Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
MAbs ; 12(1): 1838036, 2020.
Article in English | MEDLINE | ID: mdl-33146056

ABSTRACT

The extracellular ATP/adenosine axis in the tumor microenvironment (TME) has emerged as an important immune-regulatory pathway. Nucleoside triphosphate diphosphohydrolase-1 (NTPDase1), otherwise known as CD39, is highly expressed in the TME, both on infiltrating immune cells and tumor cells across a broad set of cancer indications. CD39 processes pro-inflammatory extracellular ATP to ADP and AMP, which is then processed by Ecto-5'-nucleotidase/CD73 to immunosuppressive adenosine. Directly inhibiting the enzymatic function of CD39 via an antibody has the potential to unleash an immune-mediated anti-tumor response via two mechanisms: 1) increasing the availability of immunostimulatory extracellular ATP released by damaged and/or dying cells, and 2) reducing the generation and accumulation of suppressive adenosine within the TME. Tizona Therapeutics has engineered a novel first-in-class fully human anti-CD39 antibody, TTX-030, that directly inhibits CD39 ATPase enzymatic function with sub-nanomolar potency. Further characterization of the mechanism of inhibition by TTX-030 using CD39+ human melanoma cell line SK-MEL-28 revealed an uncompetitive allosteric mechanism (α < 1). The uncompetitive mechanism of action enables TTX-030 to inhibit CD39 at the elevated ATP concentrations reported in the TME. Maximal inhibition of cellular CD39 ATPase velocity was 85%, which compares favorably to results reported for antibody inhibitors to other enzyme targets. The allosteric mechanism of TTX-030 was confirmed via mapping the epitope to a region of CD39 distant from its active site, which suggests possible models for how potent inhibition is achieved. In summary, TTX-030 is a potent allosteric inhibitor of CD39 ATPase activity that is currently being evaluated in clinical trials for cancer therapy.


Subject(s)
Adenosine Triphosphatases/drug effects , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apyrase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Antibodies, Monoclonal/chemistry , Antibody Affinity , Antineoplastic Agents/chemistry , Binding Sites, Antibody , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans
2.
Cancer Discov ; 9(12): 1754-1773, 2019 12.
Article in English | MEDLINE | ID: mdl-31699796

ABSTRACT

We explored the mechanism of action of CD39 antibodies that inhibit ectoenzyme CD39 conversion of extracellular ATP (eATP) to AMP and thus potentially augment eATP-P2-mediated proinflammatory responses. Using syngeneic and humanized tumor models, we contrast the potency and mechanism of anti-CD39 mAbs with other agents targeting the adenosinergic pathway. We demonstrate the critical importance of an eATP-P2X7-ASC-NALP3-inflammasome-IL18 pathway in the antitumor activity mediated by CD39 enzyme blockade, rather than simply reducing adenosine as mechanism of action. Efficacy of anti-CD39 activity was underpinned by CD39 and P2X7 coexpression on intratumor myeloid subsets, an early signature of macrophage depletion, and active IL18 release that facilitated the significant expansion of intratumor effector T cells. More importantly, anti-CD39 facilitated infiltration into T cell-poor tumors and rescued anti-PD-1 resistance. Anti-human CD39 enhanced human T-cell proliferation and Th1 cytokine production and suppressed human B-cell lymphoma in the context of autologous Epstein-Barr virus-specific T-cell transfer. SIGNIFICANCE: Overall, these data describe a potent and novel mechanism of action of antibodies that block mouse or human CD39, triggering an eATP-P2X7-inflammasome-IL18 axis that reduces intratumor macrophage number, enhances intratumor T-cell effector function, overcomes anti-PD-1 resistance, and potentially enhances the efficacy of adoptive T-cell transfer.This article is highlighted in the In This Issue feature, p. 1631.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents, Immunological/administration & dosage , Apyrase/antagonists & inhibitors , Inflammasomes/metabolism , Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Neoplasm Transplantation , Neoplasms/immunology , Receptors, Purinergic P2X7/metabolism , Signal Transduction
3.
MAbs ; 10(5): 796-802, 2018 07.
Article in English | MEDLINE | ID: mdl-29589989

ABSTRACT

Loss-of-function mutations of the ß-cell ATP-sensitive potassium channels (KATP) cause the most common and severe form of congenital hyperinsulinism (KATPHI), a disorder of ß-cell function characterized by severe hypoglycemia. Children with KATPHI are typically unresponsive to medical therapy and require pancreatectomy for intractable hypoglycemia. We tested the hypothesis that inhibition of insulin receptor signaling may prevent hypoglycemia in KATPHI. To test this hypothesis, we examined the effect of an antibody allosteric inhibitor of the insulin receptor, XMetD, on fasting plasma glucose in a mouse model of KATPHI (SUR-1-/- mice). SUR-1-/- and wild-type mice received twice weekly intraperitoneal injections of either XMetD or control antibody for 8 wks. Treatment with XMetD significantly decreased insulin sensitivity, and increased hepatic glucose output and fasting plasma glucose. These findings support the potential use of insulin receptor antagonists as a therapeutic approach to control the hypoglycemia in congenital hyperinsulinism.


Subject(s)
Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Hyperinsulinism/metabolism , Hypoglycemia/prevention & control , KATP Channels/metabolism , Receptor, Insulin/immunology , Animals , Antibodies, Monoclonal/immunology , Blood Glucose/metabolism , Fasting/blood , Humans , Hyperinsulinism/genetics , Hypoglycemia/genetics , Hypoglycemia/immunology , Insulin/blood , KATP Channels/genetics , Male , Mice, Inbred C57BL , Mice, Knockout
4.
Obesity (Silver Spring) ; 24(8): 1687-94, 2016 08.
Article in English | MEDLINE | ID: mdl-27330016

ABSTRACT

OBJECTIVE: Leptin (LEP) deficiency results in major metabolic perturbations, including obesity, dyslipidemia, and diabetes. Although LEP deficiency can be treated with daily injections of a recombinant LEP, generation of an antibody activating the LEP receptor (LEPR) that has both an intrinsically long half-life and low immunogenicity could be useful in the treatment of this condition. METHODS: Phage display technology coupled with flow cytometry and cell-based in vitro assays were employed to identify an allosteric agonist of the mouse LEPR. LEP-deficient Lep(ob) /Lep(ob) mice were used to compare in vivo effects of LEP to antibody administration. To evaluate hypothalamic effects of treatment, changes in mRNA levels of neuropeptide Y and proopiomelanocortin were measured. RESULTS: XPA.80.037 is a monoclonal antibody that demonstrates allosteric agonism of the mouse LEPR. Treatment of Lep(ob) /Lep(ob) mice with XPA.80.037 markedly reduced hyperphagia and body weight, normalized blood glucose and plasma insulin levels, and corrected dyslipidemia. These metabolic alterations correlated with changes in mRNA levels of neuropeptide Y and proopiomelanocortin, suggesting that XPA.80.037 had hypothalamic effects. CONCLUSIONS: Agonist allosteric monoclonal antibodies to the LEPR can correct metabolic effects associated with LEP deficiency in vivo and thereby have the potential to treat conditions of LEP deficiency.


Subject(s)
Blood Glucose/metabolism , Leptin/metabolism , Leptin/physiology , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Receptors, Leptin/metabolism , Allosteric Regulation , Animals , Body Weight , Diabetes Mellitus/metabolism , Half-Life , Hypothalamus/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Phenotype
6.
J Pharmacol Exp Ther ; 353(1): 35-43, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25613982

ABSTRACT

The monoclonal antibody XMetA is an allosteric partial agonist of the insulin receptor (IR), which activates the metabolic Akt kinase signaling pathway while having little or no effect on the mitogenic extracellular signal-regulated kinase (ERK) signaling pathway. To investigate the nature of this selective signaling, we have conducted a detailed investigation of XMetA to evaluate specific phosphorylation and activation of IR, Akt, and ERK in Chinese hamster ovary cell lines expressing either the short or long isoform of the human IR. Insulin activated both pathways, but the phosphorylation of Akt was more sensitive to the hormone than the phosphorylation of ERK. Maximally effective concentrations of XMetA elicited phosphorylation patterns similar to 40-100 pM insulin, which were sufficient for robust Akt phosphorylation, but had little effect on ERK phosphorylation. These data indicate that the preferential signaling of XMetA is due to an innate difference in pathway sensitivity of Akt versus ERK responses to IR activation and partial agonism by XMetA, rather than a separate pathway-biased mechanism. The metabolic selectivity of partial IR agonists like XMetA, if recapitulated in vivo, may be a desirable feature of therapeutic agents designed to regulate blood glucose levels while minimizing undesirable outcomes of excessive IR mitogenic activation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor, Insulin/metabolism , Allosteric Regulation , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Cricetulus , Drug Partial Agonism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Phosphorylation , Protein Binding , Protein Isoforms/agonists , Protein Isoforms/immunology , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/agonists , Receptor, Insulin/immunology , Signal Transduction
7.
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
8.
PLoS One ; 9(2): e88684, 2014.
Article in English | MEDLINE | ID: mdl-24533136

ABSTRACT

Previously we reported studies of XMetA, an agonist antibody to the insulin receptor (INSR). We have now utilized phage display to identify XMetS, a novel monoclonal antibody to the INSR. Biophysical studies demonstrated that XMetS bound to the human and mouse INSR with picomolar affinity. Unlike monoclonal antibody XMetA, XMetS alone had little or no agonist effect on the INSR. However, XMetS was a strong positive allosteric modulator of the INSR that increased the binding affinity for insulin nearly 20-fold. XMetS potentiated insulin-stimulated INSR signaling ∼15-fold or greater including; autophosphorylation of the INSR, phosphorylation of Akt, a major enzyme in the metabolic pathway, and phosphorylation of Erk, a major enzyme in the growth pathway. The enhanced signaling effects of XMetS were more pronounced with Akt than with Erk. In cultured cells, XMetS also enhanced insulin-stimulated glucose transport. In contrast to its effects on the INSR, XMetS did not potentiate IGF-1 activation of the IGF-1 receptor. We studied the effect of XMetS treatment in two mouse models of insulin resistance and diabetes. The first was the diet induced obesity mouse, a hyperinsulinemic, insulin resistant animal, and the second was the multi-low dose streptozotocin/high-fat diet mouse, an insulinopenic, insulin resistant animal. In both models, XMetS normalized fasting blood glucose levels and glucose tolerance. In concert with its ability to potentiate insulin action at the INSR, XMetS reduced insulin and C-peptide levels in both mouse models. XMetS improved the response to exogenous insulin without causing hypoglycemia. These data indicate that an allosteric monoclonal antibody can be generated that markedly enhances the binding affinity of insulin to the INSR. These data also suggest that an INSR monoclonal antibody with these characteristics may have the potential to both improve glucose metabolism in insulinopenic type 2 diabetes mellitus and correct compensatory hyperinsulinism in insulin resistant conditions.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens, CD/metabolism , Glucose/metabolism , Receptor, Insulin/metabolism , Allosteric Site , Animals , C-Peptide/chemistry , CHO Cells , Cell Separation , Cricetinae , Cricetulus , Diabetes Mellitus, Type 2/metabolism , Flow Cytometry , Humans , Hyperglycemia/metabolism , Hyperinsulinism/metabolism , Insulin/chemistry , Insulin Resistance , Mice , Mice, Inbred C57BL , Obesity/metabolism , Peptide Library , Phosphorylation , Protein Structure, Tertiary , Signal Transduction
9.
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
10.
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
11.
PLoS One ; 7(3): e33640, 2012.
Article in English | MEDLINE | ID: mdl-22479423

ABSTRACT

The second messenger lipid PIP(3) (phosphatidylinositol-3,4,5-trisphosphate) is generated by the lipid kinase PI3K (phosphoinositide-3-kinase) in the inner leaflet of the plasma membrane, where it regulates a broad array of cell processes by recruiting multiple signaling proteins containing PIP(3)-specific pleckstrin homology (PH) domains to the membrane surface. Despite the broad importance of PIP(3)-specific PH domains, the membrane docking geometry of a PH domain bound to its target PIP(3) lipid on a bilayer surface has not yet been experimentally determined. The present study employs EPR site-directed spin labeling and relaxation methods to elucidate the membrane docking geometry of GRP1 PH domain bound to bilayer-embedded PIP(3). The model target bilayer contains the neutral background lipid PC and both essential targeting lipids: (i) PIP(3) target lipid that provides specificity and affinity, and (ii) PS facilitator lipid that enhances the PIP(3) on-rate via an electrostatic search mechanism. The EPR approach measures membrane depth parameters for 18 function-retaining spin labels coupled to the PH domain, and for calibration spin labels coupled to phospholipids. The resulting depth parameters, together with the known high resolution structure of the co-complex between GRP1 PH domain and the PIP(3) headgroup, provide sufficient constraints to define an optimized, self-consistent membrane docking geometry. In this optimized geometry the PH domain engulfs the PIP(3) headgroup with minimal bilayer penetration, yielding the shallowest membrane position yet described for a lipid binding domain. This binding interaction displaces the PIP(3) headgroup from its lowest energy position and orientation in the bilayer, but the headgroup remains within its energetically accessible depth and angular ranges. Finally, the optimized docking geometry explains previous biophysical findings including mutations observed to disrupt membrane binding, and the rapid lateral diffusion observed for PIP(3)-bound GRP1 PH domain on supported lipid bilayers.


Subject(s)
Cell Membrane/chemistry , Lipid Bilayers/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Cell Membrane/metabolism , Electron Spin Resonance Spectroscopy , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Lipid Bilayers/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
12.
Diabetes ; 61(5): 1263-71, 2012 May.
Article in English | MEDLINE | ID: mdl-22403294

ABSTRACT

Many patients with diabetes mellitus (both type 1 and type 2) require therapy to maintain normal fasting glucose levels. To develop a novel treatment for these individuals, we used phage display technology to target the insulin receptor (INSR) complexed with insulin and identified a high affinity, allosteric, human monoclonal antibody, XMetA, which mimicked the glucoregulatory, but not the mitogenic, actions of insulin. Biophysical studies with cultured cells expressing human INSR demonstrated that XMetA acted allosterically and did not compete with insulin for binding to its receptor. XMetA was found to function as a specific partial agonist of INSR, eliciting tyrosine phosphorylation of INSR but not the IGF-IR. Although this antibody activated metabolic signaling, leading to enhanced glucose uptake, it neither activated Erk nor induced proliferation of cancer cells. In an insulin resistant, insulinopenic model of diabetes, XMetA markedly reduced elevated fasting blood glucose and normalized glucose tolerance. After 6 weeks, significant improvements in HbA(1c), dyslipidemia, and other manifestations of diabetes were observed. It is noteworthy that hypoglycemia and weight gain were not observed during these studies. These studies indicate, therefore, that allosteric monoclonal antibodies have the potential to be novel, ultra-long acting, agents for the regulation of hyperglycemia in diabetes.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Glucose/physiology , Diabetes Mellitus, Experimental/therapy , Receptor, Insulin/agonists , Animals , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Biomarkers , CHO Cells , Cells, Cultured , Cricetinae , Glucose Tolerance Test , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred ICR , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Specific Pathogen-Free Organisms
13.
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
14.
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
15.
Anal Biochem ; 386(2): 194-216, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19133223

ABSTRACT

To explore the variability in biosensor studies, 150 participants from 20 countries were given the same protein samples and asked to determine kinetic rate constants for the interaction. We chose a protein system that was amenable to analysis using different biosensor platforms as well as by users of different expertise levels. The two proteins (a 50-kDa Fab and a 60-kDa glutathione S-transferase [GST] antigen) form a relatively high-affinity complex, so participants needed to optimize several experimental parameters, including ligand immobilization and regeneration conditions as well as analyte concentrations and injection/dissociation times. Although most participants collected binding responses that could be fit to yield kinetic parameters, the quality of a few data sets could have been improved by optimizing the assay design. Once these outliers were removed, the average reported affinity across the remaining panel of participants was 620 pM with a standard deviation of 980 pM. These results demonstrate that when this biosensor assay was designed and executed appropriately, the reported rate constants were consistent, and independent of which protein was immobilized and which biosensor was used.


Subject(s)
Biosensing Techniques/methods , Proteins/analysis , Antibodies, Catalytic/analysis , Benchmarking , Binding Sites , Biosensing Techniques/statistics & numerical data , Glutathione Transferase/analysis , Kinetics , Ligands
16.
Dis Aquat Organ ; 74(1): 77-9; discussion 81-3, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-17425266

ABSTRACT

It is the opinion of the authors of the Comment on Do et al. (2006), that those authors incorrectly interpreted their test results, which are more likely the result of mislabeling of samples or within-laboratory contamination, and that the TSV isolates found in Penaeus vannamei in Korea in 2004 and 2005 did not originate from Hawaii as claimed by the authors, but from a country (or countries) in southeast Asia. Finally, we believe that the authors did not follow proper international guidelines, extend a professional courtesy to the supplier of the disputed shrimp sample, nor take a critical approach in interpreting their own data. It is unfortunate that the authors did not follow through with additional testing, or seek a second opinion from an independent laboratory, before implicating shrimp imported from Hawaii as the source of TSV in Korea.


Subject(s)
Penaeidae/virology , RNA Viruses/classification , RNA Viruses/pathogenicity , Animals , Aquaculture/methods , Breeding , France , Geography , Hawaii , Specific Pathogen-Free Organisms
17.
Biochemistry ; 46(14): 4322-36, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17367165

ABSTRACT

The C2 domain is a ubiquitous, conserved protein signaling motif widely found in eukaryotic signaling proteins. Although considerable functional diversity exists, most C2 domains are activated by Ca2+ binding and then dock to a specific cellular membrane. The C2 domains of protein kinase Calpha (PKCalpha) and cytosolic phospholipase A2alpha (cPLA2alpha), for example, are known to dock to different membrane surfaces during an intracellular Ca2+ signal. Ca2+ activation targets the PKCalpha C2 domain to the plasma membrane and the cPLA2alpha C2 domain to the internal membranes, with no detectable spatial overlap. It is crucial to determine how such targeting specificity is achieved at physiological bulk Ca2+ concentrations that during a typical signaling event rarely exceed 1 muM. For the isolated PKCalpha C2 domain in the presence of physiological Ca2+ levels, the target lipids phosphatidylserine (PS) and phosphatidylinositol-4,5-bisphosphate (PIP2) are together sufficient to recruit the PKCalpha C2 domain to a lipid mixture mimicking the plasma membrane inner leaflet. For the cPLA2alpha C2 domain, the target lipid phosphatidylcholine (PC) appears to be sufficient to drive membrane targeting to an internal membrane mimic at physiological Ca2+ levels, although the results do not rule out a second, unknown target molecule. Stopped-flow kinetic studies provide additional information about the fundamental molecular events that occur during Ca2+-activated membrane docking. In principle, C2 domain-directed intracellular targeting, which requires coincidence detection of multiple signals (Ca2+ and one or more target lipids), can exhibit two different mechanisms: messenger-activated target affinity (MATA) and target-activated messenger affinity (TAMA). The C2 domains studied here both utilize the TAMA mechanism, in which the C2 domain Ca2+ affinity is too low to be activated by physiological Ca2+ signals in most regions of the cell. Only when the C2 domain nears its target membrane, which provides a high local concentration of target lipid, is the effective Ca2+ affinity increased by the coupled binding equilibrium to a level that enables substantial Ca2+ activation and target docking. Overall, the findings emphasize the importance of using physiological ligand concentrations in targeting studies because super-physiological concentrations can drive docking interactions even when an important targeting molecule is missing.


Subject(s)
Calcium/metabolism , Cell Membrane/chemistry , Lipid Metabolism , Phospholipases A/chemistry , Protein Kinase C-alpha/chemistry , Animals , Calcium/physiology , Calcium Signaling , Cell Line , Cell Membrane/metabolism , Cytosol/enzymology , Fluorescent Dyes/metabolism , Glutathione Transferase/metabolism , Kinetics , Macrophages/enzymology , Mice , Phosphatidylcholines/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylserines/metabolism , Phospholipases A/genetics , Phospholipases A/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Spectrometry, Fluorescence
18.
Biochemistry ; 43(51): 16161-73, 2004 Dec 28.
Article in English | MEDLINE | ID: mdl-15610010

ABSTRACT

Pleckstrin homology (PH) domains play a central role in a wide array of signaling pathways by binding second messenger lipids of the phosphatidylinositol phosphate (PIP) lipid family. A given type of PIP lipid is formed in a specific cellular membrane where it is generally a minor component of the bulk lipid mixture. For example, the signaling lipid PI(3,4,5)P(3) (or PIP(3)) is generated primarily in the inner leaflet of the plasma membrane where it is believed to never exceed 0.02% of the bulk lipid. The present study focuses on the PH domain of the general receptor for phosphoinositides, isoform 1 (GRP1), which regulates the actin cytoskeleton in response to PIP(3) signals at the plasma membrane surface. The study systematically analyzes both the equilibrium and kinetic features of GRP1-PH domain binding to its PIP lipid target on a bilayer surface. Equilibrium binding measurements utilizing protein-to-membrane fluorescence resonance energy transfer (FRET) to detect GRP1-PH domain docking to membrane-bound PIP lipids confirm specific binding to PIP(3). A novel FRET competitive binding measurement developed to quantitate docking affinity yields a K(D) of 50 +/- 10 nM for GRP1-PH domain binding to membrane-bound PIP(3) in a physiological lipid mixture approximating the composition of the plasma membrane inner leaflet. This observed K(D) lies in a suitable range for regulation by physiological PIP(3) signals. Interestingly, the affinity of the interaction decreases at least 12-fold when the background anionic lipids phosphatidylserine (PS) and phosphatidylinositol (PI) are removed from the lipid mixture. Stopped-flow kinetic studies using protein-to-membrane FRET to monitor association and dissociation time courses reveal that this affinity decrease arises from a corresponding decrease in the on-rate for GRP1-PH domain docking with little or no change in the off-rate for domain dissociation from membrane-bound PIP(3). Overall, these findings indicate that the PH domain interacts not only with its target lipid, but also with other features of the membrane surface. The results are consistent with a previously undescribed type of two-step search mechanism for lipid binding domains in which weak, nonspecific electrostatic interactions between the PH domain and background anionic lipids facilitate searching of the membrane surface for PIP(3) headgroups, thereby speeding the high-affinity, specific docking of the domain to its rare target lipid.


Subject(s)
Lipid Metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Blood Proteins/genetics , Data Interpretation, Statistical , Fluorescence Resonance Energy Transfer , Humans , Kinetics , Phosphoproteins/genetics , Protein Binding , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/isolation & purification , Sequence Homology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...