Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Front Cell Neurosci ; 15: 814547, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35110998

RESUMO

Neurons integrate inputs over different time and space scales. Fast excitatory synapses at boutons (ms and µm), and slow modulation over entire dendritic arbors (seconds and mm) are all ultimately combined to produce behavior. Understanding the timing of signaling events mediated by G-protein-coupled receptors is necessary to elucidate the mechanism of action of therapeutics targeting the nervous system. Measuring signaling kinetics in live cells has been transformed by the adoption of fluorescent biosensors and dyes that convert biological signals into optical signals that are conveniently recorded by microscopic imaging or by fluorescence plate readers. Quantifying the timing of signaling has now become routine with the application of equations in familiar curve fitting software to estimate the rates of signaling from the waveform. Here we describe examples of the application of these methods, including (1) Kinetic analysis of opioid signaling dynamics and partial agonism measured using cAMP and arrestin biosensors; (2) Quantifying the signaling activity of illicit synthetic cannabinoid receptor agonists measured using a fluorescent membrane potential dye; (3) Demonstration of multiplicity of arrestin functions from analysis of biosensor waveforms and quantification of the rates of these processes. These examples show how temporal analysis provides additional dimensions to enhance the understanding of GPCR signaling and therapeutic mechanisms in the nervous system.

2.
Sci Rep ; 10(1): 12263, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32704081

RESUMO

In classical pharmacology, bioassay data are fit to general equations (e.g. the dose response equation) to determine empirical drug parameters (e.g. EC50 and Emax), which are then used to calculate chemical parameters such as affinity and efficacy. Here we used a similar approach for kinetic, time course signaling data, to allow empirical and chemical definition of signaling by G-protein-coupled receptors in kinetic terms. Experimental data are analyzed using general time course equations (model-free approach) and mechanistic model equations (mechanistic approach) in the commonly-used curve-fitting program, GraphPad Prism. A literature survey indicated signaling time course data usually conform to one of four curve shapes: the straight line, association exponential curve, rise-and-fall to zero curve, and rise-and-fall to steady-state curve. In the model-free approach, the initial rate of signaling is quantified and this is done by curve-fitting to the whole time course, avoiding the need to select the linear part of the curve. It is shown that the four shapes are consistent with a mechanistic model of signaling, based on enzyme kinetics, with the shape defined by the regulation of signaling mechanisms (e.g. receptor desensitization, signal degradation). Signaling efficacy is the initial rate of signaling by agonist-occupied receptor (kτ), simply the rate of signal generation before it becomes affected by regulation mechanisms, measurable using the model-free analysis. Regulation of signaling parameters such as the receptor desensitization rate constant can be estimated if the mechanism is known. This study extends the empirical and mechanistic approach used in classical pharmacology to kinetic signaling data, facilitating optimization of new therapeutics in kinetic terms.


Assuntos
Modelos Biológicos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Relação Dose-Resposta a Droga , Descoberta de Drogas , Farmacocinética , Transdução de Sinais/efeitos dos fármacos
3.
Sci Rep ; 10(1): 1766, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32019973

RESUMO

The kinetics/dynamics of signaling are of increasing value for G-protein-coupled receptor therapeutic development, including spatiotemporal signaling and the kinetic context of biased agonism. Effective application of signaling kinetics to developing new therapeutics requires reliable kinetic assays and an analysis framework to extract kinetic pharmacological parameters. Here we describe a platform for measuring arrestin recruitment kinetics to GPCRs using a high quantum yield, genetically encoded fluorescent biosensor, and a data analysis framework to quantify the recruitment kinetics. The sensor enabled high temporal resolution measurement of arrestin recruitment to the angiotensin AT1 and vasopressin V2 receptors. The analysis quantified the initial rate of arrestin recruitment (kτ), a biologically-meaningful kinetic drug efficacy parameter, by fitting time course data using routine curve-fitting methods. Biased agonism was assessed by comparing kτ values for arrestin recruitment with those for Gq signaling via the AT1 receptor. The kτ ratio values were in good agreement with bias estimates from existing methods. This platform potentially improves and simplifies assessment of biased agonism because the same assay modality is used to compare pathways (potentially in the same cells), the analysis method is parsimonious and intuitive, and kinetic context is factored into the bias measurement.


Assuntos
Técnicas Biossensoriais/métodos , Ligação Proteica/fisiologia , Transdução de Sinais/fisiologia , Angiotensina I/metabolismo , Arrestinas/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Cinética , Ligantes , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Vasopressinas/metabolismo
4.
SLAS Discov ; 23(9): 898-906, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29991302

RESUMO

Cell-based assays to detect Gαi signaling are often indirect, frequently involve complex pharmacological interventions, and are usually blind to the kinetics of the signaling. Our goal was to develop a simple, direct measure of Gαi signaling in living cells. We previously reported our fluorescent cADDis assay and showed that it reliably detects Gαs-mediated increases in cAMP levels. Agonists that stimulate a Gs-coupled receptor produce changes in the intensity of bright green or red fluorescent protein sensors that can be followed over time using automated fluorescence plate readers or fluorescence imaging systems. Since the cADDis sensors can monitor Gαs-mediated increases in adenylyl cyclase activity, in theory they should also be capable of detecting Gαi-mediated decreases. Here we apply our green fluorescent cADDis sensor to the detection of Gαi-mediated inhibition of adenylyl cyclase activity. We validated and optimized the assay in living HEK 293T cells using several known Gαi-coupled receptors and agonists, and we report robust Z' statistics and consistent EC50 responses.


Assuntos
Bioensaio/métodos , AMP Cíclico/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Adenilil Ciclases/metabolismo , Animais , Automação Laboratorial , Biomarcadores , Ativação Enzimática , Células HEK293 , Humanos , Cinética , Microscopia de Fluorescência , Imagem Óptica/métodos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 113(46): 13069-13074, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27799542

RESUMO

Protein kinase A (PKA) phosphorylates Gli proteins, acting as a negative regulator of the Hedgehog pathway. PKA was recently detected within the cilium, and PKA activity specifically in cilia regulates Gli processing. Using a cilia-targeted genetically encoded sensor, we found significant basal PKA activity. Using another targeted sensor, we measured basal ciliary cAMP that is fivefold higher than whole-cell cAMP. The elevated basal ciliary cAMP level is a result of adenylyl cyclase 5 and 6 activity that depends on ciliary phosphatidylinositol (3,4,5)-trisphosphate (PIP3), not stimulatory G protein (Gαs), signaling. Sonic Hedgehog (SHH) reduces ciliary cAMP levels, inhibits ciliary PKA activity, and increases Gli1. Remarkably, SHH regulation of ciliary cAMP and downstream signals is not dependent on inhibitory G protein (Gαi/o) signaling but rather Ca2+ entry through a Gd3+-sensitive channel. Therefore, PIP3 sustains high basal cAMP that maintains PKA activity in cilia and Gli repression. SHH activates Gli by inhibiting cAMP through a G protein-independent mechanism that requires extracellular Ca2+ entry.


Assuntos
Cálcio/metabolismo , Cílios/metabolismo , AMP Cíclico/metabolismo , Proteínas Hedgehog/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Camundongos
6.
J Biomol Screen ; 21(3): 298-305, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26657040

RESUMO

Protein-based, fluorescent biosensors power basic research on cell signaling in health and disease, but their use in automated laboratories is limited. We have now created two live-cell assays, one for diacyl glycerol and another for cAMP, that are robust (Z' > 0.7) and easily deployed on standard fluorescence plate readers. We describe the development of these assays, focusing on the parameters that were critical for optimization, in the hopes that the lessons learned can be generalized to the development of new biosensor-based assays.


Assuntos
Automação Laboratorial , Técnicas Biossensoriais , AMP Cíclico/metabolismo , Diglicerídeos/metabolismo , Baculoviridae/fisiologia , AMP Cíclico/química , Diglicerídeos/química , Células HEK293 , Humanos , Reprodutibilidade dos Testes , Transdução Genética
7.
Nat Methods ; 12(3): 195-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25622108

RESUMO

We have developed a versatile new class of genetically encoded fluorescent biosensor based on reversible exchange of the heterodimeric partners of green and red dimerization-dependent fluorescent proteins. We demonstrate the use of this strategy to construct both intermolecular and intramolecular ratiometric biosensors for qualitative imaging of caspase activity, Ca(2+) concentration dynamics and other second-messenger signaling activities.


Assuntos
Técnicas Biossensoriais/métodos , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/metabolismo , Cálcio/metabolismo , Calmodulina/metabolismo , Caspase 3/genética , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Proteínas Luminescentes/genética , Imagem Molecular/métodos , Multimerização Proteica , Proteína Vermelha Fluorescente
8.
Cells ; 3(2): 247-57, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24709960

RESUMO

Transient receptor potential canonical 6 (TRPC6) is a cation selective, DAG-regulated, Ca2+-permeable channel activated by the agonists of Gq-protein-coupled heptahelical receptors. Dysfunctions of TRPC6 are implicated in the pathogenesis of various cardiovascular and kidney conditions such as vasospasm and glomerulosclerosis. When stimulated by agonists of the histamine H1 receptor (H1R), TRPC6 activity decays to the baseline despite the continuous presence of the agonist. In this study, we examined whether H1R desensitization contributes to regulating the decay rate of TRPC6 activity upon receptor stimulation. We employed the HEK expression system and a biosensor allowing us to simultaneously detect the changes in intracellular diacylglycerol (DAG) and Ca2+ concentrations. We found that the histamine-induced DAG response was biphasic, in which a transient peak was followed by maintained elevated plateau, suggesting that desensitization of H1R takes place in the presence of histamine. The application of PKC inhibitor Gö6983 slowed the decay rate of intracellular DAG concentration. Activation of the mouse H1R mutant lacking a putative PKC phosphorylation site, Ser399, responsible for the receptor desensitization, resulted in a prolonged intracellular DAG increase and greater Mn2+ influx through the TRPC6 channel. Thus, our data support the hypothesis that PKC-dependent H1R phosphorylation leads to a reduced production of intracellular DAG that contributes to TRPC6 activity regulation.

9.
J Biomol Screen ; 18(7): 797-806, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23580666

RESUMO

There is a growing need in drug discovery and basic research to measure multiple second-messenger components of cell signaling pathways in real time and in relevant tissues and cell types. Many G-protein-coupled receptors activate the heterotrimeric protein, Gq, which in turn activates phospholipase C (PLC). PLC cleaves phosphatidylinositol 4,5-bisphosphate (PIP2) to produce two second messengers: diacylglycerol (DAG), which remains in the plasma membrane, and inositol triphosphate (IP3), which diffuses through the cytosol to release stores of intracellular calcium ions (Ca(2+)). Our goal was to create a series of multiplex sensors that would make it possible to simultaneously measure two different components of the Gq pathway in living cells. Here we describe new fluorescent sensors for DAG and PIP2 that produce robust changes in green or red fluorescence and can be combined with one another, or with existing Ca(2+) sensors, in a live-cell assay. These assays can detect multiple components of Gq signaling, simultaneously in real time, on standard fluorescent plate readers or live-cell imaging systems.


Assuntos
Bioensaio , Sinalização do Cálcio , Receptores Acoplados a Proteínas G/metabolismo , Trifosfato de Adenosina/fisiologia , Técnicas Biossensoriais , Diglicerídeos/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Células HEK293 , Humanos , Proteínas Luminescentes/biossíntese , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína Quinase C-delta/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Sistemas do Segundo Mensageiro , Espectrometria de Fluorescência , Fosfolipases Tipo C/metabolismo , Proteína Vermelha Fluorescente
10.
PLoS One ; 7(8): e42791, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912738

RESUMO

Phospholipase C produces two second messengers--diacylglycerol (DAG), which remains in the membrane, and inositol triphosphate (IP(3)), which triggers the release of calcium ions (Ca(2+)) from intracellular stores. Genetically encoded sensors based on a single circularly permuted fluorescent protein (FP) are robust tools for studying intracellular Ca(2+) dynamics. We have developed a robust sensor for DAG based on a circularly permuted green FP that can be co-imaged with the red fluorescent Ca(2+) sensor R-GECO for simultaneous measurement of both second messengers.


Assuntos
Técnicas Biossensoriais/métodos , Cálcio/metabolismo , Diglicerídeos/metabolismo , Sistemas do Segundo Mensageiro , Sobrevivência Celular , Corantes Fluorescentes/metabolismo , Células HEK293 , Humanos , Espaço Intracelular/metabolismo , Proteínas Luminescentes/química , Proteínas Luminescentes/metabolismo , Conformação Proteica , Proteína Quinase C-delta/química , Proteína Quinase C-delta/metabolismo , Fatores de Tempo
11.
Diabetes ; 60(6): 1797-804, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21536946

RESUMO

OBJECTIVE: Congenital hyperinsulinemic hypoglycemia is a group of genetic disorders of insulin secretion most commonly associated with inactivating mutations of the ß-cell ATP-sensitive K(+) channel (K(ATP) channel) genes ABCC8 (SUR1) and KCNJ11 (Kir6.2). Recessive mutations of these genes cause hyperinsulinism that is unresponsive to treatment with diazoxide, a channel agonist. Dominant K(ATP) mutations have been associated with diazoxide-responsive disease. We hypothesized that some medically uncontrollable cases with only one K(ATP) mutation might have dominant, diazoxide-unresponsive disease. RESEARCH DESIGN AND METHODS: Mutations of the K(ATP) genes were identified by sequencing genomic DNA. Effects of mutations on K(ATP) channel function in vitro were studied by expression in COSm6 cells. RESULTS: In 15 families with diazoxide-unresponsive diffuse hyperinsulism, we found 17 patients with a monoallelic missense mutation of SUR1. Nine probands had de novo mutations, two had an affected sibling or parent, and four had an asymptomatic carrier parent. Of the 13 different mutations, 12 were novel. Expression of mutations revealed normal trafficking of channels but severely impaired responses to diazoxide or MgADP. Responses were significantly lower compared with nine SUR1 mutations associated with dominant, diazoxide-responsive hyperinsulinism. CONCLUSIONS: These results demonstrate that some dominant mutations of SUR1 can cause diazoxide-unresponsive hyperinsulinism. In vitro expression studies may be helpful in distinguishing such mutations from dominant mutations of SUR1 associated with diazoxide-responsive disease.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Hiperinsulinismo Congênito/tratamento farmacológico , Hiperinsulinismo Congênito/genética , Diazóxido/uso terapêutico , Canais de Potássio Corretores do Fluxo de Internalização/genética , Receptores de Droga/genética , Anti-Hipertensivos/uso terapêutico , Humanos , Modelos Biológicos , Mutação , Mutação de Sentido Incorreto/genética , Linhagem , Receptores de Sulfonilureias
12.
J Gen Physiol ; 137(3): 299-314, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21321069

RESUMO

Functional integrity of pancreatic adenosine triphosphate (ATP)-sensitive potassium (K(ATP)) channels depends on the interactions between the pore-forming potassium channel subunit Kir6.2 and the regulatory subunit sulfonylurea receptor 1 (SUR1). Previous studies have shown that the N-terminal transmembrane domain of SUR1 (TMD0) interacts with Kir6.2 and is sufficient to confer high intrinsic open probability (P(o)) and bursting patterns of activity observed in full-length K(ATP) channels. However, the nature of TMD0-Kir6.2 interactions that underlie gating modulation is not well understood. Using two previously described disease-causing mutations in TMD0 (R74W and E128K), we performed amino acid substitutions to study the structural roles of these residues in K(ATP) channel function in the context of full-length SUR1 as well as TMD0. Our results revealed that although R74W and E128K in full-length SUR1 both decrease surface channel expression and reduce channel sensitivity to ATP inhibition, they arrive there via distinct mechanisms. Mutation of R74 uniformly reduced TMD0 protein levels, suggesting that R74 is necessary for stability of TMD0. In contrast, E128 mutations retained TMD0 protein levels but reduced functional coupling between TMD0 and Kir6.2 in mini-K(ATP) channels formed by TMD0 and Kir6.2. Importantly, E128K full-length channels, despite having a greatly reduced P(o), exhibit little response to phosphatidylinositol 4,5-bisphosphate (PIP(2)) stimulation. This is reminiscent of Kir6.2 channel behavior in the absence of SUR1 and suggests that TMD0 controls Kir6.2 gating by modulating Kir6.2 interactions with PIP(2). Further supporting this notion, the E128W mutation in full-length channels resulted in channel inactivation that was prevented or reversed by exogenous PIP(2). These results identify a critical determinant in TMD0 that controls Kir6.2 gating by controlling channel sensitivity to PIP(2). Moreover, they uncover a novel mechanism of K(ATP) channel inactivation involving aberrant functional coupling between SUR1 and Kir6.2.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Ativação do Canal Iônico , Canais KATP/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Droga/metabolismo , Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/genética , Trifosfato de Adenosina/metabolismo , Animais , Arginina , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Ácido Glutâmico , Canais KATP/química , Canais KATP/genética , Potenciais da Membrana , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Estrutura Terciária de Proteína , Transporte Proteico , Ratos , Receptores de Droga/química , Receptores de Droga/genética , Receptores de Sulfonilureias , Fatores de Tempo , Transfecção
13.
J Biol Chem ; 285(9): 6012-23, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20032456

RESUMO

The inwardly rectifying potassium channel Kir6.2 assembles with sulfonylurea receptor 1 to form the ATP-sensitive potassium (K(ATP)) channels that regulate insulin secretion in pancreatic beta-cells. Mutations in K(ATP) channels underlie insulin secretion disease. Here, we report the characterization of a heterozygous missense Kir6.2 mutation, G156R, identified in congenital hyperinsulinism. Homomeric mutant channels reconstituted in COS cells show similar surface expression as wild-type channels but fail to conduct potassium currents. The mutated glycine is in the pore-lining transmembrane helix of Kir6.2; an equivalent glycine in other potassium channels has been proposed to serve as a hinge to allow helix bending during gating. We found that mutation of an adjacent asparagine, Asn-160, to aspartate, which converts the channel from a weak to a strong inward rectifier, on the G156R background restored ion conduction in the mutant channel. Unlike N160D channels, however, G156R/N160D channels are not blocked by intracellular polyamines at positive membrane potential and exhibit wild-type-like nucleotide sensitivities, suggesting the aspartate introduced at position 160 interacts with arginine at 156 to restore ion conduction and gating. Using tandem Kir6.2 tetramers containing G156R and/or N160D in designated positions, we show that one mutant subunit in the tetramer is insufficient to abolish conductance and that G156R and N160D can interact in the same or adjacent subunits to restore conduction. We conclude that the glycine at 156 is not essential for K(ATP) channel gating and that the Kir6.2 gating defect caused by the G156R mutation could be rescued by manipulating chemical interactions between pore residues.


Assuntos
Hiperinsulinismo Congênito/genética , Mutação de Sentido Incorreto , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Eletrofisiologia , Glicina , Ativação do Canal Iônico/genética , Camundongos , Canais de Potássio Corretores do Fluxo de Internalização/química , Conformação Proteica
14.
Mech Dev ; 125(11-12): 1033-47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18718533

RESUMO

In vertebrate embryos, neural crest cells emerge from the dorsal neural tube and migrate along well defined pathways to form a wide diversity of tissues, including the majority of the peripheral nervous system (PNS). Members of the cadherin family of cell adhesion molecules play key roles during the initiation of migration, mediating the delamination of cells from the neural tube. However, a role for cadherins in the sorting and re-aggregation of the neural crest to form the PNS has not been established. We report the requirement for a protocadherin, chicken protocadherin-1 (Pcdh1), in neural crest cell sorting during the formation of the dorsal root ganglia (DRG). In embryos, cPcdh1 is highly expressed in the developing DRG, where it co-localizes with the undifferentiated and mitotically active cells along the perimeter. Pcdh1 can promote cell adhesion in vivo and disrupting Pcdh1 function in embryos results in fewer neural crest cells localizing to the DRG, with a concomitant increase in cells that migrate to the sympathetic ganglia. Furthermore, those cells that still localize to the DRG, when Pcdh1 is inhibited, are no longer found at the perimeter, but are instead dispersed throughout the DRG and are now more likely to differentiate along the sensory neuron pathway. These results demonstrate that Pcdh1-mediated cell adhesion plays an important role as neural crest cells coalesce to form the DRG, where it serves to sort cells to the mitotically active perimeter.


Assuntos
Caderinas/fisiologia , Gânglios Espinais/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/metabolismo , Sistema Nervoso Periférico/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular , Linhagem da Célula , Movimento Celular , Galinhas , Gânglios Espinais/metabolismo , Hibridização In Situ , Microscopia de Fluorescência , Mitose , Neurônios/metabolismo , Protocaderinas , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA