Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cancer Cell ; 31(3): 383-395, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28262555

ABSTRACT

The anti-FcRH5/CD3 T cell-dependent bispecific antibody (TDB) targets the B cell lineage marker FcRH5 expressed in multiple myeloma (MM) tumor cells. We demonstrate that TDBs trigger T cell receptor activation by inducing target clustering and exclusion of CD45 phosphatase from the synapse. The dimensions of the target molecule play a key role in the efficiency of the synapse formation. The anti-FcRH5/CD3 TDB kills human plasma cells and patient-derived myeloma cells at picomolar concentrations and results in complete depletion of B cells and bone marrow plasma cells in cynomolgus monkeys. These data demonstrate the potential for the anti-FcRH5/CD3 TDB, alone or in combination with inhibition of PD-1/PD-L1 signaling, in the treatment of MM and other B cell malignancies.


Subject(s)
Antibodies, Bispecific/therapeutic use , CD3 Complex/immunology , Epitopes , Immunological Synapses/physiology , Multiple Myeloma/drug therapy , Receptors, Fc/immunology , T-Lymphocytes/immunology , Animals , Cytokines/metabolism , Humans , Leukocyte Common Antigens/physiology , Lymphocyte Activation , Macaca fascicularis , Mice , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/physiology , Receptors, Antigen, T-Cell/physiology , Receptors, Fc/analysis
2.
JCI Insight ; 1(7): e86689, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27699264

ABSTRACT

Eosinophilic inflammation and Th2 cytokine production are central to the pathogenesis of asthma. Agents that target either eosinophils or single Th2 cytokines have shown benefits in subsets of biomarker-positive patients. More broadly effective treatment or disease-modifying effects may be achieved by eliminating more than one inflammatory stimulator. Here we present a strategy to concomitantly deplete Th2 T cells, eosinophils, basophils, and type-2 innate lymphoid cells (ILC2s) by generating monoclonal antibodies with enhanced effector function (19A2) that target CRTh2 present on all 4 cell types. Using human CRTh2 (hCRTh2) transgenic mice that mimic the expression pattern of hCRTh2 on innate immune cells but not Th2 cells, we demonstrate that anti-hCRTh2 antibodies specifically eliminate hCRTh2+ basophils, eosinophils, and ILC2s from lung and lymphoid organs in models of asthma and Nippostrongylus brasiliensis infection. Innate cell depletion was accompanied by a decrease of several Th2 cytokines and chemokines. hCRTh2-specific antibodies were also active on human Th2 cells in vivo in a human Th2-PBMC-SCID mouse model. We developed humanized hCRTh2-specific antibodies that potently induce antibody-dependent cell cytotoxicity (ADCC) of primary human eosinophils and basophils and replicated the in vivo depletion capacity of their murine parent. Therefore, depletion of hCRTh2+ basophils, eosinophils, ILC2, and Th2 cells with h19A2 hCRTh2-specific antibodies may be a novel and more efficacious treatment for asthma.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Asthma/therapy , Th2 Cells/cytology , Animals , Antibodies, Monoclonal, Humanized/immunology , Basophils/cytology , Cytokines , Disease Models, Animal , Eosinophils/cytology , Humans , Immunity, Innate , Lung/cytology , Lung/immunology , Lymphocytes/cytology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, SCID , Mice, Transgenic
3.
PLoS Pathog ; 12(6): e1005702, 2016 06.
Article in English | MEDLINE | ID: mdl-27351973

ABSTRACT

Broadly neutralizing antibodies targeting the stalk region of influenza A virus (IAV) hemagglutinin (HA) are effective in blocking virus infection both in vitro and in vivo. The highly conserved epitopes recognized by these antibodies are critical for the membrane fusion function of HA and therefore less likely to be permissive for virus mutational escape. Here we report three resistant viruses of the A/Perth/16/2009 strain that were selected in the presence of a broadly neutralizing stalk-binding antibody. The three resistant viruses harbor three different mutations in the HA stalk: (1) Gln387Lys; (2) Asp391Tyr; (3) Asp391Gly. The Gln387Lys mutation completely abolishes binding of the antibody to the HA stalk epitope. The other two mutations, Asp391Tyr and Asp391Gly, do not affect antibody binding at neutral pH and only slightly reduce binding at low pH. Interestingly, they enhance the fusion ability of the HA, representing a novel mechanism that allows productive membrane fusion even in the presence of antibody and hence virus escape from antibody neutralization. Therefore, these mutations illustrate two different resistance mechanisms used by IAV to escape broadly neutralizing stalk-binding antibodies. Compared to the wild type virus, the resistant viruses release fewer progeny viral particles during replication and are more sensitive to Tamiflu, suggesting reduced viral fitness.


Subject(s)
Antibodies, Neutralizing/immunology , Drug Resistance, Microbial/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immune Evasion/immunology , Influenza A virus/immunology , Animals , Antibodies, Viral/immunology , Blotting, Western , Dogs , Flow Cytometry , Humans , Immunohistochemistry , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Mice , Neutralization Tests , Orthomyxoviridae Infections/immunology , Polymerase Chain Reaction
4.
Nature ; 527(7578): 323-8, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26536114

ABSTRACT

Staphylococcus aureus is considered to be an extracellular pathogen. However, survival of S. aureus within host cells may provide a reservoir relatively protected from antibiotics, thus enabling long-term colonization of the host and explaining clinical failures and relapses after antibiotic therapy. Here we confirm that intracellular reservoirs of S. aureus in mice comprise a virulent subset of bacteria that can establish infection even in the presence of vancomycin, and we introduce a novel therapeutic that effectively kills intracellular S. aureus. This antibody-antibiotic conjugate consists of an anti-S. aureus antibody conjugated to a highly efficacious antibiotic that is activated only after it is released in the proteolytic environment of the phagolysosome. The antibody-antibiotic conjugate is superior to vancomycin for treatment of bacteraemia and provides direct evidence that intracellular S. aureus represents an important component of invasive infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteremia , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Intracellular Space/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Vancomycin/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Bacteremia/drug therapy , Bacteremia/microbiology , Carrier State/drug therapy , Carrier State/microbiology , Drug Design , Female , Immunoconjugates/chemistry , Intracellular Space/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Microbial Sensitivity Tests , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Vancomycin/therapeutic use
5.
EBioMedicine ; 2(7): 730-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26288846

ABSTRACT

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/ßKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/ßKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/ßKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/ßKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/ßKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.


Subject(s)
Adipose Tissue, Brown/metabolism , Antibodies, Bispecific/pharmacology , Insulin/pharmacology , Membrane Proteins/agonists , Receptor, Fibroblast Growth Factor, Type 1/agonists , Adiponectin/metabolism , Adipose Tissue, Brown/drug effects , Animals , Cell Line , Energy Metabolism/drug effects , Fibroblast Growth Factors/pharmacology , HEK293 Cells , Humans , Klotho Proteins , Macaca fascicularis , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Obese , Protein Binding/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Thermogenesis/drug effects , Weight Loss/drug effects
6.
Clin Cancer Res ; 21(14): 3252-62, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25862760

ABSTRACT

PURPOSE: Chemotherapies are limited by a narrow therapeutic index resulting in suboptimal exposure of the tumor to the drug and acquired tumor resistance. One approach to overcome this is through antibody-drug conjugates (ADC) that facilitate greater potency via target-specific delivery of highly potent cytotoxic agents. EXPERIMENTAL DESIGN: In this study, we used a bioinformatics approach to identify the lymphocyte antigen 6 complex locus E (LY6E), an IFN-inducible glycosylphosphatidylinositol (GPI)-linked cell membrane protein as a promising ADC target. We developed a monoclonal anti-LY6E antibody and characterized in situ LY6E expression in over 750 cancer specimens and normal tissues. Target-dependent anti-LY6E ADC killing was investigated both in vitro and in vivo using patient-derived xenograft models. RESULTS: Using in silico approaches, we found that LY6E was significantly overexpressed and amplified in a wide array of different human solid tumors. IHC analysis revealed high LY6E protein expression in a number of tumor types, such as breast, lung, gastric, ovarian, pancreatic, kidney and head/neck carcinomas. Characterization of the endocytic pathways for LY6E revealed that the LY6E-specific antibody is internalized into cells leading to lysosomal accumulation. Consistent with this, a LY6E-specific ADC inhibited in vitro cell proliferation and produced durable tumor regression in vivo in clinically relevant LY6E-expressing xenograft models. CONCLUSIONS: Our results identify LY6E as a highly promising molecular ADC target for a variety of solid tumor types with current unmet medical need.


Subject(s)
Antigens, Neoplasm/pharmacology , Antigens, Surface/immunology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Neoplasms/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Flow Cytometry , GPI-Linked Proteins/immunology , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, SCID , Polymerase Chain Reaction , RNA, Small Interfering , Transfection , Xenograft Model Antitumor Assays
7.
Cancer Res ; 74(19): 5561-71, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25228655

ABSTRACT

Clinical results from the latest strategies for T-cell activation in cancer have fired interest in combination immunotherapies that can fully engage T-cell immunity. In this study, we describe a trastuzumab-based bispecific antibody, HER2-TDB, which targets HER2 and conditionally activates T cells. HER2-TDB specifically killed HER2-expressing cancer cells at low picomolar concentrations. Because of its unique mechanism of action, which is independent of HER2 signaling or chemotherapeutic sensitivity, HER2-TDB eliminated cells refractory to currently approved HER2 therapies. HER2-TDB exhibited potent antitumor activity in four preclinical model systems, including MMTV-huHER2 and huCD3 transgenic mice. PD-L1 expression in tumors limited HER2-TDB activity, but this resistance could be reversed by anti-PD-L1 treatment. Thus, combining HER2-TDB with anti-PD-L1 yielded a combination immunotherapy that enhanced tumor growth inhibition, increasing the rates and durability of therapeutic response.


Subject(s)
Antibodies, Bispecific/immunology , Lymphocyte Activation , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Rats , Rats, Sprague-Dawley , Trastuzumab
8.
Mol Cancer Ther ; 13(11): 2630-40, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25249555

ABSTRACT

Mesothelin (MSLN) is an attractive target for antibody-drug conjugate therapy because it is highly expressed in various epithelial cancers, with normal expression limited to nondividing mesothelia. We generated novel antimesothelin antibodies and conjugated an internalizing one (7D9) to the microtubule-disrupting drugs monomethyl auristatin E (MMAE) and MMAF, finding the most effective to be MMAE with a lysosomal protease-cleavable valine-citrulline linker. The humanized (h7D9.v3) version, αMSLN-MMAE, specifically targeted mesothelin-expressing cells and inhibited their proliferation with an IC50 of 0.3 nmol/L. Because the antitumor activity of an antimesothelin immunotoxin (SS1P) in transfected mesothelin models did not translate to the clinic, we carefully selected in vivo efficacy models endogenously expressing clinically relevant levels of mesothelin, after scoring mesothelin levels in ovarian, pancreatic, and mesothelioma tumors by immunohistochemistry. We found that endogenous mesothelin in cancer cells is upregulated in vivo and identified two suitable xenograft models for each of these three indications. A single dose of αMSLN-MMAE profoundly inhibited or regressed tumor growth in a dose-dependent manner in all six models, including two patient-derived tumor xenografts. The robust and durable efficacy of αMSLN-MMAE in preclinical models of ovarian, mesothelioma, and pancreatic cancers justifies the ongoing phase I clinical trial.


Subject(s)
GPI-Linked Proteins/immunology , Immunotoxins/pharmacology , Mesothelioma/drug therapy , Oligopeptides/pharmacology , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Female , GPI-Linked Proteins/biosynthesis , Humans , Immunohistochemistry , Immunotoxins/chemistry , Immunotoxins/immunology , Mesothelin , Mice , Oligopeptides/chemistry , Random Allocation , Transfection , Xenograft Model Antitumor Assays
9.
Mol Cell ; 54(6): 987-998, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24882208

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a cellular process essential to the development and maintenance of solid tissues. In cancer, EMT suppresses apoptosis, but the mechanisms remain unclear. EMT selectively attenuated apoptosis signaling via the death receptors DR4 and DR5. Loss of the epithelial cell adhesion protein E-cadherin recapitulated this outcome, whereas homotypic E-cadherin engagement promoted apoptotic signaling via DR4/DR5, but not Fas. Depletion of α-catenin, which couples E-cadherin to the actin cytoskeleton, or actin polymerization inhibitors similarly attenuated DR4/DR5-induced apoptosis. E-cadherin bound specifically to ligated DR4/DR5, requiring extracellular cadherin domain 1 and calcium. E-cadherin augmented DR4/DR5 clustering and assembly of the death-inducing signaling complex (DISC), increasing caspase-8 activation in high molecular weight cell fractions. Conversely, EMT attenuated DR4/DR5-mediated DISC formation and caspase-8 stimulation. Consistent with these findings, epithelial cancer cell lines expressing higher E-cadherin levels displayed greater sensitivity to DR4/DR5-mediated apoptosis. These results have potential implications for tissue homeostasis as well as cancer therapy.


Subject(s)
Apoptosis/physiology , Cadherins/metabolism , Epithelial-Mesenchymal Transition/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Actin Cytoskeleton/metabolism , Antigens, CD , Apoptosis Regulatory Proteins/metabolism , Cadherins/genetics , Calcium , Caspase 8/metabolism , Cell Line, Tumor , Cytoskeleton , Fas-Associated Death Domain Protein/genetics , HEK293 Cells , Humans , RNA Interference , RNA, Small Interfering , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transforming Growth Factor beta/pharmacology , alpha Catenin/genetics
10.
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
11.
J Biol Chem ; 288(50): 36168-78, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24189067

ABSTRACT

Elevated glucagon levels and increased hepatic glucagon receptor (GCGR) signaling contribute to hyperglycemia in type 2 diabetes. We have identified a monoclonal antibody that inhibits GCGR, a class B G-protein coupled receptor (GPCR), through a unique allosteric mechanism. Receptor inhibition is mediated by the binding of this antibody to two distinct sites that lie outside of the glucagon binding cleft. One site consists of a patch of residues that are surface-exposed on the face of the extracellular domain (ECD) opposite the ligand-binding cleft, whereas the second binding site consists of residues in the αA helix of the ECD. A docking model suggests that the antibody does not occlude the ligand-binding cleft. We solved the crystal structure of GCGR ECD containing a naturally occurring G40S mutation and found a shift in the register of the αA helix that prevents antibody binding. We also found that alterations in the αA helix impact the normal function of GCGR. We present a model for the allosteric inhibition of GCGR by a monoclonal antibody that may form the basis for the development of allosteric modulators for the treatment of diabetes and other class B GPCR-related diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Receptors, Glucagon/chemistry , Receptors, Glucagon/immunology , Allosteric Regulation , Amino Acid Sequence , Animals , Crystallography, X-Ray , Extracellular Space/metabolism , Humans , Male , Mice , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Glucagon/antagonists & inhibitors
12.
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
13.
Proc Natl Acad Sci U S A ; 109(36): 14393-8, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22908259

ABSTRACT

Members of the class B family of G protein-coupled receptors (GPCRs) bind peptide hormones and have causal roles in many diseases, ranging from diabetes and osteoporosis to anxiety. Although peptide, small-molecule, and antibody inhibitors of these GPCRs have been identified, structure-based descriptions of receptor antagonism are scarce. Here we report the mechanisms of glucagon receptor inhibition by blocking antibodies targeting the receptor's extracellular domain (ECD). These studies uncovered a role for the ECD as an intrinsic negative regulator of receptor activity. The crystal structure of the ECD in complex with the Fab fragment of one antibody, mAb1, reveals that this antibody inhibits glucagon receptor by occluding a surface extending across the entire hormone-binding cleft. A second antibody, mAb23, blocks glucagon binding and inhibits basal receptor activity, indicating that it is an inverse agonist and that the ECD can negatively regulate receptor activity independent of ligand binding. Biochemical analyses of receptor mutants in the context of a high-resolution ECD structure show that this previously unrecognized inhibitory activity of the ECD involves an interaction with the third extracellular loop of the receptor and suggest that glucagon-mediated structural changes in the ECD accompany receptor activation. These studies have implications for the design of drugs to treat class B GPCR-related diseases, including the potential for developing novel allosteric regulators that target the ECDs of these receptors.


Subject(s)
Models, Molecular , Protein Conformation , Receptors, Glucagon/chemistry , Receptors, Glucagon/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cell Line , Chromatography, Affinity , Crystallography , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fab Fragments/metabolism , Molecular Sequence Data , Mutagenesis , Protein Structure, Tertiary/genetics , Receptors, Glucagon/antagonists & inhibitors
14.
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
15.
PLoS One ; 7(12): e51817, 2012.
Article in English | MEDLINE | ID: mdl-23284778

ABSTRACT

The structure of the Fab region of antibodies is critical to their function. By introducing single cysteine substitutions into various positions of the heavy and light chains of the Fab region of trastuzumab, a potent antagonist of HER2, and using thiol chemistry to link the different Fabs together, we produced a variety of monospecific F(ab')(2)-like molecules with activities spanning from activation to inhibition of breast tumor cell growth. These isomers (or bis-Fabs) of trastuzumab, with varying relative spatial arrangements between the Fv-regions, were able to either promote or inhibit cell-signaling activities through the PI3K/AKT and MAPK pathways. A quantitative phosphorylation mapping of HER2 indicated that the agonistic isomers produced a distinct phosphorylation pattern associated with activation. This study suggests that antibody geometric isomers, found both in nature and during synthetic antibody development, can have profoundly different biological activities independent of their affinities for their target molecules.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/drug therapy , Immunoglobulin Fab Fragments/pharmacology , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal, Humanized/immunology , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoglobulin Fab Fragments/immunology , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Trastuzumab , Tumor Cells, Cultured
16.
Sci Transl Med ; 3(113): 113ra126, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22174314

ABSTRACT

Clinical use of recombinant fibroblast growth factor 21 (FGF21) for the treatment of type 2 diabetes and other disorders linked to obesity has been proposed; however, its clinical development has been challenging owing to its poor pharmacokinetics. Here, we describe an alternative antidiabetic strategy using agonistic anti-FGFR1 (FGF receptor 1) antibodies (R1MAbs) that mimic the metabolic effects of FGF21. A single injection of R1MAb into obese diabetic mice induced acute and sustained amelioration of hyperglycemia, along with marked improvement in hyperinsulinemia, hyperlipidemia, and hepatosteatosis. R1MAb activated the mitogen-activated protein kinase pathway in adipose tissues, but not in liver, and neither FGF21 nor R1MAb improved glucose clearance in lipoatrophic mice, which suggests that adipose tissues played a central role in the observed metabolic effects. In brown adipose tissues, both FGF21 and R1MAb induced phosphorylation of CREB (cyclic adenosine 5'-monophosphate response element-binding protein), and mRNA expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator 1α) and the downstream genes associated with oxidative metabolism. Collectively, we propose FGFR1 in adipose tissues as a major functional receptor for FGF21, as an upstream regulator of PGC-1α, and as a compelling target for antibody-based therapy for type 2 diabetes and other obesity-associated disorders.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Type 2/therapy , Fibroblast Growth Factors/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Receptor, Fibroblast Growth Factor, Type 1/genetics , Tissue Distribution , Trans-Activators/metabolism , Transcription Factors
17.
Nat Immunol ; 12(12): 1159-66, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21993848

ABSTRACT

Interleukin 17C (IL-17C) is a member of the IL-17 family that is selectively induced in epithelia by bacterial challenge and inflammatory stimuli. Here we show that IL-17C functioned in a unique autocrine manner, binding to a receptor complex consisting of the receptors IL-17RA and IL-17RE, which was preferentially expressed on tissue epithelial cells. IL-17C stimulated epithelial inflammatory responses, including the expression of proinflammatory cytokines, chemokines and antimicrobial peptides, which were similar to those induced by IL-17A and IL-17F. However, IL-17C was produced by distinct cellular sources, such as epithelial cells, in contrast to IL-17A, which was produced mainly by leukocytes, especially those of the T(H)17 subset of helper T cells. Whereas IL-17C promoted inflammation in an imiquimod-induced skin-inflammation model, it exerted protective functions in dextran sodium sulfate-induced colitis. Thus, IL-17C is an essential autocrine cytokine that regulates innate epithelial immune responses.


Subject(s)
Autocrine Communication , Epithelial Cells/immunology , Immunity, Innate/immunology , Interleukin-17/metabolism , Animals , Cell Line , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Epithelial Cells/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Inflammation/immunology , Inflammation/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Binding , Receptors, Interleukin-17/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/pathology
18.
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
19.
Clin Cancer Res ; 17(5): 965-75, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21245091

ABSTRACT

PURPOSE: To identify and evaluate targets amenable to antibody therapy in melanoma. EXPERIMENTAL DESIGN: We searched for mRNA transcripts coding for cell-surface proteins with expression patterns similar to that of the melanoma oncogene MITF. One such candidate, the endothelin B receptor (EDNBR), was first analyzed for a functional contribution to tumor growth by conditional induction of shRNA. Second, antibodies were raised to the receptor, conjugated with monomethyl auristatin E, and tested for efficacy against melanoma tumor models generated from cell lines. RESULTS: Conditional knockdown of the receptor in tumor xenograft models resulted in only a modest impact on tumor growth. A monoclonal antibody reactive with the N-terminal tail of EDNBR was found to internalize rapidly into melanoma cells. When conjugated with monomethyl auristatin E, the antibody-drug conjugate (ADC) showed remarkable efficacy against human melanoma cell lines and xenograft tumor models that was commensurate with levels of receptor expression. Comparative immunohistochemistry revealed a range of EDNBR expression across a panel of human melanomas, with the majority expressing levels equivalent to or greater than that in the models responsive to the ADC. CONCLUSION: An ADC targeting the EDNBR is highly efficacious in preclinical models of melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Endothelin B Receptor Antagonists , Immunoconjugates/therapeutic use , Melanoma, Experimental/drug therapy , Melanoma/drug therapy , Oligopeptides/therapeutic use , Receptor, Endothelin B/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Humans , Macaca fascicularis , Melanoma/genetics , Melanoma/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Microphthalmia-Associated Transcription Factor/genetics , Oligopeptides/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Xenograft Model Antitumor Assays
20.
Dev Cell ; 20(1): 33-46, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21238923

ABSTRACT

Robo4 is an endothelial cell-specific member of the Roundabout axon guidance receptor family. To identify Robo4 binding partners, we performed a protein-protein interaction screen with the Robo4 extracellular domain. We find that Robo4 specifically binds to UNC5B, a vascular Netrin receptor, revealing unexpected interactions between two endothelial guidance receptors. We show that Robo4 maintains vessel integrity by activating UNC5B, which inhibits signaling downstream of vascular endothelial growth factor (VEGF). Function-blocking monoclonal antibodies against Robo4 and UNC5B increase angiogenesis and disrupt vessel integrity. Soluble Robo4 protein inhibits VEGF-induced vessel permeability and rescues barrier defects in Robo4(-/-) mice, but not in mice treated with anti-UNC5B. Thus, Robo4-UNC5B signaling maintains vascular integrity by counteracting VEGF signaling in endothelial cells, identifying a novel function of guidance receptor interactions in the vasculature.


Subject(s)
Blood Vessels/metabolism , Blood Vessels/pathology , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Animals , Antibodies, Blocking/pharmacology , Blood Vessels/drug effects , Blood Vessels/enzymology , Capillary Permeability/drug effects , Enzyme Activation/drug effects , Humans , Ligands , Mice , Models, Biological , Netrin Receptors , Protein Binding/drug effects , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction/drug effects , Sus scrofa , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...