Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38791511

RESUMO

G protein-coupled receptors (GPCRs) are relevant targets for health and disease as they regulate various aspects of metabolism, proliferation, differentiation, and immune pathways. They are implicated in several disease areas, including cancer, diabetes, cardiovascular diseases, and mental disorders. It is worth noting that about a third of all marketed drugs target GPCRs, making them prime pharmacological targets for drug discovery. Numerous functional assays have been developed to assess GPCR activity and GPCR signaling in living cells. Here, we review the current literature of genetically encoded cell-based assays to measure GPCR activation and downstream signaling at different hierarchical levels of signaling, from the receptor to transcription, via transducers, effectors, and second messengers. Singleplex assay formats provide one data point per experimental condition. Typical examples are bioluminescence resonance energy transfer (BRET) assays and protease cleavage assays (e.g., Tango or split TEV). By contrast, multiplex assay formats allow for the parallel measurement of multiple receptors and pathways and typically use molecular barcodes as transcriptional reporters in barcoded assays. This enables the efficient identification of desired on-target and on-pathway effects as well as detrimental off-target and off-pathway effects. Multiplex assays are anticipated to accelerate drug discovery for GPCRs as they provide a comprehensive and broad identification of compound effects.


Assuntos
Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/metabolismo , Humanos , Transdução de Sinais/efeitos dos fármacos , Desenvolvimento de Medicamentos/métodos , Descoberta de Drogas/métodos , Animais , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Bioensaio/métodos
2.
Int J Mol Sci ; 25(9)2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38732237

RESUMO

NanoLuc-mediated bioluminescence resonance energy transfer (NanoBRET) has gained popularity for its ability to homogenously measure ligand binding to G protein-coupled receptors (GPCRs), including the subfamily of chemokine receptors. These receptors, such as ACKR3, CXCR4, CXCR3, play a crucial role in the regulation of the immune system, are associated with inflammatory diseases and cancer, and are seen as promising drug targets. The aim of this study was to optimize NanoBRET-based ligand binding to NLuc-ACKR3 and NLuc-CXCR4 using different fluorescently labeled chemokine CXCL12 analogs and their use in a multiplex NanoBRET binding assay of two chemokine receptors at the same time. The four fluorescent CXCL12 analogs (CXCL12-AZD488, -AZD546, -AZD594, -AZD647) showed high-affinity saturable binding to both NLuc-ACKR3 and NLuc-CXCR4, with relatively low levels of non-specific binding. Additionally, the binding of all AZDye-labeled CXCL12s to Nluc receptors was inhibited by pharmacologically relevant unlabeled chemokines and small molecules. The NanoBRET binding assay for CXCL10-AZD488 binding to Nluc-CXCR3 was also successfully established and successfully employed for the simultaneous measurement of the binding of unlabeled small molecules to NLuc-CXCR3 and NLuc-CXCR4. In conclusion, multiplexing the NanoBRET-based competition binding assay is a promising tool for testing unlabeled (small) molecules against multiple GPCRs simultaneously.


Assuntos
Quimiocina CXCL12 , Ligação Proteica , Receptores CXCR3 , Receptores CXCR4 , Receptores CXCR , Humanos , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR/genética , Quimiocina CXCL12/metabolismo , Receptores CXCR3/metabolismo , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Ligantes , Corantes Fluorescentes/química
3.
J Biol Chem ; 299(6): 104807, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37172730

RESUMO

Here, we report a bioluminescence resonance energy transfer (BRET) assay as a novel way to investigate the binding of unlabeled ligands to the human transient receptor potential mucolipin 1 (hTRPML1), a lysosomal ion channel involved in several genetic diseases and cancer progression. This novel BRET assay can be used to determine equilibrium and kinetic binding parameters of unlabeled compounds to hTRPML1 using intact human-derived cells, thus complementing the information obtained using functional assays based on ion channel activation. We expect this new BRET assay to expedite the identification and optimization of cell-permeable ligands that interact with hTRPML1 within the physiologically relevant environment of lysosomes.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência , Canais de Potencial de Receptor Transitório , Humanos , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Ligantes , Lisossomos/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo
4.
Mol Cancer Ther ; 20(9): 1743-1754, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34158349

RESUMO

Activating mutations in RAS are found in approximately 30% of human cancers, resulting in the delivery of a persistent signal to critical downstream effectors that drive tumorigenesis. RAS-driven malignancies respond poorly to conventional cancer treatments and inhibitors that target RAS directly are limited; therefore, the identification of new strategies and/or drugs to disrupt RAS signaling in tumor cells remains a pressing therapeutic need. Taking advantage of the live-cell bioluminescence resonance energy transfer (BRET) methodology, we describe the development of a NanoBRET screening platform to identify compounds that modulate binding between activated KRAS and the CRAF kinase, an essential effector of RAS that initiates ERK cascade signaling. Using this strategy, libraries containing synthetic compounds, targeted inhibitors, purified natural products, and natural product extracts were evaluated. These efforts resulted in the identification of compounds that inhibit RAS/RAF binding and in turn suppress RAS-driven ERK activation, but also compounds that have the deleterious effect of enhancing the interaction to upregulate pathway signaling. Among the inhibitor hits identified, the majority were compounds derived from natural products, including ones reported to alter KRAS nanoclustering (ophiobolin A), to impact RAF function (HSP90 inhibitors and ROS inducers) as well as some with unknown targets and activities. These findings demonstrate the potential for this screening platform in natural product drug discovery and in the development of new therapeutic agents to target dysregulated RAS signaling in human disease states such as cancer.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Fibroblastos/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Proteínas ras/agonistas , Proteínas ras/antagonistas & inibidores , Animais , Fibroblastos/metabolismo , Humanos , Ligantes , Nanotecnologia/métodos , Proteínas Proto-Oncogênicas c-raf/química , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas ras/metabolismo
5.
Int J Mol Sci ; 22(3)2021 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-33499147

RESUMO

Receptor heteromerization is the formation of a complex involving at least two different receptors with pharmacology that is distinct from that exhibited by its constituent receptor units. Detection of these complexes and monitoring their pharmacology is crucial for understanding how receptors function. The Receptor-Heteromer Investigation Technology (Receptor-HIT) utilizes ligand-dependent modulation of interactions between receptors and specific biomolecules for the detection and profiling of heteromer complexes. Previously, the interacting biomolecules used in Receptor-HIT assays have been intracellular proteins, however in this study we have for the first time used bioluminescence resonance energy transfer (BRET) with fluorescently-labeled ligands to investigate heteromerization of receptors on the cell surface. Using the Receptor-HIT ligand binding assay with NanoBRET, we have successfully investigated heteromers between the angiotensin II type 1 (AT1) receptor and the ß2 adrenergic receptor (AT1-ß2AR heteromer), as well as between the AT1 and angiotensin II type 2 receptor (AT1-AT2 heteromer).


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Ligantes , Nanotecnologia/métodos , Receptores de Angiotensina/metabolismo , Ligação Competitiva , Compostos de Boro/química , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Cinética , Ligação Proteica , Multimerização Proteica , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais
6.
J Mater Chem B ; 9(1): 35-52, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33225338

RESUMO

It is an urgency to detect infectious pathogens or cancer biomarkers using rapid, simple, convenient and cost-effective methods in complex biological samples. Many existing approaches (traditional virus culture, ELISA or PCR) for the pathogen and biomarker assays face several challenges in the clinical applications that require lengthy time, sophisticated sample pre-treatment and expensive instruments. Due to the simple and rapid detection manner as well as no requirement of expensive equipment, many visual detection methods have been considered to resolve the aforementioned problems. Meanwhile, various new materials and colorimetric/fluorescent methods have been tried to construct new biosensors for infectious pathogens and biomarkers. However, the recent progress of these aspects is rarely reviewed, especially in terms of integration of new materials, microdevice and detection mechanism into the visual detection systems. Herein, we provide a broad field of view to discuss the recent progress in the visual detection of infectious pathogens and cancer biomarkers along with the detection mechanism, new materials, novel detection methods, special targets as well as multi-functional microdevices and systems. The novel visual approaches for the infectious pathogens and biomarkers, such as bioluminescence resonance energy transfer (BRET), metal-induced metallization and clustered regularly interspaced short palindromic repeats (CRISPR)-based biosensors, are discussed. Additionally, recent advancements in visual assays utilizing various new materials for proteins, nucleic acids, viruses, exosomes and small molecules are comprehensively reviewed. Future perspectives on the visual sensing systems for infectious pathogens and cancers are also proposed.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Biomarcadores Tumorais/metabolismo , Técnicas Biossensoriais/métodos , Neoplasias/metabolismo , Animais , Biomarcadores Tumorais/genética , Sistemas CRISPR-Cas/fisiologia , Colorimetria/métodos , Humanos , Neoplasias/diagnóstico , Ressonância de Plasmônio de Superfície/métodos
7.
Methods Mol Biol ; 2201: 117-125, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32975794

RESUMO

Quantitative measurement of receptor signaling by different ligands is important for understanding the mechanism of drug action and screening of drugs. Here, we describe a simple and cost-effective method of measuring adenylyl cyclase inhibition, one of the hallmarks of opioid receptor activation. The assay is based on bioluminescence resonance energy transfer (BRET) that involves transfection of a biosensor in human embryonic kidney (HEK)-293 cells stably transfected with µ-opioid receptor (µ receptor), enabling real-time measurement of cAMP levels.


Assuntos
Inibidores de Adenilil Ciclases/análise , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Imagem Molecular/métodos , Toxina Adenilato Ciclase , Inibidores de Adenilil Ciclases/metabolismo , Adenilil Ciclases/metabolismo , Analgésicos Opioides , Animais , Colforsina/farmacologia , AMP Cíclico , Transferência de Energia , Células HEK293 , Humanos , Receptores Opioides/química , Receptores Opioides/metabolismo , Receptores Opioides mu
8.
Sci Rep ; 10(1): 16616, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33024173

RESUMO

Monitoring of exosome dynamics in living organisms is essential to demonstrate the real functions of cancer-derived exosomes. Currently, these have been elucidated in vitro or under non-physiological conditions in vivo in most cases. To overcome these limitations, we developed an imaging method using Antares2-mediated bioluminescence resonance energy transfer (BRET) for observing long-term accumulation of exosomes in vivo. Ectopic expression of CD63-Antares2 effectively labeled exosomes with Antares2, which emitted intense, long-wavelength luminescence suitable for in vivo monitoring. Transplantation of CD63-Antares2-expressing prostate cancer cells into mice allowed determining the amount of cancer-derived exosomes released from primary tumors into the bloodstream and visualizing the long-term homing behavior of exosomes to their target organs or tissues. Interestingly, secreted exosome was decreased upon administration of low dose of dasatinib, an approved tyrosine-kinase inhibitor. The CD63-Antares2 xenograft mouse model will be useful for elucidating the dynamics of cancer-derived exosomes in vivo and evaluating the therapeutic efficacy and mechanism of exosome production inhibitors.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Transferência de Energia , Exossomos/metabolismo , Imagem Molecular/métodos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Animais , Dasatinibe/farmacologia , Xenoenxertos , Masculino , Camundongos , Transplante de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 117(35): 21723-21730, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32817560

RESUMO

G proteins are activated when they associate with G protein-coupled receptors (GPCRs), often in response to agonist-mediated receptor activation. It is generally thought that agonist-induced receptor-G protein association necessarily promotes G protein activation and, conversely, that activated GPCRs do not interact with G proteins that they do not activate. Here we show that GPCRs can form agonist-dependent complexes with G proteins that they do not activate. Using cell-based bioluminescence resonance energy transfer (BRET) and luminescence assays we find that vasopressin V2 receptors (V2R) associate with both Gs and G12 heterotrimers when stimulated with the agonist arginine vasopressin (AVP). However, unlike V2R-Gs complexes, V2R-G12 complexes are not destabilized by guanine nucleotides and do not promote G12 activation. Activating V2R does not lead to signaling responses downstream of G12 activation, but instead inhibits basal G12-mediated signaling, presumably by sequestering G12 heterotrimers. Overexpressing G12 inhibits G protein receptor kinase (GRK) and arrestin recruitment to V2R and receptor internalization. Formyl peptide (FPR1 and FPR2) and Smoothened (Smo) receptors also form complexes with G12 that are insensitive to nucleotides, suggesting that unproductive GPCR-G12 complexes are not unique to V2R. These results indicate that agonist-dependent receptor-G protein association does not always lead to G protein activation and may in fact inhibit G protein activation.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , AMP Cíclico/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Ligantes , Ligação Proteica/fisiologia , Receptores de Vasopressinas/metabolismo , Transdução de Sinais/fisiologia , Vasopressinas/metabolismo , beta-Arrestinas/metabolismo
10.
Cell ; 182(3): 770-785.e16, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32634377

RESUMO

Heterotrimeric G-proteins (Gαßγ) are the main transducers of signals from GPCRs, mediating the action of countless natural stimuli and therapeutic agents. However, there are currently no robust approaches to directly measure the activity of endogenous G-proteins in cells. Here, we describe a suite of optical biosensors that detect endogenous active G-proteins with sub-second resolution in live cells. Using a modular design principle, we developed genetically encoded, unimolecular biosensors for endogenous Gα-GTP and free Gßγ: the two active species of heterotrimeric G-proteins. This design was leveraged to generate biosensors with specificity for different heterotrimeric G-proteins or for other G-proteins, such as Rho GTPases. Versatility was further validated by implementing the biosensors in multiple contexts, from characterizing cancer-associated G-protein mutants to neurotransmitter signaling in primary neurons. Overall, the versatile biosensor design introduced here enables studying the activity of endogenous G-proteins in live cells with high fidelity, temporal resolution, and convenience.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/instrumentação , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Técnicas Biossensoriais/instrumentação , Técnicas Biossensoriais/métodos , Guanosina Trifosfato/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Motivos de Aminoácidos , Animais , Células Cultivadas , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Guanosina Trifosfato/química , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Neurônios/química , Neurônios/metabolismo , Neurônios/fisiologia , Transdução de Sinais , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo
11.
Methods Mol Biol ; 2041: 163-181, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31646488

RESUMO

Fluorescent antagonists offer the ability to interrogate G protein-coupled receptor pharmacology. With resonance energy transfer techniques, fluorescent antagonists can be implemented to monitor receptor-ligand interactions using assays originally designed for radiolabeled probes. The fluorescent nature of these antagonists also enables the localization and distribution of the receptors to be visualized in living cells. Here, we describe the generation of modified purinergic receptors with the NanoLuc luciferase or SNAP-tag, using the P1 adenosine A3 receptor as an example. We also describe the procedure of characterizing a novel fluorescent purinergic antagonist using ligand-mediated bioluminescence resonance energy transfer assays and confocal microscopy.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Microscopia de Fluorescência/métodos , Agonistas do Receptor Purinérgico P1/metabolismo , Receptor A3 de Adenosina/metabolismo , Receptores Purinérgicos P1/metabolismo , Fluorescência , Células HEK293 , Humanos , Luciferases/metabolismo , Ligação Proteica , Multimerização Proteica , Agonistas do Receptor Purinérgico P1/química , Receptor A3 de Adenosina/química , Receptores Purinérgicos P1/química , Transdução de Sinais
12.
Mol Pharmacol ; 97(1): 23-34, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31707356

RESUMO

Smoothened (SMO) is a GPCR that mediates hedgehog signaling. Hedgehog binds the transmembrane protein Patched, which in turn regulates SMO activation. Overactive SMO signaling is oncogenic and is therefore a clinically established drug target. Here we establish a nanoluciferase bioluminescence resonance energy transfer (NanoBRET)-based ligand binding assay for SMO providing a sensitive and high throughput-compatible addition to the toolbox of GPCR pharmacologists. In the NanoBRET-based binding assay, SMO is N terminally tagged with nanoluciferase (Nluc) and binding of BODIPY-cyclopamine is assessed by quantifying resonance energy transfer between receptor and ligand. The assay allowed kinetic analysis of ligand-receptor binding in living HEK293 cells, competition binding experiments using commercially available SMO ligands (SANT-1, cyclopamine-KAAD, SAG1.3 and purmorphamine), and pharmacological dissection of two BODIPY-cyclopamine binding sites. This high throughput-compatible assay is superior to commonly used SMO ligand binding assays in the separation of specific from non-specific ligand binding and, provides a suitable complement to chemical biology strategies for the discovery of novel SMO-targeting drugs. SIGNIFICANCE STATEMENT: We established a NanoBRET-based binding assay for SMO with superior sensitivity compared to fluorescence-based assays. This assay allows distinction of two separate binding sites for BODIPY-cyclopamine on the SMO transmembrane core in live cells in real time. The assay is a valuable complement for drug discovery efforts and will support a better understanding of Class F GPCR pharmacology.


Assuntos
Sítios de Ligação/genética , Bioensaio/métodos , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened/antagonistas & inibidores , Alcaloides de Veratrum/farmacologia , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Compostos de Boro/química , Cinamatos/farmacologia , Descoberta de Drogas/métodos , Técnicas de Inativação de Genes , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Ligantes , Luciferases/química , Morfolinas/farmacologia , Nanoestruturas/química , Purinas/farmacologia , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Alcaloides de Veratrum/química
13.
Chem Commun (Camb) ; 56(2): 281-284, 2019 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-31807738

RESUMO

Bioluminescence resonance energy transfer (BRET) is a commonly used assay system for studying protein-protein interactions and protein folding in vivo. Conventional BRET systems have solely depended on an overlap of the energy donor and acceptor spectra. In this study, we engineered a conceptually unique ligand-activatable BRET system (termed BRET9), where a full-length Artificial Luciferase variant 23 (ALuc23), acting as the energy donor, is sandwiched between a protein pair of interest, FRB and FKBP12, and linked to a fluorescent protein as the energy acceptor. A specific ligand, rapamycin, then activates inter- and intramolecular interactions of FRB and FKBP12, which develop molecular strain in the sandwiched ALuc23 to accelerate further folding. We found that this system greatly enhanced both the total bioluminescence spectrum and the BRET signal in the far-red (FR) region. We characterized the molecular construct by studying 18 different designs categorized into four groups. The best BRET system design allowed an approximately 5-fold enhancement of the bioluminescence intensities in the FR region. This new BRET system provides a robust ligand-activatable platform that efficiently reports FR bioluminescence signals in cells and living animal models.


Assuntos
Luciferases/química , Serina-Treonina Quinases TOR/metabolismo , Proteína 1A de Ligação a Tacrolimo/metabolismo , Animais , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Linhagem Celular Tumoral , Humanos , Ligantes , Limite de Detecção , Luciferases/genética , Proteínas Luminescentes/química , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Ligação Proteica , Sirolimo/química , Sirolimo/metabolismo
14.
Analyst ; 144(12): 3765-3772, 2019 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-31089611

RESUMO

Investigation of the functions of insulin-secreting cells in response to glucose in single-living cells is essential for improving our knowledge on the pathogenesis of diabetes. Therefore, it is desired to develop a new convenient method that enables the direct detection of insulin secreted from single-living cells. Here, insulin-sensor-cells expressing a protein-based insulin-detecting probe immobilized on the extracellular membrane were developed to evaluate the insulin-secretion response in single-living pancreatic ß cells. The protein-based insulin-detecting probe (NαLY) was composed of a bioluminescent protein (nano-luc), the αCT segment of the insulin receptor, L1 and CR domains of the insulin receptor, and a fluorescent protein (YPet). NαLY exhibited a bioluminescence resonance energy transfer (BRET) signal in response to insulin; thus, cells of Hepa1-6 line were genetically engineered to express NαLY on the extracellular membrane. The cells were found to act as insulin-sensor-cells, exhibiting a BRET signal in response to insulin. When the insulin-sensor-cells and pancreatic ß cells (MIN6 cell line) were cocultured and stimulated with glucose, insulin-sensor-cells nearby pancreatic ß cells showed the spike-shaped BRET signal response, whereas the insulin-sensor-cells close to one pancreatic ß cell did not exhibit such signal response. However, all the insulin-sensor-cells showed a gradual increase in BRET signals, which were presumably attributed to the increase in insulin concentrations in the culture dish, confirming the function of these insulin-sensor-cells. Therefore, we demonstrated that heterogenetic insulin secretion in single-living pancreatic ß cells could be measured directly using the insulin sensor cells.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Técnicas Biossensoriais/métodos , Células Secretoras de Insulina/metabolismo , Insulina/análise , Análise de Célula Única/métodos , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura/métodos , Fluorescência , Glucose/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Engenharia de Proteínas/métodos , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
15.
Cell Signal ; 54: 27-34, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30471466

RESUMO

Bioluminescence resonance energy transfer (BRET) is a versatile tool used to investigate membrane receptor signalling and function. We have recently developed a homogenous NanoBRET ligand binding assay to monitor interactions between G protein-coupled receptors and fluorescent ligands. However, this assay requires the exogenous expression of a receptor fused to the nanoluciferase (Nluc) and is thus not applicable to natively-expressed receptors. To overcome this limitation in HEK293 cells, we have utilised CRISPR/Cas9 genome engineering to insert Nluc in-frame with the endogenous ADORA2B locus this resulted in HEK293 cells expressing adenosine A2B receptors under endogenous promotion tagged on their N-terminus with Nluc. As expected, we found relatively low levels of endogenous (gene-edited) Nluc/A2B receptor expression compared to cells transiently transfected with expression vectors coding for Nluc/A2B. However, in cells expressing gene-edited Nluc/A2B receptors we observed clear saturable ligand binding of a non-specific fluorescent adenosine receptor antagonist XAC-X-BY630 (Kd = 21.4 nM). Additionally, at gene-edited Nluc/A2B receptors we derived pharmacological parameters of ligand binding; Kd as well as Kon and Koff for binding of XAC-X-BY630 by NanoBRET association kinetic binding assays. Lastly, cells expressing gene-edited Nluc/A2B were used to determine the pKi of unlabelled adenosine receptor ligands in competition ligand binding assays. Utilising CRISPR/Cas9 genome engineering here we show that NanoBRET ligand binding assays can be performed at gene-edited receptors under endogenous promotion in live cells, therefore overcoming a fundamental limitation of NanoBRET ligand assays.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Receptor A2B de Adenosina/análise , Sistemas CRISPR-Cas , Células HEK293 , Humanos , Luciferases/química
16.
Biochem Pharmacol ; 158: 232-242, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30347205

RESUMO

The type 1 angiotensin II (AngII) receptor (AT1R) transactivates the epidermal growth factor receptor (EGFR), which leads to pathological remodeling of heart, blood vessels and kidney. End-point assays are used as surrogates of EGFR activation, however these downstream readouts are not applicable to live cells, in real-time. Herein, we report the use of a bioluminescence resonance energy transfer (BRET)-based assay to assess recruitment of the EGFR adaptor protein, growth factor receptor-bound protein 2 (Grb2), to the EGFR. In a variety of cell lines, both epidermal growth factor (EGF) and AngII stimulated Grb2 recruitment to EGFR. The BRET assay was used to screen a panel of 9 G protein-coupled receptors (GPCRs) and further developed for other EGFR family members (HER2 and HER3); the AT1R was able to transactivate HER2, but not HER3. Mechanistically, AT1R-mediated ERK1/2 activation was dependent on Gq/11 and EGFR tyrosine kinase activity, whereas the recruitment of Grb2 to the EGFR was independent of Gq/11 and only partially dependent on EGFR tyrosine kinase activity. This Gq/11 independence of EGFR transactivation was confirmed using AT1R mutants and in CRISPR cell lines lacking Gq/11. EGFR transactivation was also apparently independent of ß-arrestins. Finally, we used additional BRET-based assays and confocal microscopy to provide evidence that both AngII- and EGF-stimulation promoted AT1R-EGFR heteromerization. In summary, we report an alternative approach to monitoring AT1R-EGFR transactivation in live cells, which provides a more direct and proximal view of this process, including the potential for complexes between the AT1R and EGFR.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Ativação Transcricional/fisiologia , Animais , Células CHO , Cricetulus , Receptores ErbB/análise , Receptores ErbB/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/análise , Células HEK293 , Humanos , Masculino , Camundongos , Células NIH 3T3 , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/análise
17.
Pharmacol Res Perspect ; 6(5): e00432, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30263124

RESUMO

Relaxin family peptide (RXFPs) 1-4 receptors modulate the activity of cyclic adenosine monophosphate (cAMP) to produce a range of physiological functions. RXFP1 and RXFP2 increase cAMP via Gαs, whereas RXFP3 and RXFP4 inhibit cAMP via Gαi/o. RXFP1 also shows a delayed increase in cAMP downstream of Gαi3. In this study we have assessed whether the bioluminescence resonance energy transfer (BRET)-based biosensor CAMYEL (cAMP sensor using YFP-Epac-Rluc), which allows real-time measurement of cAMP activity in live cells, will aid in understanding ligand- and cell-specific RXFP signaling. CAMYEL detected concentration-dependent changes in cAMP activity at RXFP1-4 in recombinant cell lines, using a variety of ligands with potencies comparable to those seen in conventional cAMP assays. We used RXFP2 and RXFP3 antagonists to demonstrate that CAMYEL detects dynamic changes in cAMP by reversing cAMP activation or inhibition respectively, with real-time addition of antagonist after agonist stimulation. To demonstrate the utility of CAMYEL to detect cAMP activation in native cells expressing low levels of RXFP receptor, we cloned CAMYEL into a lentiviral vector and transduced THP-1 cells, which express low levels of RXFP1. THP-1 CAMYEL cells demonstrated robust cAMP activation in response to relaxin. However, the CAMYEL assay was unable to detect the Gαi3-mediated phase of RXFP1 cAMP activation in PTX-treated THP-1 cells or HEK293A cells with knockout of Gαs. Our data demonstrate that cytoplasmically-expressed CAMYEL efficiently detects real-time cAMP activation by Gαs or inhibition by Gαi/o but may not detect cAMP generated in specific intracellular compartments such as that generated by Gαi3 upon RXFP1 activation.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Técnicas Biossensoriais/métodos , AMP Cíclico/metabolismo , Transferência de Energia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Citoplasma/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas Luminescentes/química , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Relaxina/metabolismo , Transdução de Sinais , Fatores de Tempo
18.
Anal Chem ; 90(19): 11495-11502, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30188118

RESUMO

By virtue of its self-illuminating mechanism, the bioluminescence resonance energy transfer (BRET) technique has recently emerged as a promising platform for point-of-care (POC) diagnostics. However, due to the difficulty of incorporating generic affinity elements, such as aptamers and antibodies, current BRET-based methods are still not applicable to most clinically important biomarkers. Furthermore, the inability of these methods to amplify BRET signals leads to limited sensitivity in some applications. Here, we present a modular strategy for amplified BRET detection of protein biomarkers in human peripheral blood samples. In this strategy, a DNA-templated bioluminescent module was constructed by simultaneously binding luciferase and green fluorescent protein to one DNA template in a site-specific manner. The proposed modules showed high energy transfer efficiency and could be assembled into long self-illuminating polymers. Owing to this modular design, aptamers and antibodies were rationally incorporated, enabling specific assembly of multiple bioluminescent modules on one target. This strategy realized amplified BRET assays for human α-thrombin and prostate specific antigen (PSA) with the detection limit in the picomolar range using either a spectrophotometer or a smartphone. The modularity of our strategy allowed detection of different biomarkers by simple exchange of affinity elements. Furthermore, the self-illumination and isothermal amplification performance of this strategy make it an attractive tool for POC diagnostics.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Biomarcadores/análise , DNA/química , Humanos , Imunoensaio , Limite de Detecção , Sistemas Automatizados de Assistência Junto ao Leito , Antígeno Prostático Específico/análise , Smartphone , Trombina/análise
19.
FEBS Lett ; 592(18): 3126-3138, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30117167

RESUMO

Protease-activated receptor 2 (PAR2) is a G protein-coupled receptor (GPCR) activated by endogenous proteases, in particular, trypsin. Although regulators of G protein signaling (RGS) are known to inhibit GPCR/Gα-mediated signaling, their specific effects on PAR2 are poorly understood at present. Here, we use a bioluminescence resonance energy transfer technique to investigate whether RGS16 and RGS18 bind PAR2 in live cells to regulate PAR2/Gαi/o -mediated signaling. Notably, we find that RGS16 binds to PAR2 in the presence of Gαi while RGS18 does not interact with PAR2, regardless of the presence of Gα. Both RGS16 and RGS18 inhibit PAR2/Gαi/o -mediated signaling. To our knowledge, the current study is the first to highlight the effects of RGS proteins on PAR2-mediated signaling.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Proteínas RGS/metabolismo , Receptor PAR-2/metabolismo , Animais , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia Confocal , Ligação Proteica , Proteínas RGS/genética , Receptor PAR-2/genética , Transdução de Sinais
20.
Elife ; 72018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29989546

RESUMO

The RAS family of proteins is amongst the most highly mutated in human cancers and has so far eluded drug therapy. Currently, much effort is being made to discover mutant RAS inhibitors and in vitro screening for RAS-binding drugs must be followed by cell-based assays. Here, we have developed a robust set of bioluminescence resonance energy transfer (BRET)-based RAS biosensors that enable monitoring of RAS-effector interaction inhibition in living cells. These include KRAS, HRAS and NRAS and a variety of different mutations that mirror those found in human cancers with the major RAS effectors such as CRAF, PI3K and RALGDS. We highlighted the utility of these RAS biosensors by showing a RAS-binding compound is a potent pan-RAS-effector interactions inhibitor in cells. The RAS biosensors represent a useful tool to investigate and characterize the potency of anti-RAS inhibitors in cells and more generally any RAS protein-protein interaction (PPI) in cells.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Técnicas Biossensoriais/métodos , Mutação , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Transferência de Energia , Células HEK293 , Humanos , Ligação Proteica , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA