Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
2.
Proc Natl Acad Sci U S A ; 119(32): e2122037119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35914163

ABSTRACT

Receptor-activity-modifying proteins (RAMPs) are ubiquitously expressed membrane proteins that associate with different G protein-coupled receptors (GPCRs), including the parathyroid hormone 1 receptor (PTH1R), a class B GPCR and an important modulator of mineral ion homeostasis and bone metabolism. However, it is unknown whether and how RAMP proteins may affect PTH1R function. Using different optical biosensors to measure the activation of PTH1R and its downstream signaling, we describe here that RAMP2 acts as a specific allosteric modulator of PTH1R, shifting PTH1R to a unique preactivated state that permits faster activation in a ligand-specific manner. Moreover, RAMP2 modulates PTH1R downstream signaling in an agonist-dependent manner, most notably increasing the PTH-mediated Gi3 signaling sensitivity. Additionally, RAMP2 increases both PTH- and PTHrP-triggered ß-arrestin2 recruitment to PTH1R. Employing homology modeling, we describe the putative structural molecular basis underlying our functional findings. These data uncover a critical role of RAMPs in the activation and signaling of a GPCR that may provide a new venue for highly specific modulation of GPCR function and advanced drug design.


Subject(s)
Receptor Activity-Modifying Protein 2 , Receptor, Parathyroid Hormone, Type 1 , Signal Transduction , Biosensing Techniques , Ligands , Parathyroid Hormone/metabolism , Receptor Activity-Modifying Protein 2/genetics , Receptor Activity-Modifying Protein 2/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 2/metabolism
3.
Cell ; 185(7): 1130-1142.e11, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35294858

ABSTRACT

G protein-coupled receptors (GPCRs) relay extracellular stimuli into specific cellular functions. Cells express many different GPCRs, but all these GPCRs signal to only a few second messengers such as cAMP. It is largely unknown how cells distinguish between signals triggered by different GPCRs to orchestrate their complex functions. Here, we demonstrate that individual GPCRs signal via receptor-associated independent cAMP nanodomains (RAINs) that constitute self-sufficient, independent cell signaling units. Low concentrations of glucagon-like peptide 1 (GLP-1) and isoproterenol exclusively generate highly localized cAMP pools around GLP-1- and ß2-adrenergic receptors, respectively, which are protected from cAMP originating from other receptors and cell compartments. Mapping local cAMP concentrations with engineered GPCR nanorulers reveals gradients over only tens of nanometers that define the size of individual RAINs. The coexistence of many such RAINs allows a single cell to operate thousands of independent cellular signals simultaneously, rather than function as a simple "on/off" switch.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Cell Physiological Phenomena , Cyclic AMP , Glucagon-Like Peptide 1 , Receptors, Adrenergic, beta-2 , Receptors, G-Protein-Coupled/chemistry , Second Messenger Systems
4.
Methods Mol Biol ; 2483: 181-194, 2022.
Article in English | MEDLINE | ID: mdl-35286676

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is a universal second messenger that mediates a myriad of cell functions across all kingdoms of life.The ability to monitor intracellular changes of cAMP concentration in living cells using FRET-based biosensors is proving to be of paramount importance to unraveling the sophisticated organization of cAMP signaling.Here we describe the deployment of the fruit fly Drosophila melanogaster, specifically the third instar larval stage, as an in vivo model to study the spatio-temporal dynamics of cAMP in neurons. The ubiquity of cAMP signaling and conservation of fundamental mechanisms across species ensures relevance to vertebrate neurons while providing a more structurally and ethically simple model.


Subject(s)
Drosophila melanogaster , Drosophila , Animals , Cyclic AMP , Larva , Neurons
5.
Methods Mol Biol ; 2268: 137-147, 2021.
Article in English | MEDLINE | ID: mdl-34085266

ABSTRACT

Here we describe the stepwise application of bioluminescence resonance energy transfer (BRET)-based conformational receptor biosensors to study GPCR activation in intact cells. This technology can be easily adopted to various plate reader devices and microtiter plate formats. Due to the high sensitivity of these BRET-based receptor biosensors and their ability to quantify simultaneously receptor activation/de-activation kinetics as well as compound efficacy and potency, these optical tools provide the most direct and unbiased approach to monitor GPCR activity in a high-throughput-compatible assay format, representing a novel promising tool for the discovery of potential GPCR therapeutics.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Drug Evaluation, Preclinical/methods , Fluorescent Dyes/chemistry , High-Throughput Screening Assays/methods , Luciferases/metabolism , Receptors, G-Protein-Coupled/metabolism , HEK293 Cells , Humans , Protein Conformation , Receptors, G-Protein-Coupled/chemistry
7.
Cell ; 182(6): 1519-1530.e17, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32846156

ABSTRACT

Cells relay a plethora of extracellular signals to specific cellular responses by using only a few second messengers, such as cAMP. To explain signaling specificity, cAMP-degrading phosphodiesterases (PDEs) have been suggested to confine cAMP to distinct cellular compartments. However, measured rates of fast cAMP diffusion and slow PDE activity render cAMP compartmentalization essentially impossible. Using fluorescence spectroscopy, we show that, contrary to earlier data, cAMP at physiological concentrations is predominantly bound to cAMP binding sites and, thus, immobile. Binding and unbinding results in largely reduced cAMP dynamics, which we term "buffered diffusion." With a large fraction of cAMP being buffered, PDEs can create nanometer-size domains of low cAMP concentrations. Using FRET-cAMP nanorulers, we directly map cAMP gradients at the nanoscale around PDE molecules and the areas of resulting downstream activation of cAMP-dependent protein kinase (PKA). Our study reveals that spatiotemporal cAMP signaling is under precise control of nanometer-size domains shaped by PDEs that gate activation of downstream effectors.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Single-Cell Analysis/methods , Computer Simulation , Cyclic AMP/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Cytoplasm/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Models, Molecular , Phosphoric Diester Hydrolases/chemistry , Protein Binding , Protein Domains , Recombinant Proteins , Spatio-Temporal Analysis , Spectrometry, Fluorescence
8.
Commun Biol ; 1: 105, 2018.
Article in English | MEDLINE | ID: mdl-30271985

ABSTRACT

G-protein-coupled receptors (GPCRs) represent one of the most important classes of drug targets. The discovery of new GCPR therapeutics would greatly benefit from the development of a generalizable high-throughput assay to directly monitor their activation or de-activation. Here we screened a variety of labels inserted into the third intracellular loop and the C-terminus of the α2A-adrenergic receptor and used fluorescence (FRET) and bioluminescence resonance energy transfer (BRET) to monitor ligand-binding and activation dynamics. We then developed a universal intramolecular BRET receptor sensor design to quantify efficacy and potency of GPCR ligands in intact cells and real time. We demonstrate the transferability of the sensor design by cloning ß2-adrenergic and PTH1-receptor BRET sensors and monitored their efficacy and potency. For all biosensors, the Z factors were well above 0.5 showing the suitability of such design for microtiter plate assays. This technology will aid the identification of novel types of GPCR ligands.

9.
Elife ; 62017 08 08.
Article in English | MEDLINE | ID: mdl-28784204

ABSTRACT

Adhesion-type G protein-coupled receptors (aGPCRs), a large molecule family with over 30 members in humans, operate in organ development, brain function and govern immunological responses. Correspondingly, this receptor family is linked to a multitude of diverse human diseases. aGPCRs have been suggested to possess mechanosensory properties, though their mechanism of action is fully unknown. Here we show that the Drosophila aGPCR Latrophilin/dCIRL acts in mechanosensory neurons by modulating ionotropic receptor currents, the initiating step of cellular mechanosensation. This process depends on the length of the extended ectodomain and the tethered agonist of the receptor, but not on its autoproteolysis, a characteristic biochemical feature of the aGPCR family. Intracellularly, dCIRL quenches cAMP levels upon mechanical activation thereby specifically increasing the mechanosensitivity of neurons. These results provide direct evidence that the aGPCR dCIRL acts as a molecular sensor and signal transducer that detects and converts mechanical stimuli into a metabotropic response.


Subject(s)
Action Potentials , Cyclic AMP/metabolism , Drosophila Proteins/metabolism , Mechanoreceptors/physiology , Receptors, Peptide/metabolism , Sensory Receptor Cells/physiology , Animals , Drosophila , Electrophysiological Phenomena , Optical Imaging
10.
PLoS One ; 12(4): e0174856, 2017.
Article in English | MEDLINE | ID: mdl-28406920

