Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 415: 143-56, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26277398

ABSTRACT

Insulin acts as the major regulator of the fasting-to-fed metabolic transition by altering substrate metabolism, promoting energy storage, and helping activate protein synthesis. In addition to its glucoregulatory and other metabolic properties, insulin can also act as a growth factor. The metabolic and mitogenic responses to insulin are regulated by divergent post-receptor signaling mechanisms downstream from the activated insulin receptor (IR). However, the anabolic and growth-promoting properties of insulin require tissue-specific inter-relationships between the two pathways, and the nature and scope of insulin-regulated processes vary greatly across tissues. Understanding the nuances of this interplay between metabolic and growth-regulating properties of insulin would have important implications for development of novel insulin and IR modulator therapies that stimulate insulin receptor activation in both pathway- and tissue-specific manners. This review will provide a unique perspective focusing on the roles of "metabolic" and "mitogenic" actions of insulin signaling in various tissues, and how these networks should be considered when evaluating selective pharmacologic approaches to prevent or treat metabolic disease.


Subject(s)
Insulin/metabolism , MAP Kinase Signaling System , Humans , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Organ Specificity , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/agonists , Receptor, Insulin/metabolism
2.
J Cell Biochem ; 116(9): 2109-19, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25808283

ABSTRACT

It has been proposed that monoclonal antibodies may become therapeutics for metabolic diseases such as diabetes mellitus. We have previously characterized an allosteric monoclonal antibody to the human insulin receptor (IR), XMetA, that activated metabolic signaling leading to enhanced glucose transport in cultured cells, and chronically reduced fasting blood glucose levels in mouse models of diabetes mellitus. Under acute dosing conditions, the large size of an IR-binding antibody like XMetA (∼ 150 kDa) could lead to a more rapid access into liver, an insulin sensitive tissue with well-fenestrated capillaries, when compared to other insulin sensitive tissues with non-fenestrated capillaries, such as muscle and adipose. Thus, in the present study we administered XMetA (10 mg/kg) and insulin (0.5 U/kg) via IV injection, and for 90 min compared their effects on blood glucose lowering and IR activation in three of the major insulin-sensitive tissues of the normal fasted mouse: liver, adipose, and muscle. Like insulin, XMetA lowered blood glucose levels, although the effect was less rapid. Insulin activated IR autophosphorylation and Akt phosphorylation in liver, fat, and muscle. In contrast, IR activation by XMetA was primarily observed in the liver. Both insulin and XMetA lowered ß-hydroxybutyrate levels in plasma; however, only insulin reduced both non-esterified fatty acids (NEFA) and glycerol concentrations. These data indicate that, in normal mice, acute glucose regulation by XMetA is largely mediated by its action on the liver.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Blood Glucose/drug effects , Insulin/administration & dosage , Liver/metabolism , Receptor, Insulin/agonists , 3-Hydroxybutyric Acid/blood , Adipose Tissue/metabolism , Animals , Antibodies, Monoclonal/pharmacology , CHO Cells , Cricetulus , Humans , Injections, Intravenous , Insulin/pharmacology , Male , Mice , Muscles/metabolism , Organ Specificity , Phosphorylation/drug effects , Receptor, Insulin/metabolism
3.
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
4.
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
5.
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
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 Immunol Methods ; 391(1-2): 60-71, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23454004

ABSTRACT

Phage display antibody libraries have a proven track record for the discovery of therapeutic human antibodies, increasing the demand for large and diverse phage antibody libraries for the discovery of new therapeutics. We have constructed naïve antibody phage display libraries in both Fab and scFv formats, with each library having more than 250 billion clones that encompass the human antibody repertoire. These libraries show high fidelity in open reading frame and expression percentages, and their V-gene family distribution, VH-CDR3 length and amino acid usage mirror the natural diversity of human antibodies. Both the Fab and scFv libraries show robust sequence diversity in target-specific binders and differential V-gene usage for each target tested, supporting the use of libraries that utilize multiple display formats and V-gene utilization to maximize antibody-binding diversity. For each of the targets, clones with picomolar affinities were identified from at least one of the libraries and for the two targets assessed for activity, functional antibodies were identified from both libraries.


Subject(s)
Cell Surface Display Techniques , Immunoglobulin Fab Fragments/immunology , Peptide Library , Receptor, Insulin/immunology , Receptor, TIE-2/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antibody Affinity , Antibody Specificity , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/genetics , Mitogen-Activated Protein Kinases/metabolism , Open Reading Frames , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/genetics , Receptor, TIE-2/genetics , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Transfection
8.
Am J Physiol Endocrinol Metab ; 304(11): E1175-87, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23512805

ABSTRACT

Elevated blood branched-chain amino acids (BCAA) are often associated with insulin resistance and type 2 diabetes, which might result from a reduced cellular utilization and/or incomplete BCAA oxidation. White adipose tissue (WAT) has become appreciated as a potential player in whole body BCAA metabolism. We tested if expression of the mitochondrial BCAA oxidation checkpoint, branched-chain α-ketoacid dehydrogenase (BCKD) complex, is reduced in obese WAT and regulated by metabolic signals. WAT BCKD protein (E1α subunit) was significantly reduced by 35-50% in various obesity models (fa/fa rats, db/db mice, diet-induced obese mice), and BCKD component transcripts significantly lower in subcutaneous (SC) adipocytes from obese vs. lean Pima Indians. Treatment of 3T3-L1 adipocytes or mice with peroxisome proliferator-activated receptor-γ agonists increased WAT BCAA catabolism enzyme mRNAs, whereas the nonmetabolizable glucose analog 2-deoxy-d-glucose had the opposite effect. The results support the hypothesis that suboptimal insulin action and/or perturbed metabolic signals in WAT, as would be seen with insulin resistance/type 2 diabetes, could impair WAT BCAA utilization. However, cross-tissue flux studies comparing lean vs. insulin-sensitive or insulin-resistant obese subjects revealed an unexpected negligible uptake of BCAA from human abdominal SC WAT. This suggests that SC WAT may not be an important contributor to blood BCAA phenotypes associated with insulin resistance in the overnight-fasted state. mRNA abundances for BCAA catabolic enzymes were markedly reduced in omental (but not SC) WAT of obese persons with metabolic syndrome compared with weight-matched healthy obese subjects, raising the possibility that visceral WAT contributes to the BCAA metabolic phenotype of metabolically compromised individuals.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Adipose Tissue, White/metabolism , Amino Acids, Branched-Chain/metabolism , Obesity/metabolism , Adipocytes/metabolism , Adult , Animals , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Insulin/blood , Mice , Mice, Obese , Middle Aged , Rats , Rats, Zucker
9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...