Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
bioRxiv ; 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37398498

ABSTRACT

Axon pathfinding is controlled by attractive and repulsive molecular cues that activate receptors on the axonal growth cone, but the full repertoire of axon guidance molecules remains unknown. The vertebrate DCC receptor family contains the two closely related members DCC and Neogenin with prominent roles in axon guidance and three additional, divergent members - Punc, Nope, and Protogenin - for which functions in neural circuit formation have remained elusive. We identified a secreted Punc/Nope/Protogenin ligand, WFIKKN2, which guides mouse peripheral sensory axons through Nope-mediated repulsion. In contrast, WFIKKN2 attracts motor axons, but not via Nope. These findings identify WFIKKN2 as a bifunctional axon guidance cue that acts through divergent DCC family members, revealing a remarkable diversity of ligand interactions for this receptor family in nervous system wiring. One-Sentence Summary: WFIKKN2 is a ligand for the DCC family receptors Punc, Nope, and Prtg that repels sensory axons and attracts motor axons.

2.
PLoS One ; 16(12): e0260800, 2021.
Article in English | MEDLINE | ID: mdl-34879110

ABSTRACT

The cancer-associated fibroblast (CAF) marker podoplanin (PDPN) is generally correlated with poor clinical outcomes in cancer patients and thus represents a promising therapeutic target. Despite its biomedical relevance, basic aspects of PDPN biology such as its cellular functions and cell surface ligands remain poorly uncharacterized, thus challenging drug development. Here, we utilize a high throughput platform to elucidate the PDPN cell surface interactome, and uncover the neutrophil protein CD177 as a new binding partner. Quantitative proteomics analysis of the CAF phosphoproteome reveals a role for PDPN in cell signaling, growth and actomyosin contractility, among other processes. Moreover, cellular assays demonstrate that CD177 is a functional antagonist, recapitulating the phenotype observed in PDPN-deficient CAFs. In sum, starting from the unbiased elucidation of the PDPN co-receptome, our work provides insights into PDPN functions and reveals the PDPN/CD177 axis as a possible modulator of fibroblast physiology in the tumor microenvironment.


Subject(s)
Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Isoantigens/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Tumor Microenvironment , Apoptosis , Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Isoantigens/genetics , Membrane Glycoproteins/genetics , Neutrophils/immunology , Neutrophils/metabolism , Prognosis , Receptors, Cell Surface/genetics , Survival Rate , Tumor Cells, Cultured
3.
Cell ; 182(2): 329-344.e19, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32589946

ABSTRACT

Cell surface receptors and their interactions play a central role in physiological and pathological signaling. Despite its clinical relevance, the immunoglobulin superfamily (IgSF) remains uncharacterized and underrepresented in databases. Here, we present a systematic extracellular protein map, the IgSF interactome. Using a high-throughput technology to interrogate most single transmembrane receptors for binding to 445 IgSF proteins, we identify over 500 interactions, 82% previously undocumented, and confirm more than 60 receptor-ligand pairs using orthogonal assays. Our study reveals a map of cell-type-specific interactions and the landscape of dysregulated receptor-ligand crosstalk in cancer, including selective loss of function for tumor-associated mutations. Furthermore, investigation of the IgSF interactome in a large cohort of cancer patients identifies interacting protein signatures associated with clinical outcome. The IgSF interactome represents an important resource to fuel biological discoveries and a framework for understanding the functional organization of the surfaceome during homeostasis and disease, ultimately informing therapeutic development.


Subject(s)
Immunoglobulins/metabolism , Neoplasms/pathology , Protein Interaction Maps , B7-H1 Antigen/metabolism , Carcinoembryonic Antigen/metabolism , Cell Communication , Cluster Analysis , Culture Media, Conditioned/chemistry , HEK293 Cells , Humans , Immunoglobulins/chemistry , Immunoglobulins/genetics , Ligands , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Mol Cell Proteomics ; 18(11): 2310-2323, 2019 11.
Article in English | MEDLINE | ID: mdl-31308249

ABSTRACT

Receptors expressed on the plasma membrane and their interacting partners critically regulate cellular communication during homeostasis and disease, and as such represent main therapeutic targets. Despite its importance for drug development, receptor-ligand proteomics has remained a daunting field, in part because of the challenges associated to the study of membrane-expressed proteins. Here, to enable sensitive detection of receptor-ligand interactions in high throughput, we implement a new platform, the Conditioned Media AlphaScreen, for interrogation of a library consisting of most single transmembrane human proteins. Using this method to study key immune receptors, we identify and further validate the interleukin receptor IL20RA as the first binding partner for the checkpoint inhibitor B7-H3. Further, KIR2DL5, a natural killer cell protein that had remained orphan, is uncovered as a functional binding partner for the poliovirus receptor (PVR). This interaction is characterized using orthogonal assays, which demonstrate that PVR specifically engages KIR2DL5 on natural killer cells leading to inhibition of cytotoxicity. Altogether, these results reveal unappreciated links between protein families that may importantly influence receptor-driven functions during disease. Applicable to any target of interest, this technology represents a versatile and powerful approach for elucidation of receptor-ligand interactomes, which is essential to understand basic aspects of the biology of the plasma membrane proteins and ultimately inform the development of novel therapeutic strategies.


Subject(s)
B7 Antigens/metabolism , Extracellular Matrix/metabolism , Killer Cells, Natural/metabolism , Receptors, Interleukin/metabolism , Receptors, KIR2DL5/metabolism , Receptors, Virus/metabolism , Cell Communication , HEK293 Cells , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Ligands , Protein Binding , Protein Interaction Maps
6.
Neuron ; 91(2): 328-40, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27477018

ABSTRACT

Genetic variants of TREM2, a protein expressed selectively by microglia in the brain, are associated with Alzheimer's disease (AD). Starting from an unbiased protein microarray screen, we identified a set of lipoprotein particles (including LDL) and apolipoproteins (including CLU/APOJ and APOE) as ligands of TREM2. Binding of these ligands by TREM2 was abolished or reduced by disease-associated mutations. Overexpression of wild-type TREM2 was sufficient to enhance uptake of LDL, CLU, and APOE in heterologous cells, whereas TREM2 disease variants were impaired in this activity. Trem2 knockout microglia showed reduced internalization of LDL and CLU. ß-amyloid (Aß) binds to lipoproteins and this complex is efficiently taken up by microglia in a TREM2-dependent fashion. Uptake of Aß-lipoprotein complexes was reduced in macrophages from human subjects carrying a TREM2 AD variant. These data link three genetic risk factors for AD and reveal a possible mechanism by which mutant TREM2 increases risk of AD. VIDEO ABSTRACT.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoproteins/metabolism , Brain/metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Receptors, Immunologic/metabolism , Cell Line , Humans , Membrane Glycoproteins/genetics , Protein Binding , Receptors, Immunologic/genetics
7.
Nat Commun ; 7: 12220, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27447090

ABSTRACT

Substantial efforts have been made to understand the interactions between immune checkpoint receptors and their ligands targeted in immunotherapies against cancer. To carefully characterize the complete network of interactions involved and the binding affinities between their extracellular domains, an improved kinetic assay is needed to overcome limitations with surface plasmon resonance (SPR). Here, we present a magneto-nanosensor platform integrated with a microfluidic chip that allows measurement of dissociation constants in the micromolar-range. High-density conjugation of magnetic nanoparticles with prey proteins allows multivalent receptor interactions with sensor-immobilized bait proteins, more closely mimicking natural-receptor clustering on cells. The platform has advantages over traditional SPR in terms of insensitivity of signal responses to pH and salinity, less consumption of proteins and better sensitivities. Using this platform, we characterized the binding affinities of the PD-1-PD-L1/PD-L2 co-inhibitory receptor system, and discovered an unexpected interaction between the two known PD-1 ligands, PD-L1 and PD-L2.