ABSTRACT

In their role as second messengers, cyclic nucleotides such as cAMP have a variety of intracellular effects. These complex tasks demand a highly organized orchestration of spatially and temporally confined cAMP action which should be best achieved by compartmentalization of the latter. A great body of evidence suggests that cAMP compartments may be established and maintained by cAMP degrading enzymes, e.g. phosphodiesterases (PDEs). However, the molecular and biophysical details of how PDEs can orchestrate cAMP gradients are entirely unclear. In this paper, using fusion proteins of cAMP FRET-sensors and PDEs in living cells, we provide direct experimental evidence that the cAMP concentration in the vicinity of an individual PDE molecule is below the detection limit of our FRET sensors (<100nM). This cAMP gradient persists in crude cytosol preparations. We developed mathematical models based on diffusion-reaction equations which describe the creation of nanocompartments around a single PDE molecule and more complex spatial PDE arrangements. The analytically solvable equations derived here explicitly determine how the capability of a single PDE, or PDE complexes, to create a nanocompartment depend on the cAMP degradation rate, the diffusive mobility of cAMP, and geometrical and topological parameters. We apply these generic models to our experimental data and determine the diffusive mobility and degradation rate of cAMP. The results obtained for these parameters differ by far from data in literature for free soluble cAMP interacting with PDE. Hence, restricted cAMP diffusion in the vincinity of PDE is necessary to create cAMP nanocompartments in cells.


Subject(s)
Cyclic AMP/metabolism , Cytosol/metabolism , Models, Biological , Phosphoric Diester Hydrolases/metabolism , Cell Line , Humans
11.
Cell Rep ; 17(5): 1238-1246, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27783939

ABSTRACT

The second messenger cyclic AMP (cAMP) plays an important role in synaptic plasticity. Although there is evidence for local control of synaptic transmission and plasticity, it is less clear whether a similar spatial confinement of cAMP signaling exists. Here, we suggest a possible biophysical basis for the site-specific regulation of synaptic plasticity by cAMP, a highly diffusible small molecule that transforms the physiology of synapses in a local and specific manner. By exploiting the octopaminergic system of Drosophila, which mediates structural synaptic plasticity via a cAMP-dependent pathway, we demonstrate the existence of local cAMP signaling compartments of micrometer dimensions within single motor neurons. In addition, we provide evidence that heterogeneous octopamine receptor localization, coupled with local differences in phosphodiesterase activity, underlies the observed differences in cAMP signaling in the axon, cell body, and boutons.


Subject(s)
Cyclic AMP/metabolism , Drosophila melanogaster/metabolism , Motor Neurons/metabolism , Presynaptic Terminals/metabolism , Signal Transduction , Animals , Axons/drug effects , Axons/metabolism , Cell Body/drug effects , Cell Body/metabolism , Drosophila melanogaster/drug effects , Fluorescence Resonance Energy Transfer , Iontophoresis , Motor Neurons/drug effects , Octopamine/pharmacology , Phosphoric Diester Hydrolases/metabolism , Presynaptic Terminals/drug effects , Signal Transduction/drug effects
12.
Plant Foods Hum Nutr ; 71(1): 81-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26852311

ABSTRACT

Three different flavoring methods of olive oil were tested employing two different herbs, thyme and oregano. The traditional method consist in the infusion of herbs into the oil. A second scarcely diffused method is based on the addition of herbs to the crushed olives before the malaxation step during the extraction process. The third innovative method is the implementation of the ultrasound before the olive paste malaxation. The objective of the study is to verify the effect of the treatments on the quality of the product, assessed by means of the chemical characteristics, the phenol composition and the radical scavenging activity of the resulting oils. The less favorable method was the addition of herbs directly to the oil. A positive effect was achieved by the addition of herbs to the olive paste and other advantages were attained by the employment of ultrasound. These last two methods allow to produce oils "ready to sell", instead the infused oils need to be filtered. Moreover, the flavoring methods applied during the extraction process determine a significant increment of phenolic content and radical scavenging activity of olive oils. The increments were higher when oregano is used instead of thyme. Ultrasound inhibited the olive polyphenoloxidase, the endogenous enzyme responsible for olive oil phenol oxidation. This treatment of olive paste mixed with herbs before malaxation was revealed as the most favorable method due to the best efficiency, reduced time consumption and minor labor, enhancing the product quality of flavored olive oil.


Subject(s)
Flavoring Agents/chemistry , Food Handling/methods , Olea/chemistry , Olive Oil/chemistry , Origanum/chemistry , Thymus Plant/chemistry , Catechol Oxidase/metabolism , Free Radical Scavengers/analysis , Free Radical Scavengers/chemistry , Humans , Liquid-Liquid Extraction , Ointments/chemistry , Olea/enzymology , Olive Oil/isolation & purification , Olive Oil/standards , Oxidation-Reduction , Phenols/analysis , Phenols/chemistry , Plant Leaves/chemistry , Plant Proteins/metabolism , Taste
13.
Am J Physiol Gastrointest Liver Physiol ; 309(8): G670-9, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26316590

ABSTRACT

Apical cAMP-dependent CFTR Cl(-) channels are essential for efficient vectorial movement of ions and fluid into the lumen of the colon. It is well known that Ca(2+)-mobilizing agonists also stimulate colonic anion secretion. However, CFTR is apparently not activated directly by Ca(2+), and the existence of apical Ca(2+)-dependent Cl(-) channels in the native colonic epithelium is controversial, leaving the identity of the Ca(2+)-activated component unresolved. We recently showed that decreasing free Ca(2+) concentration ([Ca(2+)]) within the endoplasmic reticulum (ER) lumen elicits a rise in intracellular cAMP. This process, which we termed "store-operated cAMP signaling" (SOcAMPS), requires the luminal ER Ca(2+) sensor STIM1 and does not depend on changes in cytosolic Ca(2+). Here we assessed the degree to which SOcAMPS participates in Ca(2+)-activated Cl(-) transport as measured by transepithelial short-circuit current (Isc) in polarized T84 monolayers in parallel with imaging of cAMP and PKA activity using fluorescence resonance energy transfer (FRET)-based reporters in single cells. In Ca(2+)-free conditions, the Ca(2+)-releasing agonist carbachol and Ca(2+) ionophore increased Isc, cAMP, and PKA activity. These responses persisted in cells loaded with the Ca(2+) chelator BAPTA-AM. The effect on Isc was enhanced in the presence of the phosphodiesterase (PDE) inhibitor 3-isobutyl-1-methylxanthine (IBMX), inhibited by the CFTR inhibitor CFTRinh-172 and the PKA inhibitor H-89, and unaffected by Ba(2+) or flufenamic acid. We propose that a discrete component of the "Ca(2+)-dependent" secretory activity in the colon derives from cAMP generated through SOcAMPS. This alternative mode of cAMP production could contribute to the actions of diverse xenobiotic agents that disrupt ER Ca(2+) homeostasis, leading to diarrhea.


Subject(s)
Calcium/metabolism , Chlorides/metabolism , Colon/metabolism , Cyclic AMP/metabolism , Cell Line, Tumor , Colon/cytology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endoplasmic Reticulum/metabolism , Fluorescence Resonance Energy Transfer , Humans , Signal Transduction
14.
Mol Pharmacol ; 88(3): 596-603, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25979002

ABSTRACT

The study of G protein-coupled receptors (GPCRs) has benefited greatly from experimental approaches that interrogate their functions in controlled, artificial environments. Working in vitro, GPCR receptorologists discovered the basic biologic mechanisms by which GPCRs operate, including their eponymous capacity to couple to G proteins; their molecular makeup, including the famed serpentine transmembrane unit; and ultimately, their three-dimensional structure. Although the insights gained from working outside the native environments of GPCRs have allowed for the collection of low-noise data, such approaches cannot directly address a receptor's native (in vivo) functions. An in vivo approach can complement the rigor of in vitro approaches: as studied in model organisms, it imposes physiologic constraints on receptor action and thus allows investigators to deduce the most salient features of receptor function. Here, we briefly discuss specific examples in which model organisms have successfully contributed to the elucidation of signals controlled through GPCRs and other surface receptor systems. We list recent examples that have served either in the initial discovery of GPCR signaling concepts or in their fuller definition. Furthermore, we selectively highlight experimental advantages, shortcomings, and tools of each model organism.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Receptors, G-Protein-Coupled/genetics
15.
Front Cell Neurosci ; 8: 350, 2014.
Article in English | MEDLINE | ID: mdl-25389388

ABSTRACT

The second messenger cyclic AMP (cAMP) is a major intracellular mediator of many hormones and neurotransmitters and regulates a myriad of cell functions, including synaptic plasticity in neurons. Whereas cAMP can freely diffuse in the cytosol, a growing body of evidence suggests the formation of cAMP gradients and microdomains near the sites of cAMP production, where cAMP signals remain apparently confined. The mechanisms responsible for the formation of such microdomains are subject of intensive investigation. The development of optical methods based on fluorescence resonance energy transfer (FRET), which allow a direct observation of cAMP signaling with high temporal and spatial resolution, is playing a fundamental role in elucidating the nature of such microdomains. Here, we will review the optical methods used for monitoring cAMP and protein kinase A (PKA) signaling in living cells, providing some examples of their application in neurons, and will discuss the major hypotheses on the formation of cAMP/PKA microdomains.

16.
Curr Opin Cell Biol ; 27: 87-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24530699

ABSTRACT

The activation of a G protein-coupled receptor is generally triggered by binding of an agonist to the receptor's binding pocket, or, in the case of rhodopsin, by light-induced changes of the pre-bound retinal. This is followed by a series of a conformational changes towards an active receptor conformation, which is capable of signalling to G proteins and other downstream proteins. In the past few years, a number of new techniques have been employed to analyze the kinetics of this activation process, including X-ray crystallographic three-dimensional structures of receptors in the inactive and the active states, NMR studies of labelled receptors, molecular simulations, and optical analyses with fluorescence resonance energy transfer (FRET). Here we review our current understanding of the activation process of GPCRs as well as open questions in the sequence of events ranging from (sub-)microsecond activation by light or agonist binding to millisecond activation of receptors by soluble ligands and the subsequent generation of an intracellular signal.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Humans , Kinetics , Ligands , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/isolation & purification , Receptors, G-Protein-Coupled/radiation effects , Rhodopsin/chemistry , Rhodopsin/metabolism , Rhodopsin/radiation effects , Signal Transduction/radiation effects
17.
J Cell Mol Med ; 16(11): 2715-25, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22681560

ABSTRACT

Diverse pathophysiological processes (e.g. obesity, lifespan determination, addiction and male fertility) have been linked to the expression of specific isoforms of the adenylyl cyclases (AC1-AC10), the enzymes that generate cyclic AMP (cAMP). Our laboratory recently discovered a new mode of cAMP production, prominent in certain cell types, that is stimulated by any manoeuvre causing reduction of free [Ca(2+) ] within the lumen of the endoplasmic reticulum (ER) calcium store. Activation of this 'store-operated' pathway requires the ER Ca(2+) sensor, STIM1, but the identity of the enzymes responsible for cAMP production and how this process is regulated is unknown. Here, we used sensitive FRET-based sensors for cAMP in single cells combined with silencing and overexpression approaches to show that store-operated cAMP production occurred preferentially via the isoform AC3 in NCM460 colonic epithelial cells. Ca(2+) entry via the plasma membrane Ca(2+) channel, Orai1, suppressed cAMP production, independent of store refilling. These findings are an important first step towards defining the functional significance and to identify the protein composition of this novel Ca(2+) /cAMP crosstalk system.


Subject(s)
Adenylyl Cyclases/metabolism , Calcium Channels/metabolism , Cyclic AMP/metabolism , Endoplasmic Reticulum/metabolism , Adenylyl Cyclases/genetics , Biosensing Techniques , Calcium/metabolism , Calcium Channels/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line/drug effects , Endoplasmic Reticulum/drug effects , Fluorescence Resonance Energy Transfer , Gene Knockdown Techniques , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , Pertussis Toxin/pharmacology , Protein Kinase Inhibitors/pharmacology , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2
18.
Cell Calcium ; 51(5): 393-401, 2012 May.
Article in English | MEDLINE | ID: mdl-22361140

ABSTRACT

Secretory granules of pancreatic ß-cells contain high concentrations of Ca2+ ions that are co-released with insulin in the extracellular milieu upon activation of exocytosis. As a consequence, an increase in the extracellular Ca2+ concentration ([Ca2+]ext) in the microenvironment immediately surrounding ß-cells should be expected following the exocytotic event. Using Ca2+-selective microelectrodes we show here that both high glucose and non-nutrient insulinotropic agents elicit a reversible increase of [Ca2+]ext within rat insulinoma (INS-1E) ß-cells pseudoislets. The glucose-induced increases in [Ca2+]ext are blocked by pretreatment with different Ca2+ channel blockers. Physiological agonists acting as positive or negative modulators of the insulin secretion and drugs known to intersect the secretory machinery at different levels also induce [Ca2+]ext changes as predicted on the basis of their described action on insulin secretion. Finally, the glucose-induced [Ca2+]ext increase is strongly inhibited after disruption of the actin web, indicating that the dynamic [Ca2+]ext changes recorded in INS-1E pseudoislets by Ca2+-selective microelectrodes occur mainly as a consequence of exocytosis of Ca2+-rich granules. In conclusion, our data directly demonstrate that the extracellular spaces surrounding ß-cells constitute a restricted domain where Ca2+ is co-released during insulin exocytosis, creating the basis for an autocrine/paracrine cell-to-cell communication system via extracellular Ca2+ sensors.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Glucose/administration & dosage , Insulin/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cell Communication/drug effects , Cellular Microenvironment/drug effects , Exocytosis , Glyburide/administration & dosage , Hydrogen-Ion Concentration , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Microelectrodes , Rats , Secretory Vesicles/drug effects
19.
J Biol Chem ; 285(45): 34850-63, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20739289

ABSTRACT

The ubiquitous bacterium Pseudomonas aeruginosa frequently causes hospital-acquired infections. P. aeruginosa also infects the lungs of cystic fibrosis (CF) patients and secretes N-(3-oxo-dodecanoyl)-S-homoserine lactone (3O-C12) to regulate bacterial gene expression critical for P. aeruginosa persistence. In addition to its effects as a quorum-sensing gene regulator in P. aeruginosa, 3O-C12 elicits cross-kingdom effects on host cell signaling leading to both pro- or anti-inflammatory effects. We find that in addition to these slow effects mediated through changes in gene expression, 3O-C12 also rapidly increases Cl(-) and fluid secretion in the cystic fibrosis transmembrane regulator (CFTR)-expressing airway epithelia. 3O-C12 does not stimulate Cl(-) secretion in CF cells, suggesting that lactone activates the CFTR. 3O-C12 also appears to directly activate the inositol trisphosphate receptor and release Ca(2+) from the endoplasmic reticulum (ER), lowering [Ca(2+)] in the ER and thereby activating the Ca(2+)-sensitive ER signaling protein STIM1. 3O-C12 increases cytosolic [Ca(2+)] and, strikingly, also cytosolic [cAMP], the known activator of CFTR. Activation of Cl(-) current by 3O-C12 was inhibited by a cAMP antagonist and increased by a phosphodiesterase inhibitor. Finally, a Ca(2+) buffer that lowers [Ca(2+)] in the ER similar to the effect of 3O-C12 also increased cAMP and I(Cl). The results suggest that 3O-C12 stimulates CFTR-dependent Cl(-) and fluid secretion in airway epithelial cells by activating the inositol trisphosphate receptor, thus lowering [Ca(2+)] in the ER and activating STIM1 and store-operated cAMP production. In CF airways, where CFTR is absent, the adaptive ability to rapidly flush the bacteria away is compromised because the lactone cannot affect Cl(-) and fluid secretion.


Subject(s)
4-Butyrolactone/analogs & derivatives , Chlorides/metabolism , Cyclic AMP/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Respiratory Mucosa/metabolism , 4-Butyrolactone/metabolism , Anions/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Line, Transformed , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Endoplasmic Reticulum/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/genetics , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Pseudomonas Infections/genetics , Quorum Sensing/drug effects , Respiratory Mucosa/microbiology , Stromal Interaction Molecule 1
20.
Nat Cell Biol ; 11(4): 433-42, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287379

ABSTRACT

Depletion of Ca(2+) from the endoplasmic reticulum (ER) results in activation of plasma membrane Ca(2+) entry channels. This 'store-operated' process requires translocation of a transmembrane ER Ca(2+) sensor protein, stromal interaction molecule 1 (STIM1), to sites closely apposed to Ca(2+) channels at the cell surface. However, it is not known whether a reduction in Ca(2+) stores is coupled to other signalling pathways by this mechanism. We found that lowering the concentration of free Ca(2+) in the ER, independently of the cytosolic Ca(2+) concentration, also led to recruitment of adenylyl cyclases. This resulted in enhanced cAMP accumulation and PKA activation, measured using FRET-based cAMP indicators. Translocation of STIM1 was required for efficient coupling of ER Ca(2+) depletion to adenylyl cyclase activity. We propose the existence of a pathway (store-operated cAMP signalling or SOcAMPS) in which the content of internal Ca(2+) stores is directly connected to cAMP signalling through a process that involves STIM1.


Subject(s)
Calcium Signaling , Cyclic AMP/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Adenylyl Cyclases/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cell Line , Chelating Agents/pharmacology , Cyclic AMP/biosynthesis , Cytosol/drug effects , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Activation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Phosphoric Diester Hydrolases/metabolism , Protein Transport/drug effects , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL