Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Protein J ; 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38824468

ABSTRACT

Renilla luciferase catalyzes the oxidation of coelenterazine to coelenteramide and results in the emission of a photon of light. Although Renilla luciferase has various applications in biotechnology, its low thermal stability limits the development of its applications. Arginine is a well-known stabilizing amino acid that plays a key role in protein stabilization against inactivation. However, its impact on enzyme properties is unpredictable. This study investigates the impact of arginine on the kinetics and thermal stability of Renilla luciferase. The enzyme's performance was significantly enhanced in the presence of arginine, with catalytic efficiency increasing by 3.31-fold and 3.08-fold when exposed to 0.2 M and 0.3 M arginine, respectively. Additionally, arginine improved the thermal stability of Renilla luciferase. Molecular dynamics simulation showed that the addition of 0.2 M arginine reduced the binding of coelenteramide, the reaction product and an enzyme inhibitor, to the active site of the Renilla luciferase. Therefore, the release of the product was accelerated, and the affinity of Renilla luciferase for coelenterazine increased. Furthermore, Molecular dynamics studies indicated an increased network of water molecules surrounding Renilla luciferase in the presence of 0.2 M arginine. This network potentially enhances the hydrophobic effect on the protein structure, ultimately improving enzyme stability. The findings of this study hold promise for the development of commercial kits incorporating Renilla luciferase.

2.
Anal Biochem ; 692: 115558, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38735426

ABSTRACT

Commercially available glow luciferase assay kits are widely popular and convenient to use. However, concerning high-throughput screening, commercial kits are limited by huge running costs. As an alternative to commercial luciferase assay kits, this study presents a cost-effective and efficient methodology of performing a simple and rapid laboratory flash luciferase assay. The proposed luciferase assay method has a versatile use ranging from screening lysates in a microplate reader for quantitative assay as well as screening live cells qualitatively or quantitatively under an imaging system.


Subject(s)
Luciferases , Luciferases/metabolism , Luciferases/chemistry , Luciferases/genetics , Humans , Luminescent Measurements/methods , Reproducibility of Results , High-Throughput Screening Assays/methods , High-Throughput Screening Assays/economics , Enzyme Assays/methods
3.
Methods Mol Biol ; 2755: 77-89, 2024.
Article in English | MEDLINE | ID: mdl-38319570

ABSTRACT

Hypoxia is a hallmark of ischemic cardiovascular diseases and solid malignant tumors. Cellular hypoxia induces numerous physiological and pathological processes, including hematopoiesis, angiogenesis, metabolic changes, cell growth, and apoptosis. Hypoxia-inducible factor-1 (HIF-1) binds to hypoxia response elements (HREs) to selectively induce the expression of various genes in response to hypoxia. Therefore, HREs have been used to develop hypoxia-targeted gene therapy.More than 70 pairs of HREs and hypoxia-inducible genes have been identified. The hypoxia-induced gene expression levels vary among HRE sequences depending on the number of HRE copies and oxygen levels. Most known HREs have not yet been thoroughly studied. Recent studies have revealed that the HRE-mediated effects of hypoxia are cell line-dependent. Herein we describe an in vitro method to investigate gene activation levels and characteristics based on varying the copy number of HREs in response to cellular hypoxia. We explain how to clone HREs into luciferase reporter constructs in the sense, antisense, and dual directions to measure luciferase expression for functional analyses.


Subject(s)
Hypoxia , Oxygen , Humans , Cell Hypoxia , Hypoxia/genetics , Apoptosis/genetics , Luciferases/genetics
4.
Biochem Biophys Rep ; 37: 101617, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38371529

ABSTRACT

Renilla luciferase catalyzes the oxidation of coelenterazine to coelenteramide, resulting in the emission of a photon of light. This study investigated the impact of sorbitol on the structural and kinetic properties of Renilla luciferase using circular dichroism, fluorescence spectroscopy, and molecular dynamics simulations. Our investigation, carried out using circular dichroism and fluorescence analyses, as well as a thermal stability assay, has revealed that sorbitol induces conformational changes in the enzyme but does not improve its thermal stability. Moreover, through kinetic studies, it has been demonstrated that at a concentration of 0.4 M, sorbitol enhances the catalytic efficiency of Renilla luciferase. However, at higher concentrations, sorbitol results in a decrease in catalytic efficiency. Additionally, molecular dynamics simulations have shown that sorbitol increases the presence of hydrophobic pockets on the enzyme's surface. These simulations have also provided evidence that at a concentration of 0.4 M, sorbitol facilitates substrate access to the active site of the enzyme. Nevertheless, at higher concentrations, sorbitol obstructs substrate trafficking, most likely due to its impact on the gateway to the active site. This study may provide insights into the kinetic changes observed in enzymes with buried active sites, such as those with α/ß hydrolase fold.

5.
World J Gastroenterol ; 29(40): 5543-5556, 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37970476

ABSTRACT

BACKGROUND: Phosphatidylinositol-3-kinases (PI3K) is a well-known route in inflammation-related cancer. Recent discovery on PI3K-related genes revealed a potential variant that links ulcerative colitis (UC) and colorectal cancer (CRC) with colitis-associated cancer (CAC). PI3K/AKT pathway has been recommended as a potential additional therapeutic option for CRC due to its substantial role in modifying cellular processes. Buparlisib is a pan-class I PI3K inhibitor previously shown to reduce tumor growth. AIM: To investigate the regulation of rs10889677 and the role of buparlisib in the PI3K signaling pathway in CAC pathogenesis. METHODS: Genomic DNA from 32 colonic samples, including CAC (n = 7), UC (n = 10) and CRC (n = 15), was sequenced for the rs10889677 mutation. The mutant and wildtype fragments were amplified and cloned in the pmirGLO vector. The luciferase activity of cloned vectors was assessed after transfection into the HT29 cell line. CAC mice were induced by a mixture of a single azoxymethane injection and three cycles of dextran sulphate sodium, then buparlisib was administered after 14 d. The excised colon was subjected to immunohistochemistry for Ki67 and Cleaved-caspase-3 markers and quantitative real-time polymerase chain reaction analysis for Pdk1 and Sgk2. RESULTS: Luciferase activity decreased by 2.07-fold in the rs10889677 mutant, confirming the hypothesis that the variant disrupted miRNA binding sites, which led to an increase in IL23R expression and the activation of the PI3K signaling pathway. Furthermore, CAC-induced mice had a significantly higher disease activity index (P < 0.05). Buparlisib treatment significantly decreased mean weight loss in CAC-induced mice (P < 0.05), reduced the percentage of proliferating cells by 5%, and increased the number of apoptotic cells. The treatment also caused a downward trend of Pdk1 expression and significantly decreased Sgk2 expression. CONCLUSION: Our findings suggested that the rs10889677 variant as a critical initiator of the PI3K signaling pathway, and buparlisib had the ability to prevent PI3K-non-AKT activation in the pathophysiology of CAC.


Subject(s)
Aminopyridines , Colitis, Ulcerative , Colitis-Associated Neoplasms , Colitis , Colonic Neoplasms , Morpholines , Mice , Animals , Phosphatidylinositol 3-Kinases/metabolism , Colitis-Associated Neoplasms/complications , Signal Transduction/genetics , Inflammation/complications , Colitis, Ulcerative/complications , Colonic Neoplasms/pathology , Phosphatidylinositols/adverse effects , Luciferases , Colitis/chemically induced , Colitis/complications , Colitis/drug therapy
6.
Biochem Biophys Res Commun ; 665: 133-140, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37163933

ABSTRACT

Coelenterazine (CTZ) is known as a light-emitting source for the bioluminescence reaction in marine organisms. CTZ has two phenolic hydroxy groups at the C2-benzyl and C6-phenyl positions, and a keto-enol type hydroxy group at the C3-position in the core structure of imidazopyrazinone (= 3,7-dihydroimidazopyrazin-3-one). These hydroxy groups in CTZ could be sulfated by sulfotransferase(s), and the sulfates of Watasenia luciferin (CTZ disulfate at the C2- and C6-positions) and Renilla pre-luciferin (CTZ 3-enol sulfate) have been identified in marine organisms. To characterize the sulfation process of CTZ, human cytosolic aryl sulfotransferase SULT1A1 (SUTase) was used as a model enzyme. The sulfated products catalyzed by SUTase with 3'-phosphoadenosine 5'-phosphosulfate (PAPS) were analyzed by LC/ESI-TOF-MS. The product was the monosulfate of CTZ and identified as the C2-benzyl sulfate of CTZ (CTZ C2-benzyl monosulfate), but CTZ disulfate, CTZ 3-enol sulfate, and CTZ C6-phenyl monosulfate were not detected. The non-enzymatic oxidation products of dehydrocoelenterazine (dCTZ, dehydrogenated derivative of CTZ), coelenteramide (CTMD), and coelenteramine (CTM) from CTZ were also identified as their monosulfates.


Subject(s)
Arylsulfotransferase , Imidazoles , Humans , Imidazoles/chemistry , Sulfotransferases , Luciferins , Sulfates
7.
Sensors (Basel) ; 23(7)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37050557

ABSTRACT

Imaging protein-protein interactions (PPIs) is a hot topic in molecular medicine in the postgenomic sequencing era. In the present study, we report bright and highly sensitive single-chain molecular strain probe templates which embed full-length Renilla luciferase 8.6-535SG (RLuc86SG) or Artificial luciferase 49 (ALuc49) as reporters. These reporters were deployed between FKBP-rapamycin binding domain (FRB) and FK506-binding protein (FKBP) as a PPI model. This unique molecular design was conceptualized to exploit molecular strains of the sandwiched reporters appended by rapamycin-triggered intramolecular PPIs. The ligand-sensing properties of the templates were maximized by interface truncations and substrate modulation. The highest fold intensities, 9.4 and 16.6, of the templates were accomplished with RLuc86SG and ALuc49, respectively. The spectra of the templates, according to substrates, revealed that the colors are tunable to blue, green, and yellow. The putative substrate-binding chemistry and the working mechanisms of the probes were computationally modeled in the presence or absence of rapamycin. Considering that the molecular strain probe templates are applicable to other PPI models, the present approach would broaden the scope of the bioassay toolbox, which harnesses the privilege of luciferase reporters and the unique concept of the molecular strain probes into bioassays and molecular imaging.


Subject(s)
Molecular Probes , Tacrolimus Binding Proteins , Protein Binding , Luciferases/genetics , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/metabolism , Sirolimus/chemistry , Sirolimus/metabolism
8.
RNA Biol ; 19(1): 1050-1058, 2022 01.
Article in English | MEDLINE | ID: mdl-36093925

ABSTRACT

Delicate variances in the translational machinery affect how efficiently different organisms approach protein synthesis. Determining the scale of this effect, however, requires knowledge on the differences of mistranslation levels. Here, we used a dual-luciferase reporter assay cloned into a broad host range plasmid to reveal the translational fidelity profiles of Pseudomonas putida, Pseudomonas aeruginosa and Escherichia coli. We observed that these profiles are surprisingly different, whereas species more prone to translational frameshifting are not necessarily more prone to stop codon readthrough. As tRNA modifications are among the factors that have been implicated to affect translation accuracy, we also show that translational fidelity is context-specifically influenced by pseudouridines in the anticodon stem-loop of tRNA, but the effect is not uniform between species.


Subject(s)
Anticodon , Pseudouridine , Anticodon/genetics , Codon , Escherichia coli/genetics , RNA Processing, Post-Transcriptional , RNA, Transfer/genetics
9.
Methods Mol Biol ; 2525: 281-287, 2022.
Article in English | MEDLINE | ID: mdl-35836076

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in multiple tissues, such as bone marrow, adipose tissue, umbilical cord, and amniotic fluid. MSCs can differentiate into multilineage cells under defined conditions in vitro and in vivo. MSCs have been shown to have therapeutic effects on various types of diseases. Noninvasive in vivo monitoring of MSCs is considered one of the important techniques for developing cell therapy. In this protocol, we introduce strategized MSCs derived from bone marrow (BM-MSCs) of knock-in mouse model expressing mCherry-Renilla luciferase (mCherry-RLuc) for noninvasive bioluminescence imaging (BLI) of injected BM-MSCs in vivo.


Subject(s)
Bone Marrow Cells , Mesenchymal Stem Cells , Animals , Bone Marrow , Cell Differentiation , Cell Proliferation , Mice
10.
Methods Mol Biol ; 2524: 17-36, 2022.
Article in English | MEDLINE | ID: mdl-35821460

ABSTRACT

Bioluminescence (BL), the emission light resulting from the enzyme-catalyzed oxidative reaction, is a powerful imaging modality for monitoring biological phenomena both in vitro and in vivo. Coelenterazine (CTZ), the known widespread luciferin found in bioluminescent organisms, develops bioluminescence imaging (BLI). Here, we describe an approach to synthesize a series of novel CTZ derivatives for diversifying the toolbox of the BL substrates. Furthermore, we exemplify some of them display excellent BL signals in vitro and in vivo, and thus should be noted as one of the ideal substrates for in vivo BLI compared with a well-known conventional substrate, DeepBlueC.


Subject(s)
Luminescent Measurements , Pyrazines , Diagnostic Imaging , Imidazoles/chemistry , Luminescent Measurements/methods , Pyrazines/chemistry
11.
Methods Mol Biol ; 2524: 209-221, 2022.
Article in English | MEDLINE | ID: mdl-35821474

ABSTRACT

Aberrant splicing of precursor messenger RNA (pre-mRNA) can generate abnormal transcripts, and most of the human diseases have been shown to associate with abnormal splicing of pre-mRNA. Conventional methods require sample lysis and thus cannot be used for monitoring pre-mRNA splicing in real time. This chapter guides how to develop an intron-retained bioluminescence (BL) reporter, which simulates the splicing process of pre-mRNA in vitro and in vivo noninvasively. In the following, we illustrate the design and construction of RLuc-intron and the methods of BL experiments in vitro and in vivo. The exemplified results show that our reporter is suitable for high-throughput screening of splicing inhibitors for the therapies of the diseases caused by aberrant splicing.


Subject(s)
RNA Precursors , RNA Splicing , Diagnostic Imaging , Humans , Introns/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Methods Mol Biol ; 2524: 457-469, 2022.
Article in English | MEDLINE | ID: mdl-35821492

ABSTRACT

Angiogenesis is a prerequisite for tumor growth and invasion, and anti-angiogenesis has become a highlight in tumor treatment research. However, so far, there is no reliable solution for how to simultaneously visualize the relationship between tumor progression and angiogenesis. Bioluminescence imaging (BLI) has been broadly utilized and is a very promising non-invasive imaging technique with the advantages of low cost, high sensitivity, and robust specificity. In this chapter, we describe a dual bioluminescence imaging BLI protocol for tumor progression and angiogenesis through implanting murine breast cancer cell line 4T1 which stably expressing Renilla luciferase (RLuc) into the transgenic mice with angiogenesis-induced firefly luciferase (FLuc) expression. This modality enables us to synchronously monitor the tumor progression and angiogenesis in the same mouse, which has broad applicability in oncology studies.


Subject(s)
Luminescent Measurements , Neovascularization, Pathologic , Animals , Luciferases, Firefly , Luminescent Measurements/methods , Mice , Mice, Transgenic , Molecular Imaging , Neovascularization, Pathologic/metabolism
13.
Arch Toxicol ; 96(9): 2501-2510, 2022 09.
Article in English | MEDLINE | ID: mdl-35678845

ABSTRACT

The activity of nuclear receptors (e.g., pregnane x receptor, PXR) can be assessed by luminescence-based dual reporter gene assays. Under most conditions, receptor-activated firefly luminescence is normalized to Renilla luminescence, which is triggered by a constitutively active promoter. Simultaneous damage to the cells can however disrupt these signals and thus impair the interpretation of the data. Consequently, this study addressed three important aspects: First, idealized models were described, each highlighting crucial characteristics and important pitfalls of dual PXR reporter gene assays used to evaluate PXR activation or inhibition. Second, these models were supported by experimental data obtained with a strong PXR activator (rifampicin) with low cytotoxicity, a PXR activator with high cytotoxicity (dovitinib), a proposed PXR inhibitor that reportedly has no toxic effects (triptolide), and a cytotoxic control (oxaliplatin). Data were evaluated for relative PXR activity data, individual firefly or Renilla luminescence, and anti-proliferative effects of the compounds (assessed by crystal violet staining). Finally, a step-by-step guide is proposed to avoid misleading set-up of the assay or misinterpretation of the data obtained. Key considerations here include (1) omission of drug concentrations beyond 10-20% proliferation inhibition; (2) observation of Renilla luminescence, because this tends to indicate 'false PXR activation' when it inexplicably decreases; (3) parallel decrease of relative PXR activity and proliferation below baseline levels in conjunction with a sharp decrease in Renilla luminescence indicates 'false PXR antagonism'; (4) non-sigmoidal relationships suggest the absence of concentration dependency.


Subject(s)
Receptors, Steroid , Cytochrome P-450 CYP3A/genetics , Genes, Reporter , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Rifampin/pharmacology
14.
Acta Pharm Sin B ; 12(2): 637-650, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35256936

ABSTRACT

Receptor activity-modulating proteins (RAMPs) are accessory molecules that form complexes with specific G protein-coupled receptors (GPCRs) and modulate their functions. It is established that RAMP interacts with the glucagon receptor family of GPCRs but the underlying mechanism is poorly understood. In this study, we used a bioluminescence resonance energy transfer (BRET) approach to comprehensively investigate such interactions. In conjunction with cAMP accumulation, Gα q activation and ß-arrestin1/2 recruitment assays, we not only verified the GPCR-RAMP pairs previously reported, but also identified new patterns of GPCR-RAMP interaction. While RAMP1 was able to modify the three signaling events elicited by both glucagon receptor (GCGR) and glucagon-like peptide-1 receptor (GLP-1R), and RAMP2 mainly affected ß-arrestin1/2 recruitment by GCGR, GLP-1R and glucagon-like peptide-2 receptor, RAMP3 showed a widespread negative impact on all the family members except for growth hormone-releasing hormone receptor covering the three pathways. Our results suggest that RAMP modulates both G protein dependent and independent signal transduction among the glucagon receptor family members in a receptor-specific manner. Mapping such interactions provides new insights into the role of RAMP in ligand recognition and receptor activation.

15.
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
16.
Acta Pharm Sin B ; 12(4): 1636-1651, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34745850

ABSTRACT

SARS-CoV-2 main protease (Mpro) is one of the most extensively exploited drug targets for COVID-19. Structurally disparate compounds have been reported as Mpro inhibitors, raising the question of their target specificity. To elucidate the target specificity and the cellular target engagement of the claimed Mpro inhibitors, we systematically characterize their mechanism of action using the cell-free FRET assay, the thermal shift-binding assay, the cell lysate Protease-Glo luciferase assay, and the cell-based FlipGFP assay. Collectively, our results have shown that majority of the Mpro inhibitors identified from drug repurposing including ebselen, carmofur, disulfiram, and shikonin are promiscuous cysteine inhibitors that are not specific to Mpro, while chloroquine, oxytetracycline, montelukast, candesartan, and dipyridamole do not inhibit Mpro in any of the assays tested. Overall, our study highlights the need of stringent hit validation at the early stage of drug discovery.

17.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203212

ABSTRACT

Firefly luciferase is susceptible to inhibition and stabilization by compounds under investigation for biological activity and toxicity. This can lead to false-positive results in in vitro cell-based assays. However, firefly luciferase remains one of the most commonly used reporter genes. Here, we evaluated isoflavonoids for inhibition of firefly luciferase. These natural compounds are often studied using luciferase reporter-gene assays. We used a quantitative structure-activity relationship (QSAR) model to compare the results of in silico predictions with a newly developed in vitro assay that enables concomitant detection of inhibition of firefly and Renilla luciferases. The QSAR model predicted a moderate to high likelihood of firefly luciferase inhibition for all of the 11 isoflavonoids investigated, and the in vitro assays confirmed this for seven of them: daidzein, genistein, glycitein, prunetin, biochanin A, calycosin, and formononetin. In contrast, none of the 11 isoflavonoids inhibited Renilla luciferase. Molecular docking calculations indicated that isoflavonoids interact favorably with the D-luciferin binding pocket of firefly luciferase. These data demonstrate the importance of reporter-enzyme inhibition when studying the effects of such compounds and suggest that this in vitro assay can be used to exclude false-positives due to firefly or Renilla luciferase inhibition, and to thus define the most appropriate reporter gene.


Subject(s)
Genes, Reporter/physiology , Isoflavones/metabolism , Luciferases, Renilla/metabolism , Animals , Fireflies , Genes, Reporter/genetics , Isoflavones/chemistry , Luciferases, Renilla/chemistry , Protein Structure, Secondary
18.
Methods Mol Biol ; 2274: 111-126, 2021.
Article in English | MEDLINE | ID: mdl-34050467

ABSTRACT

Coelenterazine (CTZ) is a common substrate to most marine luciferases and photoproteins. The present protocol introduces mammalian cell imaging with nine novel dye- and azide-conjugated CTZ analogues, which were synthesized by conjugating a series of fluorescent dyes or an azide group to the C-2 or C-6 position of CTZ backbone. The investigation on the optical properties revealed that azide-conjugated CTZs emit greatly selective bioluminescence (BL) to artificial luciferases (ALucs) and ca. 130 nm blue-shifted BL with Renilla luciferase variant 8.6 (RLuc8.6) in mammalian cells. The corresponding kinetic study explains that azide-conjugated CTZ exerts higher catalytic efficiency than CTZ. Nile red-conjugated CTZ completely showed red-shifted CRET spectra and characteristic BRET spectra with artificial luciferase 16 (ALuc16). The present protocol shows that the minimal spectral overlap occurs among the pairs of [Furimazine/NanoLuc], [6-N3-CTZ/ALuc26], [6-pi-OH-CTZ/RLuc8.6], and [6-N3-CTZ/RLuc8.6] because of the substrate-driven luciferase specificity or color shifts, convincing a cross talk-free multiplex bioassay platform. The present protocol introduces a new toolbox to bioassays and multiplex molecular imaging platforms for mammalian cells.


Subject(s)
Azides/chemistry , Imidazoles/chemistry , Luciferases/metabolism , Luminescent Agents/chemistry , Luminescent Measurements/methods , Molecular Imaging/methods , Pyrazines/chemistry , Animals , COS Cells , Chlorocebus aethiops
19.
J Adv Res ; 34: 1-12, 2021 12.
Article in English | MEDLINE | ID: mdl-35024177

ABSTRACT

Introduction: Alzheimer's disease (AD) is a progressive brain disorder, and one of the most common causes of dementia and amnesia. Due to the complex pathogenesis of AD, the underlying mechanisms remain unclear. Although scientists have made increasing efforts to develop drugs for AD, no effective therapeutic agents have been found. Objectives: Natural products and their constituents have shown promise for treating neurodegenerative diseases, including AD. Thus, in-depth study of medical plants, and the main active ingredients thereof against AD, is necessary to devise therapeutic agents. Methods: In this study, N2a/APP cells and SAMP8 mice were employed as in vitro and in vivo models of AD. Multiple molecular biological methods were used to investigate the potential therapeutic actions of oxyphylla A, and the underlying mechanisms. Results: Results showed that oxyphylla A, a novel compound extracted from Alpinia oxyphylla, could reduce the expression levels of amyloid precursor protein (APP) and amyloid beta (Aß) proteins, and attenuate cognitive decline in SAMP8 mice. Further investigation of the underlying mechanisms showed that oxyphylla A exerted an antioxidative effect through the Akt-GSK3ß and Nrf2-Keap1-HO-1 pathways.Conclusions.Taken together, our results suggest a new horizon for the discovery of therapeutic agents for AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Caproates , Cognition , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cresols , Disease Models, Animal , Glycogen Synthase Kinase 3 beta , Kelch-Like ECH-Associated Protein 1 , Mice , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-akt
20.
Methods Mol Biol ; 2201: 15-26, 2021.
Article in English | MEDLINE | ID: mdl-32975785

ABSTRACT

MOR expression levels at a specific cell type or tissue significantly contribute to its role in pain transmission and in other responses involving opioid receptors. Therefore, molecular processes regulating MOR levels have gained more and more interest. Recently, posttranscriptional regulation mechanisms have been shown to play a relevant role in influencing MOR expression levels, with polymorphisms and mutations within OPRM1 3'-UTR region impacting the differential opioid-mediated response observed within individuals. Here we report a Renilla luciferase reporter assay format suitable for dissecting the contribution of different and distinct OPRM1 3'-UTR elements to MOR expression levels in a model of glial cells, both under basal conditions and following specific treatments.


Subject(s)
Gene Expression Profiling/methods , Receptors, Opioid, mu/genetics , 3' Untranslated Regions/genetics , Animals , Gene Expression/genetics , Gene Expression Regulation/genetics , Genes, Reporter/genetics , Humans , Luciferases, Renilla/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , Receptors, Opioid, mu/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL