Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
Biotechnol Lett ; 43(8): 1585-1594, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33945054

ABSTRACT

OBJECTIVES: Firefly luciferase, one of the most extensively studied enzymes, has numerous applications. However, luciferase activity is inhibited by sodium chloride. This study was aimed at obtaining mutant luciferase enzymes resistant to the sodium chloride inhibition. RESULTS: We first obtained two mutant luciferase enzymes whose inhibition were alleviated and determined the mutations to be Val288Ile and Glu488Val. Under medical dialysis condition (140 mM sodium chloride), the wild type was inhibited to 44% of its original activity level. In contrast, the single mutants, Val288Ile and Glu488Val, retained 67% and 79% of their original activity, respectively. Next, we introduced Val288Ile and Glu488Val mutations into wild-type luciferase to create a double mutant using site-directed mutagenesis. Notably, the double mutant retained its activity more than 95% of that in the absence of sodium chloride. CONCLUSIONS: The mutant luciferase, named luciferase CR, was found to retain its activity in various concentrations of sodium chloride. The luciferase CR may be extensively useful in any bioassay which includes firefly luciferase and is employed in the presence of sodium chloride.


Subject(s)
Luciferases, Firefly/antagonists & inhibitors , Sodium Chloride/pharmacology , Animals , Escherichia coli , Fireflies/enzymology , Fireflies/genetics , Luciferases, Firefly/genetics , Luminescent Measurements , Mutagenesis, Site-Directed , Mutant Proteins/antagonists & inhibitors
2.
Bioorg Chem ; 94: 103395, 2020 01.
Article in English | MEDLINE | ID: mdl-31733898

ABSTRACT

Firefly luciferase (FLuc) is a powerful tool for molecular and cellular biology, and popular in high-throughput screening and drug discovery. However, FLuc assays have been plagued with positive and negative artefacts due to stabilisation and inhibition by small molecules from a range of chemical classes. Here we disclose Phase II clinical compound SMT C1100 for the treatment of Duchenne muscular dystrophy as an FLuc inhibitor (KD of 0.40 ±â€¯0.15 µM). Enzyme kinetic studies using SMT C1100 and other non-competitive inhibitors including resveratrol and NFκBAI4 identified previously undescribed modes of inhibition with respect to FLuc's luciferyl adenylate intermediate. Employing a photoaffinity strategy to identify SMT C1100's binding site, a photolabelled SMT C1100 probe instead underwent FLuc-dependent photooxidation. Our findings support novel binding sites on FLuc for non-competitive inhibitors.


Subject(s)
Benzoxazoles/pharmacology , Enzyme Inhibitors/pharmacology , Fireflies/enzymology , Luciferases, Firefly/antagonists & inhibitors , Animals , Benzoxazoles/chemical synthesis , Benzoxazoles/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Kinetics , Luciferases, Firefly/metabolism , Molecular Structure , Structure-Activity Relationship
3.
Biotechnol Appl Biochem ; 66(5): 755-762, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31021480

ABSTRACT

The therapeutic potential of microRNA-21 (miR-21) small-molecule inhibitors has been of particular interest to medicinal chemists. Moreover, the development of more facile screening methods is lacking. In the present study, two potential screening strategies for miR-21 small-molecule inhibitor including the stem-loop reverse transcription-quantitative PCR and dual luciferase reporter assay system were demonstrated and discussed in detail. A pmirGLO-miR21cswt plasmid and its two different mutants were constructed for dual luciferase reporter assay system. In addition, the sensitivity and specificity of these two methods were validated. Our results demonstrated that both strategies are decent choices for the screening of small-molecule inhibitors for miR-21 and possibly other miRNAs. Eventually, we applied our optimized strategy to discover and characterize several promising compounds such as azobenzene derivate A, enoxacin, and norfloxacin for their potential impact on intracellular miR-21 concentration.


Subject(s)
Genes, Reporter/drug effects , Luciferases, Firefly/antagonists & inhibitors , MicroRNAs/pharmacology , Real-Time Polymerase Chain Reaction , Small Molecule Libraries/pharmacology , Drug Evaluation, Preclinical , Genes, Reporter/genetics , HeLa Cells , High-Throughput Screening Assays , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Tumor Cells, Cultured
4.
Anal Chem ; 89(11): 6099-6105, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28492074

ABSTRACT

Chalcone refers to an aromatic ketone and an enone that constitutes the central core for various important biological compounds in drug discovery. Moreover, the firefly luciferase (Fluc) as the bioluminescent reporter has been widely used in life science research and high-throughput screening (HTS). However, Fluc might suffer from direct inhibition by HTS compounds resulting in the occurrence of "false positives." In the current research, we discovered a series of chalcone compounds as Fluc inhibitors with favorable potency both in vitro and in vivo. Moreover, our compound 3i showed remarkable systemic inhibition in transgenic mice. Both enzymatic kinetics study and cocrystal structure demonstrated that compound 3i is competitive for substrate aminoluciferin, while noncompetitive for ATP. Besides, compound 3i exhibited excellent selectivity as a promising quenching agent in a simulated dual-luciferase reporter assay. We believed that our research would contribute to improving scientists' awareness of the Fluc inhibitors, pay attention to the bias results, and even expand the utilization of bioluminescence in life science research.


Subject(s)
Chalcones/pharmacology , Enzyme Inhibitors/pharmacology , Luciferases, Firefly/antagonists & inhibitors , Luminescence , Animals , Cell Line, Tumor , Chalcones/chemistry , Enzyme Inhibitors/chemistry , Female , Fireflies , Luciferases, Firefly/isolation & purification , Luciferases, Firefly/metabolism , Luminescent Measurements , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Molecular Structure
5.
Photochem Photobiol Sci ; 15(2): 244-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26789132

ABSTRACT

The luciferase reporter gene assay system is broadly applied in various biomedical aspects, including signaling pathway dissection, transcriptional activity analysis, and genetic toxicity testing. It significantly improves the experimental accuracy and reduces the experimental error by the addition of an internal control. In the current research, we discovered some specific ions that could selectively inhibit firefly luciferase while having a negligible effect on renilla luciferase in vitro in the dual-reporter gene assay. We showed that these ionic compounds had a high potential of being utilized as quench-and-activate reagents in the dual-reporter assay. Furthermore, results from kinetic studies on ion-mediated quenching effects indicated that different ions have distinct inhibition modes. Our study is anticipated to guide a more affordable design of quench-and-activate reagents in biomedicine and pharmaceutical analysis.


Subject(s)
Fireflies/enzymology , Ions/metabolism , Luciferases, Firefly/metabolism , Luciferases, Renilla/metabolism , Luminescent Agents/metabolism , Renilla/enzymology , Animals , Enzyme Assays , Fireflies/genetics , Genes, Reporter , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Firefly/genetics , Luciferases, Renilla/antagonists & inhibitors , Luciferases, Renilla/genetics , Luminescence , Renilla/genetics
6.
Anal Biochem ; 487: 45-8, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26212314

ABSTRACT

Reporter gene assays are widely used for the assessment of transcription factor activation following xenobiotic exposure of cells. A critical issue with such assays is the possibility of interference of test compounds with the test system, for example, by direct inhibition of the reporter enzyme. Here we show that the pyrrolizidine alkaloid heliotrine interferes with reporter signals derived from GAL4-based nuclear receptor transactivation assays by a mechanism independent of luciferase enzyme inhibition. These data highlight the necessity to conduct proper control experiments in order to avoid perturbation of reporter assays by test chemicals.


Subject(s)
Genes, Reporter/drug effects , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Renilla/antagonists & inhibitors , Pyrrolizidine Alkaloids/pharmacology , Animals , Fireflies , Genes, Reporter/genetics , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Luciferases, Renilla/genetics , Luciferases, Renilla/metabolism , Pyrrolizidine Alkaloids/chemistry , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renilla
7.
Bioconjug Chem ; 25(1): 4-10, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24341748

ABSTRACT

In this paper, novel firefly luciferase-specific inhibitor compounds (FLICs) are evaluated as potential tools for cellular trafficking of transporter conjugates. As a proof-of-concept, we designed FLICs that were suitable for solid phase peptide synthesis and could be covalently conjugated to peptides via an amide bond. The spacer between inhibitor and peptide was optimized to gain efficient inhibition of recombinant firefly luciferase (FLuc) without compromising the activity of the model peptides. The hypothesis of using FLICs as tools for cellular trafficking studies was ensured with U87Fluc glioblastoma cells expressing firefly luciferase. Results show that cell penetrating peptide (penetratin) FLIC conjugate 9 inhibited FLuc penetrated cells efficiently (IC50 = 1.6 µM) and inhibited bioluminescence, without affecting the viability of the cells. Based on these results, peptide-FLIC conjugates can be used for the analysis of cellular uptake of biomolecules in a new way that can at the same time overcome some downsides seen with other methods. Thus, FLICs can be considered as versatile tools that broaden the plethora of methods that take advantage of the bioluminescence phenomena.


Subject(s)
Carrier Proteins/chemistry , Fireflies/enzymology , Isoxazoles/chemistry , Isoxazoles/pharmacology , Luminescence , Animals , Carrier Proteins/metabolism , Cell-Penetrating Peptides , Dose-Response Relationship, Drug , Humans , Isoxazoles/pharmacokinetics , Kinetics , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Firefly/metabolism , Luminescent Measurements , Molecular Structure , Structure-Activity Relationship , Time Factors , Tissue Distribution
8.
Nanotechnology ; 24(34): 345101, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23899823

ABSTRACT

We report on the dose-dependent inhibition of firefly luciferase activity induced by exposure of the enzyme to 20 nm citrate-coated silver nanoparticles (AgNPs). The inhibition mechanism was examined by characterizing the physicochemical properties and biophysical interactions of the enzyme and the AgNPs. Consistently, binding of the enzyme induced an increase in zeta potential from -22 to 6 mV for the AgNPs, triggered a red-shift of 44 nm in the absorbance peak of the AgNPs, and rendered a 'protein corona' of 20 nm in thickness on the nanoparticle surfaces. However, the secondary structures of the enzyme were only marginally affected upon formation of the protein corona, as verified by circular dichroism spectroscopy measurement and multiscale discrete molecular dynamics simulations. Rather, inductively coupled plasma mass spectrometry measurement revealed a significant ion release from the AgNPs. The released silver ions could readily react with the cysteine residues and N-groups of the enzyme to alter the physicochemical environment of their neighboring catalytic site and subsequently impair the enzymatic activity.


Subject(s)
Luciferases, Firefly/metabolism , Metal Nanoparticles/chemistry , Silver/metabolism , Gold/analysis , Ions , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Firefly/chemistry , Metal Nanoparticles/ultrastructure , Molecular Dynamics Simulation , Protein Structure, Secondary , Silver/analysis , Spectrophotometry, Ultraviolet , Static Electricity
9.
Proc Natl Acad Sci U S A ; 107(11): 4878-83, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20194791

ABSTRACT

Firefly luciferase (FLuc), an ATP-dependent bioluminescent reporter enzyme, is broadly used in chemical biology and drug discovery assays. PTC124 (Ataluren; (3-[5-(2-fluorophenyl)-1,2,4-oxadiazol-3-yl]benzoic acid) discovered in an FLuc-based assay targeting nonsense codon suppression, is an unusually potent FLuc-inhibitor. Paradoxically, PTC124 and related analogs increase cellular FLuc activity levels by posttranslational stabilization. In this study, we show that FLuc inhibition and stabilization is the result of an inhibitory product formed during the FLuc-catalyzed reaction between its natural substrate, ATP, and PTC124. A 2.0 A cocrystal structure revealed the inhibitor to be the acyl-AMP mixed-anhydride adduct PTC124-AMP, which was subsequently synthesized and shown to be a high-affinity multisubstrate adduct inhibitor (MAI; K(D) = 120 pM) of FLuc. Biochemical assays, liquid chromatography/mass spectrometry, and near-attack conformer modeling demonstrate that formation of this novel MAI is absolutely dependent upon the precise positioning and reactivity of a key meta-carboxylate of PTC124 within the FLuc active site. We also demonstrate that the inhibitory activity of PTC124-AMP is relieved by free coenzyme A, a component present at high concentrations in luciferase detection reagents used for cell-based assays. This explains why PTC124 can appear to increase, instead of inhibit, FLuc activity in cell-based reporter gene assays. To our knowledge, this is an unusual example in which the "off-target" effect of a small molecule is mediated by an MAI mechanism.


Subject(s)
Luciferases, Firefly/metabolism , Models, Molecular , Oxadiazoles/metabolism , Adenosine Monophosphate/metabolism , Cell Line , Coenzyme A/metabolism , Crystallography, X-Ray , Enzyme Assays , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Stability/drug effects , Humans , Isomerism , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Firefly/chemistry , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Solutions , Substrate Specificity/drug effects , Temperature
10.
Bioorg Med Chem Lett ; 22(20): 6433-41, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22981335

ABSTRACT

2-Substituted pyrrolo[2,3-b]quinoxalines having free NH were prepared directly from 3-alkynyl-2-chloroquinoxalines in a single pot by using readily available and inexpensive methane sulfonamide (or p-toluene sulfonamide) as an ammonia surrogate. The reaction proceeded in the presence of Cu(OAc)(2) affording the desired product in moderate yield. The crystal structure analysis of a representative compound and its supramolecular interactions are presented. Some of the compounds synthesized exhibited inhibitory activities against luciferase that was supported by the predictive binding mode of these compounds with luciferase enzyme through molecular docking studies. The key observations disclosed here can alert users of luciferase reporter gene assays for possible false positive results due to the direct inhibition of luciferase.


Subject(s)
Fireflies/enzymology , Luciferases, Firefly/antagonists & inhibitors , Quinoxalines/chemistry , Quinoxalines/pharmacology , Animals , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Genes, Reporter/drug effects , Models, Molecular , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacology , Quinoxalines/chemical synthesis
11.
Bioorg Med Chem ; 20(18): 5473-82, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22898255

ABSTRACT

A novel firefly luciferase inhibitor (3a) with a pyrrolo[2,3-d]pyrimidine core was identified in a cell-based NF-κB luciferase reporter gene assay. It potently inhibited the firefly luciferase derived from Photinus pyralis with an IC(50) value of 0.36 ± 0.05 µM. Kinetic analysis of 3a inhibition showed that it is predominantly competitive with respect to D-luciferin and uncompetitive with respect to ATP. Therefore, several pyrrolo[2,3-d]pyrimidine analogues were prepared to further investigate the structure-activity relationship (SAR) for luciferase inhibition. The most potent inhibitor of this series was 4c, which showed an IC(50) value of 0.06 ± 0.01 µM. In addition, molecular docking studies suggested that both 3a and 4c could be accommodated in the D-luciferin binding pocket, which is expected for a predominantly competitive inhibitor with respect to D-luciferin.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Luciferases, Firefly/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Luciferases, Firefly/metabolism , Models, Molecular , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship
12.
Photochem Photobiol Sci ; 10(6): 1039-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21409209

ABSTRACT

The inhibition mechanisms of the firefly luciferase (Luc) by three of the most important inhibitors of the reactions catalysed by Luc, dehydroluciferyl-coenzyme A (L-CoA), dehydroluciferin (L) and L-luciferin (L-LH(2)) were investigated. Light production in the presence and absence of these inhibitors (0.5 to 2 µM) has been measured in 50 mM Hepes buffer (pH = 7.5), 10 nM Luc, 250 µM ATP and D-luciferin (D-LH(2), from 3.75 up to 120 µM). Nonlinear regression analysis with the appropriate kinetic models (Henri-Michaelis-Menten and William-Morrison equations) reveals that L-CoA is a non-competitive inhibitor of Luc (K(i) = 0.88 ± 0.03 µM), L is a tight-binding uncompetitive inhibitor (K(i) = 0.00490 ± 0.00009 µM) and L-LH(2) acts as a mixed-type non-competitive-uncompetitive inhibitor (K(i) = 0.68 ± 0.14 µM and αK(i) = 0.34 ± 0.16 µM). The K(m) values obtained for L-CoA, L and L-LH(2) were 16.1 ± 1.0, 16.6 ± 2.3 and 14.4 ± 0.96 µM, respectively. L and L-LH(2) are strong inhibitors of Luc, which may indicate an important role for these compounds in Luc characteristic flash profile. L-CoA K(i) supports the conclusion that CoA can stimulate the light emission reaction by provoking the formation of a weaker inhibitor.


Subject(s)
Coenzyme A/chemistry , Firefly Luciferin/chemistry , Luciferases, Firefly/antagonists & inhibitors , Animals , Fireflies/enzymology , Kinetics , Luciferases, Firefly/metabolism , Protein Binding , Stereoisomerism
13.
J Nat Prod ; 74(6): 1500-2, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21639131

ABSTRACT

A prefractionated Streptomyces-derived extract was initially identified as being active using a luciferase-based AMP-activated protein kinase (AMPK) assay. Bioassay-guided fractionation led to the isolation of the new compound quinazolin-4(3H)-one (1) as the active component. However, 1 was shown to have potent firefly luciferase inhibitory activity with no effect on AMPK. This is the first report of a natural luciferase inhibitor.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Biological Products/isolation & purification , Biological Products/pharmacology , Luciferases, Firefly/antagonists & inhibitors , Quinazolinones/isolation & purification , Quinazolinones/pharmacology , Streptomyces/chemistry , Animals , Biological Products/chemistry , Luciferases, Firefly/metabolism , Molecular Structure , Quinazolinones/chemistry
15.
J Photochem Photobiol B ; 210: 111980, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32745950

ABSTRACT

The three hypoxia-inducible factor (HIF) prolyl-4-hydroxylase domain (PHD) 1-3 enzymes confer oxygen sensitivity to the HIF pathway and are novel therapeutic targets for treatment of renal anemia. Inhibition of the PHDs may further be beneficial in other hypoxia-associated diseases, including ischemia and chronic inflammation. Several pharmacologic PHD inhibitors (PHIs) are available, but our understanding of their selectivity and its chemical basis is limited. We here report that the PHI JNJ-42041935 (JNJ-1935) is structurally similar to the firefly luciferase substrate D-luciferin. Our results demonstrate that JNJ-1935 is a novel inhibitor of firefly luciferase enzymatic activity. In contrast, the PHIs FG-4592 (roxadustat) and FG-2216 (ICA, BIQ, IOX3, YM 311) did not affect firefly luciferase. The JNJ-1935 mode of inhibition is competitive with a Ki of 1.36 µM. D-luciferin did not inhibit the PHDs, despite its structural similarity to JNJ-1935. This study provides insights into a previously unknown JNJ-1935 off-target effect as well as into the chemical requirements for firefly luciferase and PHD inhibitors and may inform the development of novel compounds targeting these enzymes.


Subject(s)
Luciferases, Firefly/metabolism , Prolyl-Hydroxylase Inhibitors/chemistry , Animals , Benzothiazoles/metabolism , Binding, Competitive , Fireflies/enzymology , Glycine/analogs & derivatives , Glycine/chemistry , Glycine/metabolism , Isoquinolines/chemistry , Isoquinolines/metabolism , Kinetics , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Firefly/genetics , Prolyl-Hydroxylase Inhibitors/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Renilla/enzymology
16.
BMC Immunol ; 9: 49, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18710581

ABSTRACT

BACKGROUND: Interleukin 1 beta (IL-1beta) plays an important role in a number of chronic and acute inflammatory diseases. To understand the role of IL-1beta in disease processes and develop an in vivo screening system for anti-inflammatory drugs, a transgenic mouse line was generated which incorporated the transgene firefly luciferase gene driven by a 4.5-kb fragment of the human IL-1beta gene promoter. Luciferase gene expression was monitored in live mice under anesthesia using bioluminescence imaging in a number of inflammatory disease models. RESULTS: In a LPS-induced sepsis model, dramatic increase in luciferase activity was observed in the mice. This transgene induction was time dependent and correlated with an increase of endogenous IL-1beta mRNA and pro-IL-1beta protein levels in the mice. In a zymosan-induced arthritis model and an oxazolone-induced skin hypersensitivity reaction model, luciferase expression was locally induced in the zymosan injected knee joint and in the ear with oxazolone application, respectively. Dexamethasone suppressed the expression of luciferase gene both in the acute sepsis model and in the acute arthritis model. CONCLUSION: Our data suggest that the transgenic mice model could be used to study transcriptional regulation of the IL-1beta gene expression in the inflammatory process and evaluation the effect of anti-inflammatory drug in vivo.


Subject(s)
Interleukin-1beta/genetics , Luciferases, Firefly/genetics , Luminescent Proteins/genetics , Transcriptional Activation/genetics , Animals , Dexamethasone/administration & dosage , Disease Models, Animal , Gene Expression , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Luciferases, Firefly/antagonists & inhibitors , Luminescent Proteins/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Rheumatic Fever/chemically induced , Rheumatic Fever/genetics , Rheumatic Fever/immunology , Sepsis/chemically induced , Sepsis/genetics , Sepsis/immunology , Zymosan/administration & dosage , Zymosan/adverse effects
17.
Sci Rep ; 8(1): 4694, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549271

ABSTRACT

RNA interference (RNAi)-based gene therapy has great potential in cancer and infectious disease treatment to correct abnormal up-regulation of gene expression. We show a new original method uses synthetic microRNAs combined with a thermo-inducible promoter to reduce specific gene expression. The targeted gene is the luciferase firefly reporter gene overexpressed in a subcutaneous tumor which allows the RNAi monitoring by bioluminescence imaging (BLI). The inducible inhibition was first demonstrated in vitro using genetically modified cells lines and then in vivo using the corresponding xenograft model in mice. Achieving spatio-temporal control, we demonstrate the feasibility to induce, in vivo, a specific gene inhibition on demand. Future applications of this RNAi-based gene therapy, which can be restricted to pathological tissue, would offer wide-ranging potential for disease treatment.


Subject(s)
Fever , Gene Silencing , Glioblastoma/pathology , Luciferases, Firefly/metabolism , Luminescent Measurements , MicroRNAs/genetics , Optical Imaging/methods , Animals , Female , Glioblastoma/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Firefly/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Promoter Regions, Genetic , RNA, Small Interfering , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Biochim Biophys Acta ; 1721(1-3): 124-9, 2005 Jan 18.
Article in English | MEDLINE | ID: mdl-15652187

ABSTRACT

We reported that anesthetics increased the partial molal volume of firefly luciferase (FFL), while long-chain fatty acids (LCFA) decreased it. The present study measured the actions of dodecanol (neutral), dodecanoic acid (negatively charged), and dodecylamine (positively charged) hydrophobic molecules on FFL. The interaction modes are measured by (1) ATP-induced bioluminescence of FFL and (2) fluorescence of 2-(p-toluidino)naphthalene-6-sulfonate (TNS). TNS fluoresces brightly in hydrophobic media. It competes with the substrate luciferin on the FFL binding. From the Scatchard plot of TNS titration, the maximum binding number of TNS was 0.83, and its binding constant was 8.27 x 10(5) M(-1). Job's plot also showed that the binding number is 0.89. From the TNS titration of FFL, the binding constant was estimated to be 8.8 x 10(5) M(-1). Dodecanoic acid quenched the TNS fluorescence entirely. Dodecanol quenched about 25% of the fluorescence, whereas dodecylamine increased it. By comparing the fluorescence of TNS and bioluminescence of FFL, the binding modes and the inhibition mechanisms of these dodecane analogues are classified in three different modes: competitive (dodecanoic acid), noncompetitive (dodecylamine), and mixed (dodecanol).


Subject(s)
Amines/pharmacology , Anesthetics/pharmacology , Dodecanol/pharmacology , Lauric Acids/pharmacology , Luciferases, Firefly/antagonists & inhibitors , Binding Sites , Fluorescence , Protein Denaturation , Temperature
20.
Neurosci Lett ; 403(1-2): 103-8, 2006 Jul 31.
Article in English | MEDLINE | ID: mdl-16701950

ABSTRACT

The AMPA receptor subunit GluR2 is downregulated in neurons following a wide range of neurological insults. Here we report that suppression of GluR2 gene promoter activity is associated with kainate (KA)-induced downregulation of GluR2 subunit levels in primary cultured cortical neurons. RT-PCR and Northern blotting showed a significant decrease in GluR2 mRNA in cultured neurons after KA exposure. Transfection of cultured neurons with an expression vector pGL3-GluR2(-298/+283), where the reporter gene firefly luciferase was driven by the GluR2 promoter, revealed that KA exposure suppressed the transcriptional activation of the GluR2 promoter. Furthermore, the expression of the RE1-silencing transcription factor (REST) was increased in KA-exposed cortical neurons; enhanced binding of REST to RE1-like silencer element in the proximal promoter of the GluR2 subunit gene was evidenced by electrophoresis mobility shift assay. Chromatin immunoprecipitation showed that suppressed activity of the GluR2 promoter in cultured neurons after KA exposure was related to deacetylation of histone H4. These results indicate that REST as a crucial factor binds to RE1-like silencer element in the GluR2 promoter, suppressing transcription of the GluR2 subunit gene during KA exposure. Our data suggest that transcriptional suppression of the GluR2 subunit gene may contribute at least in part to downregulation of GluR2 subunit protein in neurons during KA exposure. Because our experiments showed a reduction of glutamate release in KA-exposed cortical neurons, REST may play a latent role in delayed neuronal death or in seizure-induced tolerance.


Subject(s)
Cerebral Cortex/metabolism , Kainic Acid/metabolism , Neurons/metabolism , Receptors, AMPA/antagonists & inhibitors , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Acetylation , Animals , Blotting, Northern , Cells, Cultured , Cerebral Cortex/cytology , Chromatin Immunoprecipitation , Down-Regulation , Electrophoretic Mobility Shift Assay , Genes, Reporter , Histones/metabolism , Kainic Acid/toxicity , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Firefly/genetics , Neurons/drug effects , Promoter Regions, Genetic , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , RNA, Messenger/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Silencer Elements, Transcriptional
SELECTION OF CITATIONS
SEARCH DETAIL