Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Am Chem Soc ; 146(20): 13875-13885, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38718165

ABSTRACT

Bioluminescence is a fascinating natural phenomenon, wherein organisms produce light through specific biochemical reactions. Among these organisms, Renilla luciferase (RLuc) derived from the sea pansy Renilla reniformis is notable for its blue light emission and has potential applications in bioluminescent tagging. Our study focuses on RLuc8, a variant of RLuc with eight amino acid substitutions. Recent studies have shown that the luminescent emitter coelenteramide can adopt multiple protonation states, which may be influenced by nearby residues at the enzyme's active site, demonstrating a complex interplay between protein structure and bioluminescence. Herein, using the quantum mechanical consistent force field method and the semimacroscopic protein dipole-Langevin dipole method with linear response approximation, we show that the phenolate state of coelenteramide in RLuc8 is the primary light-emitting species in agreement with experimental results. Our calculations also suggest that the proton transfer (PT) from neutral coelenteramide to Asp162 plays a crucial role in the bioluminescence process. Additionally, we reproduced the observed emission maximum for the amide anion in RLuc8-D120A and the pyrazine anion in the presence of a Na+ counterion in RLuc8-D162A, suggesting that these are the primary emitters. Furthermore, our calculations on the neutral emitter in the engineered AncFT-D160A enzyme, structurally akin to RLuc8-D162A but with a considerably blue-shifted emission peak, aligned with the observed data, possibly explaining the variance in emission peaks. Overall, this study demonstrates an effective approach to investigate chromophores' bimolecular states while incorporating the PT process in emission spectra calculations, contributing valuable insights for future studies of PT in photoproteins.


Subject(s)
Pyrazines , Quantum Theory , Pyrazines/chemistry , Pyrazines/metabolism , Renilla/enzymology , Luciferases/chemistry , Luciferases/metabolism , Luminescence , Animals , Imidazoles/chemistry , Benzeneacetamides
2.
Int J Mol Sci ; 24(3)2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36768474

ABSTRACT

Ca2+-triggered coelenterazine-binding protein (CBP) is a natural form of the luciferase substrate involved in the Renilla bioluminescence reaction. It is a stable complex of coelenterazine and apoprotein that, unlike coelenterazine, is soluble and stable in an aquatic environment and yields a significantly higher bioluminescent signal. This makes CBP a convenient substrate for luciferase-based in vitro assay. In search of a similar substrate form for the luciferase NanoLuc, a furimazine-apoCBP complex was prepared and verified against furimazine, coelenterazine, and CBP. Furimazine-apoCBP is relatively stable in solution and in a frozen or lyophilized state, but as distinct from CBP, its bioluminescence reaction with NanoLuc is independent of Ca2+. NanoLuc turned out to utilize all the four substrates under consideration. The pairs of CBP-NanoLuc and coelenterazine-NanoLuc generate bioluminescence with close efficiency. As for furimazine-apoCBP-NanoLuc pair, the efficiency with which it generates bioluminescence is almost twice lower than that of the furimazine-NanoLuc. The integral signal of the CBP-NanoLuc pair is only 22% lower than that of furimazine-NanoLuc. Thus, along with furimazine as the most effective NanoLuc substrate, CBP can also be recommended as a substrate for in vitro analytical application in view of its water solubility, stability, and Ca2+-triggering "character".


Subject(s)
Carrier Proteins , Luminescent Measurements , Animals , Carrier Proteins/metabolism , Luciferases/metabolism , Renilla , Calcium/metabolism
3.
Biochem Biophys Res Commun ; 587: 24-28, 2022 01 08.
Article in English | MEDLINE | ID: mdl-34864391

ABSTRACT

Coelenterazine (CTZ) is known as luciferin (a substrate) for the luminescence reaction with luciferase (an enzyme) in marine organisms and is unstable in aqueous solutions. The dehydrogenated form of CTZ (dehydrocoelenterazine, dCTZ) is stable and thought to be a storage form of CTZ and a recycling intermediate from the condensation reaction of coelenteramine and 4-hydroxyphenylpyruvic acid to CTZ. In this study, the enzymatic conversion of dCTZ to CTZ was successfully achieved using NAD(P)H:FMN oxidoreductase from the bioluminescent bacterium Vibrio fischeri ATCC 7744 (FRase) in the presence of NADH (the FRase-NADH reaction). CTZ reduced from dCTZ in the FRase-NADH reaction was identified by HPLC and LC/ESI-TOF-MS analyses. Thus, dCTZ can be enzymatically converted to CTZ in vitro. Furthermore, the concentration of dCTZ could be determined by the luminescence activity using the CTZ-utilizing luciferases (Gaussia luciferase or Renilla luciferase) coupled with the FRase-NADH reaction.


Subject(s)
Aliivibrio fischeri/enzymology , Bacterial Proteins/metabolism , Imidazoles/metabolism , Luciferases/metabolism , NADH, NADPH Oxidoreductases/metabolism , Pyrazines/metabolism , Renilla/enzymology , Aliivibrio fischeri/genetics , Animals , Bacterial Proteins/genetics , Biocatalysis , Biotransformation , Chromatography, High Pressure Liquid , Flavin Mononucleotide/metabolism , Gene Expression , Kinetics , Luciferases/genetics , Luminescence , Luminescent Measurements , NADH, NADPH Oxidoreductases/genetics , Phenylpyruvic Acids/metabolism , Renilla/genetics
4.
J Virol ; 93(23)2019 12 01.
Article in English | MEDLINE | ID: mdl-31511391

ABSTRACT

The nonstructural protein NS5A of hepatitis C virus (HCV) is a phosphorylated protein that is indispensable for viral replication and assembly. We previously showed that NS5A undergoes sequential serine S232/S235/S238 phosphorylation resulting in NS5A transition from a hypo- to a hyperphosphorylated state. Here, we studied functions of S229 with a newly generated antibody specific to S229 phosphorylation. In contrast to S232, S235, or S238 phosphorylation detected only in the hyperphosphorylated NS5A, S229 phosphorylation was found in both hypo- and hyperphosphorylated NS5A, suggesting that S229 phosphorylation initiates NS5A sequential phosphorylation. Immunoblotting showed an inverse relationship between S229 phosphorylation and S235 phosphorylation. When S235 was phosphorylated as in the wild-type NS5A, the S229 phosphorylation level was low; when S235 could not be phosphorylated as in the S235A mutant NS5A, the S229 phosphorylation level was high. These results suggest an intrinsic feedback regulation between S229 phosphorylation and S235 phosphorylation. It has been known that NS5A distributes in large static and small dynamic intracellular structures and that both structures are required for the HCV life cycle. We found that S229A or S229D mutation was lethal to the virus and that both increased NS5A in large intracellular structures. Similarly, the lethal S235A mutation also increased NS5A in large structures. Likewise, the replication-compromised S235D mutation also increased NS5A in large structures, albeit to a lesser extent. Our data suggest that S229 probably cycles through phosphorylation and dephosphorylation to maintain a delicate balance of NS5A between hypo- and hyperphosphorylated states and the intracellular distribution necessary for the HCV life cycle.IMPORTANCE This study joins our previous efforts to elucidate how NS5A transits between hypo- and hyperphosphorylated states via phosphorylation on a series of highly conserved serine residues. Of the serine residues, serine 229 is the most interesting since phosphorylation-mimicking and phosphorylation-ablating mutations at this serine residue are both lethal. With a new high-quality antibody specific to serine 229 phosphorylation, we concluded that serine 229 must remain wild type so that it can dynamically cycle through phosphorylation and dephosphorylation that govern NS5A between hypo- and hyperphosphorylated states. Both are required for the HCV life cycle. When phosphorylated, serine 229 signals phosphorylation on serine 232 and 235 in a sequential manner, leading NS5A to the hyperphosphorylated state. As serine 235 phosphorylation is reached, serine 229 is dephosphorylated, stopping signal for hyperphosphorylation. This balances NS5A between two phosphorylation states and in intracellular structures that warrant a productive HCV life cycle.


Subject(s)
Hepacivirus/metabolism , Serine/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , HEK293 Cells , Hepatitis C/virology , Humans , Phosphorylation , Protein Kinases/metabolism , Proteomics , Renilla , Viral Nonstructural Proteins/chemistry , Virus Replication/physiology
5.
Int J Mol Sci ; 22(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374392

ABSTRACT

Two G-quadruplex forming oligonucleotides [d(TG4T)4 and d(TG6T)4] were selected as two tetramolecular quadruplex nanostructures because of their demonstrated ability to be modified with hydrophobic molecules. This allowed us to synthesize two series of G-quadruplex conjugates that differed in the number of G-tetrads, as well as in the terminal position of the lipid modification. Both solution and solid-phase syntheses were carried out to yield the corresponding lipid oligonucleotide conjugates modified at their 3'- and 5'-termini, respectively. Biophysical studies confirmed that the presence of saturated alkyl chains with different lengths did not affect the G-quadruplex integrity, but increased the stability. Next, the G-quadruplex domain was added to an 18-mer antisense oligonucleotide. Gene silencing studies confirmed the ability of such G-rich oligonucleotides to facilitate the inhibition of target Renilla luciferase without showing signs of toxicity in tumor cell lines.


Subject(s)
G-Quadruplexes , Lipids/chemistry , Nanostructures/chemistry , Oligonucleotides/genetics , Animals , Biophysics , Cell Line, Tumor , Circular Dichroism , HEK293 Cells , HeLa Cells , Humans , Luciferases/metabolism , Microscopy, Fluorescence , Nucleic Acid Conformation , Oligonucleotides/chemistry , Oligonucleotides, Antisense , Renilla/enzymology , Transfection
6.
Pharm Dev Technol ; 25(7): 855-864, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32188321

ABSTRACT

G protein-coupled receptor (GPCR) 87, is overexpressed in various cancer cells especially pancreatic cancer and plays a critical role in tumor cell survival. Nano-particles (NP) have become the essential vehicles for nucleotide internalization to the cell, due to the negative charge of nucleotides and their poor stability in blood circulation. In this study, the HEK293T cell linewas transfected with GPR87-plasmid after which the double-stranded RNA molecules targeting the GPR87 gene were prepared and purified. 1.1B4 cancer cell lines were used as model pancreatic cancer cells. Produced siRNA molecules were encapsulated in Poly(Lactic-Co-Glycolic Acid) (PLGA) nano-micelles using three different methods, two of which were according to literature with (siR-PLGA-S) or without (siR-PLGA-V) sonication. However, a new method was suggested to overcome problems such as poly-dispersity and large sizes of siR-PLGA-S and siR-PLGA-V. The new method consists of encapsulating siRNA using mild agitation to the pre-made PLGA NPs. The latter method provided mono-dispersed particles (siR-P-PLGA) with 92 nm size and desired Encapsulation Efficiency (EE%). siR-P-PLGA was able to silence the GPR-87 gene in a ratio of 83.9%, almost 41 times more effective than siR-PLGA-S and siR-PLGA-V in HEK 293 T cells. siR-P-PLGA was able to show a mild cytotoxic effect on 1.1B4 pancreatic cancer cells within 48 h.


Subject(s)
Gene Targeting/methods , Nanoparticles , Pancreatic Neoplasms/genetics , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Small Interfering/genetics , Receptors, Lysophosphatidic Acid/genetics , Animals , Cell Line, Tumor , Gene Silencing/physiology , Genetic Engineering/methods , HEK293 Cells , Humans , Nanoparticles/administration & dosage , Pancreatic Neoplasms/therapy , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , RNA, Small Interfering/administration & dosage , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Renilla
7.
Arch Toxicol ; 93(11): 3141-3152, 2019 11.
Article in English | MEDLINE | ID: mdl-31515601

ABSTRACT

The chemical warfare agent sulfur mustard (SM) alkylates a multitude of biomacromolecules including DNA and proteins. Cysteine residues and nucleophilic nitrogen atoms in purine DNA bases are typical targets of SM but potentially every nucleophilic structure may be alkylated by SM. In the present study, we analyzed potential SM-induced alkylation of glucocorticoid (GC) hormones and functional consequences thereof. Hydrocortisone (HC), the synthetic betamethasone (BM) and dexamethasone (DEX) were chosen as representative GCs. Structural modifications were assessed by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy. The hypothesized alkylation was verified and structurally allocated to the OH-group of the C21 atom. The biological function of SM-alkylated GCs was investigated using GC-regulated dual-luciferase reporter gene assays and an ex vivo GC responsiveness assay coupled with real-time quantitative polymerase chain reaction (RT-qPCR). For the reporter gene assays, HEK293-cells were transiently transfected with a dual-luciferase reporter gene that is transcriptional regulated by a GC-response element. These cells were then incubated either with untreated or SM-derivatized HC, BM or DEX. Firefly-luciferase (Fluc) activity was determined 24 h after stimulation. Fluc-activity significantly decreased after stimulation with SM-pre-exposed GC dependent on the SM concentration. The ex vivo RT-qPCR-based assay for human peripheral leukocyte responsiveness to DEX revealed a transcriptional dysregulation of GC-regulated genes (FKBP5, IL1R2, and GILZ) after stimulation with SM-alkylated DEX. Our results present GCs as new biological targets of SM associated with a disturbance of hormone function.


Subject(s)
Alkylating Agents/toxicity , Chemical Warfare Agents/toxicity , Gene Expression Regulation/drug effects , Glucocorticoids/metabolism , Mustard Gas/toxicity , Animals , Betamethasone/pharmacology , Cotinine/analogs & derivatives , Cotinine/pharmacology , Dexamethasone/pharmacology , Genes, Reporter , Glucocorticoids/genetics , HEK293 Cells , Humans , Luciferases/genetics , Renilla , Transfection
8.
Anal Chem ; 90(21): 12986-12993, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30234965

ABSTRACT

Sensitive and selective quantification of individual sugars in complex media is technically challenging and usually requires HPLC separation. Accurate measurement without the need for separation would be highly desirable. The measurement of trace levels of lactose in lactose-reduced milk exemplifies the problem, with the added challenge that trace lactose must be measured in the presence of ≈140 mM glucose and galactose, the products of lactase digestion of lactose. Biosensing is an alternative to HPLC, but current biosensing methods, based on coupled-enzyme assays, tend to have poor sensitivity and complex biochemistry and can be time-consuming. We explored a fundamentally different approach, based on identifying a lactose-specific binding protein compatible with photonic transduction. We identified the BgaR transcriptional regulator of Clostridium perfringens, which is highly selective for lactose, as a suitable ligand binding domain and combined it with a bioluminescence energy resonance transfer transduction system. This BRET-based biosensor showed a 27% decrease in the BRET ratio in the presence of saturating (1 mM) lactose. Using a 5 min assay, the half maximal effective concentration (EC50) for lactose in phosphate-buffered saline (PBS) was 12 µM. The biosensor was 200 times more sensitive to lactose than to glucose or galactose. Sensitivity and selectivity were not significantly affected by the presence of 10% (v/v) dialyzed milk. The biosensor is suitable for direct determination of residual lactose in lactase-treated milk, with a limit of detection of 0.2 µM, 100 times below the most stringent lactose-free standard and without the need to remove fat or protein from the sample.


Subject(s)
Bacterial Proteins/chemistry , Biosensing Techniques/methods , Lactose/analysis , Milk/chemistry , Transcription Factors/chemistry , Agrobacterium tumefaciens/chemistry , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Clostridium perfringens/chemistry , Energy Transfer , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lactose/metabolism , Ligands , Limit of Detection , Luminescence , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Renilla/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Chembiochem ; 19(13): 1409-1413, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29656613

ABSTRACT

(2'S)-2'-Deoxy-2'-C-methyluridine and (2'R)-2'-deoxy-2'-C-methyluridine were incorporated in the 3'-overhang region of the sense and antisense strands and in positions 2 and 5 of the seed region of siRNA duplexes directed against Renilla luciferase, whereas (2'S)-2'-deoxy-2'-C-methylcytidine was incorporated in the 6-position of the seed region of the same constructions. A dual luciferase reporter assay in transfected HeLa cells was used as a model system to measure the IC50 values of 24 different modified duplexes. The best results were obtained by the substitution of one thymidine unit in the antisense 3'-overhang region by (2'S)- or (2'R)-2'-deoxy-2'-C-methyluridine, reducing IC50 to half of the value observed for the natural control. The selectivity of the modified siRNA was measured, it being found that modifications in positions 5 and 6 of the seed region had a positive effect on the ON/OFF activity.


Subject(s)
RNA, Small Interfering/chemistry , Uridine/analogs & derivatives , Animals , Enzyme Assays , HeLa Cells , Humans , Inhibitory Concentration 50 , Luciferases, Renilla/genetics , RNA Stability , RNA, Small Interfering/chemical synthesis , RNA, Small Interfering/genetics , Renilla/enzymology , Stereoisomerism , Temperature , Uridine/chemistry
10.
Protein Expr Purif ; 145: 39-44, 2018 05.
Article in English | MEDLINE | ID: mdl-29288731

ABSTRACT

Luciferase from Renilla reniformis (RLuc) is a good research tool as a reporter protein and bioimaging probes, yielding blue light using the substrate coelenterazine. However, the applications are limited since RLuc is unstable under various conditions. Therefore, an attempt was made to increase RLuc thermostability. In this study, 5 mutations reported previously [1] and one mutation obtained using site-directed mutagenesis were combined. As a result of this combination, the thermostability effect increased, with the mutant showing approximately 10 °C higher stability. Furthermore, the mutant simultaneously improved a tolerance for protease digestion, e.g. trypsin and proteinase K, and for organic solvent. Residual activity of the mutant after treatment with 10% 2-propanol, 10% DMF and 20% DMSO at 35 °C for 1 h was 29.4, 24.8 and 91.3%, respectively, whereas that of the wild type was 0.4, 0.1 and 24.3%, respectively.


Subject(s)
Hot Temperature , Luciferases, Renilla/metabolism , Mutagenesis, Site-Directed , Renilla/enzymology , Animals , Enzyme Stability , Luciferases, Renilla/chemistry , Luciferases, Renilla/genetics , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 112(14): 4352-6, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25831507

ABSTRACT

Fluorescence live imaging has become an essential methodology in modern cell biology. However, fluorescence requires excitation light, which can sometimes cause potential problems, such as autofluorescence, phototoxicity, and photobleaching. Furthermore, combined with recent optogenetic tools, the light illumination can trigger their unintended activation. Because luminescence imaging does not require excitation light, it is a good candidate as an alternative imaging modality to circumvent these problems. The application of luminescence imaging, however, has been limited by the two drawbacks of existing luminescent protein probes, such as luciferases: namely, low brightness and poor color variants. Here, we report the development of bright cyan and orange luminescent proteins by extending our previous development of the bright yellowish-green luminescent protein Nano-lantern. The color change and the enhancement of brightness were both achieved by bioluminescence resonance energy transfer (BRET) from enhanced Renilla luciferase to a fluorescent protein. The brightness of these cyan and orange Nano-lanterns was ∼20 times brighter than wild-type Renilla luciferase, which allowed us to perform multicolor live imaging of intracellular submicron structures. The rapid dynamics of endosomes and peroxisomes were visualized at around 1-s temporal resolution, and the slow dynamics of focal adhesions were continuously imaged for longer than a few hours without photobleaching or photodamage. In addition, we extended the application of these multicolor Nano-lanterns to simultaneous monitoring of multiple gene expression or Ca(2+) dynamics in different cellular compartments in a single cell.


Subject(s)
Luciferases/chemistry , Luminescence , Luminescent Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Calcium/metabolism , Cell Line , DNA/chemistry , Dogs , Embryonic Stem Cells/cytology , Endosomes/metabolism , Fluorescence Resonance Energy Transfer , Focal Adhesions , Gene Expression Regulation , Luciferases, Renilla/metabolism , Mice , Molecular Sequence Data , Oligonucleotides/chemistry , Peroxisomes/metabolism , Promoter Regions, Genetic , Renilla , Vinculin/chemistry
12.
J Neuroinflammation ; 14(1): 111, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28577576

ABSTRACT

BACKGROUND: Sphingosine 1-phosphate (S1P) signals through G protein-coupled receptors to elicit a wide range of cellular responses. In CNS injury and disease, the blood-brain barrier is compromised, causing leakage of S1P from blood into the brain. S1P can also be locally generated through the enzyme sphingosine kinase-1 (Sphk1). Our previous studies demonstrated that S1P activates inflammation in murine astrocytes. The S1P1 receptor subtype has been most associated with CNS disease, particularly multiple sclerosis. S1P3 is most highly expressed and upregulated on astrocytes, however, thus we explored the involvement of this receptor in inflammatory astrocytic responses. METHODS: Astrocytes isolated from wild-type (WT) or S1P3 knockout (KO) mice were treated with S1P3 selective drugs or transfected with short interfering RNA to determine which receptor subtypes mediate S1P-stimulated inflammatory responses. Interleukin-6 (IL-6), and vascular endothelial growth factor A (VEGFa) messenger RNA (mRNA) and cyclooxygenase-2 (COX-2) mRNA and protein were assessed by q-PCR and Western blotting. Activation of RhoA was measured using SRE.L luciferase and RhoA implicated in S1P signaling by knockdown of Gα12/13 proteins or by inhibiting RhoA activation with C3 exoenzyme. Inflammation was simulated by in vitro scratch injury of cultured astrocytes. RESULTS: S1P3 was highly expressed in astrocytes and further upregulated in response to simulated inflammation. Studies using S1P3 knockdown and S1P3 KO astrocytes demonstrated that S1P3 mediates activation of RhoA and induction of COX-2, IL-6, and VEGFa mRNA, with some contribution from S1P2. S1P induces expression of all of these genes through coupling to the Gα12/13 proteins which activate RhoA. Studies using S1P3 selective agonists/antagonists as well as Fingolimod (FTY720) confirmed that stimulation of S1P3 induces COX-2 expression in astrocytes. Simulated inflammation increased expression of Sphk1 and consequently activated S1P3, demonstrating an autocrine pathway through which S1P is formed and released from astrocytes to regulate COX-2 expression. CONCLUSIONS: S1P3, through its ability to activate RhoA and its upregulation in astrocytes, plays a unique role in inducing inflammatory responses and should be considered as a potentially important therapeutic target for CNS disease progression.


Subject(s)
Astrocytes/metabolism , Gene Expression/physiology , Receptors, Lysosphingolipid/metabolism , Signal Transduction/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Cyclooxygenase 2/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression/drug effects , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Lysosphingolipid/genetics , Renilla , Signal Transduction/drug effects , Transfection , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , rhoA GTP-Binding Protein/genetics
15.
Org Biomol Chem ; 15(48): 10238-10244, 2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29177293

ABSTRACT

The prodrug or caged-luciferin strategy affords an excellent platform for persistent bioluminescence imaging. In the current work, we designed and synthesized ten novel pro-substrates for Renilla luciferase by introducing ester protecting groups of different sizes into the carbonyl group of the free luciferin 1. Taking advantage of intracellular esterases, lipases, and nucleophilic substances, the ester protecting groups were hydrolyzed, resulting in the release of a free luciferin and a bioluminescence signal turn-on. Among the tested pro-substrates, the butyryloxymethyl luciferin 7 exhibited low cytotoxicity and a prolonged luminescence signal both in cellulo and in vivo. Therefore, the butyryloxymethyl luciferin 7 can act as a promising substrate for noninvasive extended imaging in diagnostic and therapeutic fields.


Subject(s)
Firefly Luciferin/chemistry , Luciferases/analysis , Luminescent Measurements , Renilla/enzymology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Female , Firefly Luciferin/chemical synthesis , Firefly Luciferin/pharmacology , Humans , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Substrate Specificity
16.
Bioconjug Chem ; 27(2): 354-62, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26322739

ABSTRACT

Optical imaging of protein-protein interactions (PPIs) facilitates comprehensive elucidation of intracellular molecular events. We demonstrate an optical measure for visualizing molecular tension triggered by any PPI in mammalian cells. Twenty-three kinds of candidate designs were fabricated, in which a full-length artificial luciferase (ALuc) was sandwiched between two model proteins of interest, e.g., FKBP and FRB. One of the designs greatly enhanced the bioluminescence in response to varying concentrations of rapamycin. It is confirmed with negative controls that the elevated bioluminescence is solely motivated from the molecular tension. The probe design was further modified toward eliminating the C-terminal end of ALuc and was found to improve signal-to-background ratios, named "a combinational probe". The utilities were elucidated with detailed substrate selectivity, bioluminescence imaging of live cells, and different PPI models. This study expands capabilities of luciferases as a tool for analyses of molecular dynamics and cell signaling in living subjects.


Subject(s)
Luciferases, Renilla/metabolism , Molecular Probes/metabolism , Protein Interaction Mapping/methods , TOR Serine-Threonine Kinases/metabolism , Tacrolimus Binding Proteins/metabolism , Amino Acid Sequence , Animals , Biomechanical Phenomena , COS Cells , Chlorocebus aethiops , Humans , Luciferases, Renilla/chemistry , Luminescent Measurements/methods , Molecular Probes/chemistry , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Renilla/chemistry , Renilla/enzymology , TOR Serine-Threonine Kinases/chemistry , Tacrolimus Binding Proteins/chemistry
17.
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
18.
Biochem Biophys Res Commun ; 458(3): 543-548, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25677617

ABSTRACT

Luciferase assay has become an increasingly important technique to monitor a wide range of biological processes. However, the mainstay protocols require a luminometer to acquire and process the data, therefore limiting its application to specialized research labs. To overcome this limitation, we have developed an alternative protocol that utilizes a commonly available cooled charge-coupled device (CCCD), instead of a luminometer for data acquiring and processing. By measuring activities of different luciferases, we characterized their substrate specificity, assay linearity, signal-to-noise levels, and fold-changes via CCCD. Next, we defined the assay parameters that are critical for appropriate use of CCCD for different luciferases. To demonstrate the usefulness in cultured mammalian cells, we conducted a case study to examine NFκB gene activation in response to inflammatory signals in human embryonic kidney cells (HEK293 cells). We found that data collected by CCCD camera was equivalent to those acquired by luminometer, thus validating the assay protocol. In comparison, The CCCD-based protocol is readily amenable to live-cell and high-throughput applications, offering fast simultaneous data acquisition and visual and quantitative data presentation. In conclusion, the CCCD-based protocol provides a useful alternative for monitoring luciferase reporters. The wide availability of CCCD will enable more researchers to use luciferases to monitor and quantify biological processes.


Subject(s)
Luciferases, Firefly/analysis , Luciferases, Renilla/analysis , Luminescent Agents/analysis , Luminescent Measurements/instrumentation , Animals , Fireflies/enzymology , Genes, Reporter , HEK293 Cells , High-Throughput Screening Assays/instrumentation , Humans , Luciferases, Firefly/genetics , Luciferases, Renilla/genetics , Luminescent Agents/metabolism , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Renilla/enzymology , Transfection
19.
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
20.
J Neurochem ; 129(1): 99-106, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24215460

ABSTRACT

Protein aggregation is a common feature of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration. How protein aggregates are formed and contribute to neurodegeneration, however, is not clear. Mutation of Ubiquilin 2 (UBQLN2) has recently been linked to ALS and frontotemporal lobar degeneration. Therefore, we examined the effect of ALS-linked UBQLN2 mutation on endoplasmic reticulum-associated protein degradation (ERAD). Compared to its wild-type counterpart, mutated UBQLN2 caused greater accumulation of the ERAD substrate Hong Kong variant of α-1-antitrypsin, although ERAD was disturbed by both UBQLN2 over-expression and knockdown. Also, UBQLN2 interacted with ubiquitin regulatory X domain-containing protein 8 (UBXD8) in vitro and in vivo, and this interaction was impaired by pathogenic mutation of UBQLN2. As UBXD8 is an endoplasmic membrane protein involved in the translocation of ubiquitinated ERAD substrates, UBQLN2 likely cooperates with UBXD8 to transport defective proteins from the endoplasmic reticulum to the cytosol for degradation, and this cell-protective function is disturbed by pathogenic mutation of UBQLN2.


Subject(s)
Blood Proteins/metabolism , Cell Cycle Proteins/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Mutation/physiology , Proteolysis , Ubiquitins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autophagy-Related Proteins , Blood Proteins/genetics , Cell Cycle Proteins/genetics , Cell Survival/physiology , Cells, Cultured , Chick Embryo , Chickens , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Membrane Proteins/genetics , Protein Binding/physiology , Protein Transport/physiology , Renilla , Ubiquitins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL