RESUMO
G protein-coupled receptors (GPCRs) play pivotal roles in cellular signaling and can regulate several cellular functions such as proliferation, secretion, protein expression, and cellular metabolism. Coupling of GPCRs to members of the Gq/11 protein family results in activation of inositol trisphosphate (IP3) and accumulation of calcium intracellularly. This protocol chapter outlines a step-by-step guide for utilizing the inositol phosphate-1 (IP1) accumulation assay, a time-resolved fluorescence resonance energy transfer (TR-FRET) method, to investigate Gq-IP3 signaling. The assay serves as a valuable tool for those conducting pharmacological investigations and compound screening targeting this critical cellular pathway. This protocol chapter covers experimental setup, sample preparation, and data analysis, providing researchers with an in-depth guide to explore the pharmacology of Gq-coupled receptors.
Assuntos
Transferência Ressonante de Energia de Fluorescência , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Transdução de Sinais , Transferência Ressonante de Energia de Fluorescência/métodos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Células HEK293 , Cálcio/metabolismo , Fosfatos de Inositol/metabolismoRESUMO
Mitochondria are crucial powerhouses and central organelles for maintaining normal physiological activities in eukaryotic cells. The use of highly specific optical biosensors to monitor mitochondrial autophagy (mitophagy) is an important way for detecting mitochondrial abnormalities. Herein, we report a pH responsive G-quadruplex (G4) structure folded by the oligonucleotide named P24. P24 is composed of four GGCCTG repeating units, and the high guanine content allows it to form an antiparallel G4 topology at pH 4.5 (lysosomal pH). However, when pH increases to around 7.4 (mitochondrial pH), P24 further transforms into a double-stranded structure. Unlike most oligonucleotides that enter lysosomes, P24 highly targets mitochondria in live cells. These characteristics enable P24 to construct a pH responsive optical biosensor by linking a pair of fluorescence resonance energy transfer (FRET) fluorophores. The P24 based biosensor demonstrates reliable applications in detecting mitophagy in live cells.
Assuntos
Técnicas Biossensoriais , Transferência Ressonante de Energia de Fluorescência , Quadruplex G , Mitocôndrias , Técnicas Biossensoriais/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Concentração de Íons de Hidrogênio , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/química , DNA/química , Mitofagia , Células HeLa , Corantes Fluorescentes/química , AutofagiaRESUMO
Multiplex biosensors are highly sought-after tools in disease diagnosis. This technique involves the simultaneous sensing of multiple biomarkers, whose levels and ratios can provide a more comprehensive assessment of disease conditions compared to single biomarker detection. In most diseases like cancer due to its complexity, several biomarkers are involved in their occurrence. On the other hand, a single biomarker may be implicated in various diseases. Multiplex sensing employs various techniques, such as optical, electrochemical, and electrochemiluminescence methods. This comprehensive review focuses on optical multiplex sensing techniques, including surface plasmon resonance, localized surface plasmon resonance, fluorescence resonance energy transfer, chemiluminescence, surface-enhanced Raman spectroscopy, and photonic crystal sensors. The review delves into their mechanisms, materials utilized, and strategies for biomarker detection.
Assuntos
Biomarcadores , Técnicas Biossensoriais , Técnicas Biossensoriais/métodos , Biomarcadores/análise , Humanos , Análise Espectral Raman/métodos , Ressonância de Plasmônio de Superfície/métodos , Transferência Ressonante de Energia de Fluorescência/métodosRESUMO
γ-Secretase plays a pivotal role in the central nervous system. Our recent development of genetically encoded Förster resonance energy transfer (FRET)-based biosensors has enabled the spatiotemporal recording of γ-secretase activity on a cell-by-cell basis in live neurons in culture. Nevertheless, how γ-secretase activity is regulated in vivo remains unclear. Here, we employ the near-infrared (NIR) C99 720-670 biosensor and NIR confocal microscopy to quantitatively record γ-secretase activity in individual neurons in living mouse brains. Intriguingly, we uncovered that γ-secretase activity may influence the activity of γ-secretase in neighboring neurons, suggesting a potential 'cell non-autonomous' regulation of γ-secretase in mouse brains. Given that γ-secretase plays critical roles in important biological events and various diseases, our new assay in vivo would become a new platform that enables dissecting the essential roles of γ-secretase in normal health and diseases.
Assuntos
Secretases da Proteína Precursora do Amiloide , Encéfalo , Transferência Ressonante de Energia de Fluorescência , Animais , Secretases da Proteína Precursora do Amiloide/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Encéfalo/metabolismo , Camundongos , Transferência Ressonante de Energia de Fluorescência/métodos , Neurônios/metabolismo , Técnicas Biossensoriais/métodos , Microscopia ConfocalRESUMO
Optical investigations of nanometer distances between proteins, their subunits, or other biomolecules have been the exclusive prerogative of Förster resonance energy transfer (FRET) microscopy for decades. In this work, we show that MINFLUX fluorescence nanoscopy measures intramolecular distances down to 1 nanometer-and in planar projections down to 1 angstrom-directly, linearly, and with angstrom precision. Our method was validated by quantifying well-characterized 1- to 10-nanometer distances in polypeptides and proteins. Moreover, we visualized the orientations of immunoglobulin subunits, applied the method in human cells, and revealed specific configurations of a histidine kinase PAS domain dimer. Our results open the door for examining proximities and interactions by direct position measurements at the intramacromolecular scale.
Assuntos
Microscopia de Fluorescência , Proteínas , Humanos , Transferência Ressonante de Energia de Fluorescência/métodos , Histidina Quinase/química , Microscopia de Fluorescência/métodos , Peptídeos/química , Domínios Proteicos , Multimerização Proteica , Subunidades Proteicas/química , Proteínas/química , Proteínas/metabolismoRESUMO
Branched-chain amino acids (BCAAs) play an important role in the functioning of mammalian cells and the central nervous system. However, available genetically encoded indicators for BCAAs are based on Förster resonance energy transfer and have a limited dynamic range. We developed a single fluorescent protein-based sensor for BCAAs, called NeIle, which is composed of circularly permutated mNeonGreen protein inserted into the leucine-isoleucine-valine binding protein (LIVBP) from Escherichia coli bacteria. In solution, the NeIle indicator displayed a positive fluorescence response to adding isoleucine, leucine, and valin amino acids with high ΔF/F dynamic ranges of 27-, 19-, and 11-fold and the corresponding affinity values of 5.0, 2.9, and 75 mM, respectively. The spectral and biochemical properties of the NeIle indicator were characterized in solution. We characterized the brightness of the NeIle indicator in living mammalian cells, including cultured neurons. Using the NeIle indicator, we successfully visualized the dynamics of isoleucine transients in different organelles of mammalian cells. We obtained and analyzed the X-ray crystal structure of the NeIle indicator in an isoleucine-bound state. Structure-guided directed mutagenesis of the NeIle indicator revealed the basis of its fluorescence response and selectivity to isoleucine.
Assuntos
Aminoácidos de Cadeia Ramificada , Humanos , Aminoácidos de Cadeia Ramificada/química , Aminoácidos de Cadeia Ramificada/metabolismo , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Animais , Escherichia coli/genética , Escherichia coli/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HEK293RESUMO
ADP-ribosylation, the transfer of ADP-ribose (ADPr) from nicotinamide adenine dinucleotide (NAD+) groups to proteins, is a conserved post-translational modification (PTM) that occurs most prominently in response to DNA damage. ADP-ribosylation is a dynamic PTM regulated by writers (PARPs), erasers (ADPr hydrolases), and readers (ADPR binders). PARP1 is the primary DNA damage-response writer responsible for adding a polymer of ADPR to proteins (PARylation). Real-time monitoring of PARP1-mediated PARylation, especially in live cells, is critical for understanding the spatial and temporal regulation of this unique PTM. Here, we describe a genetically encoded FRET probe (pARS) for semiquantitative monitoring of PARylation dynamics. pARS feature a PAR-binding WWE domain flanked with turquoise and Venus. With a ratiometric readout and excellent signal-to-noise characteristics, we show that pARS can monitor PARP1-dependent PARylation temporally and spatially in real-time. pARS provided unique insights into PARP1-mediated PARylation kinetics in vitro and high-sensitivity detection of PARylation in live cells, even under mild DNA damage. We also show that pARS can be used to determine the potency of PARP inhibitors in vitro and, for the first time, in live cells in response to DNA damage. The robustness and ease of use of pARS make it an important tool for the PARP field.
Assuntos
Transferência Ressonante de Energia de Fluorescência , Poli ADP Ribosilação , Humanos , Transferência Ressonante de Energia de Fluorescência/métodos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Técnicas Biossensoriais/métodos , Células HEK293RESUMO
A sustainable operation for harvesting metals in the lanthanide series is needed to meet the rising demand for rare earth elements across diverse global industries. However, existing methods are limited in their capacity for detection and capture at environmentally and industrially relevant lanthanide concentrations. Supercharged fluorescent proteins have solvent-exposed, negatively charged residues that potentially create multiple direct chelation pockets for free lanthanide cations. Here, we demonstrate that negatively supercharged proteins can bind and quantitatively report concentrations of lanthanides via an underutilized lanthanide-to-chromophore pathway of energy transfer. The top-performing sensors detect lanthanides in the micromolar to millimolar range and remain unperturbed by environmentally significant concentrations of competing metals. As a demonstration of the versatility and adaptability of this energy transfer method, we show proximity and signal transmission between the lanthanides and a supramolecular assembly of supercharged proteins, paving the way for the detection of lanthanides via programmable protein oligomers and materials.
Assuntos
Elementos da Série dos Lantanídeos , Proteínas Luminescentes , Elementos da Série dos Lantanídeos/química , Elementos da Série dos Lantanídeos/metabolismo , Proteínas Luminescentes/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/química , Transferência de Energia , Transferência Ressonante de Energia de Fluorescência/métodosRESUMO
Phosphatase of regenerating liver (PRL) is an oncogenic protein that promotes tumor progression by directly binding to cyclin M (CNNM) membrane proteins and inhibiting their Mg2+ efflux activity. In this study, we have developed a high-throughput screening system to detect the interactions between PRL and CNNM proteins based on homogenous time-resolved fluorescence resonance energy transfer (HTR-FRET, HTRF). We optimized the tag sequences attached to the recombinant proteins of the CNNM4 CBS domains and PRL3 lacking the carboxyl terminal CAAX motif, and successfully detected the interaction by observing the FRET signal in the mixture of the tagged proteins and fluorophore-conjugated antibodies. Moreover, we performed compound library screening using this system and discovered several compounds that could efficiently inhibit the PRL-CNNM interaction. Characterization of one candidate compound revealed that it was relatively stable compared with thienopyridone, a known inhibitor of the PRL-CNNM interaction. The candidate compound can also inhibit PRL function in cells: suppression of CNNM-dependent Mg2+ efflux, and has sufficient in vitro drug metabolism and pharmacokinetic properties. Overall, these results demonstrate the effectiveness of this screening system for identifying novel inhibitors of the PRL-CNNM interaction, which could contribute to the development of novel anti-cancer drugs.
Assuntos
Transferência Ressonante de Energia de Fluorescência , Ensaios de Triagem em Larga Escala , Proteínas Tirosina Fosfatases , Humanos , Ensaios de Triagem em Larga Escala/métodos , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Animais , Ligação Proteica , Magnésio/metabolismo , Células HEK293 , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/antagonistas & inibidoresRESUMO
In humans, DNA polymerase δ (pol δ) holoenzymes, comprised of pol δ and the processivity sliding clamp, proliferating cell nuclear antigen (PCNA), carry out DNA synthesis during lagging strand replication, the initiation of leading strand DNA replication as well as most of the major DNA damage repair pathways. In each of these contexts, pol δ holoenzymes are assembled at primer/template (P/T) junctions and initiate DNA synthesis in a stepwise process that involves the PCNA clamp loader, replication factor C and, depending on the DNA synthesis pathway, the major single strand DNA-binding protein complex, replication protein A (RPA). In a recent report from our laboratory, we designed and utilized direct, ensemble Förster Resonance Energy Transfer approaches to monitor the transient state kinetics of pol δ holoenzyme assembly and initiation of DNA synthesis on P/T junctions engaged by RPA. In this chapter, we detail the original approaches and discuss adaptations that can be utilized to monitor fast kinetic reactions in the millisecond (ms) timescale. All approaches described in this chapter utilize a commercially-available fluorescence spectrophotometer, can be readily evolved for alternative DNA polymerases and P/T DNA substrates, and permit incorporation of protein posttranslational modifications, accessory factors, DNA covalent modifications, accessory factors, enzymes, etc. Hence, these approaches are widely accessible and broadly applicable for characterizing DNA polymerase holoenzyme assembly and initiation of DNA synthesis during any PCNA-dependent DNA synthesis pathway.
Assuntos
DNA Polimerase III , Replicação do DNA , DNA , Transferência Ressonante de Energia de Fluorescência , Antígeno Nuclear de Célula em Proliferação , Transferência Ressonante de Energia de Fluorescência/métodos , DNA Polimerase III/metabolismo , DNA Polimerase III/química , Humanos , Cinética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Antígeno Nuclear de Célula em Proliferação/química , Antígeno Nuclear de Célula em Proliferação/genética , Holoenzimas/metabolismo , Holoenzimas/química , DNA/metabolismo , DNA/química , Proteína de Replicação A/metabolismo , Proteína de Replicação A/química , Proteína de Replicação C/metabolismo , Proteína de Replicação C/genética , Proteína de Replicação C/químicaRESUMO
In recent years, the connection between APOBEC3 cytosine deaminases and cancer mutagenesis has become ever more apparent. This growing awareness and lack of inhibitory drugs has created a distinct need for biochemical tools that can be used to identify and characterize potential inhibitors of this family of enzymes. In response to this challenge, we have developed a Real-time APOBEC3-mediated DNA Deamination (RADD) assay. The RADD assay provides a rapid, real-time fluorescence readout of APOBEC3 DNA deamination and serves as a crucial addition to the existing APOBEC3 biochemical and cellular toolkit. This method improves upon contemporary DNA deamination assays by offering a more rapid and quantifiable readout as well as providing a platform that is readily adaptable to a high-throughput format for inhibitor discovery. In this chapter we provide a detailed guide for the usage of the RADD assay for the characterization of APOBEC3 enzymes and potential inhibitors.
Assuntos
DNA , Transferência Ressonante de Energia de Fluorescência , Humanos , Transferência Ressonante de Energia de Fluorescência/métodos , DNA/metabolismo , Desaminação , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Ensaios Enzimáticos/métodos , Inibidores Enzimáticos/farmacologia , Desaminases APOBEC/metabolismoRESUMO
BACKGROUND: Dichlorvos (DDVP) is an efficient and highly toxic organophosphorus pesticide. Considering its effects on human health and ecosystems, pesticide residue and pollution monitoring is of great significance. Traditional methods like chromatography with mass spectrometry are not portable or rapid because they use large instruments and complex pre-processing methods. Compared with other optional on-site detection technologies, like enzymatic and antibody methods, aptamers are advantageous because they are stable, readily modified, and inexpensive. Therefore, screening and developing a specific adapter for DDVP detection is necessary and will be of practical value. RESULTS: We screened, modified, and compared two dual-labeled aptamer probes (Cy3-DV55-Cy5 and Cy3-DV65-Cy5). The kinetics studied showed that 5 min was sufficient for the detection reaction. Both aptamers showed selectivity for DDVP but DV55 was superior to DV65. To research the binding stabilities and the mechanism between the aptamers and DDVP, the secondary structures, melting temperatures, fluorescence quenching types, and constants were investigated. Which showed that DV55 was specific for DDVP and showed better binding than DV65. Comparison of the UV absorption and FRET for DV55 and the truncated structures suggested that loop 3 in DV55 might play an important role in the binding of DV55 to DDVP. The Cy3-DV55-Cy5 aptamer had a linear range of 0-100 µM for DDVP detection and the limit of detection was 150 nM. Simulated pesticide residue detection experiments showed that the method was simple, fast, and had acceptable recovery (89.8%-105.2 %). SIGNIFICANCE: Pesticide detection is important but on-site detection methods are usually not portable or rapid. We developed two dual-labeled aptamer probes that could feasibly be practically applied to rapid on-site DDVP detection of pesticide residues and pollutants. This research provides experimental and theoretical data for the development and design of similar pesticide probes.
Assuntos
Aptâmeros de Nucleotídeos , Diclorvós , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes , Aptâmeros de Nucleotídeos/química , Transferência Ressonante de Energia de Fluorescência/métodos , Diclorvós/análise , Corantes Fluorescentes/química , Carbocianinas/química , Limite de DetecçãoRESUMO
The diagnoses of retroviruses are essential for controlling the rapid spread of pandemics. However, the real-time Reverse Transcriptase quantitative Polymerase Chain Reaction (RT-qPCR), which has been the gold standard for identifying viruses such as SARS-CoV-2 in the early stages of infection, is associated with high costs and logistical challenges. To innovate in viral RNA detection a novel molecular approach for detecting SARS-CoV-2 viral RNA, as a proof of concept, was developed. This method combines specific viral gene analysis, trans-acting ribozymes, and Fluorescence Resonance Energy Transfer (FRET)-based hybridization of fluorescent DNA hairpins. In this molecular mechanism, SARS-CoV-2 RNA is specifically recognized and cleaved by ribozymes, releasing an initiator fragment that triggers a hybridization chain reaction (HCR) with DNA hairpins containing fluorophores, leading to a FRET process. A consensus SARS-CoV-2 RNA target sequence was identified, and specific ribozymes were designed and transcribed in vitro to cleave the viral RNA into fragments. DNA hairpins labeled with Cy3/Cy5 fluorophores were then designed and synthesized for HCR-FRET assays targeting the RNA fragment sequences resulting from ribozyme cleavage. The results demonstrated that two of the three designed ribozymes effectively cleaved the target RNA within 10 minutes. Additionally, DNA hairpins labeled with Cy3/Cy5 pairs efficiently detected target RNA specifically and triggered detectable HCR-FRET reactions. This method is versatile and can be adapted for use with other viruses. Furthermore, the design and construction of a DIY photo-fluorometer prototype enabled us to explore the development of a simple and cost-effective point-of-care detection method based on digital image analysis.
Assuntos
Transferência Ressonante de Energia de Fluorescência , RNA Catalítico , RNA Viral , SARS-CoV-2 , Transferência Ressonante de Energia de Fluorescência/métodos , RNA Viral/genética , RNA Catalítico/genética , RNA Catalítico/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , Humanos , COVID-19/virologia , COVID-19/diagnóstico , Hibridização de Ácido Nucleico/métodos , Carbocianinas/químicaRESUMO
A dual-emission fluorescent biosensing method was developed for simultaneous determination of CaMV35S and NOS in genetically modified (GM) plants. Two designed hairpin DNA (H1, H2) sequences were used as templates to synthesize H1-AgNCs (λex = 570 nm, λem = 625 nm) and H2-AgNCs (λex = 470 nm, λem = 555 nm). By using H1-AgNCs and H2-AgNCs as dual-signal tags, combined with signal amplification strategy of magnetic separation to reduce background signal and an enzyme-free catalytic hairpin assembly (CHA) signal amplification strategy, a novel multi-target fluorescent biosensor was fabricated to detect multiple targets based on FRET between signal tags (donors) and magnetic Fe3O4 modified graphene oxide (Fe3O4@GO, acceptors). In the presence of the target NOS and CaMV35S, the hairpin structures of H1 and H2 can be opened respectively, and the exposed sequences will hybridize with the G-rich hairpin sequences HP1 and HP2 respectively, displacing the target sequences to participate in the next round of CHA cycle. Meanwhile, H1-HP1 and H2-HP2 double-stranded DNA sequences (dsDNA) were formed, resulting in the desorption of dsDNA from the surface of Fe3O4@GO due to weak π-π interaction between dsDNA and Fe3O4@GO and leading to the fluorescence recovery of AgNCs. Under optimal conditions, the linear ranges of this fluorescence sensor were 5 ~ 300 nmol L-1 for NOS and 5 ~ 200 nmol L-1 CaMV35S, and the LODs were 0.14 nmol L-1 and 0.18 nmol L-1, respectively. In addition, the fluorescence sensor has good selectivity for the detection of NOS and CaMV35S in GM soybean samples, showing the potential applications in GM screening.
