Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
PLoS One ; 16(12): e0257972, 2021.
Article in English | MEDLINE | ID: mdl-34972111

ABSTRACT

Cancer immunotherapies, such as checkpoint blockade of programmed cell death protein-1 (PD-1), represents a breakthrough in cancer treatment, resulting in unprecedented results in terms of overall and progression-free survival. Discovery and development of novel anti PD-1 inhibitors remains a field of intense investigation, where novel monoclonal antibodies (mAbs) and novel antibody formats (e.g., novel isotype, bispecific mAb and low-molecular-weight compounds) are major source of future therapeutic candidates. HLX10, a fully humanized IgG4 monoclonal antibody against PD-1 receptor, increased functional activities of human T-cells and showed in vitro, and anti-tumor activity in several tumor models. The combined inhibition of PD-1/PDL-1 and angiogenesis pathways using anti-VEGF antibody may enhance a sustained suppression of cancer-related angiogenesis and tumor elimination. To elucidate HLX10's mode of action, we solved the structure of HLX10 in complex with PD-1 receptor. Detailed epitope analysis showed that HLX10 has a unique mode of recognition compared to the clinically approved PD1 antibodies Pembrolizumab and Nivolumab. Notably, HLX10's epitope was closer to Pembrolizumab's epitope than Nivolumab's epitope. However, HLX10 and Pembrolizumab showed an opposite heavy chain (HC) and light chain (LC) usage, which recognizes several overlapping amino acid residues on PD-1. We compared HLX10 to Nivolumab and Pembrolizumab and it showed similar or better bioactivity in vitro and in vivo, providing a rationale for clinical evaluation in cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/immunology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition/drug effects , Epitopes/immunology , Humans , Immunoglobulin Fab Fragments/metabolism , Interferon-gamma/metabolism , Interleukin-2/metabolism , Ligands , Macaca fascicularis , Mice, Inbred NOD , Mice, SCID , Models, Molecular , Neoplasms/drug therapy , Nivolumab/chemistry , Nivolumab/therapeutic use , Protein Binding , Rats , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
2.
Expert Opin Biol Ther ; 21(11): 1491-1507, 2021 11.
Article in English | MEDLINE | ID: mdl-34632911

ABSTRACT

BACKGROUND: Cetuximab, the first approved EGFR targeting therapeutic antibody, is currently used to treat colorectal cancer and head and neck cancer. While effective, cetuximab is associated with a higher rate of skin rash, infusion reactions, and gastrointestinal toxicity, which was suggested to be linked to the presence of heterogeneous glycan contents on the Fab of the SP2/0-produced cetuximab. OBJECTIVE AND METHODS: To improve efficacy and minimize toxicity of EGFR inhibition treatment, we re-engineered cetuximab by humanizing its Fab regions and minimizing its glycan contents to generate HLX07. RESULTS: HLX07 binds to EGFR with similar affinity as cetuximab and shows better bioactivity compared to cetuximab in vitro. In vivo studies demonstrated that HLX07 significantly inhibited the growth of A431, FaDu, NCI-H292, and WiDr tumor cells and synergized them with chemotherapeutics and immune simulator agents such as anti-PD-1. In cynomolgus monkeys, 13-week repeat-dose GLP toxicokinetic studies showed minimal-to-mild toxicities in the dose range of up to 60 mg/kg/wk. In the preliminary phase 1 dose-escalation study, HLX07 had showed lower incidence of skin rashes with grade >2 severities. CONCLUSION: HLX07 is currently under phase 1/2 clinical development. We believe HLX07 would potentially be an alternative for patients who have been suffering from cetuximab-mediated toxicity.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Antibodies, Monoclonal , Antineoplastic Agents/adverse effects , Cell Line, Tumor , Cetuximab , ErbB Receptors , Humans
3.
Oncotarget ; 9(77): 34459-34470, 2018 Oct 02.
Article in English | MEDLINE | ID: mdl-30349641

ABSTRACT

Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) has been reported to mediate both tumorigenic and anti-tumor effects in vivo. Blockade of the CEACAM1 signaling pathway has recently been implicated as a novel mechanism for cancer immunotherapy. CC1, a mouse anti-CEACAM1 monoclonal antibody (mAb), has been widely used as a pharmacological tool in preclinical studies to inform on CEACAM1 pathway biology although limited data are available on its CEACAM1 blocking characteristics or pharmacodynamic-pharmacokinetic profiles. We sought to investigate CEACAM1 expression on mouse tumor and immune cells, characterize CC1 mAb binding, and evaluate CC1 in syngeneic mouse oncology models as a monotherapy and in combination with an anti-PD-1 mAb. CEACAM1 expression was observed at high levels on neutrophils, NK cells and myeloid-derived suppressor cells (MDSCs), while the expression on tumor-infiltrating CD8+ T cells was low. Unexpectedly, rather than blocking, CC1 facilitated binding of soluble CEACAM1 to CEACAM1 expressing cells. No anti-tumor effects were observed in CT26, MBT2 or A20 models when tested up to 30 mg/kg dose, a dose that was estimated to achieve >90% target engagement in vivo. Taken together, tumor infiltrating CD8+ T cells express low levels of CEACAM1 and CC1 Ab mediates no or minimal anti-tumor effects in vivo, as a monotherapy or in combination with anti-PD-1 treatment.

4.
Nutrients ; 9(1)2016 Dec 28.
Article in English | MEDLINE | ID: mdl-28036028

ABSTRACT

High-fat (HF) diets typically promote diet-induced obesity (DIO) and metabolic dysfunction (i.e., insulin resistance, hypertriglyceridemia, and hepatic steatosis). Dysfunction of triacylglycerol (TAG) metabolism may contribute to the development of hepatic steatosis, via increased de novo lipogenesis or repackaging of circulating nonesterified fatty acids (NEFAs). Hepatic TAG production (HTP) rate can be assessed through injecting mice with nonionic detergents that inhibit tissue lipoprotein lipase. Potential confounding effects of detergent-based HTP tests (HTPTs) used in longitudinal studies-including the impact on food intake, energy balance, and weight gain-have not been reported. To examine this, male C57BL/6J mice were fed a 10% or 60% kcal diet. After 4 weeks, the mice underwent an HTPT via poloxamer 407 intraperitoneal injections (1000 mg/kg). Weight gain, energy intake, and postabsorptive TAG levels normalized 7-10 days post-HTPT. The post-HTPT recovery of body weight and energy intake suggest that, in metabolic phenotyping studies, any additional sample collection should occur at least 7-10 days after the HTPT to reduce confounding effects. Diet-specific effects on HTP were also observed: HF-fed mice had reduced HTP, plasma TAG, and NEFA levels compared to controls. In conclusion, the current study highlights the procedural and physiological complexities associated with studying lipid metabolism using a HTPT in the DIO mouse model.


Subject(s)
Diet, High-Fat/adverse effects , Liver/metabolism , Triglycerides/biosynthesis , Animals , Disease Models, Animal , Energy Intake , Energy Metabolism , Fatty Acids, Nonesterified/blood , Fatty Liver/blood , Fatty Liver/etiology , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/etiology , Triglycerides/blood , Weight Gain
5.
J Diabetes Sci Technol ; 8(4): 865-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24876415

ABSTRACT

Many therapeutic monoclonal antibodies act as antagonists to receptors by targeting and blocking the natural ligand binding site (orthosteric site). In contrast, the use of antibodies to target receptors at allosteric sites (distinct from the orthosteric site) has not been extensively studied. This approach is especially important in metabolic diseases in which endogenous ligand levels are dysregulated. Herein, we review our investigations of 3 categories of human monoclonal antibodies that bind allosterically to the insulin receptor (INSR) and affect its activity: XMetA, XMetS and XMetD. XMetA directly activates the INSR either alone or in combination with insulin. XMetS, in contrast, does not directly activate the INSR but markedly enhances the receptor's ability to bind insulin and potentiate insulin signaling. Both XMetA and XMetS are effective in controlling hyperglycemia in mouse models of diabetes. A third allosteric antibody, XMetD, is an inhibitor of INSR signaling. This antibody reverses insulin-induced hypoglycemia in a mouse model of hyperinsulinemia. These studies indicate, therefore, that allosteric antibodies to INSR can modulate its signaling and correct conditions of glucose dysregulation. These studies also raise the possibility that the use of allosteric antibodies can be expanded to other receptors for the treatment of metabolic disorders.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hyperglycemia/drug therapy , Hypoglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Receptor, Insulin/immunology , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Insulin/blood , Insulin/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation
6.
MAbs ; 6(1): 262-72, 2014.
Article in English | MEDLINE | ID: mdl-24423625

ABSTRACT

Novel therapies are needed for the treatment of hypoglycemia resulting from both endogenous and exogenous hyperinsulinema. To provide a potential new treatment option, we identified XMetD, an allosteric monoclonal antibody to the insulin receptor (INSR) that was isolated from a human antibody phage display library. To selectively obtain antibodies directed at allosteric sites, panning of the phage display library was conducted using the insulin-INSR complex. Studies indicated that XMetD bound to the INSR with nanomolar affinity. Addition of insulin reduced the affinity of XMetD to the INSR by 3-fold, and XMetD reduced the affinity of the INSR for insulin 3-fold. In addition to inhibiting INSR binding, XMetD also inhibited insulin-induced INSR signaling by 20- to 100-fold. These signaling functions included INSR autophosphorylation, Akt activation and glucose transport. These data indicated that XMetD was an allosteric antagonist of the INSR because, in addition to inhibiting the INSR via modulation of binding affinity, it also inhibited the INSR via modulation of signaling efficacy. Intraperitoneal injection of XMetD at 10 mg/kg twice weekly into normal mice induced insulin resistance. When sustained-release insulin implants were placed into normal mice, they developed fasting hypoglycemia in the range of 50 mg/dl. This hypoglycemia was reversed by XMetD treatment. These studies demonstrate that allosteric monoclonal antibodies, such as XMetD, can antagonize INSR signaling both in vitro and in vivo. They also suggest that this class of allosteric monoclonal antibodies has the potential to treat hyperinsulinemic hypoglycemia resulting from conditions such as insulinoma, congenital hyperinsulinism and insulin overdose.


Subject(s)
Antibodies, Monoclonal/immunology , Congenital Hyperinsulinism/immunology , Receptor, Insulin/antagonists & inhibitors , Single-Chain Antibodies/immunology , Animals , Antibodies, Monoclonal/pharmacology , Biological Transport, Active/drug effects , Biological Transport, Active/immunology , CHO Cells , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/pathology , Cricetinae , Cricetulus , Glucose/immunology , Insulin Resistance/immunology , Mice , Rats , Receptor, Insulin/immunology , Single-Chain Antibodies/pharmacology
7.
J Pharmacol Exp Ther ; 348(1): 202-15, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24194526

ABSTRACT

Interleukin-1ß (IL-1ß) is a proinflammatory cytokine that is implicated in many autoinflammatory disorders, but is also important in defense against pathogens. Thus, there is a need to safely and effectively modulate IL-1ß activity to reduce pathology while maintaining function. Gevokizumab is a potent anti-IL-1ß antibody being developed as a treatment for diseases in which IL-1ß has been associated with pathogenesis. Previous data indicated that gevokizumab negatively modulates IL-1ß signaling through an allosteric mechanism. Because IL-1ß signaling is a complex, dynamic process involving multiple components, it is important to understand the kinetics of IL-1ß signaling and the impact of gevokizumab on this process. In the present study, we measured the impact of gevokizumab on the IL-1ß system using Schild analysis and surface plasmon resonance studies, both of which demonstrated that gevokizumab decreases the binding affinity of IL-1ß for the IL-1 receptor type I (IL-1RI) signaling receptor, but not the IL-1 counter-regulatory decoy receptor (IL-1 receptor type II). Gevokizumab inhibits both the binding of IL-1ß to IL-1RI and the subsequent recruitment of IL-1 accessory protein primarily by reducing the association rates of these interactions. Based on this information and recently published structural data, we propose that gevokizumab decreases the association rate for binding of IL-1ß to its receptor by altering the electrostatic surface potential of IL-1ß, thus reducing the contribution of electrostatic steering to the rapid association rate. These data indicate, therefore, that gevokizumab is a unique inhibitor of IL-1ß signaling that may offer an alternative to current therapies for IL-1ß-associated autoinflammatory diseases.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Receptors, Interleukin-1/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/immunology , HeLa Cells , Humans , Interleukin-1beta/antagonists & inhibitors , Protein Binding/immunology , Receptors, Interleukin-1/metabolism
8.
J Immunol Methods ; 394(1-2): 10-21, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23624043

ABSTRACT

Improper protein folding or aggregation can frequently be responsible for low expression and poor functional activity of antibody fragments secreted into the Escherichia coli periplasm. Expression issues also can affect selection of antibody candidates from phage libraries, since antibody fragments displayed on phage also are secreted into the E. coli periplasm. To improve secretion of properly folded antibody fragments into the periplasm, we have developed a novel approach that involves co-expressing the antibody fragments with the peptidyl prolyl cis-trans isomerase, FkpA, lacking its signal sequence (cytFkpA) which consequently is expressed in the E. coli cytosol. Cytoplasmic expression of cytFkpA improved secretion of functional Fab fragments into the periplasm, exceeding even the benefits from co-expressing Fab fragments with native, FkpA localized in the periplasm. In addition, panning and subsequent screening of large Fab and scFv naïve phage libraries in the presence of cytFkpA significantly increased the number of unique clones selected, as well as their functional expression levels and diversity.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/immunology , Immunoglobulin Fab Fragments/metabolism , Membrane Proteins/physiology , Peptidylprolyl Isomerase/physiology , Periplasm/metabolism , Enzyme-Linked Immunosorbent Assay , Peptide Library , Protein Folding
9.
Atherosclerosis ; 216(2): 313-20, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21411094

ABSTRACT

OBJECTIVE: Atherosclerosis is a condition that is increasingly contributing to worldwide mortality through complications such as stroke and myocardial infarction. IL-1ß plays multiple direct, local roles in the formation and stability of the atheroma by eliciting the production of additional cytokines and proteolytic enzymes from macrophages, endothelial cells (EC) and smooth muscle cells (SMC). We therefore tested whether an anti-IL-1ß antibody, XOMA 052, might inhibit the secretion of pro-atherogenic cytokines from macrophages in vitro and affect a positive outcome in the Apolipoprotein E-deficient mouse (ApoE(-/-)) model of atherosclerosis in vivo. METHODS AND RESULTS: In an in vitro co-culture model, XOMA 052 inhibited macrophage-induced secretion of key atherogenic cytokines from EC and SMC, including IL-6, IL-8, MCP-1 and TNFα. The release of degradative enzymes, such as the matrix metalloproteinases MMP-3 and MMP-9, was also decreased by XOMA 052. In addition, XOMA 052 inhibited the secretion of IL-7 from EC and IL-4 from SMC, cytokines not previously reported to be driven by IL-1ß in this context. In vivo, XMA052 MG1K, a chimeric murine version of XOMA 052, inhibited the formation of atherosclerotic lesions in the ApoE(-/-) model at all three doses tested. This effect was comparable to that reported for complete genetic ablation of IL-1ß or IL-1R1 on an ApoE(-/-) background and was associated with decreases in plasma non-HDL/HDL cholesterol ratio and plaque lipid content and macrophage infiltration. CONCLUSIONS: These results demonstrate for the first time that an antibody targeting IL-1ß can inhibit the progression of atherosclerosis in vivo, highlighting the importance of this key cytokine in cardiovascular disease.


Subject(s)
Antibodies, Monoclonal/metabolism , Apolipoproteins E/genetics , Atherosclerosis/blood , Biomarkers/metabolism , Interleukin-1beta/metabolism , Plaque, Atherosclerotic/blood , Animals , Apolipoproteins E/blood , Atherosclerosis/immunology , Body Weight , Coculture Techniques , Cytokines/metabolism , Endothelial Cells/cytology , Humans , Lipids/chemistry , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Plaque, Atherosclerotic/immunology
10.
MAbs ; 3(1): 49-60, 2011.
Article in English | MEDLINE | ID: mdl-21048425

ABSTRACT

Interleukin-1ß (IL-1ß) is a potent mediator of inflammatory responses and plays a role in the differentiation of a number of lymphoid cells. In several inflammatory and autoimmune diseases, serum levels of IL-1ß are elevated and correlate with disease development and severity. The central role of the IL-1 pathway in several diseases has been validated by inhibitors currently in clinical development or approved by the FDA. However, the need to effectively modulate IL-1ß-mediated local inflammation with the systemic delivery of an efficacious, safe and convenient drug still exists. To meet these challenges, we developed XOMA 052 (gevokizumab), a potent anti-IL-1ß neutralizing antibody that was designed in silico and humanized using Human Engineering™ technology. XOMA 052 has a 300 femtomolar binding affinity for human IL-1ß and an in vitro potency in the low picomolar range. XOMA 052 binds to a unique IL-1ß epitope where residues critical for binding have been identified. We have previously reported that XOMA 052 is efficacious in vivo in a diet-induced obesity mouse model thought to be driven by low levels of chronic inflammation. We report here that XOMA 052 also reduces acute inflammation in vivo, neutralizing the effect of exogenously administered human IL-1ß and blocking peritonitis in a mouse model of acute gout. Based on its high potency, novel mechanism of action, long half-life, and high affinity, XOMA 052 provides a new strategy for the treatment of a number of inflammatory, autoimmune and metabolic diseases in which the role of IL-1ß is central to pathogenesis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody Affinity/immunology , Inflammation/prevention & control , Interleukin-1beta/pharmacology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Cell Line , Cross Reactions/immunology , Dose-Response Relationship, Drug , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Inflammation/blood , Inflammation/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Interleukin-6/metabolism , Kinetics , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Protein Binding/immunology , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid
11.
J Biol Chem ; 285(27): 20607-14, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20410301

ABSTRACT

Many therapeutic antibodies act as antagonists to competitively block cellular signaling pathways. We describe here an approach for the therapeutic use of monoclonal antibodies based on context-dependent attenuation to reduce pathologically high activity while allowing homeostatic signaling in biologically important pathways. Such attenuation is achieved by modulating the kinetics of a ligand binding to its various receptors and regulatory proteins rather than by complete blockade of signaling pathways. The anti-interleukin-1beta (IL-1beta) antibody XOMA 052 is a potent inhibitor of IL-1beta activity that reduces the affinity of IL-1beta for its signaling receptor and co-receptor but not for its decoy and soluble inhibitory receptors. This mechanism shifts the effective dose response of the cytokine so that the potency of IL-1beta bound by XOMA 052 is 20-100-fold lower than that of IL-1beta in the absence of antibody in a variety of in vitro cell-based assays. We propose that by decreasing potency of IL-1beta while allowing binding to its clearance and inhibitory receptors, XOMA 052 treatment will attenuate IL-1beta activity in concert with endogenous regulatory mechanisms. Furthermore, the ability to bind the decoy receptor may reduce the potential for accumulation of antibody.target complexes. Regulatory antibodies like XOMA 052, which selectively modulate signaling pathways, may represent a new mechanistic class of therapeutic antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interleukin-1beta/physiology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bioengineering , Fibroblasts/cytology , Fibroblasts/physiology , HeLa Cells/drug effects , HeLa Cells/physiology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Interleukin-1/physiology , Interleukin-1beta/drug effects , Kidney/drug effects , Kidney/physiology , Kinetics , Ligands , Luciferases/genetics , Lung/cytology , Lung/physiology , NF-kappa B/physiology , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Receptors, Interleukin-1/drug effects , Receptors, Interleukin-1/physiology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology
12.
J Biol Chem ; 280(48): 40210-5, 2005 Dec 02.
Article in English | MEDLINE | ID: mdl-16199535

ABSTRACT

Arrestins are important proteins, which regulate the function of serpentine heptahelical receptors and contribute to multiple signaling pathways downstream of receptors. The ubiquitous beta-arrestins are believed to function exclusively as monomers, although self-association is assumed to control the activity of visual arrestin in the retina, where this isoform is particularly abundant. Here the oligomerization status of beta-arrestins was investigated using different approaches, including co-immunoprecipitation of epitope-tagged beta-arrestins and resonance energy transfer (BRET and FRET) in living cells. At steady state and at physiological concentrations, beta-arrestins constitutively form both homo- and hetero-oligomers. Co-expression of beta-arrestin2 and beta-arrestin1 prevented beta-arrestin1 accumulation into the nucleus, suggesting that hetero-oligomerization may have functional consequences. Our data clearly indicate that beta-arrestins can exist as homo- and hetero-oligomers in living cells and raise the hypothesis that the oligomeric state may regulate their subcellular distribution and functions.


Subject(s)
Arrestins/chemistry , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Dimerization , Dose-Response Relationship, Drug , Fluorescence Resonance Energy Transfer , Genetic Vectors , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Microscopy, Fluorescence , Models, Biological , Protein Binding , Protein Isoforms , Retina/chemistry , Signal Transduction , beta-Arrestins
13.
J Biol Chem ; 277(38): 34666-73, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12089144

ABSTRACT

Although homo-oligomerization has been reported for several G protein-coupled receptors, this phenomenon was not studied at low concentrations of receptors. Furthermore, it is not clear whether homo-oligomerization corresponds to an intrinsic property of nascent receptors or if it is a consequence of receptor activation. Here CCR5 receptor oligomerization was studied by bioluminescence resonance energy transfer (BRET) in cells expressing physiological levels of receptors. A strong energy transfer could be observed, in the absence of ligands, in whole cells and in both endoplasmic reticulum and plasma membrane subfractions, supporting the hypothesis of a constitutive oligomerization that occurs early after biosynthesis. No change in BRET was observed upon agonist binding, indicating that the extent of oligomerization is unrelated to the activation state of the receptor. In contrast, a robust increase of BRET, induced by a monoclonal antibody known to promote receptor clustering, suggests that microaggregation of preformed receptor homo-oligomers can occur. Taken together, our data indicate that constitutive receptor homo-oligomerization has a biologically relevant significance and might be involved in the process of receptor biosynthesis.


Subject(s)
Receptors, CCR5/metabolism , Biopolymers , Cell Membrane/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Energy Transfer , Fluorescent Antibody Technique , Humans , Luminescent Measurements , Receptors, CCR5/agonists
14.
Mol Biol Cell ; 13(2): 723-37, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11854425

ABSTRACT

CC-chemokine receptor 5 (CCR5) is the principal coreceptor for macrophage-tropic strains of human immunodeficiency virus type 1 (HIV-1). We have generated a set of anti-CCR5 monoclonal antibodies and characterized them in terms of epitope recognition, competition with chemokine binding, receptor activation and trafficking, and coreceptor activity. MC-4, MC-5, and MC-7 mapped to the amino-terminal domain, MC-1 to the second extracellular loop, and MC-6 to a conformational epitope covering multiple extracellular domains. MC-1 and MC-6 inhibited regulated on activation normal T cell expressed and secreted (RANTES), macrophage inflammatory polypeptide-1beta, and Env binding, whereas MC-5 inhibited macrophage inflammatory polypeptide-1beta and Env but not RANTES binding. MC-6 induced signaling in different functional assays, suggesting that this monoclonal antibody stabilizes an active conformation of CCR5. Flow cytometry and real-time confocal microscopy showed that MC-1 promoted strong CCR5 endocytosis. MC-1 but not its monovalent isoforms induced an increase in the transfer of energy between CCR5 molecules. Also, its monovalent isoforms bound efficiently, but did not internalize the receptor. In contrast, MC-4 did not prevent RANTES binding or subsequent signaling, but inhibited its ability to promote CCR5 internalization. These results suggest the existence of multiple active conformations of CCR5 and indicate that CCR5 oligomers are involved in an internalization process that is distinct from that induced by the receptor's agonists.


Subject(s)
Receptors, CCR5/physiology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Binding Sites , CCR5 Receptor Antagonists , Cricetinae , Endocytosis , HIV-1/physiology , Humans , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary/physiology , Receptors, CCR5/immunology , Signal Transduction , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL