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










Publication year range
2.
Front Immunol ; 5: 233, 2014.
Article in English | MEDLINE | ID: mdl-24904582

ABSTRACT

SLE is a complex autoimmune inflammatory disease characterized by pathogenic autoantibody production as a consequence of uncontrolled T-B cell activity and immune-complex deposition in various organs, including kidney, leading to tissue damage and function loss. There is a high unmet need for better treatment options other than corticosteroids and immunosuppressants. Phosphoinositol-3 kinase δ (PI3Kδ) is a promising target in this respect as it is essential in mediating B- and T-cell function in mouse and human. We report the identification of selective PI3Kδ inhibitors that blocked B-, T-, and plasmacytoid dendritic cell activities in human peripheral blood and in primary cell co-cultures (BioMAP(®)) without detecting signs of undesired toxicity. In an IFNα-accelerated mouse SLE model, our PI3Kδ inhibitors blocked nephritis development, whether administered at the onset of autoantibody appearance or the onset of proteinuria. Disease amelioration correlated with normalized immune cell numbers in the spleen, reduced immune-complex deposition as well as reduced inflammation, fibrosis, and tissue damage in the kidney. Improvements were similar to those achieved with a frequently prescribed drug for lupus nephritis, the potent immunosuppressant mycophenolate mofetil. Finally, we established a pharmacodynamics/pharmacokinetic/efficacy model that revealed that a sustained PI3Kδ inhibition of 50% is sufficient to achieve full efficacy in our disease model. These data demonstrate the therapeutic potential of PI3Kδ inhibitors in SLE and lupus nephritis.

4.
Nat Chem Biol ; 6(2): 117-24, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20081827

ABSTRACT

Deregulation of the phosphoinositide-3-OH kinase (PI(3)K) pathway has been implicated in numerous pathologies including cancer, diabetes, thrombosis, rheumatoid arthritis and asthma. Recently, small-molecule and ATP-competitive PI(3)K inhibitors with a wide range of selectivities have entered clinical development. In order to understand the mechanisms underlying the isoform selectivity of these inhibitors, we developed a new expression strategy that enabled us to determine to our knowledge the first crystal structure of the catalytic subunit of the class IA PI(3)K p110 delta. Structures of this enzyme in complex with a broad panel of isoform- and pan-selective class I PI(3)K inhibitors reveal that selectivity toward p110 delta can be achieved by exploiting its conformational flexibility and the sequence diversity of active site residues that do not contact ATP. We have used these observations to rationalize and synthesize highly selective inhibitors for p110 delta with greatly improved potencies.


Subject(s)
Catalytic Domain , Phosphatidylinositol 3-Kinases/chemistry , Protein Kinase Inhibitors/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Cell Line , Computer Simulation , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Domains and Motifs , Spodoptera , Structure-Activity Relationship , Substrate Specificity
5.
Assay Drug Dev Technol ; 4(5): 525-33, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17115923

ABSTRACT

Type II diabetes and its associated complications are a major health concern of the developed world. One of the hallmarks of diabetes is insulin resistance, where secreted insulin no longer has any effect on its target tissues, namely, liver, muscle, and fat. An important therapeutic strategy is to modulate blood glucose levels using pharmacological agents. Glycogen synthase kinase-3 (GSK3) is a serine-threonine protein kinase that plays important roles in regulating glucose metabolism. It is a key negative regulator of insulin action and is an important contributing factor to insulin resistance in liver, muscle, and adipose tissue. We describe the development of a cell-based assay designed to measure glucose production in rat hepatoma cell line H4IIE liver cells in response to treatment with small molecule inhibitors, including GSK3 inhibitors. The assay is set up in a 96-well format, and glucose production is assessed using a convenient fluorescence-based readout. This disease-relevant cellular assay is a valuable tool for the progression of small molecules that modulate glucose production.


Subject(s)
Biological Assay/methods , Glucose/metabolism , Glycogen Synthase Kinase 3/metabolism , Insulin/administration & dosage , Liver Neoplasms, Experimental/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Design , Metabolic Clearance Rate/drug effects , Rats
6.
Assay Drug Dev Technol ; 4(4): 451-60, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16945017

ABSTRACT

Glycogen synthase kinase-3 (GSK3) is a serine-threonine protein kinase that exists as two isozymes, GSK3alpha and GSK3beta. It plays important roles in regulating cell structure, function, and survival, and dysregulation of its function is linked to disorders such as Alzheimer's disease and type II diabetes. In resting cells, GSK3 is active and regulates the function of many downstream targets, including beta-catenin. We describe the development of a cell-based assay designed to measure the activity of GSK3 by directly measuring the accumulation of beta-catenin in Chinese hamster ovary clone K1 (CHOK1) cells. Beta-catenin levels were assessed using an antibody-based staining protocol with a luminometric readout. The assay is set up in a 96-well format. The use of GSK3 inhibitors demonstrated that this assay could be used to compare the effects of various small molecules on GSK3 inhibition in CHOK1 cells.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , beta Catenin/metabolism , Aminophenols/pharmacology , Animals , Benzazepines/pharmacology , Benzimidazoles/pharmacology , Blotting, Western , CHO Cells , Clone Cells , Cricetinae , Cricetulus , Dimethyl Sulfoxide/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2B/metabolism , Glycogen Synthase Kinase 3/chemistry , Humans , Imidazoles/pharmacology , Immunoblotting , Indoles/pharmacology , Lithium Chloride/pharmacology , Luminescent Measurements/methods , Maleimides/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Reproducibility of Results , beta Catenin/chemistry
7.
J Med Chem ; 48(14): 4596-607, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15999997

ABSTRACT

Several lines of evidence support the hypothesis that c-Jun N-terminal kinase (JNKs) plays a critical role in a wide range of diseases including cell death (apoptosis)-related disorders (neurodegenerative diseases, brain, heart, and renal ischemia, epilepsy) and inflammatory disorders (multiple sclerosis, rheumatoid arthritis, inflammatory bowel diseases). Screening of our internal compound collection for inhibitors of JNK3 led to the identification of (benzothiazol-2-yl)acetonitrile derivatives as potent and selective JNK1, -2, -3 inhibitors. Starting from initial hit 1 (AS007149), the chemistry and initial structure-activity relationship (SAR) of this novel and unique kinase inhibitor template were explored. Investigation of the SAR rapidly revealed that the benzothiazol-2-ylacetonitrile pyrimidine core was crucial to retain a good level of potency on rat JNK3. Therefore, compound 6 was further optimized by exploring a number of distal combinations in place of the chlorine atom. This led to the observation that the presence of an aromatic group, two carbons away from the aminopyrimidine moiety and bearing substituents conferring hydrogen bond acceptor (HBA) properties, could improve the potency. Further improvements to the biological and biopharmaceutical profile of the most promising compounds were performed, resulting in the discovery of compound 59 (AS601245). The in vitro and in vivo anti-inflammatory potential of this new JNK inhibitor was investigated and found to demonstrate efficacy per oral route in an experimental model of rheumatoid arthritis (RA).


Subject(s)
Acetonitriles/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Thiazoles/chemical synthesis , Acetonitriles/chemistry , Acetonitriles/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antirheumatic Agents/chemical synthesis , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Benzothiazoles , Humans , JNK Mitogen-Activated Protein Kinases/chemistry , Jurkat Cells , Male , Mice , Mice, Inbred C3H , Mice, Inbred DBA , Rats , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
8.
J Neurochem ; 92(5): 1054-60, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15715656

ABSTRACT

c-Jun N-terminal kinase (JNK), a member of the mitogen-activated protein kinase family, is activated in response to a number of extracellular stimuli, including inflammatory cytokines, UV irradiation and ischaemia. A large body of evidence supports a role for JNK signalling in stress-induced apoptosis. It has been hypothesized that JNK may contribute to the apoptotic response by regulating the intrinsic cell death pathway involving the mitochondria. Here, we examined the role of the JNK signalling pathway in hippocampal CA1 apoptotic neurones following transient ischaemia in gerbils. We showed early activation of death receptor-dependent apoptosis (caspase-8 activation 2 days after ischaemia) and a biphasic activation of caspase-3 and caspase-9 after ischaemia. Activation of the mitochondrial pathway, as measured by cytochrome c release, appeared as a late event (5-7 days after ischaemia). AS601245, a novel JNK inhibitor, antagonized activation of both pathways and significantly protected CA1 neurones from cell death. Our results suggest a key role of JNK in the control of death receptor and mitochondrial-dependent apoptosis after transient ischaemia.


Subject(s)
Apoptosis , Hippocampus/pathology , Ischemic Attack, Transient/pathology , JNK Mitogen-Activated Protein Kinases/physiology , Mitochondria/metabolism , Neurons/metabolism , Acetonitriles/pharmacology , Acetonitriles/therapeutic use , Analysis of Variance , Animals , Benzothiazoles , Caspases/classification , Caspases/metabolism , Cytochromes c/metabolism , Enzyme Activation/drug effects , Gerbillinae , In Situ Nick-End Labeling/methods , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/enzymology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mitochondria/drug effects , Neurons/drug effects , Reperfusion Injury/prevention & control , Thiazoles/pharmacology , Thiazoles/therapeutic use , Time Factors
9.
Br J Pharmacol ; 142(6): 953-60, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15210584

ABSTRACT

1 Myocardial ischemia/reperfusion is associated with inflammation, apoptosis and necrosis. During this process, c-jun N-terminal kinase is activated in cardiac myocytes resulting in apoptosis. 2 This study investigates the effects of AS601245, a nonpeptide ATP competitive JNK inhibitor, on infarct size caused by myocardial ischemia/reperfusion in anaesthetized rats. The left descending coronary artery of anaesthetized rats was occluded for 30 min and then reperfused for 3 h. AS601245 was administered 5 min before the end of the ischemia period as an i.v. bolus (1.5, 4.5 or 15 mg kg(-1) i.v.) followed by continuous i.v. infusion (18, 55 and 183 microg kg(-1) min(-1), respectively) during reperfusion. Controls received saline only. 3-Aminobenzamide, a poly(ADP-ribose) polymerase inhibitor, was used as reference compound at 10 mg kg(-1) i.v. bolus plus 0.17 mg kg(-1) min(-1) continuous infusion. 3 AS601245 significantly reduced infarct size at 4.5 mg kg(-1) (-44%; P<0.001) and 15 mg kg(-1) i.v. (-40.3%; P<0.001) similarly to 3-aminobenzamide (-44.2%; P<0.001). This protective effect was obtained without affecting hemodynamics or reducing ST-segment displacement. 4 The beneficial effects on infarct size correlated well with the reduction of c-jun phosphorylation (-85%; P<0.001 versus control) and of TUNEL-positive cells (-82.1%; P<0.001) in post-ischemic cardiomyocytes. No change in the phosphorylation state of p38 MAPK and ERK in post-ischemic heart was observed in the presence of AS601245 in comparison to the vehicle-treated group. 5 These results demonstrate that blocking the JNK pathway may represent a novel therapeutic approach for treating myocardial ischemia/reperfusion-induced cardiomyocyte death.


Subject(s)
Acetonitriles/pharmacology , Apoptosis/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myocardial Infarction/prevention & control , Myocytes, Cardiac/drug effects , Thiazoles/pharmacology , Anesthesia , Animals , Benzothiazoles , Blood Pressure/drug effects , Blotting, Western , Coronary Disease/physiopathology , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , Heart Rate/drug effects , Hemodynamics/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Pharmacol Exp Ther ; 310(1): 25-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14988419

ABSTRACT

Recent evidence suggests that activation of the c-Jun NH2-terminal protein kinase (JNK) signal transduction pathway may play a role in ischemia-induced cell death. Thus, preventing the activation of JNK, or c-Jun phosphorylation could be neuroprotective. In the current study, we report that a small molecule, AS601245 (1,3-benzothiazol-2-yl (2-[[2-(3-pyridinyl) ethyl] amino]-4 pyrimidinyl) acetonitrile), which has been shown to inhibit the JNK signaling pathway, promotes cell survival after cerebral ischemia. In vivo, AS601245 (40, 60, and 80 mg/kg) administered i.p. provided significant protection against the delayed loss of hippocampal CA1 neurons in a gerbil model of transient global ischemia. This effect is mediated by JNK inhibition and therefore by c-Jun expression and phosphorylation. A significant neuroprotective effect of AS601245 administered either by i.p. injection (6, 18, and 60 mg/kg) or as i.v. bolus (1 mg/kg) followed by an i.v. infusion (0.6 mg/kg/h) was also observed in rats after focal cerebral ischemia. These data suggest that the use of JNK inhibitors such as AS601245 may be a relevant strategy in the therapy of ischemic insults.


Subject(s)
Acetonitriles/pharmacology , Brain Ischemia/prevention & control , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Thiazoles/pharmacology , Acetonitriles/therapeutic use , Animals , Benzothiazoles , Brain Ischemia/complications , Brain Ischemia/etiology , Disease Models, Animal , Gerbillinae , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Mitogen-Activated Protein Kinases/metabolism , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , Rats , Rats, Wistar , Thiazoles/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL