Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biosci Biotechnol Biochem ; 84(7): 1322-1331, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32264779

ABSTRACT

NADH-quinone oxidoreductase (respiratory complex I) is a key player in mitochondrial energy metabolism. The enzyme couples electron transfer from NADH to quinone with the translocation of protons across the membrane, providing a major proton-motive force that drives ATP synthesis. Recently, X-ray crystallography and cryo-electron microscopy provided further insights into the structure and functions of the enzyme. However, little is known about the mechanism of quinone reduction, which is a crucial step in the energy coupling process. A variety of complex I inhibitors targeting the quinone-binding site have been indispensable tools for mechanistic studies on the enzyme. Using biorationally designed inhibitor probes, the author has accumulated a large amount of experimental data characterizing the actions of complex I inhibitors. On the basis of comprehensive interpretations of the data, the author reviews the structural features of the binding pocket of quinone/inhibitors in bovine mitochondrial complex I. ABBREVIATIONS: ATP: adenosine triphosphate; BODIPY: boron dipyrromethene; complex I: proton-translocating NADH-quinone oxidoreductase; DIBO: dibenzocyclooctyne; EM: electron microscopy; FeS: iron-sulfur; FMN: flavin adenine mononucleotide; LDT: ligand-directed tosylate; NADH: nicotinamide adenine dinucleotide; ROS: reactive oxygen species; SMP: submitochondrial particle; TAMRA: 6-carboxy-N,N,N',N'-tetramethylrhodamine; THF: tetrahydrofuran; TMH: transmembrane helix.


Subject(s)
Benzoquinones/antagonists & inhibitors , Benzoquinones/metabolism , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Acetogenins/pharmacology , Amiloride/pharmacology , Animals , Benzoates/pharmacology , Benzoquinones/chemistry , Binding Sites/drug effects , Cattle , Electron Transport , Electron Transport Complex I/chemistry , Humans , Oxidative Phosphorylation , Pyrazoles/pharmacology , Quinazolines/pharmacology , Reactive Oxygen Species/metabolism
2.
Article in English | MEDLINE | ID: mdl-31508379

ABSTRACT

This study aimed to determine the antimicrobial activity of thymoquinone (TQ) against Listeria monocytogenes, and to examine its inhibitory effects on biofilm formation, motility, hemolysin production, and attachment-invasion of host cells. The minimum inhibitory concentrations (MICs) of TQ against eight different L. monocytogenes strains ranged from 6.25-12.50 µg/mL. Crystal violet staining showed that TQ clearly reduced biofilm biomass at sub-MICs in a dose-dependent manner. Scanning electron microscopy suggested that TQ inhibited biofilm formation on glass slides and induced an apparent collapse of biofilm architecture. At sub-MICs, TQ effectively inhibited the motility of L. monocytogenes ATCC 19115, and significantly impacted adhesion to and invasion of human colon adenocarcinoma cells as well as the secretion of listeriolysin O. Supporting these findings, real-time quantitative polymerase chain reaction analysis revealed that TQ down-regulated the transcription of genes associated with motility, biofilm formation, hemolysin secretion, and attachment-invasion in host cells. Overall, these findings confirm that TQ has the potential to be used to combat L. monocytogenes infection.


Subject(s)
Benzoquinones/antagonists & inhibitors , Biofilms/drug effects , Listeria monocytogenes/drug effects , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Toxins/metabolism , Biofilms/growth & development , Biomass , Genes, Bacterial/genetics , Heat-Shock Proteins/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeriosis/microbiology , Microbial Sensitivity Tests , Real-Time Polymerase Chain Reaction , Virulence/drug effects , Virulence/genetics
3.
Microb Pathog ; 132: 51-58, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31028862

ABSTRACT

Porcine Circovirus Type 2 (PCV2) is a pathogen that has the ability to cause devastating disease manifestations in pig populations with major economic implications. Our previous research found that Hsp90 is required for PCV2 production in PK-15 and 3D4/31 cells. The aim of this study was to evaluate the effect of Hsp90 inhibitor regulating PCV2 replication and to explore its underlying mechanism. In PK-15 and 3D4/31 cells treated with 17-AAG after viral adsorption, replication of PCV2 was attenuated as assessed by quantitating the expression of viral protein. Following NF-κB activation it was observed that 24hpi with PCV2 was significantly inhibited in the presence of 17-AAG. The expression of Hsp90 associated client proteins in PCV2-infected cells were also reduced in the presence of 17-AAG. However, treatment with MG-132 failed to rescue 17-AAG mediated reduction of PCV2 production in host cells. Thus, Hsp90 regulates PCV2 by modulating cellular signaling proteins. These results highlight the importance of cellular proteins during PCV2 infection and the possibility of targeting cellular chaperones for developing new anti-rotaviral strategies.


Subject(s)
Benzoquinones/antagonists & inhibitors , Circovirus/drug effects , HSP90 Heat-Shock Proteins/drug effects , HSP90 Heat-Shock Proteins/genetics , Lactams, Macrocyclic/antagonists & inhibitors , Virus Replication/drug effects , Animals , Benzoquinones/chemistry , Cell Line , Cell Survival/drug effects , Circoviridae Infections/drug therapy , Circoviridae Infections/virology , HSP90 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions/drug effects , Lactams, Macrocyclic/chemistry , Leupeptins/antagonists & inhibitors , NF-kappa B/drug effects , Swine , Swine Diseases/virology
4.
PLoS One ; 12(5): e0177706, 2017.
Article in English | MEDLINE | ID: mdl-28542269

ABSTRACT

Many types of cancer express high levels of heat shock proteins (HSPs) that are molecular chaperones regulating protein folding and stability ensuring protection of cells from potentially lethal stress. HSPs in cancer cells promote survival, growth and spreading even in situations of growth factors deprivation by associating with oncogenic proteins responsible for cell transformation. Hence, it is not surprising that the identification of potent inhibitors of HSPs, notably HSP90, has been the primary research focus, in recent years. Exposure of cancer cells to HSP90 inhibitors, including 17-AAG, has been shown to cause resistance to chemotherapeutic treatment mostly attributable to induction of the heat shock response and increased cellular levels of pro-survival chaperones. In this study, we show that treatment of glioblastoma cells with 17-AAG leads to HSP90 inhibition indicated by loss of stability of the EGFR client protein, and significant increase in HSP70 expression. Conversely, co-treatment with the small-molecule kinase inhibitor D11 leads to suppression of the heat shock response and inhibition of HSF1 transcriptional activity. Beside HSP70, Western blot and differential mRNA expression analysis reveal that combination treatment causes strong down-regulation of the small chaperone protein HSP27. Finally, we demonstrate that incubation of cells with both agents leads to enhanced cytotoxicity and significantly high levels of LC3-II suggesting autophagy induction. Taken together, results reported here support the notion that including D11 in future treatment regimens based on HSP90 inhibition can potentially overcome acquired resistance induced by the heat shock response in brain cancer cells.


Subject(s)
Benzoquinones/antagonists & inhibitors , Brain Neoplasms/pathology , Glioblastoma/pathology , Glucosides/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Lactams, Macrocyclic/antagonists & inhibitors , Lignans/pharmacology , Protein Kinase Inhibitors/pharmacology , Up-Regulation/drug effects , Benzoquinones/pharmacology , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Drug Interactions , Drug Resistance, Neoplasm/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heat Shock Transcription Factors , Humans , Lactams, Macrocyclic/pharmacology , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcriptome/drug effects
5.
Food Funct ; 6(1): 219-29, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25375233

ABSTRACT

Foods of plant origin contain flavonoids. In the adzuki bean, (+)-catechin, quercetin 3-O-rutinoside (rutin), and quercetin 7-O-ß-D-glucopyranoside (Q7G) are the major flavonoids. During mastication of foods prepared from the adzuki bean, the flavonoids are mixed with saliva and swallowed into the stomach. Here we investigated the interactions between Q7G and (+)-catechin at pH 2, which may proceed in the stomach after the ingestion of foods prepared from the adzuki bean. Q7G reacted with nitrous acid producing nitric oxide (˙NO) and a glucoside of 2-(3,4-dihydroxybenzoyl)-2,4,6-trihydroxy-3(2H)-benzofuranone. (+)-Catechin reacted with nitrous acid producing ˙NO and 6,8-dinitrosocatechin. The production of the dinitrosocatechin was partly suppressed by Q7G, and the suppression resulted in the enhancement of Q7G oxidation. 6,8-Dinitrosocatechin reacted further with nitrous acid generating the o-quinone, and the quinone formation was effectively suppressed by Q7G. In the flavonoids investigated, the suppressive effect decreased in the order Q7G≈quercetin>kaempferol>quercetin 4'-O-glucoside>rutin. Essentially the same results were obtained when (-)-epicatechin was used instead of (+)-catechin. The results indicate that nitrous acid-induced formation of 6,8-dinitrosocatechins and the o-quinones can be suppressed by flavonols in the stomach, and that both a hydroxyl group at C3 and ortho-hydroxyl groups in the B-ring are required for efficient suppression.


Subject(s)
Anticarcinogenic Agents/metabolism , Carcinogens/antagonists & inhibitors , Catechin/analogs & derivatives , Digestion , Glucosides/metabolism , Models, Biological , Nitroso Compounds/antagonists & inhibitors , Quercetin/analogs & derivatives , Animals , Anticarcinogenic Agents/chemistry , Benzofurans/chemistry , Benzofurans/metabolism , Benzoquinones/antagonists & inhibitors , Benzoquinones/chemistry , Benzoquinones/metabolism , Carcinogens/chemistry , Carcinogens/metabolism , Catechin/antagonists & inhibitors , Catechin/chemistry , Catechin/metabolism , Dietary Supplements , Fabaceae/chemistry , Functional Food/analysis , Gastric Juice/chemistry , Gastric Juice/enzymology , Gastric Juice/metabolism , Glucosides/chemistry , Humans , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitroso Compounds/chemistry , Nitroso Compounds/metabolism , Nitrous Acid/chemistry , Nitrous Acid/metabolism , Quercetin/chemistry , Quercetin/metabolism , Quinones/chemistry , Quinones/metabolism , Saliva/chemistry , Saliva/enzymology , Saliva/metabolism , Seeds/chemistry , Stereoisomerism
6.
Pest Manag Sci ; 70(11): 1641-5, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24616024

ABSTRACT

Rust fungi are major pathogens of many annual and perennial crops. Crop protection is largely based on genetic and chemical control. Fungicide resistance is a significant issue that has affected many crop pathogens. Some pathogens have rapidly developed resistance and hence are regarded as high-risk species. Rust fungi have been classified as being low risk, in spite of sharing many relevant features with high-risk pathogens. An examination of the evidence suggests that rust fungi may be wrongly classified as low risk. Of the nine classes of fungicide to which resistance has developed, six are inactive against rusts. The three remaining classes are quinone outside inhibitors (QoIs), demethylation inhibitors (DMIs) and succinate dehydrogenase inhibitors (SDHIs). QoIs have been protected by a recently discovered intron that renders resistant mutants unviable. Low levels of resistance have developed to DMIs, but with limited field significance. Older SDHI fungicides were inactive against rusts. Some of the SDHIs introduced since 2003 are active against rusts, so it may be that insufficient time has elapsed for resistance to develop, especially as SDHIs are generally sold in mixtures with other actives. It would therefore seem prudent to increase the level of vigilance for possible cases of resistance to established and new fungicides in rusts.


Subject(s)
Basidiomycota/drug effects , Drug Resistance, Fungal , Fungicides, Industrial/pharmacology , Basidiomycota/genetics , Basidiomycota/metabolism , Benzoquinones/antagonists & inhibitors , Methylation/drug effects , Plant Diseases/microbiology , Succinate Dehydrogenase/antagonists & inhibitors
7.
Phytopathology ; 104(4): 347-56, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24168041

ABSTRACT

Respiration inhibitors such as the succinate dehydrogenase inhibitors (SDHIs) and the quinone outside inhibitors (QoIs) are fungicide classes with increasing relevance in gray mold control. However, recent studies have shown that dual resistance to both fungicide classes is a common trait in Botrytis cinerea populations from several hosts throughout the world. Resistance of B. cinerea to SDHIs is associated with several mutations in the sdhB, sdhC, and sdhD genes, while resistance to QoIs, in most cases, is associated with the G143A mutation in the cytb gene. The objective of the current study was to investigate the fitness and the competitive ability of B. cinerea field strains possessing one of the H272Y/R/L, N230I, or P225F sdhB substitutions and the G143A mutation of cytb. Fitness parameters measured were (i) mycelial growth and conidia germination in vitro, (ii) aggressiveness and sporulation capacity in vivo, (iii) sclerotia production in vitro and sclerotia viability under different storage conditions, and (iv) sensitivity to oxidative stress imposed by diquat treatments. The competitive ability of the resistant isolates was measured in the absence and presence of the SDHI fungicides boscalid and fluopyram selection pressure. The measurements of individual fitness components showed that the H272R/G143A isolates had the lower differences compared with the sensitive isolates. In contrast, the groups of H272Y/L/G143A, N230I/G143A, and P225F/G143A isolates showed reduced fitness values compared with the sensitive isolates. Isolates possessing only the cytb G143A substitution did not show any fitness cost. The competition experiments showed that, in the absence of fungicide selection pressure, after four disease cycles on apple fruit, the sensitive isolates dominated in the population in all the mixtures tested. In contrast, when the competition experiment was conducted under the selection pressure of boscalid, a gradual decrease in the frequency of sensitive isolates was observed, whereas the frequency of H272L and P225F isolates was increased. When the competition experiment was conducted in the presence of fluopyram, the sensitive isolates were eliminated even after the first disease cycle and the P225F mutants dominated in the population. Such results suggest that the sdhB mutations may have adverse effects on the mutants. The observed dominance of sensitive isolates in the competition experiments conducted in the absence of fungicides suggest that the application of SDHIs in alternation schemes may delay the selection or reduce the frequency of SDHI-resistant mutants.


Subject(s)
Botrytis/genetics , Drug Resistance, Fungal , Fungal Proteins/genetics , Fungicides, Industrial/pharmacology , Malus/microbiology , Plant Diseases/microbiology , Benzoquinones/antagonists & inhibitors , Botrytis/drug effects , Botrytis/growth & development , Botrytis/physiology , Fluocinolone Acetonide/pharmacology , Fruit/microbiology , Fungal Proteins/metabolism , Genotype , Microbial Sensitivity Tests , Mutation, Missense , Mycelium , Oxidative Stress , Phenotype , Spores, Fungal , Succinate Dehydrogenase/antagonists & inhibitors
9.
Proc Natl Acad Sci U S A ; 110(11): 4297-302, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23440205

ABSTRACT

As much as two-thirds of the proton gradient used for transmembrane free energy storage in oxygenic photosynthesis is generated by the cytochrome b6f complex. The proton uptake pathway from the electrochemically negative (n) aqueous phase to the n-side quinone binding site of the complex, and a probable route for proton exit to the positive phase resulting from quinol oxidation, are defined in a 2.70-Å crystal structure and in structures with quinone analog inhibitors at 3.07 Å (tridecyl-stigmatellin) and 3.25-Å (2-nonyl-4-hydroxyquinoline N-oxide) resolution. The simplest n-side proton pathway extends from the aqueous phase via Asp20 and Arg207 (cytochrome b6 subunit) to quinone bound axially to heme c(n). On the positive side, the heme-proximal Glu78 (subunit IV), which accepts protons from plastosemiquinone, defines a route for H(+) transfer to the aqueous phase. These pathways provide a structure-based description of the quinone-mediated proton transfer responsible for generation of the transmembrane electrochemical potential gradient in oxygenic photosynthesis.


Subject(s)
Benzoquinones/chemistry , Chlamydomonas reinhardtii/enzymology , Cytochrome b6f Complex/chemistry , Heme/chemistry , Protons , Benzoquinones/antagonists & inhibitors , Benzoquinones/metabolism , Cytochrome b6f Complex/metabolism , Heme/metabolism , Ion Transport/physiology , Membrane Potentials/physiology , Protein Structure, Quaternary , Protein Structure, Tertiary
10.
J Radiat Res ; 53(4): 545-50, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22843619

ABSTRACT

The purpose of this study is to clarify the effect of a heat shock protein 90 inhibitor, 17-allylamino-17-demethoxygeldanamycin (17-AAG), in combination with X-rays or carbon-ion beams on cell killing in human oral squamous cell carcinoma LMF4 cells. Cell survival was measured by colony formation assay. Cell-cycle distribution was analyzed by flow cytometry. Expression of DNA repair-related proteins was investigated by western blotting. The results showed 17-AAG to have synergistic effects on cell lethality with X-rays, but not with carbon-ion beams. The 17-AAG decreased G(2)/M arrest induced by X-rays, but not by carbon-ion beams. Both X-ray and carbon-ion irradiation up-regulated expression of non-homologous end-joining-associated proteins, Ku70 and Ku80, but 17-AAG inhibited only X-ray-induced up-regulation of these proteins. These results show that 17-AAG with X-rays releases G(2)/M phase arrest; cells carrying misrepaired DNA damage then move on to the G(1) phase. We demonstrate, for the first time, that the radiosensitization effect of 17-AAG is not seen with carbon-ion beams because 17-AAG does not affect these changes.


Subject(s)
Benzoquinones/antagonists & inhibitors , Carbon/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Ions/therapeutic use , Lactams, Macrocyclic/antagonists & inhibitors , Mouth Neoplasms/drug therapy , Mouth Neoplasms/radiotherapy , Cell Cycle , Cell Division , Cell Line, Tumor , Combined Modality Therapy/methods , Dose-Response Relationship, Radiation , G2 Phase , Humans , Time Factors , X-Rays
11.
Biochim Biophys Acta ; 1817(2): 287-97, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22037395

ABSTRACT

Functional roles of an anionic lipid phosphatidylglycerol (PG) were studied in pgsA-gene-inactivated and cdsA-gene-inactivated/phycobilisome-less mutant cells of a cyanobacterium Synechocystis sp. PCC 6803, which can grow only in PG-supplemented media. 1) A few days of PG depletion suppressed oxygen evolution of mutant cells supported by p-benzoquinone (BQ). The suppression was recovered slowly in a week after PG re-addition. Measurements of fluorescence yield indicated the enhanced sensitivity of Q(B) to the inactivation by BQ. It is assumed that the loss of low-affinity PG (PG(L)) enhances the affinity for BQ that inactivates Q(B). 2) Oxygen evolution without BQ, supported by the endogenous electron acceptors, was slowly suppressed due to the direct inactivation of Q(B) during 10 days of PG depletion, and was recovered rapidly within 10h upon the PG re-addition. It is concluded that the loss of high-affinity PG (PG(H)) displaces Q(B) directly. 3) Electron microscopy images of PG-depleted cells showed the specific suppression of division of mutant cells, which had developed thylakoid membranes attaching phycobilisomes (PBS). 4) Although the PG-depletion for 14 days decreased the chlorophyll/PBS ratio to about 1/4, flourescence spectra/lifetimes were not modified indicating the flexible energy transfer from PBS to different numbers of PSII. Longer PG-depletion enhanced allophycocyanin fluorescence at 683nm with a long 1.2ns lifetime indicating the suppression of energy transfer from PBS to PSII. 5) Action sites of PG(H), PG(L) and other PG molecules on PSII structure are discussed.


Subject(s)
Phosphatidylglycerols/metabolism , Photosystem II Protein Complex/chemistry , Photosystem II Protein Complex/metabolism , Plastoquinone/chemistry , Plastoquinone/metabolism , Benzoquinones/antagonists & inhibitors , Benzoquinones/metabolism , Binding Sites/drug effects , Catalysis/drug effects , Cell Shape/drug effects , Crystallography, X-Ray , Electron Transport/drug effects , Models, Biological , Models, Molecular , Organisms, Genetically Modified , Oxygen/metabolism , Oxygen/pharmacology , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/pharmacology , Pigments, Biological/chemistry , Pigments, Biological/metabolism , Protein Binding/drug effects , Synechocystis/cytology , Synechocystis/drug effects , Synechocystis/genetics , Synechocystis/metabolism
12.
Br J Pharmacol ; 161(3): 541-54, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20880395

ABSTRACT

BACKGROUND AND PURPOSE: Constitutive activation of the signal transducer and activator of transcription 3 (STAT3) pathway is frequently encountered in several human cancers including multiple myeloma (MM). Thus, agents that suppress STAT3 phosphorylation have a potential for treatment of MM. In the present report, we investigated whether thymoquinone (TQ), the main component isolated from the medicinal plant Nigella sativa, modulated the STAT3 signalling pathway in MM cells. EXPERIMENTAL APPROACH: The effect of TQ on both constitutive and IL-6-induced STAT3 activation, associated protein kinases, STAT3-regulated gene products involved in proliferation, survival and angiogenesis, cellular proliferation and apoptosis in MM cells, was investigated. KEY RESULTS: We found that TQ inhibited both constitutive and IL-6-inducible STAT3 phosphorylation which correlated with the inhibition of c-Src and JAK2 activation. Vanadate reversed the TQ-induced down-regulation of STAT3 activation, suggesting the involvement of a protein tyrosine phosphatase. Indeed, we found that TQ can induce the expression of Src homology-2 phosphatase 2 that correlated with suppression of STAT3 activation. TQ also down-regulated the expression of STAT3-regulated gene products, such as cyclin D1, Bcl-2, Bcl-xL, survivin, Mcl-1 and vascular endothelial growth factor. Finally, TQ induced the accumulation of cells in sub-G1 phase, inhibited proliferation and induced apoptosis, as indicated by poly ADP ribose polymerase cleavage. TQ also significantly potentiated the apoptotic effects of thalidomide and bortezomib in MM cells. CONCLUSIONS AND IMPLICATIONS: Our study has identified STAT3 signalling as a target of TQ and has thus raised its potential application in the prevention and treatment of MM and other cancers.


Subject(s)
Apoptosis/drug effects , Benzoquinones/pharmacology , Cell Proliferation/drug effects , Multiple Myeloma/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Benzoquinones/antagonists & inhibitors , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Down-Regulation/drug effects , Humans , Inositol Polyphosphate 5-Phosphatases , Interleukin-6/pharmacology , Janus Kinase 2/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Pyrazines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Thalidomide/pharmacology , Vanadates/pharmacology
13.
J Biol Chem ; 283(12): 7580-9, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18178560

ABSTRACT

Nonesterified fatty acids may influence mitochondrial function by alterations in gene expression, metabolism, and/or mitochondrial Ca(2+) ([Ca(2+)](m)) homeostasis. We have previously reported that polyunsaturated fatty acids induce Ca(2+) efflux from mitochondria, an action that may deplete [Ca(2+)](m) and thus contribute to nonesterified fatty acid-responsive mitochondrial dysfunction. Here we show that the chaperone protein heat shock protein 90 beta1 (hsp90beta1) is required for polyunsaturated fatty acid-induced mitochondrial Ca(2+) efflux (PIMCE). Retinoic acid induced differentiation of human teratocarcinoma NT2 cells in association with attenuation of PIMCE. Proteomic analysis of mitochondrial proteins revealed that hsp90beta1, among other proteins, was reduced in retinoic acid-differentiated cells. Blockade of PIMCE in NT2 cells by 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin, a known inhibitor of the chaperone activity of hsp90, and hsp90beta1 RNA interference demonstrated that hsp90beta1 is essential for PIMCE. We also show localization of hsp90beta1 in mitochondria by Western blot and immunofluorescence. Distinctive effects of inhibitors binding to the N or C terminus of hsp90 on PIMCE in isolated mitochondria suggested that the C terminus of hsp90beta1 plays a critical role in PIMCE.


Subject(s)
Calcium/metabolism , Fatty Acids, Unsaturated/metabolism , HSP90 Heat-Shock Proteins/metabolism , Homeostasis/physiology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Antineoplastic Agents/pharmacology , Benzoquinones/antagonists & inhibitors , Benzoquinones/pharmacology , Cell Line, Tumor , Fatty Acids, Unsaturated/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Homeostasis/drug effects , Humans , Lactams, Macrocyclic/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Mitochondria/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Proteomics , RNA Interference , Tretinoin/pharmacology
14.
Biochemistry ; 44(18): 6920-8, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15865437

ABSTRACT

The photosynthetic reaction center (RC) from purple bacteria converts light into chemical energy. Although the RC shows two nearly structurally symmetric branches, A and B, light-induced electron transfer in the native RC occurs almost exclusively along the A-branch to a primary quinone electron acceptor Q(A). Subsequent electron and proton transfer to a mobile quinone molecule Q(B) converts it to a quinol, Q(B)H(2). We report the construction and characterization of a series of mutants in Rhodobacter sphaeroides designed to reduce Q(B) via the B-branch. The quantum efficiency to Q(B) via the B-branch Phi(B) ranged from 0.4% in an RC containing the single mutation Ala-M260 --> Trp to 5% in a quintuple mutant which includes in addition three mutations to inhibit transfer along the A-branch (Gly-M203 --> Asp, Tyr-M210 --> Phe, Leu-M214 --> His) and one to promote transfer along the B-branch (Phe-L181 --> Tyr). Comparing the value of 0.4% for Phi(B) obtained in the AW(M260) mutant, which lacks Q(A), to the 100% quantum efficiency for Phi(A) along the A-branch in the native RC, we obtain a ratio for A-branch to B-branch electron transfer of 250:1. We determined the structure of the most effective (quintuple) mutant RC at 2.25 A (R-factor = 19.6%). The Q(A) site did not contain a quinone but was occupied by the side chain of Trp-M260 and a Cl(-). In this structure a nonfunctional quinone was found to occupy a new site near M258 and M268. The implications of this work to trap intermediate states are discussed.


Subject(s)
Benzoquinones/chemistry , Benzoquinones/metabolism , Photosynthetic Reaction Center Complex Proteins/genetics , Photosynthetic Reaction Center Complex Proteins/metabolism , Rhodobacter sphaeroides/genetics , Rhodobacter sphaeroides/metabolism , Bacteriochlorophylls/chemistry , Bacteriochlorophylls/genetics , Bacteriochlorophylls/metabolism , Benzoquinones/antagonists & inhibitors , Binding Sites/genetics , Crystallization , Crystallography, X-Ray , Electron Transport/genetics , Kinetics , Lasers , Models, Chemical , Mutagenesis, Site-Directed , Oxidation-Reduction , Pheophytins/chemistry , Pheophytins/genetics , Pheophytins/metabolism , Photolysis , Photosynthetic Reaction Center Complex Proteins/isolation & purification , Rhodobacter sphaeroides/growth & development , Spectrophotometry
15.
Bioorg Med Chem ; 13(9): 3185-95, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15809154

ABSTRACT

Various 2-thiopyrimidine derivatives have been synthesized by an efficient, one-pot reaction of functionalized amines with either 4-isothiocyanato-4-methyl-2-pentanone or 3-isothiocyanatobutanal. All the synthesized compounds were fully characterized by elemental analysis (CHN), FT-IR, (1)H NMR, and mass spectral data. One of the compounds, 7,7,8a-trimethyl-hexahydro-thiazolo[3,2-c]pyrimidine-5-thione (17) showed good anti-inflammatory (37.4% at 100 mg/kg p.o.) and analgesic activity (75% at 100 mg/kg p.o.). 7-(1-Mercapto-3,3,4a-trimethyl-4,4a,5,9b-tetrahydro-3H-pyrido[4,3-b]indol-7-yl)-3,3,4a-trimethyl-3,4,4a,5-tetrahydro-benzo[4,5]imidazo[1,2-c]pyrimidine-1-thiol (3) showed moderate activity against CDK-1 (IC(50)=5 microM). The other compounds showed moderate anti-inflammatory (5-20%), analgesic (25-75%) and protein kinase (CDK-5, GSK-3) inhibitory activities (IC(50)> 10 microM).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzoquinones/antagonists & inhibitors , Benzoquinones/pharmacology , Carrageenan/antagonists & inhibitors , Carrageenan/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinases/antagonists & inhibitors , Mice , Molecular Structure , Pyrimidines/chemistry , Rats , Structure-Activity Relationship , Sulfur Compounds/chemical synthesis , Sulfur Compounds/chemistry , Sulfur Compounds/pharmacology
16.
Biochemistry ; 43(16): 4755-63, 2004 Apr 27.
Article in English | MEDLINE | ID: mdl-15096044

ABSTRACT

In Rhodobacter sphaeroides reaction centers containing the mutation Ala M260 to Trp (AM260W), transmembrane electron transfer along the A-branch of cofactors is prevented by the loss of the QA ubiquinone. Reaction centers that contain this AM260W mutation are proposed to photoaccumulate the P(+)QB- radical pair following transmembrane electron transfer along the B-branch of cofactors (Wakeham, M. C., Goodwin, M. G., McKibbin, C., and Jones, M. R. (2003) Photoaccumulation of the P(+)QB- radical pair state in purple bacterial reaction centers that lack the QA ubiquinone. FEBS Lett. 540, 234-240). The yield of the P(+)QB- state appears to depend upon which additional mutations are present. In the present paper, Fourier transform infrared (FTIR) difference spectroscopy was used to demonstrate that photooxidation of the reaction center's primary donor in QA-deficient reaction centers results in formation of a semiquinone at the QB site by B-branch electron transfer. Reduction of QB by the B-branch pathway still occurs at 100 K, with a yield of approximately 10% relative to that at room temperature, in contrast to the QA- to QB reaction in the wild-type reaction center, which is not active at cryogenic temperatures. These FTIR results suggest that the conformational changes that "gate" the QA- to QB reaction do not necessarily have the same influence on QB reduction when the electron donor is the HB anion, at least in a minority of reaction centers.


Subject(s)
Benzoquinones/chemistry , Photosynthetic Reaction Center Complex Proteins/chemistry , Rhodobacter sphaeroides/chemistry , Alanine/genetics , Anti-Bacterial Agents/chemistry , Benzoquinones/antagonists & inhibitors , Benzoquinones/metabolism , Binding Sites/genetics , Electron Transport/genetics , Freezing , Light , Mutagenesis, Site-Directed , Oxidation-Reduction , Photosynthetic Reaction Center Complex Proteins/antagonists & inhibitors , Photosynthetic Reaction Center Complex Proteins/genetics , Polyenes/chemistry , Rhodobacter sphaeroides/genetics , Spectroscopy, Fourier Transform Infrared , Tryptophan/genetics , Ubiquinone/genetics , Ubiquinone/metabolism
18.
J Ethnopharmacol ; 88(1): 11-4, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12902046

ABSTRACT

Croton malambo (K.) bark aqueous extract, popularly known in Venezuela as "palomatias" or "torco" was tested for acute toxicity and for its anti-inflammatory and antinociceptive effects using tail flick and writhing syndrome tests models, respectively. Croton malambo aqueous extract (6.15 mg/kg i.p.) administered intraperitoneally had a significant antinociceptive and anti-inflammatory effects compared to acetylsalicylic acid (200mg/kg p.o.) and sodium diclofenac (5.64 mg/kg p.o.). Studies to determine correlation between chemical composition and pharmacological activity are underway.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Croton , Nociceptors/drug effects , Pain/drug therapy , Phytotherapy , Plant Bark/chemistry , Plant Extracts/therapeutic use , Albumins/adverse effects , Analgesics, Non-Narcotic/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzoquinones/adverse effects , Benzoquinones/antagonists & inhibitors , Edema/chemically induced , Edema/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Male , Medicine, Traditional , Mice , Morphine/pharmacology , Morphine/therapeutic use , Rats , Rats, Sprague-Dawley , Toxicity Tests, Acute , Venezuela
19.
Free Radic Res ; 30(2): 93-103, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10193577

ABSTRACT

In low concentrations, benzene and its metabolite hydroquinone are known to have diverse biological effects on cells, including the synergistic stimulation with GM-CSF of hematopoietic colony formation in vitro, stimulation of granulocytic differentiation in vitro and in vivo, and general suppression of hematopoiesis in vivo. These chemicals are also known to be active in the induction of active oxygen species. We used several assays to determine the effects of benzene metabolites (hydroquinone, benzenetriol, benzoquinone) and active oxygen species (xanthine/xanthine oxidase) on cell growth and cell cycle kinetics of the human myeloid cell line HL-60. HL-60 cells treated with these chemicals for 2 h in PBS showed increased growth over untreated controls in a subsequent 18h growth period in complete media. Incorporation of 3H-thymidine was also increased proportionately by these treatments. Catalase treatment abrogated the increased cell growth of all chemicals, suggesting an oxidative mechanism for the effect of all treatments alike. Cell cycle kinetics assays showed that the growth increase was caused by an increased recruitment of cells from G0/G1 to S-phase for both hydroquinone and active oxygen, rather than a decrease in the length of the cell cycle. Benzene metabolite's enhancement of growth of myeloid cells through an active oxygen mechanism may be involved in a number of aspects of benzene toxicity, including enhanced granulocytic growth and differentiation, stimulation of GM-CSF-induced colony formation, apoptosis inhibition, and stimulation of progenitor cell mitogenesis in the bone marrow. These effects in sum may be involved in the benzene-induced "promotion" of a clonal cell population to the fully leukemic state.


Subject(s)
Benzene/metabolism , Cell Division/drug effects , Hydroquinones/pharmacology , Reactive Oxygen Species/metabolism , Benzoquinones/antagonists & inhibitors , Benzoquinones/pharmacology , Catalase/metabolism , Cell Cycle/drug effects , DNA/biosynthesis , Dose-Response Relationship, Drug , Flow Cytometry , HL-60 Cells , Humans , Hydroquinones/antagonists & inhibitors , Oxygen/metabolism , Time Factors , Xanthine/metabolism , Xanthine Oxidase/metabolism
20.
Drugs ; 52 Suppl 5: 24-45; discussion 45-6, 1996.
Article in English | MEDLINE | ID: mdl-8922555

ABSTRACT

Dexketoprofen trometamol is a water-soluble salt of the dextrorotatory enantiomer of the nonsteroidal anti-inflammatory drug (NSAID) ketoprofen. Racemic ketoprofen is used as an analgesic and an anti-inflammatory agent, and is one of the most potent in vitro inhibitors of prostaglandin synthesis. This effect is due to the S(+)-enantiomer (dexketoprofen), while the R(-)-enantiomer is devoid of such activity. The pharmacokinetic profile of ketoprofen and its enantiomers was assessed in several animals species and in human volunteers. In humans, the relative bioavailability of oral dexketoprofen trometamol (12.5 and 25 mg, respectively) is similar to that of oral racemic ketoprofen (25 and 50 mg, respectively), as measured in all cases by the area under the concentration-time curve values for S(+)-ketoprofen. Dexketoprofen trometamol, given as a tablet, is rapidly absorbed, with a time to maximum plasma concentration (tmax) of between 0.25 and 0.75 hours, whereas the tmax for the S-enantiomer after the racemic drug, administered as tablets or capsules prepared with the free acid, is between 0.5 and 3 hours. Peak plasma concentrations of 1.4 and 3.1 mg/L are reached after administration of dexketoprofen trometamol 12.5 and 25 mg, respectively. From 70 to 80% of the administered dose is recovered in the urine during the first 12 hours, mainly as the acyl-glucuronoconjugated parent drug. No R(-)-ketoprofen is found in the urine after administration of dexketoprofen [S(+)-ketoprofen], confirming the absence of bioinversion of the S(+)-enantiomer in humans. in animal studies, the anti-inflammatory potency of dexketoprofen was always equivalent to that demonstrated by twice the dose of ketoprofen. Similarly, animal studies showed a high analgesic potency for dexketoprofen trometamol. The R(-)-enantiomer demonstrated a much lower potency, its analgesic action being apparent only in conditions where the metabolic bioinversion to the S(+)-enantiomer was significant. The gastric ulcerogenic effect of dexketoprofen at various oral doses (1.5 to 6 mg/kg) in the rat do not differ from those of the corresponding double doses (3 to 12 mg/kg) of racemic ketoprofen. Repeated (5-day) oral administration of dexketoprofen as the trometamol salt causes less gastric ulceration than was observed after the acid form of both dexketoprofen and the racemate. In addition, single dose dexketoprofen as the free acid at 10 to 20 mg/kg does not show a significant intestinal ulcerogenic effect in rats, while racemic ketoprofen 20 or 40 mg/kg is clearly ulcerogenic to the small intestine. The analgesic efficacy of oral dexketoprofen trometamol 10 to 20 mg is superior to that of placebo and similar to that of ibuprofen 400 mg in patients with moderate to serve pain after third molar extraction. The time to onset of pain relief appeared to be shorter in patients treated with dexketoprofen trometamol than in those treated with ibuprofen 400 mg. Dexketoprofen trometamol was well tolerated, with a reported incidence of adverse events similar to that of placebo.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Ketoprofen/therapeutic use , Analgesia , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoquinones/antagonists & inhibitors , Cyclooxygenase Inhibitors/pharmacology , Digestive System/drug effects , Ethacrynic Acid/antagonists & inhibitors , Humans , Inflammation/drug therapy , Intestinal Diseases/chemically induced , Intestine, Small/drug effects , Ketoprofen/chemistry , Ketoprofen/pharmacokinetics , Ketoprofen/pharmacology , Mice , Pain/chemically induced , Pain/drug therapy , Rats , Stereoisomerism , Stomach Ulcer/chemically induced , Ulcer/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...