways through monotonic activation of receptors. We describe a severe anemia resulting from a homozygous mutation (R150Q) in the cytokine erythropoietin (EPO). Surprisingly, the EPO R150Q mutant shows only a mild reduction in affinity for its receptor but has altered binding kinetics. The EPO mutant is less effective at stimulating erythroid cell proliferation and differentiation, even at maximally potent concentrations. While the EPO mutant can stimulate effectors such as STAT5 to a similar extent as the wild-type ligand, there is reduced JAK2-mediated phosphorylation of select downstream targets. This impairment in downstream signaling mechanistically arises from altered receptor dimerization dynamics due to extracellular binding changes. These results demonstrate how variation in a single cytokine can lead to biased downstream signaling and can thereby cause human disease. Moreover, we have defined a distinct treatable form of anemia through mutation identification and functional studies.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/pathology , Erythropoietin/genetics , Mutation, Missense , Signal Transduction , Anemia, Diamond-Blackfan/therapy , Child , Consanguinity , Enzyme Activation , Erythropoiesis , Erythropoietin/chemistry , Female , Humans , Janus Kinase 2/metabolism , Kinetics , Male , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism
3.
Cell ; 160(6): 1196-208, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25728669

ABSTRACT

Most cell-surface receptors for cytokines and growth factors signal as dimers, but it is unclear whether remodeling receptor dimer topology is a viable strategy to "tune" signaling output. We utilized diabodies (DA) as surrogate ligands in a prototypical dimeric receptor-ligand system, the cytokine Erythropoietin (EPO) and its receptor (EpoR), to dimerize EpoR ectodomains in non-native architectures. Diabody-induced signaling amplitudes varied from full to minimal agonism, and structures of these DA/EpoR complexes differed in EpoR dimer orientation and proximity. Diabodies also elicited biased or differential activation of signaling pathways and gene expression profiles compared to EPO. Non-signaling diabodies inhibited proliferation of erythroid precursors from patients with a myeloproliferative neoplasm due to a constitutively active JAK2V617F mutation. Thus, intracellular oncogenic mutations causing ligand-independent receptor activation can be counteracted by extracellular ligands that re-orient receptors into inactive dimer topologies. This approach has broad applications for tuning signaling output for many dimeric receptor systems.


Subject(s)
Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Cell Line , Crystallography, X-Ray , Dimerization , Erythropoietin/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Point Mutation , Protein Engineering , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/antagonists & inhibitors , Sequence Alignment
4.
Proc Natl Acad Sci U S A ; 120(20): e2301908120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155863

ABSTRACT

The endosomal system of eukaryotic cells represents a central sorting and recycling compartment linked to metabolic signaling and the regulation of cell growth. Tightly controlled activation of Rab GTPases is required to establish the different domains of endosomes and lysosomes. In metazoans, Rab7 controls endosomal maturation, autophagy, and lysosomal function. It is activated by the guanine nucleotide exchange factor (GEF) complex Mon1-Ccz1-Bulli (MCBulli) of the tri-longin domain (TLD) family. While the Mon1 and Ccz1 subunits have been shown to constitute the active site of the complex, the role of Bulli remains elusive. We here present the cryo-electron microscopy (cryo-EM) structure of MCBulli at 3.2 Å resolution. Bulli associates as a leg-like extension at the periphery of the Mon1 and Ccz1 heterodimers, consistent with earlier reports that Bulli does not impact the activity of the complex or the interactions with recruiter and substrate GTPases. While MCBulli shows structural homology to the related ciliogenesis and planar cell polarity effector (Fuzzy-Inturned-Wdpcp) complex, the interaction of the TLD core subunits Mon1-Ccz1 and Fuzzy-Inturned with Bulli and Wdpcp, respectively, is remarkably different. The variations in the overall architecture suggest divergent functions of the Bulli and Wdpcp subunits. Based on our structural analysis, Bulli likely serves as a recruitment platform for additional regulators of endolysosomal trafficking to sites of Rab7 activation.


Subject(s)
Vesicular Transport Proteins , rab GTP-Binding Proteins , Animals , Vesicular Transport Proteins/metabolism , Cryoelectron Microscopy , Protein Transport , rab GTP-Binding Proteins/metabolism , Endosomes/metabolism , Guanine Nucleotide Exchange Factors/metabolism
5.
EMBO Rep ; 23(10): e55450, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35920255

ABSTRACT

Interleukin 27 (IL-27) is a heterodimeric cytokine that elicits potent immunosuppressive responses. Comprised of EBI3 and p28 subunits, IL-27 binds GP130 and IL-27Rα receptor chains to activate the JAK/STAT signaling cascade. However, how these receptors recognize IL-27 and form a complex capable of phosphorylating JAK proteins remains unclear. Here, we used cryo electron microscopy (cryoEM) and AlphaFold modeling to solve the structure of the IL-27 receptor recognition complex. Our data show how IL-27 serves as a bridge connecting IL-27Rα (domains 1-2) with GP130 (domains 1-3) to initiate signaling. While both receptors contact the p28 component of the heterodimeric cytokine, EBI3 stabilizes the complex by binding a positively charged surface of IL-27Rα and Domain 1 of GP130. We find that assembly of the IL-27 receptor recognition complex is distinct from both IL-12 and IL-6 cytokine families and provides a mechanistic blueprint for tuning IL-27 pleiotropic actions.


Subject(s)
Cytokine Receptor gp130 , Interleukin-27 , Receptors, Interleukin , Cytokine Receptor gp130/chemistry , Humans , Interleukin-12 , Interleukin-27/chemistry , Interleukin-6 , Interleukins , Receptors, Interleukin/chemistry
6.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33384332

ABSTRACT

Thrombopoietin (TPO) and the TPO-receptor (TPO-R, or c-MPL) are essential for hematopoietic stem cell (HSC) maintenance and megakaryocyte differentiation. Agents that can modulate TPO-R signaling are highly desirable for both basic research and clinical utility. We developed a series of surrogate protein ligands for TPO-R, in the form of diabodies (DBs), that homodimerize TPO-R on the cell surface in geometries that are dictated by the DB receptor binding epitope, in effect "tuning" downstream signaling responses. These surrogate ligands exhibit diverse pharmacological properties, inducing graded signaling outputs, from full to partial TPO agonism, thus decoupling the dual functions of TPO/TPO-R. Using single-cell RNA sequencing and HSC self-renewal assays we find that partial agonistic diabodies preserved the stem-like properties of cultured HSCs, but also blocked oncogenic colony formation in essential thrombocythemia (ET) through inverse agonism. Our data suggest that dampening downstream TPO signaling is a powerful approach not only for HSC preservation in culture, but also for inhibiting oncogenic signaling through the TPO-R.


Subject(s)
Receptors, Thrombopoietin/metabolism , Thrombopoietin/metabolism , Cell Differentiation/physiology , Cell Membrane/metabolism , Epitopes/immunology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Ligands , Megakaryocytes/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Cytokine/metabolism , Receptors, Thrombopoietin/immunology , Receptors, Thrombopoietin/physiology , Signal Transduction/physiology , Thrombocythemia, Essential/metabolism , Thrombopoietin/physiology
7.
Anal Chem ; 86(17): 8593-602, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25148216

ABSTRACT

Unraveling the spatiotemporal organization of signaling complexes within the context of plasma membrane nanodomains has remained a highly challenging task. Here, we have applied super-resolution image correlation based on tracking and localization microscopy (TALM) for probing transient confinement as well as ligand binding and intracellular effector recruitment of the type I interferon (IFN) receptor in the plasma membrane of live cells. Ligand and receptor were labeled with monofunctional quantum dots, thus allowing long-term tracking with very high spatial and temporal resolution without an artificial receptor cross-linking at the cell surface. Dual-color TALM was employed for visualizing protein-protein interactions involved in IFN signaling at both sides of the plasma membrane with high spatial and temporal resolution. By pair correlation analyses based on time-lapse TALM images (pcTALM), complex assembly within dynamic submicroscopic zones was identified. Strikingly, recruitment of the IFN effector protein signal transducer and activator of transcription 2 (STAT2) into these dynamic signaling zones could be observed. The results suggest that confined diffusion zones in the plasma membrane are employed as transient platforms for the assembly of signaling complexes.


Subject(s)
Microscopy, Fluorescence , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Biotin/chemistry , Biotin/metabolism , Cell Membrane/metabolism , HeLa Cells , Humans , Oligopeptides/chemistry , Oligopeptides/metabolism , Protein Interaction Maps , Quantum Dots/chemistry , Receptor, Interferon alpha-beta/genetics , STAT2 Transcription Factor/metabolism , Time-Lapse Imaging
8.
Curr Opin Cell Biol ; 83: 102177, 2023 08.
Article in English | MEDLINE | ID: mdl-37327649

ABSTRACT

Rab GTPases are molecular switches with essential roles in mediating vesicular trafficking and establishing organelle identity. The conversion from the inactive, cytosolic to the membrane-bound, active species and back is tightly controlled by regulatory proteins. Recently, the roles of membrane properties and lipid composition of different target organelles in determining the activity state of Rabs have come to light. The investigation of several Rab guanine nucleotide exchange factors (GEFs) has revealed principles of how the recruitment via lipid interactions and the spatial confinement on the membrane surface contribute to spatiotemporal specificity in the Rab GTPase network. This paints an intricate picture of the control mechanisms in Rab activation and highlights the importance of the membrane lipid code in the organization of the endomembrane system.


Subject(s)
Guanine Nucleotide Exchange Factors , rab GTP-Binding Proteins , Guanine Nucleotide Exchange Factors/metabolism , Organelles/metabolism , Membrane Lipids
9.
Angew Chem Int Ed Engl ; 51(20): 4868-71, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22488831

ABSTRACT

In living color: efficient intracellular covalent labeling of proteins with a photoswitchable dye using the HaloTag for dSTORM super-resolution imaging in live cells is described. The dynamics of cellular nanostructures at the plasma membrane were monitored with a time resolution of a few seconds. In combination with dual-color FPALM imaging, submicroscopic receptor organization within the context of the membrane skeleton was resolved.


Subject(s)
Cell Membrane/chemistry , Green Fluorescent Proteins/chemistry , Microscopy, Fluorescence/methods , Actins/chemistry , Actins/metabolism , Cell Membrane/metabolism , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Nanostructures/chemistry , Transfection
10.
Cell Rep Methods ; 2(2): 100165, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35474965

ABSTRACT

Localization and tracking of individual receptors by single-molecule imaging opens unique possibilities to unravel the assembly and dynamics of signaling complexes in the plasma membrane. We present a comprehensive workflow for imaging and analyzing receptor diffusion and interaction in live cells at single molecule level with up to four colors. Two engineered, monomeric GFP variants, which are orthogonally recognized by anti-GFP nanobodies, are employed for efficient and selective labeling of target proteins in the plasma membrane with photostable fluorescence dyes. This labeling technique enables us to quantitatively resolve the stoichiometry and dynamics of the interferon-γ (IFNγ) receptor signaling complex in the plasma membrane of living cells by multicolor single-molecule imaging. Based on versatile spatial and spatiotemporal correlation analyses, we identify ligand-induced receptor homo- and heterodimerization. Multicolor single-molecule co-tracking and quantitative single-molecule Förster resonance energy transfer moreover reveals transient assembly of IFNγ receptor heterotetramers and confirms its structural architecture.


Subject(s)
Fluorescence Resonance Energy Transfer , Single Molecule Imaging , Single Molecule Imaging/methods , Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer/methods , Proteins/chemistry , Fluorescent Dyes/chemistry
11.
Sci Immunol ; 7(78): eade5686, 2022 12 09.
Article in English | MEDLINE | ID: mdl-36459543

ABSTRACT

Cytokines interact with their receptors in the extracellular space to control immune responses. How the physicochemical properties of the extracellular space influence cytokine signaling is incompletely elucidated. Here, we show that the activity of interleukin-2 (IL-2), a cytokine critical to T cell immunity, is profoundly affected by pH, limiting IL-2 signaling within the acidic environment of tumors. Generation of lactic acid by tumors limits STAT5 activation, effector differentiation, and antitumor immunity by CD8+ T cells and renders high-dose IL-2 therapy poorly effective. Directed evolution enabled selection of a pH-selective IL-2 mutein (Switch-2). Switch-2 binds the IL-2 receptor subunit IL-2Rα with higher affinity, triggers STAT5 activation, and drives CD8+ T cell effector function more potently at acidic pH than at neutral pH. Consequently, high-dose Switch-2 therapy induces potent immune activation and tumor rejection with reduced on-target toxicity in normal tissues. Last, we show that sensitivity to pH is a generalizable property of a diverse range of cytokines with broad relevance to immunity and immunotherapy in healthy and diseased tissues.


Subject(s)
Interleukin-2 , Neoplasms , Humans , STAT5 Transcription Factor , CD8-Positive T-Lymphocytes , Cytokines , Hydrogen-Ion Concentration
12.
Elife ; 102021 04 19.
Article in English | MEDLINE | ID: mdl-33871355

ABSTRACT

Cytokines elicit pleiotropic and non-redundant activities despite strong overlap in their usage of receptors, JAKs and STATs molecules. We use IL-6 and IL-27 to ask how two cytokines activating the same signaling pathway have different biological roles. We found that IL-27 induces more sustained STAT1 phosphorylation than IL-6, with the two cytokines inducing comparable levels of STAT3 phosphorylation. Mathematical and statistical modeling of IL-6 and IL-27 signaling identified STAT3 binding to GP130, and STAT1 binding to IL-27Rα, as the main dynamical processes contributing to sustained pSTAT1 levels by IL-27. Mutation of Tyr613 on IL-27Rα decreased IL-27-induced STAT1 phosphorylation by 80% but had limited effect on STAT3 phosphorgylation. Strong receptor/STAT coupling by IL-27 initiated a unique gene expression program, which required sustained STAT1 phosphorylation and IRF1 expression and was enriched in classical Interferon Stimulated Genes. Interestingly, the STAT/receptor coupling exhibited by IL-6/IL-27 was altered in patients with systemic lupus erythematosus (SLE). IL-6/IL-27 induced a more potent STAT1 activation in SLE patients than in healthy controls, which correlated with higher STAT1 expression in these patients. Partial inhibition of JAK activation by sub-saturating doses of Tofacitinib specifically lowered the levels of STAT1 activation by IL-6. Our data show that receptor and STATs concentrations critically contribute to shape cytokine responses and generate functional pleiotropy in health and disease.


Subject(s)
Cytokine Receptor gp130/agonists , Interleukin-27/pharmacology , Interleukin-6/pharmacology , Receptors, Interleukin/agonists , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Th1 Cells/drug effects , Amino Acid Motifs , Binding, Competitive , Case-Control Studies , Cells, Cultured , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Humans , Interferon Regulatory Factor-1/metabolism , Interleukin-27/metabolism , Interleukin-6/metabolism , Kinetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Models, Biological , Mutation , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism
13.
Cell Rep ; 33(12): 108545, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33357429

ABSTRACT

Cytokines are highly pleiotropic ligands that regulate the immune response. Here, using interleukin-6 (IL-6) as a model system, we perform detailed phosphoproteomic and transcriptomic studies in human CD4+ T helper 1 (Th-1) cells to address the molecular bases defining cytokine functional pleiotropy. We identify CDK8 as a negative regulator of STAT3 transcriptional activities, which interacts with STAT3 upon IL-6 stimulation. Inhibition of CDK8 activity, using specific small molecule inhibitors, reduces the IL-6-induced phosphoproteome by 23% in Th-1 cells, including STAT3 S727 phosphorylation. STAT3 binding to target DNA sites in the genome is increased upon CDK8 inhibition, which results in a concomitant increase in STAT3-mediated transcriptional activity. Importantly, inhibition of CDK8 activity under Th-17 polarizing conditions results in an enhancement of Th-17 differentiation. Our results support a model where CDK8 regulates STAT3 transcriptional processivity by modulation of its gene loci resident time, critically contributing to diversification of IL-6 responses.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Chromosome Mapping/methods , Cyclin-Dependent Kinase 8/genetics , Humans , Phosphorylation , Signal Transduction
14.
Sci Signal ; 13(649)2020 09 15.
Article in English | MEDLINE | ID: mdl-32934073

ABSTRACT

Interleukin-10 (IL-10) is a dimeric cytokine with both immunosuppressive and immunostimulatory activities; however, IL-10-based therapies have shown only marginal clinical benefits. Here, we explored whether the stability of the IL-10 receptor complex contributes to the immunomodulatory potency of IL-10. We generated an IL-10 mutant with enhanced affinity for its IL-10Rß receptor using yeast surface display. Compared to the wild-type cytokine, the affinity-enhanced IL-10 variants recruited IL-10Rß more efficiently into active cell surface signaling complexes and triggered greater STAT1 and STAT3 activation in human monocytes and CD8+ T cells. These effects, in turn, led to more robust induction of IL-10-mediated gene expression programs at low ligand concentrations in both human cell subsets. IL-10-regulated genes are involved in monocyte energy homeostasis, migration, and trafficking and in CD8+ T cell exhaustion. At nonsaturating doses, IL-10 did not induce key components of its gene expression program, which may explain its lack of efficacy in clinical settings. Our engineered IL-10 variant showed a more robust bioactivity profile than that of wild-type IL-10 at low doses in monocytes and CD8+ T cells. Moreover, CAR-modified T cells expanded with the engineered IL-10 variant displayed superior cytolytic activity than those expanded with wild-type IL-10. Our study provides insights into how IL-10 receptor complex stability fine-tunes IL-10 biology and opens new opportunities to revitalize failed IL-10 therapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-10/immunology , Monocytes/immunology , Mutation/immunology , Signal Transduction/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Energy Metabolism/genetics , Energy Metabolism/immunology , Gene Expression Profiling/methods , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Ligands , Monocytes/cytology , Monocytes/metabolism , Protein Binding , Receptors, Interleukin-10/genetics , Receptors, Interleukin-10/immunology , Receptors, Interleukin-10/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Sf9 Cells , Signal Transduction/genetics , Spodoptera
15.
Science ; 367(6478): 643-652, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32029621

ABSTRACT

Homodimeric class I cytokine receptors are assumed to exist as preformed dimers that are activated by ligand-induced conformational changes. We quantified the dimerization of three prototypic class I cytokine receptors in the plasma membrane of living cells by single-molecule fluorescence microscopy. Spatial and spatiotemporal correlation of individual receptor subunits showed ligand-induced dimerization and revealed that the associated Janus kinase 2 (JAK2) dimerizes through its pseudokinase domain. Oncogenic receptor and hyperactive JAK2 mutants promoted ligand-independent dimerization, highlighting the formation of receptor dimers as the switch responsible for signal activation. Atomistic modeling and molecular dynamics simulations based on a detailed energetic analysis of the interactions involved in dimerization yielded a mechanistic blueprint for homodimeric class I cytokine receptor activation and its dysregulation by individual mutations.


Subject(s)
Carcinogenesis/genetics , Cell Membrane/chemistry , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Protein Multimerization , Receptors, Erythropoietin/chemistry , Receptors, Somatotropin/chemistry , Receptors, Thrombopoietin/chemistry , Amino Acid Substitution/genetics , HeLa Cells , Humans , Janus Kinase 2/antagonists & inhibitors , Ligands , Microscopy, Fluorescence , Models, Molecular , Mutation , Nitriles , Phenylalanine/genetics , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction , Single Molecule Imaging , Valine/genetics
16.
Elife ; 82019 11 27.
Article in English | MEDLINE | ID: mdl-31774398

ABSTRACT

Cytokines activate signaling via assembly of cell surface receptors, but it is unclear whether modulation of cytokine-receptor binding parameters can modify biological outcomes. We have engineered IL-6 variants with different affinities to gp130 to investigate how cytokine receptor binding dwell-times influence functional selectivity. Engineered IL-6 variants showed a range of signaling amplitudes and induced biased signaling, with changes in receptor binding dwell-times affecting more profoundly STAT1 than STAT3 phosphorylation. We show that this differential signaling arises from defective translocation of ligand-gp130 complexes to the endosomal compartment and competitive STAT1/STAT3 binding to phospho-tyrosines in gp130, and results in unique patterns of STAT3 binding to chromatin. This leads to a graded gene expression response and differences in ex vivo differentiation of Th17, Th1 and Treg cells. These results provide a molecular understanding of signaling biased by cytokine receptors, and demonstrate that manipulation of signaling thresholds is a useful strategy to decouple cytokine functional pleiotropy.


Subject(s)
Cytokine Receptor gp130/chemistry , Interleukin-6/chemistry , STAT1 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Th17 Cells/metabolism , Binding Sites , Cell Differentiation , Cloning, Molecular , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Endosomes/chemistry , Endosomes/metabolism , Gene Expression , HeLa Cells , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Kinetics , Models, Molecular , Phosphorylation , Primary Cell Culture , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Engineering/methods , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology
17.
Front Immunol ; 9: 2143, 2018.
Article in English | MEDLINE | ID: mdl-30319612

ABSTRACT

Cytokines comprise a large family of secreted ligands that are critical for the regulation of immune homeostasis. Cytokines initiate signaling via dimerization or oligomerization of the cognate receptor subunits, triggering the activation of the Janus Kinases (JAKs)/ signal transducer and activator of transcription (STATs) pathway and the induction of specific gene expression programs and bioactivities. Deregulation of cytokines or their downstream signaling pathways are at the root of many human disorders including autoimmunity and cancer. Identifying and understanding the mechanistic principles that govern cytokine signaling will, therefore, be highly important in order to harness the therapeutic potential of cytokines. In this review, we will analyze how biophysical (ligand-receptor binding geometry and affinity) and cellular (receptor trafficking and intracellular abundance of signaling molecules) parameters shape the cytokine signalosome and cytokine functional pleiotropy; from the initial cytokine binding to its receptor to the degradation of the cytokine receptor complex in the proteasome and/or lysosome. We will also discuss how combining advanced protein engineering with detailed signaling and functional studies has opened promising avenues to tackle complex questions in the cytokine signaling field.


Subject(s)
Cytokines/genetics , Homeostasis/immunology , Protein Engineering , Receptors, Cytokine/metabolism , Signal Transduction/immunology , Cytokines/immunology , Cytokines/metabolism , Humans , Janus Kinases/metabolism , Receptors, Cytokine/agonists , Receptors, Cytokine/immunology , STAT Transcription Factors/metabolism
18.
Nat Struct Mol Biol ; 24(3): 279-289, 2017 03.
Article in English | MEDLINE | ID: mdl-28165510

ABSTRACT

Type I interferons (IFNs) are multifunctional cytokines that regulate immune responses and cellular functions but also can have detrimental effects on human health. A tight regulatory network therefore controls IFN signaling, which in turn may interfere with medical interventions. The JAK-STAT signaling pathway transmits the IFN extracellular signal to the nucleus, thus resulting in alterations in gene expression. STAT2 is a well-known essential and specific positive effector of type I IFN signaling. Here, we report that STAT2 is also a previously unrecognized, crucial component of the USP18-mediated negative-feedback control in both human and mouse cells. We found that STAT2 recruits USP18 to the type I IFN receptor subunit IFNAR2 via its constitutive membrane-distal STAT2-binding site. This mechanistic coupling of effector and negative-feedback functions of STAT2 may provide novel strategies for treatment of IFN-signaling-related human diseases.


Subject(s)
Endopeptidases/metabolism , Interferon Type I/metabolism , STAT2 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Feedback, Physiological , Humans , Immunoblotting , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Domains , Receptor, Interferon alpha-beta/metabolism , STAT2 Transcription Factor/chemistry , Two-Hybrid System Techniques , Ubiquitin Thiolesterase
19.
Nat Commun ; 8: 15976, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28706306

ABSTRACT

The spatiotemporal organization of cytokine receptors in the plasma membrane is still debated with models ranging from ligand-independent receptor pre-dimerization to ligand-induced receptor dimerization occurring only after receptor uptake into endosomes. Here, we explore the molecular and cellular determinants governing the assembly of the type II interleukin-4 receptor, taking advantage of various agonists binding the receptor subunits with different affinities and rate constants. Quantitative kinetic studies using artificial membranes confirm that receptor dimerization is governed by the two-dimensional ligand-receptor interactions and identify a critical role of the transmembrane domain in receptor dimerization. Single molecule localization microscopy at physiological cell surface expression levels, however, reveals efficient ligand-induced receptor dimerization by all ligands, largely independent of receptor binding affinities, in line with the similar STAT6 activation potencies observed for all IL-4 variants. Detailed spatiotemporal analyses suggest that kinetic trapping of receptor dimers in actin-dependent microcompartments sustains robust receptor dimerization and signalling.


Subject(s)
Cell Membrane/metabolism , Receptors, Interleukin-4, Type II/metabolism , Actin Cytoskeleton , Cell Compartmentation , Dimerization , HeLa Cells , Humans , Ligands , Receptors, Interleukin-4, Type II/agonists , STAT6 Transcription Factor/metabolism
20.
Sci Adv ; 2(12): e1600452, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27957535

ABSTRACT

The interaction dynamics of signaling complexes is emerging as a key determinant that regulates the specificity of cellular responses. We present a combined experimental and computational study that quantifies the consequences of plasma membrane microcompartmentalization for the dynamics of type I interferon receptor complexes. By using long-term dual-color quantum dot (QD) tracking, we found that the lifetime of individual ligand-induced receptor heterodimers depends on the integrity of the membrane skeleton (MSK), which also proved important for efficient downstream signaling. By pair correlation tracking and localization microscopy as well as by fast QD tracking, we identified a secondary confinement within ~300-nm-sized zones. A quantitative spatial stochastic diffusion-reaction model, entirely parameterized on the basis of experimental data, predicts that transient receptor confinement by the MSK meshwork allows for rapid reassociation of dissociated receptor dimers. Moreover, the experimentally observed apparent stabilization of receptor dimers in the plasma membrane was reproduced by simulations of a refined, hierarchical compartment model. Our simulations further revealed that the two-dimensional association rate constant is a key parameter for controlling the extent of MSK-mediated stabilization of protein complexes, thus ensuring the specificity of this effect. Together, experimental evidence and simulations support the hypothesis that passive receptor confinement by MSK-based microcompartmentalization promotes maintenance of signaling complexes in the plasma membrane.


Subject(s)
Cell Membrane/physiology , Cytokines/metabolism , Protein Multimerization , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Cell Line , Diffusion , Humans , Microscopy, Fluorescence , Quantum Dots
SELECTION OF CITATIONS
SEARCH DETAIL