Subject(s)
B7-H1 Antigen/metabolism , Magnetics/methods , Nanoparticles/chemistry , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Protein Interaction Mapping , Computer Systems , Humans , Hydrogen-Ion Concentration , Kinetics , Protein Binding , Salinity
8.
Nat Commun ; 7: 11473, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27145901

ABSTRACT

Viruses encode secreted and cell-surface expressed proteins essential to modulate host immune defenses and establish productive infections. However, to date there has been no systematic study of the extracellular interactome of any human virus. Here we utilize the E3 proteins, diverse and rapidly evolving transmembrane-containing proteins encoded by human adenoviruses, as a model system to survey the extracellular immunomodulatory landscape. From a large-scale protein interaction screen against a microarray of more than 1,500 human proteins, we find and validate 51 previously unidentified virus-host interactions. Our results uncover conserved strategies as well as substantial diversity and multifunctionality in host targeting within and between viral species. Prominent modulation of the leukocyte immunoglobulin-like and signalling lymphocyte activation molecule families and a number of inhibitory receptors were identified as hubs for viral perturbation, suggesting unrecognized immunoregulatory strategies. We describe a virus-host extracellular interaction map of unprecedented scale that provides new insights into viral immunomodulation.


Subject(s)
Adenoviruses, Human/immunology , Immunomodulation/immunology , Protein Interaction Maps/immunology , Viral Proteins/immunology , A549 Cells , Adenoviruses, Human/metabolism , Adenoviruses, Human/physiology , Animals , CHO Cells , Cell Line, Tumor , Cells, Cultured , Cricetulus , Extracellular Space/immunology , Extracellular Space/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Jurkat Cells , K562 Cells , Protein Binding , Proteome/immunology , Proteome/metabolism , Viral Proteins/metabolism
9.
Science ; 350(6263): 961-5, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26586761

ABSTRACT

Axon pathfinding is orchestrated by numerous guidance cues, including Slits and their Robo receptors, but it remains unclear how information from multiple cues is integrated or filtered. Robo3, a Robo family member, allows commissural axons to reach and cross the spinal cord midline by antagonizing Robo1/2-mediated repulsion from midline-expressed Slits and potentiating deleted in colorectal cancer (DCC)-mediated midline attraction to Netrin-1, but without binding either Slits or Netrins. We identified a secreted Robo3 ligand, neural epidermal growth factor-like-like 2 (NELL2), which repels mouse commissural axons through Robo3 and helps steer them to the midline. These findings identify NELL2 as an axon guidance cue and establish Robo3 as a multifunctional regulator of pathfinding that simultaneously mediates NELL2 repulsion, inhibits Slit repulsion, and facilitates Netrin attraction to achieve a common guidance purpose.


Subject(s)
Axons/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Spinal Cord/embryology , Animals , Axons/metabolism , Ligands , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Netrin-1 , Neurogenesis/genetics , Receptors, Cell Surface , Receptors, Immunologic/metabolism , Tumor Suppressor Proteins/metabolism , Roundabout Proteins
10.
Anal Biochem ; 479: 1-5, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25797350

ABSTRACT

When used in conjunction with multivalent protein probes, protein microarrays offer a robust technology for discovery of low-affinity extracellular protein-protein interactions. Probes for receptor-matching screens generally consist of purified extracellular domains fused to affinity tags. Given that approximately two-thirds of extracellular proteins are transmembrane domain-containing proteins, it would be desirable to develop a system to express and display probe receptors in a native-like membrane environment. Toward this end, we evaluated baculovirus display as a platform for generating multivalent probes for protein microarray screens. Virion particles were generated displaying single-transmembrane domain receptors BTLA, CD200, and EFNB2, representing a range of affinities for their interacting partners. Virions directly labeled with Cy5 fluorophore were screened against a microarray containing more than 600 extracellular proteins, and the results were compared with data derived from soluble Fc protein or probe-coated protein A microbeads. An optimized protocol employing a blocking step with a nonrelated probe-expressing control baculovirus allowed identification of the expected interactions with a signal-to-noise ratio similar to or higher than those obtained with the other formats. Our results demonstrate that baculovirus display is suitable for detection of high- and low-affinity extracellular protein-protein interactions on protein microarrays. This platform eliminates the need for protein purification and provides a native-like lipid environment for membrane-associated receptors.


Subject(s)
Antigens, CD/metabolism , Baculoviridae/genetics , Ephrin-B2/metabolism , Protein Array Analysis/methods , Receptors, Immunologic/metabolism , Animals , Antigens, CD/genetics , Baculoviridae/metabolism , Cell Line , Ephrin-B2/genetics , Humans , Ligands , Protein Binding , Receptors, Immunologic/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic , Signal-To-Noise Ratio , Virion/genetics , Virion/metabolism
11.
PLoS One ; 9(1): e84823, 2014.
Article in English | MEDLINE | ID: mdl-24400119

ABSTRACT

The growth and guidance of many axons in the developing nervous system require Netrin-mediated activation of Deleted in Colorectal Cancer (DCC) and other still unknown signaling cues. Commissural axon guidance defects are more severe in DCC mutant mice than Netrin-1 mutant mice, suggesting additional DCC activating signals besides Netrin-1 are involved in proper axon growth. Here we report that interaction screens on extracellular protein microarrays representing over 1,000 proteins uniquely identified Cerebellin 4 (CBLN4), a member of the C1q-tumor necrosis factor (TNF) family, and Netrin-1 as extracellular DCC-binding partners. Immunofluorescence and radio-ligand binding studies demonstrate that Netrin-1 competes with CBLN4 binding at an overlapping site within the membrane-proximal fibronectin domains (FN) 4-6 of DCC and binds with ∼5-fold higher affinity. CBLN4 also binds to the DCC homolog, Neogenin-1 (NEO1), but with a lower affinity compared to DCC. CBLN4-null mice did not show a defect in commissural axons of the developing spinal cord but did display a transient increase in the number of wandering axons in the brachial plexus, consistent with a role in axon guidance. Overall, the data solidifies CBLN4 as a bona fide DCC ligand and strengthens its implication in axon guidance.


Subject(s)
Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Axons/metabolism , Carrier Proteins , DCC Receptor , Embryonic Development/genetics , Humans , Kinetics , Ligands , Mice , Mutation , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Netrin-1 , Neurogenesis/genetics , Neurons/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Precursors/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
12.
Science ; 341(6151): 1246-9, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23887873

ABSTRACT

Gram-negative bacteria including Escherichia coli, Citrobacter rodentium, Salmonella typhimurium, and Shigella flexneri are sensed in an ill-defined manner by an intracellular inflammasome complex that activates caspase-11. We show that macrophages loaded with synthetic lipid A, E. coli lipopolysaccharide (LPS), or S. typhimurium LPS activate caspase-11 independently of the LPS receptor Toll-like receptor 4 (TLR4). Consistent with lipid A triggering the noncanonical inflammasome, LPS containing a divergent lipid A structure antagonized caspase-11 activation in response to E. coli LPS or Gram-negative bacteria. Moreover, LPS-mutant E. coli failed to activate caspase-11. Tlr4(-/-) mice primed with TLR3 agonist polyinosinic:polycytidylic acid [poly(I:C)] to induce pro-caspase-11 expression were as susceptible as wild-type mice were to sepsis induced by E. coli LPS. These data unveil a TLR4-independent mechanism for innate immune recognition of LPS.


Subject(s)
Immunity, Innate , Inflammasomes/immunology , Lipid A/immunology , Macrophages/immunology , Toll-Like Receptor 4/immunology , Animals , Caspases/biosynthesis , Caspases, Initiator , Cholera Toxin/immunology , Disease Models, Animal , Escherichia coli/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Lipid A/genetics , Mice , Mice, Mutant Strains , Mutation , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Sepsis/immunology
13.
Curr Protoc Protein Sci ; Chapter 27: Unit 27.3, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23546621

ABSTRACT

Functional protein microarrays offer the capability for high-throughput protein interaction analysis and have long promised to be a powerful tool for understanding protein interactions at the proteome scale. Although popular techniques for protein-protein interaction mapping like yeast-two-hybrid and affinity-purification mass spectrometry have performed well for identifying intracellular protein-protein interactions, the study of interactions between extracellular proteins has remained challenging for these methods. Instead, the use of protein microarrays appears to be a robust and efficient method for the identification of interactions among the members of this class of protein. This unit describes methods for extracellular protein microarray production, screening, and analysis. A protocol is described for enhanced detection of low-affinity interactions by generating multivalent complexes using Fc-fusion bait proteins and protein A microbeads, along with a statistical method for hit scoring and identification of nonspecific interactions.


Subject(s)
Protein Array Analysis/instrumentation , Protein Array Analysis/methods , Protein Interaction Mapping/instrumentation , Protein Interaction Mapping/methods , Proteins/analysis , Animals , Extracellular Space/chemistry , Extracellular Space/metabolism , Humans , Protein Binding , Proteins/metabolism
14.
Biochem J ; 452(2): 313-20, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23496764

ABSTRACT

The mutant forms of KRas, NRas and HRas drive the initiation and progression of a number of human cancers, but less is known about the role of WT (wild-type) Ras alleles and isoforms in cancer. We used zinc-finger nucleases targeting HRas and NRas to modify both alleles of these genes in the mutant KRas-driven Hec1A endometrial cancer cell line, which normally expresses WT copies of these genes. The disruption of either WT isoform of Ras compromised growth-factor-dependent signalling through the ERK (extracellular-signal-regulated kinase) pathway. In addition, the disruption of HRas hindered the activation of Akt and subsequent downstream signalling. This was associated with decreased proliferation, increased apoptosis and decreased anchorage-independent growth in the HRas-disrupted cells. However, xenograft tumour growth was not significantly affected by the disruption of either NRas or HRas. As expected, deleting the mutant allele of KRas abolished tumour growth, whereas deletion of the remaining WT copy of KRas increased the tumorigenic properties of these cells; deleting a single copy of either HRas or NRas did not mimic this effect. The present study demonstrates that the WT copies of HRas, NRas and KRas play unique roles in the context of mutant KRas-driven tumours.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , ras Proteins/chemistry , ras Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Cell Transformation, Neoplastic/chemistry , Female , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics
15.
Autoimmunity ; 46(2): 157-67, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23167594

ABSTRACT

Multiple Sclerosis (MS) is a neurodegenerative autoimmune disorder caused by chronic inflammation and demyelination within the central nervous system (CNS). Clinical studies in MS patients have demonstrated efficacy with B cell targeted therapies such as anti-CD20. However, the exact role that B cells play in the disease process is unclear. Activation Induced cytidine deaminase (AID) is an essential enzyme for the processes of antibody affinity maturation and isotype switching. To evaluate the impact of affinity maturation and isotype switching, we have interrogated the effect of AID-deficiency in an animal model of MS. Here, we show that the severity of experimental autoimmune encephalomyelitis (EAE) induced by the extracellular domain of human myelin oligodendrocyte glycoprotein (MOG1-125) is significantly reduced in Aicda deficient mice, which, unlike wild-type mice, lack serum IgG to myelin associated antigens. MOG specific T cell responses are comparable between wild-type and Aicda knockout mice suggesting an active role for antigen experienced B cells. Thus affinity maturation and/or class switching are critical processes in the pathogenesis of EAE.


Subject(s)
Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Animals , Antibody Affinity/immunology , Autoantibodies/immunology , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Gene Order , Gene Targeting , Genetic Predisposition to Disease , Humans , Immunoglobulin G/immunology , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/metabolism , T-Lymphocytes/immunology
16.
PLoS One ; 8(12): e83958, 2013.
Article in English | MEDLINE | ID: mdl-24409221

ABSTRACT

BACKGROUND: Macrophage stimulating protein (MSP) is a serum growth factor that binds to and activates the receptor tyrosine kinase, Recepteur d'Origine Nantais (RON). A non-synonymous coding variant in MSP (689C) has been associated with genetic susceptibility to both Crohn's disease and ulcerative colitis, two major types of inflammatory bowel disease (IBD) characterized by chronic inflammation of the digestive tract. We investigated the consequences of this polymorphism for MSP-RON pathway activity and IBD pathogenesis. METHODS: RON expression patterns were examined on mouse and human cells and tissues under normal and disease conditions to identify cell types regulated by MSP-RON. Recombinant MSP variants were tested for their ability to bind and stimulate RON and undergo proteolytic activation. MSP concentrations were quantified in the serum of individuals carrying the MSP 689R and 689C alleles. RESULTS: In intestinal tissue, RON was primarily expressed by epithelial cells under normal and disease conditions. The 689C polymorphism had no impact on the ability of MSP to bind to or signal through RON. In a cohort of normal individuals and IBD patients, carriers of the 689C polymorphism had lower concentrations of MSP in their serum. CONCLUSIONS: By reducing the quantities of circulating MSP, the 689C polymorphism, or a variant in linkage disequilibrium with this polymorphism, may impact RON ligand availability and thus receptor activity. Given the known functions of RON in regulating wound healing and our analysis of RON expression patterns in human intestinal tissue, these data suggest that decreased RON activity may impact the efficiency of epithelial repair and thus underlie the increased IBD susceptibility associated with the MSP 689C allele.


Subject(s)
Alleles , Genetic Predisposition to Disease , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Polymorphism, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Hepatocyte Growth Factor/blood , Humans , Inflammatory Bowel Diseases/pathology , Intestines/pathology , Mice , Models, Molecular , Protein Conformation , Proteolysis , Proto-Oncogene Proteins/blood , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
17.
Methods ; 57(4): 448-58, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22728035

ABSTRACT

Approximately one quarter of all human genes encode proteins that function in the extracellular space or serve to bridge the extracellular and intracellular environments. Physical associations between these secretome proteins serve to regulate a wide range of biological activities and consequently represent important therapeutic targets. Moreover, some extracellular proteins are targeted by pathogens to allow host access or immune evasion. Despite the importance of extracellular protein-protein interactions, our knowledge in this area has remained sparse. Weak affinities and low abundance have often hindered efforts to identify these interactions using traditional methods such as biochemical purification and cDNA library expression cloning. Moreover, current large-scale protein-protein interaction mapping techniques largely under represent extracellular protein-protein interactions. This review highlights emerging biosensor and protein microarray technology, along with more traditional cell-based techniques, that are compatible with secretome-wide screens for extracellular protein-protein interaction discovery. A combination of these approaches will serve to rapidly expand our knowledge of the extracellular protein-protein interactome.


Subject(s)
Protein Array Analysis , Protein Interaction Mapping/methods , Proteome/metabolism , Surface Plasmon Resonance , Animals , Cloning, Molecular , Flow Cytometry , Humans , Interferometry , Peptide Library , Protein Binding
18.
J Biol Chem ; 287(19): 15837-50, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22396535

ABSTRACT

Paired immunoglobulin-like receptor (PILR) α is an inhibitory receptor that recognizes several ligands, including mouse CD99, PILR-associating neural protein, and Herpes simplex virus-1 glycoprotein B. The physiological function(s) of interactions between PILRα and its cellular ligands are not well understood, as are the molecular determinants of PILRα/ligand interactions. To address these uncertainties, we sought to identify additional PILRα ligands and further define the molecular basis for PILRα/ligand interactions. Here, we identify two novel PILRα binding partners, neuronal differentiation and proliferation factor-1 (NPDC1), and collectin-12 (COLEC12). We find that sialylated O-glycans on these novel PILRα ligands, and on known PILRα ligands, are compulsory for PILRα binding. Sialylation-dependent ligand recognition is also a property of SIGLEC1, a member of the sialic acid-binding Ig-like lectins. SIGLEC1 Ig domain shares ∼22% sequence identity with PILRα, an identity that includes a conserved arginine localized to position 97 in mouse and human SIGLEC1, position 133 in mouse PILRα and position 126 in human PILRα. We observe that PILRα/ligand interactions require conserved PILRα Arg-133 (mouse) and Arg-126 (human), in correspondence with a previously reported requirement for SIGLEC1 Arg-197 in SIGLEC1/ligand interactions. Homology modeling identifies striking similarities between PILRα and SIGLEC1 ligand binding pockets as well as at least one set of distinctive interactions in the galactoxyl-binding site. Binding studies suggest that PILRα recognizes a complex ligand domain involving both sialic acid and protein motif(s). Thus, PILRα is evolved to engage multiple ligands with common molecular determinants to modulate myeloid cell functions in anatomical settings where PILRα ligands are expressed.


Subject(s)
Evolution, Molecular , Membrane Glycoproteins/metabolism , N-Acetylneuraminic Acid/metabolism , Receptors, Immunologic/metabolism , 12E7 Antigen , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/metabolism , Arginine/chemistry , Arginine/genetics , Arginine/metabolism , Binding Sites/genetics , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chlorocebus aethiops , Collectins/chemistry , Collectins/genetics , Collectins/metabolism , Conserved Sequence/genetics , HEK293 Cells , Humans , Ligands , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , N-Acetylneuraminic Acid/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Sequence Homology, Amino Acid , Sialic Acid Binding Ig-like Lectin 1 , Vero Cells
19.
Anal Biochem ; 420(2): 127-38, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21982860

ABSTRACT

Characterization of the extracellular protein interactome has lagged far behind that of intracellular proteins, where mass spectrometry and yeast two-hybrid technologies have excelled. Improved methods for identifying receptor-ligand and extracellular matrix protein interactions will greatly accelerate biological discovery in cell signaling and cellular communication. These technologies must be able to identify low-affinity binding events that are often observed between membrane-bound coreceptor molecules during cell-cell or cell-extracellular matrix contact. Here we demonstrate that functional protein microarrays are particularly well-suited for high-throughput screening of extracellular protein interactions. To evaluate the performance of the platform, we screened a set of 89 immunoglobulin (Ig)-type receptors against a highly diverse extracellular protein microarray with 686 genes represented. To enhance detection of low-affinity interactions, we developed a rapid method to assemble bait Fc fusion proteins into multivalent complexes using protein A microbeads. Based on these screens, we developed a statistical methodology for hit calling and identification of nonspecific interactions on protein microarrays. We found that the Ig receptor interactions identified using our methodology are highly specific and display minimal off-target binding, resulting in a 70% true-positive to false-positive hit ratio. We anticipate that these methods will be useful for a wide variety of functional protein microarray users.


Subject(s)
Extracellular Space/metabolism , Protein Array Analysis/methods , Protein Interaction Mapping/methods , Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , False Positive Reactions , Gene Library , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Microspheres , Proteins/chemistry , Receptors, Immunologic/metabolism , Reproducibility of Results , Substrate Specificity
20.
J Biol Chem ; 286(21): 18969-81, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21454693

ABSTRACT

Psoriasis is a human skin condition characterized by epidermal hyperproliferation and infiltration of multiple leukocyte populations. In characterizing a novel insulin growth factor (IGF)-like (IGFL) gene in mice (mIGFL), we found transcripts of this gene to be most highly expressed in skin with enhanced expression in models of skin wounding and psoriatic-like inflammation. A possible functional ortholog in humans, IGFL1, was uniquely and significantly induced in psoriatic skin samples. In vitro IGFL1 expression was up-regulated in cultured primary keratinocytes stimulated with tumor necrosis factor α but not by other psoriasis-associated cytokines. Finally, using a secreted and transmembrane protein library, we discovered high affinity interactions between human IGFL1 and mIGFL and the TMEM149 ectodomain. TMEM149 (renamed here as IGFLR1) is an uncharacterized gene with structural similarity to the tumor necrosis factor receptor family. Our studies demonstrate that IGFLR1 is expressed primarily on the surface of mouse T cells. The connection between mIGFL and IGFLR1 receptor suggests mIGFL may influence T cell biology within inflammatory skin conditions.


Subject(s)
Insulin-Like Growth Factor I/biosynthesis , Psoriasis/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Skin/metabolism , Up-Regulation , Wounds and Injuries/metabolism , Animals , Disease Models, Animal , Humans , Insulin-Like Growth Factor I/genetics , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary , Psoriasis/genetics , Psoriasis/pathology , Receptors, Tumor Necrosis Factor/genetics , Skin/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Wounds and Injuries/genetics , Wounds and Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...