Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 63: 116743, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35436748

ABSTRACT

The voltage-gated sodium channel Nav1.7 is an attractive target for the treatment of pain based on the high level of target validation with genetic evidence linking Nav1.7 to pain in humans. Our effort to identify selective, CNS-penetrant Nav1.7 blockers with oral activity, improved selectivity, good drug-like properties, and safety led to the discovery of 2-substituted quinolines and quinolones as potent small molecule Nav1.7 blockers. The design of these molecules focused on maintaining potency at Nav1.7, improving selectivity over the hERG channel, and overcoming phospholipidosis observed with the initial leads. The structure-activity relationship (SAR) studies leading to the discovery of (R)-(3-fluoropyrrolidin-1-yl)(6-((5-(trifluoromethyl)pyridin-2-yl)oxy)quinolin-2-yl)methanone (ABBV-318) are described herein. ABBV-318 displayed robust in vivo efficacy in both inflammatory and neuropathic rodent models of pain. ABBV-318 also inhibited Nav1.8, another sodium channel isoform that is an active target for the development of new pain treatments.


Subject(s)
Pain , Sodium Channels , Humans , Pain/drug therapy , Pain Management , Protein Isoforms , Sodium Channels/metabolism , Structure-Activity Relationship
2.
J Med Chem ; 59(7): 3373-91, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27015369

ABSTRACT

The genetic validation for the role of the Nav1.7 voltage-gated ion channel in pain signaling pathways makes it an appealing target for the potential development of new pain drugs. The utility of nonselective Nav blockers is often limited due to adverse cardiovascular and CNS side effects. We sought more selective Nav1.7 blockers with oral activity, improved selectivity, and good druglike properties. The work described herein focused on a series of 3- and 4-substituted indazoles. SAR studies of 3-substituted indazoles yielded analog 7 which demonstrated good in vitro and in vivo activity but poor rat pharmacokinetics. Optimization of 4-substituted indazoles yielded two compounds, 27 and 48, that exhibited good in vitro and in vivo activity with improved rat pharmacokinetic profiles. Both 27 and 48 demonstrated robust activity in the acute rat monoiodoacetate-induced osteoarthritis model of pain, and subchronic dosing of 48 showed a shift to a lower EC50 over 7 days.


Subject(s)
Analgesics/pharmacology , Imidazolidines/pharmacology , Indazoles/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Osteoarthritis/drug therapy , Pain/drug therapy , Pyrroles/pharmacology , Sodium Channel Blockers/pharmacology , Analgesics/chemistry , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Electrophysiology , Evoked Potentials , Imidazolidines/chemistry , Indazoles/chemistry , Iodoacetic Acid/toxicity , Molecular Structure , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Osteoarthritis/chemically induced , Osteoarthritis/metabolism , Pain/metabolism , Pain/pathology , Pain Measurement , Pyrroles/chemistry , Rats , Sodium Channel Blockers/chemistry , Structure-Activity Relationship
3.
Purinergic Signal ; 8(Suppl 1): 41-56, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086553

ABSTRACT

Enhanced sensitivity to noxious stimuli and the perception of non-noxious stimuli as painful are hallmark sensory perturbations associated with chronic pain. It is now appreciated that ATP, through its actions as an excitatory neurotransmitter, plays a prominent role in the initiation and maintenance of chronic pain states. Mechanistically, the ability of ATP to drive nociceptive sensitivity is mediated through direct interactions at neuronal P2X3 and P2X2/3 receptors. Extracellular ATP also activates P2X4, P2X7, and several P2Y receptors on glial cells within the spinal cord, which leads to a heightened state of neural-glial cell interaction in ongoing pain states. Following the molecular identification of the P2 receptor superfamilies, selective small molecule antagonists for several P2 receptor subtypes were identified, which have been useful for investigating the role of specific P2X receptors in preclinical chronic pain models. More recently, several P2X receptor antagonists have advanced into clinical trials for inflammation and pain. The development of orally bioavailable blockers for ion channels, including the P2X receptors, has been traditionally difficult due to the necessity of combining requirements for target potency and selectivity with suitable absorption distribution, metabolism, and elimination properties. Recent studies on the physicochemical properties of marketed orally bioavailable drugs, have identified several parameters that appear critical for increasing the probability of achieving suitable bioavailability, central nervous system exposure, and acceptable safety necessary for clinical efficacy. This review provides an overview of the antinociceptive pharmacology of P2X receptor antagonists and the chemical diversity and drug-like properties for emerging antagonists of P2X3, P2X2/3, P2X4, and P2X7 receptors.

4.
ACS Chem Biol ; 6(3): 234-44, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21090814

ABSTRACT

Inhibition of protein kinases has validated therapeutic utility for cancer, with at least seven kinase inhibitor drugs on the market. Protein kinase inhibition also has significant potential for a variety of other diseases, including diabetes, pain, cognition, and chronic inflammatory and immunologic diseases. However, as the vast majority of current approaches to kinase inhibition target the highly conserved ATP-binding site, the use of kinase inhibitors in treating nononcology diseases may require great selectivity for the target kinase. As protein kinases are signal transducers that are involved in binding to a variety of other proteins, targeting alternative, less conserved sites on the protein may provide an avenue for greater selectivity. Here we report an affinity-based, high-throughput screening technique that allows nonbiased interrogation of small molecule libraries for binding to all exposed sites on a protein surface. This approach was used to screen both the c-Jun N-terminal protein kinase Jnk-1 (involved in insulin signaling) and p38α (involved in the formation of TNFα and other cytokines). In addition to canonical ATP-site ligands, compounds were identified that bind to novel allosteric sites. The nature, biological relevance, and mode of binding of these ligands were extensively characterized using two-dimensional (1)H/(13)C NMR spectroscopy, protein X-ray crystallography, surface plasmon resonance, and direct enzymatic activity and activation cascade assays. Jnk-1 and p38α both belong to the MAP kinase family, and the allosteric ligands for both targets bind similarly on a ledge of the protein surface exposed by the MAP insertion present in the CMGC family of protein kinases and distant from the active site. Medicinal chemistry studies resulted in an improved Jnk-1 ligand able to increase adiponectin secretion in human adipocytes and increase insulin-induced protein kinase PKB phosphorylation in human hepatocytes, in similar fashion to Jnk-1 siRNA and to rosiglitazone treatment. Together, the data suggest that these new ligand series bind to a novel, allosteric, and physiologically relevant site and therefore represent a unique approach to identify kinase inhibitors.


Subject(s)
Drug Discovery , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Binding Sites/drug effects , Crystallography, X-Ray , High-Throughput Screening Assays , Humans , Mitogen-Activated Protein Kinase 8/chemistry , Mitogen-Activated Protein Kinase 8/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Bioorg Med Chem Lett ; 17(3): 668-72, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17107797

ABSTRACT

A new series of 4-anilinopyrimidines has been synthesized and evaluated as JNK1 inhibitors. SAR studies led to the discovery of potent JNK1 inhibitors with good enzymatic activity as well as cellular potency represented by compound 2b. Kinase selectivity profile and the crystal structure of 2b are also described.


Subject(s)
Aniline Compounds/chemical synthesis , Aniline Compounds/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Crystallography, X-Ray , Indicators and Reagents , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 16(22): 5723-30, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16971120

ABSTRACT

The structure-activity relationships of 5,6-positions of aminopyridine carboxamide-based c-Jun N-terminal Kinase (JNK) inhibitors were explored to expand interaction with the kinase specificity and ribose-binding pockets. The syntheses of analogues and the impact of structural modification on in vitro potency and cellular activity are described.


Subject(s)
Amides/pharmacology , Aminopyridines/pharmacology , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Amides/chemistry , Aminopyridines/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Inhibitory Concentration 50 , Protein Binding , Ribose/metabolism , Structure-Activity Relationship , Substrate Specificity
7.
J Med Chem ; 49(15): 4455-8, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16854050

ABSTRACT

C-Jun NH2 terminal kinases (JNKs) are important cell signaling enzymes. JNK1 plays a central role in linking obesity and insulin resistance. JNK2 and JNK3 may be involved in inflammatory and neurological disorders, respectively. Small-molecule JNK inhibitors could be valuable tools to study the therapeutic benefits of inhibiting these enzymes and as leads for potential drugs targeting JNKs. In this report, we disclose a series of potent and highly selective JNK inhibitors with good pharmacokinetic profiles.


Subject(s)
Amides/chemical synthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pyridines/chemical synthesis , Administration, Oral , Amides/pharmacokinetics , Amides/pharmacology , Animals , Biological Availability , Crystallography, X-Ray , Humans , In Vitro Techniques , Mice , Microsomes/metabolism , Models, Molecular , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Structure-Activity Relationship , Thermodynamics
8.
J Med Chem ; 49(12): 3563-80, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16759099

ABSTRACT

The c-Jun N-terminal kinases (JNK-1, -2, and -3) are members of the mitogen activated protein (MAP) kinase family of enzymes. They are activated in response to certain cytokines, as well as by cellular stresses including chemotoxins, peroxides, and irradiation. They have been implicated in the pathology of a variety of different diseases with an inflammatory component including asthma, stroke, Alzheimer's disease, and type 2 diabetes mellitus. In this work, high-throughput screening identified a JNK inhibitor with an excellent kinase selectivity profile. Using X-ray crystallography and biochemical screening to guide our lead optimization, we prepared compounds with inhibitory potencies in the low-double-digit nanomolar range, activity in whole cells, and pharmacokinetics suitable for in vivo use. The new compounds were over 1,000-fold selective for JNK-1 and -2 over other MAP kinases including ERK2, p38alpha, and p38delta and showed little inhibitory activity against a panel of 74 kinases.


Subject(s)
Aminopyridines/chemical synthesis , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Aminopyridines/chemistry , Aminopyridines/pharmacology , Animals , Biological Availability , Cell Line, Tumor , Crystallography, X-Ray , Half-Life , Humans , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/chemistry , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Models, Molecular , Phosphorylation , Protein Conformation , Rats , Rats, Sprague-Dawley
9.
Bioorg Med Chem Lett ; 16(10): 2590-4, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16527482

ABSTRACT

A novel class of 1,9-dihydro-9-hydroxypyrazolo[3,4-b]quinolin-4-ones as c-Jun-N-terminal kinase (JNK) inhibitors is described. These compounds were synthesized via the condensation of 2-nitrobenzaldehydes and hydroxypyrazoles. The structure-activity relationships (SAR) and kinase selectivity profile of the inhibitors are also discussed. Compound 16 was identified as a potent JNK inhibitor with good cellular potency.


Subject(s)
Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Quinolones/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Molecular Structure , Quinolones/chemistry , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 16(7): 1807-10, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16446092

ABSTRACT

A series of novel benzoxazole benzenesulfonamides was synthesized as inhibitors of fructose-1,6-bisphosphatase (FBPase-1). Extensive SAR studies led to a potent inhibitor, 53, with an IC(50) of 0.57microM. Compound 17 exhibited excellent bioavailability and a good pharmacokinetic profile in rats.


Subject(s)
Enzyme Inhibitors/pharmacology , Fructose-Bisphosphatase/antagonists & inhibitors , Sulfonamides/pharmacology , Allosteric Regulation , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Models, Molecular , Rats , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics
11.
Bioorg Med Chem Lett ; 16(7): 1811-5, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16442285

ABSTRACT

We have identified benzoxazole benzenesulfonamide 1 as a novel allosteric inhibitor of fructose-1,6-bisphosphatase (FBPase-1). X-ray crystallographic and biological studies of 1 indicate a distinct binding mode that recapitulates features of several previously reported FBPase-1 inhibitor classes.


Subject(s)
Benzoxazoles/chemistry , Enzyme Inhibitors/pharmacology , Fructose-Bisphosphatase/antagonists & inhibitors , Sulfonamides/pharmacology , Allosteric Regulation , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Fructose-Bisphosphatase/metabolism , Models, Molecular , Protein Binding , Sulfonamides/chemistry , Sulfonamides/metabolism
12.
Biochem Biophys Res Commun ; 323(2): 652-9, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15369800

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) inhibition increases insulin sensitivity and normalizes blood glucose levels in animals. The molecular events associated with PTP1B inhibition that increase insulin sensitivity remain controversial. Insulin resistant, diabetic ob/ob mice, dosed with PTP1B antisense for 3 weeks exhibited a decrease in PTP1B protein levels and a change in the expression level of p85alpha isoforms in liver, characterized by a reduction in p85alpha and an upregulation of the p50alpha and p55alpha isoforms. Transfection of mouse hepatocytes with PTP1B antisense caused a downregulation PTP1B and p85alpha protein levels. Furthermore, transfection of mouse hepatocytes with PTP1B siRNA downregulated p85alpha protein expression and enhanced insulin-induced PKB phosphorylation. Treatment of mouse hepatocytes with p85alpha antisense oligonucleotide caused a reduction of p85alpha and an increase in p50alpha and p55alpha isoforms and enhanced insulin-stimulated PKB activation. These results demonstrate that PTP1B inhibition causes a direct differential regulation of p85alpha isoforms of PI3-kinase in liver and that reduction of p85alpha may be one mechanism by which PTP1B inhibition improves insulin sensitivity and glucose metabolism in insulin-resistant states.


Subject(s)
Adipose Tissue/enzymology , Gene Expression Regulation, Enzymologic/physiology , Hepatocytes/metabolism , Liver/enzymology , Oligoribonucleotides, Antisense/administration & dosage , Phosphatidylinositol 3-Kinases/metabolism , Protein Tyrosine Phosphatases/deficiency , Animals , Gene Silencing , Isoenzymes/metabolism , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Transfection/methods
13.
Mol Cell Endocrinol ; 203(1-2): 155-68, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12782412

ABSTRACT

Protein tyrosine phosphatases are important regulators of insulin signal transduction. Our studies have shown that in insulin resistant and diabetic ob/ob and db/db mice, reducing the levels of protein tyrosine phosphatase 1B (PTP1B) protein by treatment with a PTP1B antisense oligonucleotide resulted in improved insulin sensitivity and normalized plasma glucose levels. The mechanism by which PTP1B inhibition improves insulin sensitivity is not fully understood. We have used microarray analysis to compare gene expression changes in adipose tissue, liver and muscle of PTP1B antisense-treated ob/ob mice. Our results show that treatment with PTP1B antisense resulted in the downregulation of genes involved in lipogenesis in both fat and liver, and a downregulation of genes involved in adipocyte differentiation in fat, suggesting that PTP1B antisense acts through a different mechanism than thiazolidinedione (TZD) treatment. In summary, microarray results suggest that reduction of PTP1B may alleviate hyperglycemia and enhance insulin sensitivity by a different mechanism than TZD treatment.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Lipids/biosynthesis , Liver/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Tyrosine Phosphatases/physiology , Adipose Tissue/cytology , Animals , Blood Glucose/drug effects , Cell Differentiation/drug effects , Down-Regulation/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Insulin Resistance , Mice , Mice, Obese , Muscles/metabolism , Oligonucleotides, Antisense/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/antagonists & inhibitors
14.
Mol Endocrinol ; 17(6): 1131-43, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12649327

ABSTRACT

Phosphorylation of stress-activated kinase p38, a MAPK family member, was increased in liver of ob/ob diabetic mice relative to lean littermates. Treatment of ob/ob mice with protein tyrosine phosphatase 1B (PTP1B) antisense oligonucleotides (ASO) reduced phosphorylation of p38 in liver-to below lean littermate levels-and normalized plasma glucose while reducing plasma insulin. Phosphorylation of ERK, but not JNK, was also decreased in ASO-treated mice. PTP1B ASO decreased TNFalpha protein levels and phosphorylation of the transcription factor cAMP response element binding protein (CREB) in liver, both of which can occur through decreased phosphorylation of p38 and both of which have been implicated in insulin resistance or hyperglycemia. Decreased p38 phosphorylation was not directly due to decreased phosphorylation of the kinases that normally phosphorylate p38-MKK3 and MKK6. Additionally, p38 phosphorylation was not enhanced in liver upon insulin stimulation of ASO-treated ob/ob mice (despite increased activation of other signaling molecules) corroborating that p38 is not directly affected via the insulin receptor. Instead, decreased phosphorylation of p38 may be due to increased expression of MAPK phosphatases, particularly the p38/ERK phosphatase PAC1 (phosphatase of activated cells). This study demonstrates that reduction of PTP1B protein using ASO reduces activation of p38 and its substrates TNFalpha and CREB in liver of diabetic mice, which correlates with decreased hyperglycemia and hyperinsulinemia.


Subject(s)
Diabetes Mellitus/metabolism , Liver/metabolism , Mitogen-Activated Protein Kinases/metabolism , Obesity , Oligonucleotides, Antisense/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Blood Glucose/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dual Specificity Phosphatase 2 , Insulin/metabolism , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Phosphatase 2 , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Random Allocation , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
15.
Diabetes ; 52(1): 21-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502489

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of insulin receptor (IR) signal transduction and a drug target for treatment of type 2 diabetes. Using PTP1B antisense oligonucleotides (ASOs), effects of decreased PTP1B levels on insulin signaling in diabetic ob/ob mice were examined. Insulin stimulation, prior to sacrifice, resulted in no significant activation of insulin signaling pathways in livers from ob/ob mice. However, in PTP1B ASO-treated mice, in which PTP1B protein was decreased by 60% in liver, similar stimulation with insulin resulted in increased tyrosine phosphorylation of the IR and IR substrate (IRS)-1 and -2 by threefold, fourfold, and threefold, respectively. IRS-2-associated phosphatidylinositol 3-kinase activity was also increased threefold. Protein kinase B (PKB) serine phosphorylation was increased sevenfold in liver of PTP1B ASO-treated mice upon insulin stimulation, while phosphorylation of PKB substrates, glycogen synthase kinase (GSK)-3alpha and -3beta, was increased more than twofold. Peripheral insulin signaling was increased by PTP1B ASO, as evidenced by increased phosphorylation of PKB in muscle of insulin-stimulated PTP1B ASO-treated animals despite the lack of measurable effects on muscle PTP1B protein. These results indicate that reduction of PTP1B is sufficient to increase insulin-dependent metabolic signaling and improve insulin sensitivity in a diabetic animal model.


Subject(s)
Diabetes Mellitus/physiopathology , Insulin/physiology , Obesity , Protein Serine-Threonine Kinases , Protein Tyrosine Phosphatases/antagonists & inhibitors , Signal Transduction/physiology , Animals , Blood Glucose/analysis , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin/blood , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Liver/enzymology , Mice , Mice, Inbred C57BL/genetics , Muscle, Skeletal/enzymology , Oligonucleotides, Antisense/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/metabolism , Serine/metabolism , Tyrosine/metabolism
16.
Proc Natl Acad Sci U S A ; 99(17): 11357-62, 2002 Aug 20.
Article in English | MEDLINE | ID: mdl-12169659

ABSTRACT

The role of protein-tyrosine phosphatase 1B (PTP1B) in diabetes was investigated using an antisense oligonucleotide in ob/ob and db/db mice. PTP1B antisense oligonucleotide treatment normalized plasma glucose levels, postprandial glucose excursion, and HbA(1C). Hyperinsulinemia was also reduced with improved insulin sensitivity. PTP1B protein and mRNA were reduced in liver and fat with no effect in skeletal muscle. Insulin signaling proteins, insulin receptor substrate 2 and phosphatidylinositol 3 (PI3)-kinase regulatory subunit p50alpha, were increased and PI3-kinase p85alpha expression was decreased in liver and fat. These changes in protein expression correlated with increased insulin-stimulated protein kinase B phosphorylation. The expression of liver gluconeogenic enzymes, phosphoenolpyruvate carboxykinase, and fructose-1,6-bisphosphatase was also down-regulated. These findings suggest that PTP1B modulates insulin signaling in liver and fat, and that therapeutic modalities targeting PTP1B inhibition may have clinical benefit in type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus/blood , Obesity , Oligodeoxyribonucleotides, Antisense/pharmacology , Protein Tyrosine Phosphatases/genetics , Adipose Tissue/anatomy & histology , Animals , Base Sequence , Blood Glucose/drug effects , Crosses, Genetic , Diabetes Mellitus/drug therapy , Glucose Tolerance Test , Insulin/blood , Insulin/pharmacology , Liver/anatomy & histology , Mice , Mice, Obese , Organ Size/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/metabolism , RNA, Complementary/genetics , Reference Values
17.
Diabetes ; 51(8): 2405-11, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12145151

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) has been implicated as a negative regulator of insulin action. Overexpression of PTP1B protein has been observed in insulin-resistant states associated with obesity. Mice lacking a functional PTP1B gene exhibit increased insulin sensitivity and are resistant to weight gain. To investigate the role of PTP1B in adipose tissue from obese animals, hyperglycemic obese (ob/ob) mice were treated with PTP1B antisense oligonucleotide (ISIS-113715). A significant reduction in adiposity correlated with a decrease of PTP1B protein levels in fat. Antisense treatment also influenced the triglyceride content in adipocytes, correlating with a downregulation of genes encoding proteins involved in lipogenesis, such as sterol regulatory element-binding protein 1 and their downstream targets spot14 and fatty acid synthase, as well as other adipogenic genes, lipoprotein lipase, and peroxisome proliferator-activated receptor gamma. In addition, an increase in insulin receptor substrate-2 protein and a differential regulation of the phosphatidylinositol 3-kinase regulatory subunit (p85alpha) isoforms expression were found in fat from antisense-treated animals, although increased insulin sensitivity measured by protein kinase B phosphorylation was not observed. These results demonstrate that PTP1B antisense treatment can modulate fat storage and lipogenesis in adipose tissue and might implicate PTP1B in the enlargement of adipocyte energy stores and development of obesity.


Subject(s)
Adipose Tissue/physiopathology , Gene Expression Regulation/physiology , Insulin/physiology , Lipids/biosynthesis , Oligodeoxyribonucleotides, Antisense/pharmacology , Phosphoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Weight Loss/physiology , Adipocytes/metabolism , Adipose Tissue/drug effects , Alternative Splicing , Animals , DNA Primers , Gene Expression Regulation/drug effects , Genetic Variation , Homeostasis , Hyperglycemia/enzymology , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Isoenzymes/genetics , Mice , Mice, Obese , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Time Factors , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...