Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Thorax ; 68(6): 506-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23457038

ABSTRACT

BACKGROUND: Expression of the T-cell-associated chemokine receptor CCR8 and its ligand CCL1 have been demonstrated to be elevated in patients with asthma. CCR8 deficiency or inhibition in models of allergic airway disease in mice resulted in conflicting data. OBJECTIVE: To investigate the effects of a selective small molecule CCR8 inhibitor (ML604086) in a primate model of asthma. METHODS: ML604086 and vehicle were administered by intravenous infusion to 12 cynomolgus monkeys during airway challenge with Ascaris suum. Samples were collected throughout the study to measure pharmacokinetics (PK) and systemic CCR8 inhibition, as well as inflammation, T helper 2 (Th2) cytokines and mucus in bronchoalveolar lavage (BAL). Airway resistance and compliance were measured before and after allergen challenge, and in response to increasing concentrations of methacholine. RESULTS: ML604086 inhibited CCL1 binding to CCR8 on circulating T-cells>98% throughout the duration of the study. However, CCR8 inhibition had no significant effect on allergen-induced BAL eosinophilia and the induction of the Th2 cytokines IL-4, IL-5, IL-13 and mucus levels in BAL. Changes in airway resistance and compliance induced by allergen provocation and increasing concentrations of methacholine were also not affected by ML604086. CONCLUSIONS: These results clearly demonstrate a dispensable role for CCR8 in ameliorating allergic airway disease in atopic primates, and suggest that strategies other than CCR8 antagonism should be considered for the treatment of asthma.


Subject(s)
Airway Resistance/physiology , Asthma/immunology , Biological Factors/pharmacokinetics , Receptors, CCR8/antagonists & inhibitors , Th2 Cells/immunology , Animals , Asthma/metabolism , Asthma/physiopathology , Biological Factors/administration & dosage , Bronchoalveolar Lavage Fluid/chemistry , Chemokine CCL1/antagonists & inhibitors , Chemokine CCL1/biosynthesis , Chemokine CCL1/immunology , Disease Models, Animal , Female , Infusions, Intravenous , Lung/drug effects , Lung/physiopathology , Lung Compliance , Macaca fascicularis , Male , Receptors, CCR8/biosynthesis , Receptors, CCR8/immunology , Th2 Cells/metabolism
2.
ACS Chem Neurosci ; 2(8): 402-10, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-22860169

ABSTRACT

Intellectual disability (ID; mental retardation) is considered an immutable condition. Current medical practices are aimed at relieving symptoms and not at altering the underlying cognitive deficits. Scientific advancements from the past decade have led to the exciting possibility that ID may now be treatable. Moreover, pharmaceutical therapies targeting the most common form of inherited ID, Fragile X syndrome (FXS), may become the new benchmark for central nervous system (CNS) drug discovery: seeking cures for neurodevelopmental disorders.


Subject(s)
Central Nervous System Agents/chemistry , Central Nervous System Agents/therapeutic use , Drug Discovery/trends , Fragile X Syndrome/drug therapy , Receptors, Metabotropic Glutamate/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Baclofen/analogs & derivatives , Baclofen/chemistry , Baclofen/pharmacology , Baclofen/therapeutic use , Central Nervous System Agents/pharmacology , Fragile X Syndrome/metabolism , Fragile X Syndrome/psychology , Humans , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Treatment Outcome
3.
Pharmacol Ther ; 127(1): 78-93, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20303363

ABSTRACT

Fragile X is the leading inherited cause of mental retardation and autism. Recent advances in our mechanistic understanding of the disease have led to the identification of the metabotropic glutamate receptor (mGluR) as a therapeutic target for the disease. These studies have revealed that core defects in multiple animal models can be corrected by down regulation of mGluR5 signaling. Although it remains to be seen if mGluR5 antagonists or related approaches will succeed in humans with fragile X, the progress in fragile X stands as a strong testament to the power of applying knowledge of basic neurobiology to understand pathophysiology in a genetically validated model of human psychiatric disease. These breakthroughs and several of the resulting drug development efforts are reviewed.


Subject(s)
Fragile X Syndrome/drug therapy , Fragile X Syndrome/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Clinical Trials as Topic , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/pathology , Fragile X Syndrome/physiopathology , Humans , Male , Mice , Neuronal Plasticity , Rats , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/drug effects
4.
Autoimmunity ; 40(5): 355-65, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17612897

ABSTRACT

Identification and quantitation of peripheral blood non-invasive, cell-surface markers of EAE disease activity and drug response would facilitate the preclinical development of potential therapeutics. Towards this end, we characterized the influx of immune mediators into spinal cords of diseased rats to establish the kinetics of T cell and monocyte-mediated inflammation. We then examined the periphery for regulation of T cell and monocyte activation. We report increased CD80 and VLA-4 expression on peripheral blood monocytes (PBM) during the onset and peak of experimental disease scores. Increased CD4+, CD62L - and CD4+, CD134+ T cells were detected only at disease peak, not during disease onset. PBM CD80 expression was significantly inhibited in CSA-treated animals, but increased in Dex-treated animals. PBM VLA-4 expression was unaffected by drug treatment. Both CSA and Dex inhibited CD62L shedding and CD134 expression on peripheral CD4+ T cells. These results identify quantitative, peripheral markers of disease activity and drug response.


Subject(s)
Antigens, CD/immunology , Cyclosporine/therapeutic use , Cytokines/blood , Dexamethasone/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4beta1/blood , Animals , Anti-Inflammatory Agents/therapeutic use , Antigens, CD/blood , Biomarkers , Cyclosporine/immunology , Cytokines/immunology , Dexamethasone/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunosuppressive Agents/therapeutic use , Integrin alpha4beta1/immunology , Monocytes/immunology , Rats , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology
5.
Bioorg Med Chem Lett ; 17(11): 3141-5, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17395464

ABSTRACT

The identification, optimization, and structure-activity relationship (SAR) of small-molecule CCR4 antagonists is described. An initial screening hit with micromolar potency was identified that was optimized to sub-micromolar binding potency by enantiomer resolution, halogenation of the naphthalene ring, and extension of the alkyl chain linker between the central piperidine ring and the terminal aryl group. An antagonist was identified that showed good cross-reactivity against the mouse receptor and inhibited CCR4-based cell recruitment in dose-dependent fashion.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Naphthalenes/chemistry , Naphthalenes/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Sulfonamides/chemistry , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Mice , Mice, Inbred BALB C , Naphthalenes/chemical synthesis , Receptors, CCR4 , Structure-Activity Relationship , Sulfonamides/chemical synthesis
6.
J Med Chem ; 50(3): 566-84, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-17266208

ABSTRACT

The design, synthesis, and structure-activity relationship development of naphthalene-derived human CCR8 antagonists is described. In vitro binding assay results of these investigations are reported, critical interactions of the antagonists with CCR8 are defined, and preliminary physicochemical and pharmacokinetic data for the naphthalene scaffold are presented.


Subject(s)
Naphthalenes/chemical synthesis , Receptors, Chemokine/antagonists & inhibitors , Sulfonamides/chemical synthesis , Administration, Oral , Animals , Biological Availability , Biological Transport , Calcium/metabolism , Cell Line , Cricetinae , Cricetulus , Drug Design , Humans , Naphthalenes/pharmacokinetics , Naphthalenes/pharmacology , Rats , Receptors, CCR8 , Solubility , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
7.
Biochem Biophys Res Commun ; 352(3): 609-16, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17141195

ABSTRACT

Interleukin-1 stimulation leads to the recruitment of MyD88, interleukin-1 receptor-associated kinase 1 (IRAK-1) and interleukin-1 receptor-associated kinase 4 (IRAK-4) to the IL-1 receptor. The formation of the IL-1 receptor complex triggers a series of IRAK-1 autophosphorylations, which result in activation. IRAK-4 is upstream of IRAK-1 and may act as IRAK-1 kinase to transmit the signal. To date, there is no upstream kinase reported for IRAK-4; the activation mechanism of IRAK-4 remains poorly understood. Here, for the first time, we report three autophosphorylation sites that are responsible for IRAK-4 kinase activity. LC-MS/MS analysis has identified phosphorylations at T342, T345, and S346, which reside within the activation loop. Site-directed mutants at these positions exhibit significant reductions in the catalytic activity of IRAK-4 (T342A: 57%; T345A: 66%; S346A: 50%). The absence of phosphorylation in kinase-dead IRAK-4 indicates that phosphorylations in the activation loop result from autophosphorylation rather than from phosphorylation by an upstream kinase. Finally, we demonstrate that autophosphorylation is an intramolecular event as wild-type IRAK-4 failed to transphosphorylate kinase-inactive IRAK-4. The present data indicate that the kinase activity of IRAK-4 is dependent on the autophosphorylations at T342, T345, and S346 in the activation loop.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/chemistry , Interleukin-1 Receptor-Associated Kinases/metabolism , Kidney/enzymology , Signal Transduction/physiology , Amino Acid Sequence , Binding Sites , Cell Line , Enzyme Activation , Feedback/physiology , Humans , Molecular Sequence Data , Protein Binding , Structure-Activity Relationship
8.
Arthritis Rheum ; 56(1): 117-28, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17195214

ABSTRACT

OBJECTIVE: The NF-kappaB signaling pathway promotes the immune response in rheumatoid arthritis (RA) and in rodent models of RA. NF-kappaB activity is regulated by the IKK-2 kinase during inflammatory responses. To elucidate how IKK-2 inhibition suppresses disease development, we used a combination of in vivo imaging, transcription profiling, and histopathology technologies to study mice with antibody-induced arthritis. METHODS: ML120B, a potent, small molecule inhibitor of IKK-2, was administered to arthritic animals, and disease activity was monitored. NF-kappaB activity in diseased joints was quantified by in vivo imaging. Quantitative reverse transcriptase-polymerase chain reaction was used to evaluate gene expression in joints. Protease-activated near-infrared fluorescence (NIRF) in vivo imaging was applied to assess the amounts of active proteases in the joints. RESULTS: Oral administration of ML120B suppressed both clinical and histopathologic manifestations of disease. In vivo imaging demonstrated that NF-kappaB activity in inflamed arthritic paws was inhibited by ML120B, resulting in significant suppression of multiple genes in the NF-kappaB pathway, i.e., KC, epithelial neutrophil-activating peptide 78, JE, intercellular adhesion molecule 1, CD3, CD68, tumor necrosis factor alpha, interleukin-1beta, interleukin-6, inducible nitric oxide synthase, cyclooxygenase 2, matrix metalloproteinase 3, cathepsin B, and cathepsin K. NIRF in vivo imaging demonstrated that ML120B treatment dramatically reduced the amount of active proteases in the joints. CONCLUSION: Our data demonstrate that IKK-2 inhibition in the murine model of antibody-induced arthritis suppresses both inflammation and joint destruction. In addition, this study highlights how gene expression profiling can facilitate the identification of surrogate biomarkers of disease activity and treatment response in an experimental model of arthritis.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Carbolines/pharmacology , Enzyme Inhibitors/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Niacinamide/analogs & derivatives , Spectroscopy, Near-Infrared/methods , Administration, Oral , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/pathology , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , I-kappa B Kinase/metabolism , Joints/drug effects , Joints/metabolism , Joints/pathology , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/metabolism , Niacinamide/pharmacology , RNA, Messenger/metabolism , Spectrometry, Fluorescence/methods , Up-Regulation/drug effects
9.
Arthritis Rheum ; 54(10): 3163-73, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17009244

ABSTRACT

OBJECTIVE: The IKK complex regulates NF-kappaB activation, an important pathway implicated in the rheumatoid arthritis (RA) disease process. This study was undertaken to assess the efficacy of N-(6-chloro-7-methoxy-9H-beta-carbolin-8-yl)-2-methylnicotinamide (ML120B), a potent and selective small molecule inhibitor of IKKbeta. METHODS: Polyarthritis was induced in rats by injection of Freund's complete adjuvant into the hind footpad. ML120B was administered orally twice daily, either prophylactically or therapeutically. Paw volumes and body weights were measured every 2-3 days throughout the study. We assessed bone erosions by several methods: histologic evaluation, quantitative micro-computed tomography (micro-CT) imaging analysis, and measurement of type I collagen fragments in the serum. Quantitative polymerase chain reaction was used to evaluate expression of messenger RNA for genes related to inflammation and to bone and cartilage integrity. RESULTS: Oral administration of ML120B inhibited paw swelling in a dose-dependent manner (median effective dosage 12 mg/kg twice daily) and offered significant protection against arthritis-induced weight loss as well as cartilage and bone erosion. We were able to directly demonstrate that NF-kappaB activity in arthritic joints was reduced after ML120B administration. Also, we observed that down-regulation of the NF-kappaB pathway via IKKbeta inhibition dampened the chronic inflammatory process associated with rat adjuvant-induced arthritis. CONCLUSION: The results of the present study suggest that IKKbeta inhibition is an effective therapeutic approach to treat both the inflammation and the bone/cartilage destruction observed in RA. Methods for the determination of serum markers for bone and cartilage destruction, as well as micro-CT analysis, may aid in predicting and evaluating the therapeutic efficacy of IKKbeta inhibition therapy in humans.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Bone and Bones/pathology , Cartilage/pathology , I-kappa B Kinase/antagonists & inhibitors , Animals , Arthritis, Experimental , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/metabolism , Bone and Bones/metabolism , Carbolines/pharmacology , Cartilage/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Freund's Adjuvant , Gene Expression Regulation , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Rats , Rats, Inbred Lew , Tumor Necrosis Factor-alpha/metabolism
10.
J Pharmacol Exp Ther ; 317(3): 989-1001, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16525037

ABSTRACT

IkappaB kinase (IKK) beta is essential for inflammatory cytokine-induced activation of nuclear factor kappaB (NF-kappaB). NF-kappaB plays a pivotal role in the function of major cell types that contribute to the pathophysiological process of rheumatoid arthritis (RA). Here, we report the mechanism and the effect of the IKKbeta inhibitor N-(6-chloro-7-methoxy-9H-beta-carbolin-8-yl)-2-methylnicotinamide (ML120B), a beta-carboline derivative, on NF-kappaB signaling and gene activation in RA-relevant cell systems. ML120B is a potent, selective, reversible, and ATP-competitive inhibitor of IKKbeta with an IC50 of 60 nM when evaluated in an IkappaBalpha kinase complex assay. ML120B does not inhibit other IKK isoforms or a panel of other kinases. ML120B concentration-dependently inhibits tumor necrosis factor alpha (TNFalpha)-stimulated NF-kappaB signaling via inhibition of IkappaBalpha phosphorylation, degradation, and NF-kappaB translocation into the nucleus. For the first time, we have demonstrated that in human fibroblast-like synoviocytes, TNFalpha- or interleukin (IL)-1beta-induced monocyte chemoattractant protein-1 regulated on activation, normal T cell expressed and secreted and production is IKKbeta-dependent. In addition, for the first time, we have demonstrated that lipopolysaccharide- or peptidoglycan-induced cytokine production in human cord blood-derived mast cells is IKKbeta-dependent. In addition, in human chondrocytes, ML120B inhibited IL-1beta-induced matrix metalloproteinase production with an IC50 of approximately 1 microM. ML120B also blocked IL-1beta-induced prostaglandin E2 production. In summary, ML120B blocked numerous NF-kappaB-regulated cell responses that are involved in inflammation and destructive processes in the RA joint. Our findings support the evaluation of IKKbeta inhibitors as anti-inflammatory agents for the treatment of RA.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Connective Tissue Cells , Enzyme Inhibitors/pharmacology , I-kappa B Kinase/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Chondrocytes/drug effects , Chondrocytes/enzymology , Chondrocytes/immunology , Connective Tissue Cells/drug effects , Connective Tissue Cells/enzymology , Connective Tissue Cells/immunology , Cytokines/immunology , Dinoprostone/immunology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/immunology , HeLa Cells , Humans , Mast Cells/drug effects , Mast Cells/enzymology , Mast Cells/immunology , Molecular Structure , NF-kappa B/immunology , Signal Transduction/drug effects , Synovial Membrane/cytology
11.
Prostaglandins Other Lipid Mediat ; 76(1-4): 133-47, 2005 May.
Article in English | MEDLINE | ID: mdl-15967168

ABSTRACT

Chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2), a G protein-coupled receptor activated by prostaglandin D(2) (PGD(2)), has been identified as a receptor expressed on cell types critical to the pathogenesis of asthma. The cDNA encoding guinea pig CRTH2 was cloned and mRNA expression examined in selected tissues. Transcript profiling of guinea pig CRTH2 indicated relatively high levels of expression in bone marrow, intermediate levels in brain and relatively low levels in lung, spleen, thymus, lymph node, etc. Characterization of the molecular pharmacology of guinea pig CRTH2 revealed that guinea pig CRTH2 exhibited a greater affinity for Delta(12)-PGJ(2), a stable PGD(2) metabolite relative to human CRTH2. The CRTH2 selective agonists 13,14-dihydro-15-keto PGD(2) and Delta(12)-PGJ(2) induced the recruitment of eosinophils following intradermal administration of these ligands in guinea pigs. Chemotaxis of guinea pig eosinophils was elicited by either PGD(2) or Delta(12)-PGJ(2), and was abolished by a CRTH2-specific antagonist. These results indicate that PGD(2) and the stable metabolite, Delta(12)-PGJ(2), play important roles in CRTH2 activation in the guinea pig.


Subject(s)
Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Animals , Base Sequence , Calcium/metabolism , Cell Line , Chemotaxis, Leukocyte , Cloning, Molecular , DNA Primers , DNA, Complementary , Eosinophils/cytology , Guinea Pigs , Humans , Lung/metabolism , Lymph Nodes/metabolism , Male , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Spleen/metabolism , Thymus Gland/metabolism
12.
Biochem Biophys Res Commun ; 331(4): 1114-9, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15882992

ABSTRACT

CARD12 (Ipaf/Clan) is an important regulator of caspase-1 activation. It belongs to the family of the nucleotide-binding site and leucine-rich repeat (NBS-LRR) proteins. The NBS domain of the NBS-LRR proteins contains putative ATP/GTPase-specific P-loop and Mg2+-binding site motifs. However, the nucleotide-binding properties and the function of the NBS domain are unknown. We developed a nucleotide-binding assay and investigated nucleotide binding to CARD12. We find that the NBS domain of CARD12 contains a nucleotide-binding pocket with specificity for ATP/dATP. A point mutation in the P-loop (K175R) of the NBS domain abolishes ATP/dATP binding. We further demonstrate that the nucleotide-binding site is required for CARD12-mediated caspase-1 activation. CARD12 self-association and association with procaspase-1 in transfected cells were markedly decreased by the P-loop mutation K175R. Furthermore, the P-loop mutation greatly reduced caspase-1 activation-dependent proIL-1beta processing. Thus, CARD12 function is dependent on the nucleotide-binding site. Our data provide insights into the molecular mechanisms of CARD12-mediated caspase-1 activation.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium-Binding Proteins/metabolism , Caspase 1/metabolism , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Enzyme Activation , Point Mutation , Protein Binding
13.
J Biol Chem ; 280(16): 16278-83, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15691841

ABSTRACT

Rip2 (Rick, Cardiak, CCK2, and CARD3) is a serine/threonine kinase containing a caspase recruitment domain (CARD) at the C terminus. Previous reports have shown that Rip2 is involved in multiple receptor signaling pathways that are important for innate and adaptive immune responses. However, it is not known whether Rip2 kinase activity is required for its function. Here we confirm that Rip2 participates in lipopolysaccharide (LPS)/Toll-like receptor (TLR4) signaling and demonstrate that its kinase activity is not required. Upon LPS stimulation, Rip2 was transiently recruited to the TLR4 receptor complex and associated with key TLR signaling mediators IRAK1 and TRAF6. Furthermore, Rip2 kinase activity was induced by LPS treatment. These data indicate that Rip2 is directly involved in the LPS/TLR4 signaling. Whereas macrophages from Rip2-deficient mice showed impaired NF-kappaB and p38 mitogen-activated protein kinase activation and reduced cytokine production in response to LPS stimulation, LPS signaling was intact in macrophages from mice that express Rip2 kinase-dead mutant. These results demonstrate that Rip2-mediated LPS signaling is independent of its kinase activity. Our findings strongly suggest that Rip2 functions as an adaptor molecule in transducing signals from immune receptors.


Subject(s)
Lipopolysaccharides/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Animals , I-kappa B Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , NF-KappaB Inhibitor alpha , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases , Receptors, Immunologic/metabolism , Toll-Like Receptor 4 , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Eukaryot Cell ; 2(2): 256-64, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12684375

ABSTRACT

A genetic approach utilizing the yeast Saccharomyces cerevisiae was used to identify the target of antifungal compounds. This analysis led to the identification of small molecule inhibitors of RNA polymerase (Pol) III from Saccharomyces cerevisiae. Three lines of evidence show that UK-118005 inhibits cell growth by targeting RNA Pol III in yeast. First, a dominant mutation in the g domain of Rpo31p, the largest subunit of RNA Pol III, confers resistance to the compound. Second, UK-118005 rapidly inhibits tRNA synthesis in wild-type cells but not in UK-118005 resistant mutants. Third, in biochemical assays, UK-118005 inhibits tRNA gene transcription in vitro by the wild-type but not the mutant Pol III enzyme. By testing analogs of UK-118005 in a template-specific RNA Pol III transcription assay, an inhibitor with significantly higher potency, ML-60218, was identified. Further examination showed that both compounds are broad-spectrum inhibitors, displaying activity against RNA Pol III transcription systems derived from Candida albicans and human cells. The identification of these inhibitors demonstrates that RNA Pol III can be targeted by small synthetic molecules.


Subject(s)
Antifungal Agents/pharmacology , Enzyme Inhibitors/pharmacology , RNA Polymerase III/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Candida albicans/drug effects , Candida albicans/enzymology , Candida albicans/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Drug Resistance, Fungal/genetics , Humans , Molecular Sequence Data , Molecular Weight , Mutation/genetics , Protein Subunits/genetics , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , RNA, Transfer/biosynthesis , RNA, Transfer/genetics , Reaction Time/drug effects , Reaction Time/genetics , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...