Assuntos
Técnicas Biossensoriais , Limite de Detecção , Nanopartículas Metálicas , Prata , Prata/química , Técnicas Biossensoriais/métodos , Nanopartículas Metálicas/química , Transferência Ressonante de Energia de Fluorescência/métodos , Grafite/química , Sequências Repetidas Invertidas , Plantas Geneticamente Modificadas/genética , Catálise , Corantes Fluorescentes/química , Caulimovirus/genética , Técnicas de Amplificação de Ácido Nucleico/métodos , Proteínas Virais/química , Proteínas Virais/genética , Aminoácido OxirredutasesRESUMO
DNA damage occurs when cells encounter exogenous and endogenous stresses such as long periods of desiccation, ionizing radiation and genotoxic chemicals. Efforts have been made to detect DNA damage in vivo and in vitro to characterize or quantify the damage level. It is well accepted that single-stranded DNA (ssDNA) is one of the important byproducts of DNA damage to trigger the downstream regulation. A recent study has revealed that PprI efficiently recognizes ssDNA and cleaves DdrO at a specific site on the cleavage site region (CSR) loop in the presence of ssDNA, which enables the radiation resistance of Deinococcus. Leveraging this property, we developed a quantitative DNA damage detection method in vitro based on fluorescence resonance energy transfer (FRET). DdrO protein was fused with eYFP and eCFP on the N-terminal and C-terminal respectively, between which the FRET efficiency serves as an indicator of cleavage efficiency as well as the concentration of ssDNA. The standard curve between the concentration of ssDNA and the FRET efficiency was constructed, and application examples were tested, validating the effectiveness of this method.
Assuntos
Dano ao DNA , DNA de Cadeia Simples , Deinococcus , Transferência Ressonante de Energia de Fluorescência , Transferência Ressonante de Energia de Fluorescência/métodos , Deinococcus/genética , Deinococcus/efeitos da radiação , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismoRESUMO
Although several imaging strategies for dual fluorescence (or Förster) resonance energy transfer (FRET) biosensors have been reported, their implementation is challenging because of the limited performance of fluorescent proteins and the spectral overlap of FRET biosensors. These processes often require additional data calibration to eliminate artifacts. Many CFP/YFP FRET biosensors have been developed. In this study, we introduced the mKate3/HT7(JFX650) FRET pair, which effectively formed two pairs of FRET pairs for dual-FRET imaging when combined with the CFP/YFP FRET pair. The FRET donor mKate3 exhibited higher brightness than its predecessor mKate. The FRET acceptor, HT7(JFX650), is a HaloTag7 protein covalently conjugated with a far-red JFX650-THL ligand. The pair comprising mKate3 and HT7(JFX650) represents an excellent FRET dyad, exhibiting a high FRET efficiency ratio. To use the FRET pair for dual FRET biosensor imaging, we constructed PKA and K+ biosensors based on the mKate3/HT7(JFX650) FRET pair. These biosensors can be used along with CFP/YFP biosensors to simultaneously detect the responses of intracellular PKA/Src, PKA/Ca2+, and K+/Ca2+ under different stimuli. The findings revealed that dual FRET biosensors, which are based on the combination of CFP/YFP and mKate3/HT7 (JFX650), exhibit adequate compatibility and can be used to visualize multiple molecular activities in a live cell.
Assuntos
Técnicas Biossensoriais , Transferência Ressonante de Energia de Fluorescência , Proteínas Luminescentes , Transferência Ressonante de Energia de Fluorescência/métodos , Humanos , Técnicas Biossensoriais/métodos , Proteínas Luminescentes/química , Proteínas de Fluorescência Verde/química , Células HEK293 , Proteínas Quinases Dependentes de AMP Cíclico/metabolismoRESUMO
A ratiometric fluorescence sensor (Fe-MIL-88-NH2/curcumin) based on luminescent metal-organic frameworks (LMOFs) for the determination of curcumin was constructed. Upon the addition of curcumin, the 535-nm emission of curcumin was enhanced, while the fluorescence emission at 438 nm was quenched, under 367-nm excitation. This sensor demonstrated a broad linear range from 1.5 to 40 µM, a low detection limit of 35 nM, and a fast response time of at most 30 s. We verified the Förster resonance energy transfer (FRET) mechanism between donor (Fe-MIL-88-NH2) and acceptor (curcumin), which further proved the selectivity of the approach. The sensing system enabled the detection of curcumin in the traditional Chinese medicine (TCM) Turmeric. A smartphone-assisted sensing platform was prepared to visually detect curcumin in a portable manner. This study represents the first attempt to fabricate LMOFs for ratiometric fluorescence detection of curcumin, which has promising potential for application in TCM.
Assuntos
Curcumina , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes , Limite de Detecção , Smartphone , Curcumina/química , Transferência Ressonante de Energia de Fluorescência/métodos , Corantes Fluorescentes/química , Estruturas Metalorgânicas/química , Medicina Tradicional Chinesa , Curcuma/química , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/análiseRESUMO
The determination of the dissociation constant (Kd) is pivotal in biochemistry and pharmacology for understanding binding affinities in chemical reactions, which is crucial for drug development and comprehending biological systems. Here, we introduce a single-molecule fluorescence resonance energy transfer-based method for determining Kd, alongside the conventional electrophoretic mobility shift assay method of Kd, offering insights into thermodynamic interactions between proteins and substrates. The single-molecule fluorescence resonance energy transfer approach is highlighted for its ability to accurately measure binding and dissociation kinetics through fluorescence labeling and the intrinsic nature of protein-DNA interactions, representing a significant advancement in the fields of molecular biology and pharmacology.
Assuntos
Transferência Ressonante de Energia de Fluorescência , Transferência Ressonante de Energia de Fluorescência/métodos , Cinética , DNA/metabolismo , DNA/química , Ligação Proteica , Termodinâmica , Imagem Individual de Molécula/métodos , Proteínas/química , Proteínas/metabolismoRESUMO
Unraveling the concentration-dependent spatiotemporal organization of receptors in the plasma membrane is crucial to understand cell signal initiation. A paradigm of this process is the oligomerization of CD95 during apoptosis signaling, with different oligomerization models being discussed. Here, we establish the molecular-sensitive approach cell lifetime Förster resonance energy transfer image spectroscopy to determine CD95 configurations in live cells. These data are corroborated by stimulated emission depletion microscopy, confocal photobleaching step analysis, and fluorescence correlation spectroscopy. We probed CD95 interactions for concentrations of ~10 to 1000 molecules per square micrometer, over nanoseconds to hours, and molecular to cellular scales. Quantitative benchmarking was achieved establishing high-fidelity monomer and dimer controls. While CD95 alone is primarily monomeric (~96%) and dimeric (4%), the addition of ligand induces oligomerization to dimers/trimers (~15%) leading to cell death. This study highlights molecular concentration effects and oligomerization dynamics. It reveals a minimal model, where small CD95 oligomers suffice to efficiently initiate signaling.
Assuntos
Transdução de Sinais , Receptor fas , Receptor fas/metabolismo , Receptor fas/química , Humanos , Transferência Ressonante de Energia de Fluorescência/métodos , Multimerização Proteica , Apoptose , Proteína Ligante Fas/metabolismo , Proteína Ligante Fas/química , Membrana Celular/metabolismoRESUMO
Here, we present a protocol to comprehensively quantify autophagy initiation using the readout of the microtubule associated protein 1 light chain 3 beta (LC3B) Förster's resonance energy transfer (FRET) biosensor. We describe steps for cell seeding, transfection, FRET/FLIM (fluorescence lifetime imaging microscopy) imaging, and image analysis. This protocol can be useful in any physiology- or disease-related paradigm where the LC3B biosensor can be expressed to determine whether autophagy has been initiated or is stalled. The analysis pipeline presented here can be applied to any other genetically encoded FRET sensor imaged using FRET/FLIM. For complete details on the use and execution of this protocol, please refer to Gökerküçük et al.1.