Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Med ; 215(5): 1315-1325, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29549113

ABSTRACT

p38α activation of multiple effectors may underlie the failure of global p38α inhibitors in clinical trials. A unique inhibitor (CDD-450) was developed that selectively blocked p38α activation of the proinflammatory kinase MK2 while sparing p38α activation of PRAK and ATF2. Next, the hypothesis that the p38α-MK2 complex mediates inflammasome priming cues was tested. CDD-450 had no effect on NLRP3 expression, but it decreased IL-1ß expression by promoting IL-1ß mRNA degradation. Thus, IL-1ß is regulated not only transcriptionally by NF-κB and posttranslationally by the inflammasomes but also posttranscriptionally by p38α-MK2. CDD-450 also accelerated TNF-α and IL-6 mRNA decay, inhibited inflammation in mice with cryopyrinopathy, and was as efficacious as global p38α inhibitors in attenuating arthritis in rats and cytokine expression by cells from patients with cryopyrinopathy and rheumatoid arthritis. These findings have clinical translation implications as CDD-450 offers the potential to avoid tachyphylaxis associated with global p38α inhibitors that may result from their inhibition of non-MK2 substrates involved in antiinflammatory and housekeeping responses.


Subject(s)
Inflammasomes/metabolism , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction , Animals , Arthritis/pathology , Bone and Bones/pathology , Cytokines/biosynthesis , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Joints/pathology , Male , Mice , Mitogen-Activated Protein Kinase 14/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , RNA Stability , Rats, Inbred Lew
2.
Int Immunopharmacol ; 10(10): 1170-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20609399

ABSTRACT

In this report, we show that apoptosis signal-regulating kinase 1(-/-) (ASK1 KO) mice were resistant to inflammatory arthritis induced in the K/BxN serum transfer model of rheumatoid arthritis (RA). The p38 inhibitor, SD-0006 was administered to wild type (WT) mice as a comparator. Both ASK1 KO and p38 inhibition resulted in marked attenuation of edema, cartilage damage, bone resorption, and general inflammatory responses. Transcriptional profiling of mRNA prepared from paw tissue demonstrated that the production of many proinflammatory genes including cytokines, chemokines, and extracellular matrix degradative enzymes were maintained at basal levels by either ASK1 KO or prophylactic p38 MAPK inhibition. In the mouse whole blood (MWB) assay, tumor necrosis factor-α (TNF-α)-induced KC and CCL2 levels and also LPS-induced interleukin-6 (IL-6), CCL2, and KC levels in MWB from ASK1 KO were significantly lower than those from WT. Furthermore, both p38 and JNK were activated by TNF-α in human synovial fibroblasts isolated from RA patients (RASF). SD-0006 or SP600125, a JNK inhibitor, partially blocked the elevation of IL-6 production in RASF following stimulation with TNF-α. In contrast, dual inhibition with both p38/JNK inhibitors almost completely abolished TNF-α-induced IL-6 production from these cells. Ablation of ASK1 expression in RASF using siRNA for ASK1 resulted in inhibition of TNF-α-induced IL-6 and PGE(2) production. This study is the first to suggest that ASK1 is critical for the development of RA and that ASK1 may be involved in the production of proinflammatory mediators in response to TNF-α stimulation in the RA joint.


Subject(s)
Apoptosis/physiology , Arthritis/chemically induced , MAP Kinase Kinase Kinase 5/metabolism , Animals , Arthritis/pathology , Arthritis, Rheumatoid/metabolism , Cells, Cultured , Dinoprostone/genetics , Dinoprostone/metabolism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Interleukin-6 , MAP Kinase Kinase Kinase 5/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrazoles/toxicity , Pyrimidines/toxicity , Tumor Necrosis Factor-alpha/pharmacology
3.
J Pharmacol Exp Ther ; 333(3): 797-807, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20237073

ABSTRACT

Activation of the p38 kinase pathway in immune cells leads to the transcriptional and translational regulation of proinflammatory cytokines. Mitogen-activated protein kinase-activated protein kinase 2 (MK2), a direct downstream substrate of p38 kinase, regulates lipopolysaccharide (LPS)-stimulated tumor necrosis factor alpha (TNFalpha) and interleukin-6 (IL-6) production through modulating the stability and translation of these mRNAs. Developing small-molecule inhibitors of MK2 may yield anti-inflammatory efficacy with a different safety profile relative to p38 kinase inhibitors. This article describes the pharmacologic properties of a benzothiophene MK2 inhibitor, PF-3644022 [(10R)-10-methyl-3-(6-methylpyridin-3-yl)-9,10,11,12-tetrahydro-8H-[1,4]diazepino[5',6':4,5]thieno[3,2-f]quinolin-8-one]. PF-3644022 is a potent freely reversible ATP-competitive compound that inhibits MK2 activity (K(i) = 3 nM) with good selectivity when profiled against 200 human kinases. In the human U937 monocytic cell line or peripheral blood mononuclear cells, PF-3644022 potently inhibits TNFalpha production with similar activity (IC(50) = 160 nM). PF-3644022 blocks TNFalpha and IL-6 production in LPS-stimulated human whole blood with IC(50) values of 1.6 and 10.3 microM, respectively. Inhibition of TNFalpha in U937 cells and blood correlates closely with inhibition of phospho-heat shock protein 27, a target biomarker of MK2 activity. PF-3644022 displays good pharmacokinetic parameters in rats and is orally efficacious in both the rat acute LPS-induced TNFalpha model and the chronic streptococcal cell wall-induced arthritis model. Dose-dependent inhibition of TNFalpha production in the acute model and inhibition of paw swelling in the chronic model is observed with ED(50) values of 6.9 and 20 mg/kg, respectively. PF-3644022 efficacy in the chronic inflammation model is strongly correlated with maintaining a C(min) higher than the EC(50) measured in the rat LPS-induced TNFalpha model.


Subject(s)
Anti-Inflammatory Agents , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammation/drug therapy , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Acute Disease , Adenosine Triphosphate/metabolism , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Binding, Competitive/drug effects , Cell Wall/chemistry , Chronic Disease , Dose-Response Relationship, Drug , Female , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Humans , Inflammation/chemically induced , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Protein Kinase Inhibitors/pharmacokinetics , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Streptococcus , U937 Cells , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
4.
J Pharmacol Exp Ther ; 333(2): 445-53, 2010 May.
Article in English | MEDLINE | ID: mdl-20164299

ABSTRACT

Antagonists of the cannabinoid receptor 1 (CB1) impart anti-inflammatory activity even though, paradoxically, CB2 receptors are more predominant on cells of the immune system. We attempted to understand the mechanism of this activity by using an acute model of lipopolysaccharide-induced inflammation/stress in both rat and mouse, with selective antagonists to CB1 receptors. We demonstrate that the ability of a CB1 antagonist to inhibit release of proinflammatory cytokines is not dependent on either adrenal-derived catecholamines or corticosteroids or input from the pituitary or thymus glands but does involve the spleen. Furthermore, we show that the anti-inflammatory activity is retained without communication from the central nervous system following ganglionic blockade, suggesting a peripheral site of action. Finally, we show that the anti-inflammatory activity can be inhibited with the use of a selective beta2-adrenoceptor antagonist.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, Adrenergic, beta-2/physiology , Adrenalectomy , Adrenergic beta-2 Receptor Antagonists , Animals , Arthritis/chemically induced , Arthritis/drug therapy , Arthritis/physiopathology , Benzoxazines/pharmacology , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred DBA , Morpholines/pharmacology , Naphthalenes/pharmacology , Rats , Rats, Inbred Lew , Receptor, Cannabinoid, CB1/physiology , Receptors, Adrenergic, beta-2/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
5.
Biochem Pharmacol ; 79(10): 1445-54, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20067770

ABSTRACT

Inflammation-induced microsomal prostaglandin E synthase-1 (mPGES-1) is the terminal enzyme that synthesizes prostaglandin E(2) (PGE(2)) downstream of cyclooxygenase-2 (COX-2). The efficacy of nonsteroidal anti-inflammatory drugs and COX-2 inhibitors in the treatment of the signs and symptoms of osteoarthritis, rheumatoid arthritis and inflammatory pain, largely attributed to the inhibition of PGE(2) synthesis, provides a rationale for exploring mPGES-1 inhibition as a potential novel therapy for these diseases. Toward this aim, we identified PF-9184 as a novel mPGES-1 inhibitor. PF-9184 potently inhibited recombinant human (rh) mPGES-1 (IC(50)=16.5+/-3.8nM), and had no effect against rhCOX-1 and rhCOX-2 (>6500-fold selectivity). In inflammation and clinically relevant biological systems, mPGES-1 expression, like COX-2 expression was induced in cell context- and time-dependent manner, consistent with the kinetics of PGE(2) synthesis. In rationally designed cell systems ideal for determining direct effects of the inhibitors on mPGES-1 function, but not its expression, PF-9184 inhibited PGE(2) synthesis (IC(50) in the range of 0.5-5 microM in serum-free cell and human whole blood cultures, respectively) while sparing the synthesis of 6-keto-PGF(1alpha) (PGF(1alpha)) and PGF(2alpha). In contrast, as expected, the selective COX-2 inhibitor, SC-236, inhibited PGE(2), PGF(1alpha) and PGF(2alpha) synthesis. This profile of mPGES-1 inhibition, distinct from COX-2 inhibition in cells, validates mPGES-1 as an attractive target for therapeutic intervention.


Subject(s)
Cyclic S-Oxides/antagonists & inhibitors , Cyclooxygenase 2 Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Thiazines/antagonists & inhibitors , Animals , Arthritis, Rheumatoid/metabolism , Carrageenan/pharmacology , Cells, Cultured , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression/drug effects , Humans , Immunoblotting , Interleukin-1beta/pharmacology , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/metabolism , Microsomes/drug effects , Microsomes/enzymology , Prostaglandin-E Synthases , Rats , Reverse Transcriptase Polymerase Chain Reaction
6.
Nitric Oxide ; 20(3): 150-6, 2009 May.
Article in English | MEDLINE | ID: mdl-19146971

ABSTRACT

The contribution of inducible nitric oxide synthase (iNOS) to oxidative/nitrative stress is well-documented in inflammation, but difficult to quantify. Using a novel, recently developed assay for 3-nitrotyrosine (3-NT), we characterized iNOS activity and its inhibition in preclinical models of inflammation. In particular, we utilized the 3-NT assay to assess the role of iNOS in the disease pathology as well as for proof of pharmacology of iNOS inhibitors in an acute endotoxin challenge model, in models of rheumatoid arthritis (RA) such as rat adjuvant- and collagen-induced arthritis (AIA and CIA) and a model of osteoarthritis (OA) such as rat sodium monoiodoacetate-induced arthritis (MIA). Quantification of nitrotyrosine was performed using immuno-affinity 2-D LC-MS/MS assay. This assay is a very specific and reproducible and is amenable to a number of biological fluids. Plasma levels of 3-NT were significantly elevated in an acute model of inflammation (rat LPS) and in models of rheumatoid arthritis (adjuvant- and collagen-induced arthritis), and osteoarthritis (monoiodoacetate-induced arthritis). Plasma 3-NT correlated with the severity of the inflammatory response; thus, a 20-fold increase was observed in the rat LPS model, a 10-fold increase in AIA, and only a 2.5-fold elevation in CIA. Pharmacological intervention with iNOS inhibitors decreased 3-NT levels and associated pathology. 3-NT determination allowed for better elucidation of the role of iNOS in RA and OA disease pathology and provided proof of pharmacology for NOS inhibitors in animal models of RA and OA.


Subject(s)
Nitric Oxide Synthase Type II/physiology , Tyrosine/analogs & derivatives , Animals , Arthritis, Experimental , Arthritis, Rheumatoid , Biomarkers/blood , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Inflammation , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis , Rats , Severity of Illness Index , Tyrosine/blood
7.
J Med Chem ; 50(23): 5712-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17948975

ABSTRACT

A series of pyrazole inhibitors of p38 mitogen-activated protein (MAP) kinase were designed using a binding model based on the crystal structure of 1 (SC-102) bound to p38 enzyme. New chemistry using dithietanes was developed to assemble nitrogen-linked substituents at the 5-position of pyrazoles. Calculated log D was used in tandem with structure-based design to guide medicinal chemistry strategy and improve the in vivo activity of a series of molecules. The crystal structure of an optimized inhibitor, 4 (SC-806), in complex with p38 enzyme was obtained to confirm the hypothesis that the addition of a basic nitrogen to the molecule induces an interaction with Asp112 of p38 alpha. A compound identified from this series was efficacious in an animal model of rheumatic disease.


Subject(s)
Antirheumatic Agents/chemical synthesis , Piperazines/chemical synthesis , Pyrazoles/chemical synthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Collagen , Crystallography, X-Ray , Male , Mice , Mice, Inbred DBA , Models, Molecular , Piperazines/chemistry , Piperazines/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Rats, Inbred Lew , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/chemistry
8.
Am J Ther ; 2(9): 648-659, 1995 Sep.
Article in English | MEDLINE | ID: mdl-11854842

ABSTRACT

Prostaglandin E(2) (PGE(2)) is known to inhibit in vitro T-cell responses to mitogenic and antigenic stimuli. Interaction of PGE(2) with a G protein-coupled receptor activates adenylyl cyclase, leading to cAMP formation and inhibition of interleukin-2 (IL-2) production and T-cell proliferation. Despite these effects, the application of PGE(2) as an anti-inflammatory agent has been compromised by its unfavorable pharmacodynamic and side-effect profile. Because of the potential utility of synthetic analogs as prostaglandin-based therapeutics, we evaluated the effect of misoprostol and over 100 structural analogs on cAMP formation and T-cell activation. Our results indicate that micromolar concentrations of misoprostol and particular analogs elicited a rapid and substantial rise in cAMP levels in human peripheral blood mononuclear cells. Analogs which increased cAMP also suppressed IL-2 production and T-cell growth in vitro, whereas those devoid of suppressive activity weakly induced nucleotide synthesis. Despite extensive chemical alteration of the prostanoid structure, no single analog was superior to misoprostol in inducing cAMP or modulating T-cell activity. Misoprostol and suppressive analogs were also evaluated in vivo in a murine model of antigen-induced T-cell proliferation. Prostaglandins, administered at maximum tolerable doses, were ineffective in blocking a T-cell response to alloantigenic stimulation, whereas cyclosporine and prednisolone were potent inhibitors of this response. Overall, our results indicate that misoprostol and related analogs suppress T-cell activation in vitro but require concentrations 1000-fold greater than the low nanomolar plasma levels achieved with clinical doses of misoprostol. Whether misoprostol analogs of sufficient potency can be developed for pharmacologic attentuation of T-cell activation in vivo remains to be determined.

SELECTION OF CITATIONS
SEARCH DETAIL
...