Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Biophys J ; 121(8): 1435-1448, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35300969

ABSTRACT

The patch-clamp method, which was awarded the Nobel Prize in 1991, is a well-established and indispensable method to study ion channels in living cells and to biophysically characterize non-voltage-gated ion channels, which comprise about 70% of all ion channels in the human genome. To investigate the biophysical properties of non-voltage-gated ion channels, whole-cell measurements with application of continuous voltage ramps are routinely conducted to obtain current-voltage (IV) relationships. However, adequate tools for detailed and quantitative analysis of IV curves are still missing. We use the example of the transient receptor potential classical (TRPC) channel family to elucidate whether the normalized slope conductance (NSC) is an appropriate tool for reliable discrimination of the IV curves of diverse TRPC channels that differ in their individual curve progression. We provide a robust calculation method for the NSC, and, by applying this method, we find that TRPC channel activators and modulators can evoke different NSC progressions independent from their expression levels, which points to distinguishable active channel states. TRPC6 mutations in patients with focal segmental glomerulosclerosis resulted in distinct NSC progressions, suggesting that the NSC is suitable for investigating structure-function relations and might help unravel the unknown pathomechanisms leading to focal segmental glomerulosclerosis. The NSC is an effective algorithm for extended biophysical characterization of non-voltage-gated ion channels.


Subject(s)
Glomerulosclerosis, Focal Segmental , Biophysics , Female , Humans , Ion Channels , Male , Patch-Clamp Techniques
2.
STAR Protoc ; 2(2): 100527, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34027485

ABSTRACT

Small molecular probes designed for photopharmacology and opto-chemogenetics are rapidly gaining widespread recognition for investigations of transient receptor potential canonical (TRPC) channels. This protocol describes the use of three photoswitchable diacylglycerol analogs-PhoDAG-1, PhoDAG-3, and OptoDArG-for ultrarapid activation and deactivation of native TRPC2 channels in mouse vomeronasal sensory neurons and olfactory type B cells, as well as heterologously expressed human TRPC6 channels. Photoconversion can be achieved in mammalian tissue slices and enables all-optical stimulation and shutoff of TRPC channels. For complete details on the use and execution of this protocol, please refer to Leinders-Zufall et al. (2018).


Subject(s)
Cytological Techniques/methods , Diglycerides , Photochemical Processes , Transient Receptor Potential Channels , Animals , Cells, Cultured , Diglycerides/chemistry , Diglycerides/pharmacology , Mice , Olfactory Receptor Neurons/cytology , Transient Receptor Potential Channels/analysis , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism , Vomeronasal Organ/cytology
3.
Cell Calcium ; 97: 102414, 2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33930818

ABSTRACT

The transient receptor potential classical or canonical (TRPC) 5 channel is a non-selective calcium-permeable cation channel that recently emerged as a promising target for the treatment of various diseases such as mental disorders and kidney diseases. Thus, detailed insight into the structural properties of TRPC5 channels is of utmost importance to further advance TRPC5 channels as drug targets. Recently, Song et al. (2021) have presented cryo-EM structures of the human TRPC5 channel alone or in complex with two different inhibitors thereby revealing two new distinct drug binding sites. Moreover, a binding site for the second messenger diacylglycerol (DAG) has been identified commensurate with a key role of DAG for TRPC5 channel activation.

4.
ChemMedChem ; 15(7): 566-570, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32162837

ABSTRACT

Molecular shape and pharmacological function are interconnected. To capture shape, the fractal dimensionality concept was employed, providing a natural similarity measure for the virtual screening of de novo generated small molecules mimicking the structurally complex natural product (-)-englerin A. Two of the top-ranking designs were synthesized and tested for their ability to modulate transient receptor potential (TRP) cation channels which are cellular targets of (-)-englerin A. Intracellular calcium assays and electrophysiological whole-cell measurements of TRPC4 and TRPM8 channels revealed potent inhibitory effects of one of the computer-generated compounds. Four derivatives of this identified hit compound had comparable effects on TRPC4 and TRPM8. The results of this study corroborate the use of fractal dimensionality as an innovative shape-based molecular representation for molecular scaffold-hopping.


Subject(s)
Drug Design , Sesquiterpenes, Guaiane/pharmacology , TRPC Cation Channels/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors , HEK293 Cells , Humans , Models, Molecular , Molecular Structure , Sesquiterpenes, Guaiane/chemical synthesis , Sesquiterpenes, Guaiane/chemistry , TRPC Cation Channels/metabolism , TRPM Cation Channels/metabolism
5.
Nat Commun ; 10(1): 5784, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31857598

ABSTRACT

G-protein coupled receptors (GPCRs) are versatile cellular sensors for chemical stimuli, but also serve as mechanosensors involved in various (patho)physiological settings like vascular regulation, cardiac hypertrophy and preeclampsia. However, the molecular mechanisms underlying mechanically induced GPCR activation have remained elusive. Here we show that mechanosensitive histamine H1 receptors (H1Rs) are endothelial sensors of fluid shear stress and contribute to flow-induced vasodilation. At the molecular level, we observe that H1Rs undergo stimulus-specific patterns of conformational changes suggesting that mechanical forces and agonists induce distinct active receptor conformations. GPCRs lacking C-terminal helix 8 (H8) are not mechanosensitive, and transfer of H8 to non-responsive GPCRs confers, while removal of H8 precludes, mechanosensitivity. Moreover, disrupting H8 structural integrity by amino acid exchanges impairs mechanosensitivity. Altogether, H8 is the essential structural motif endowing GPCRs with mechanosensitivity. These findings provide a mechanistic basis for a better understanding of the roles of mechanosensitive GPCRs in (patho)physiology.


Subject(s)
Cell Membrane/physiology , Mechanotransduction, Cellular/physiology , Receptors, Histamine H1/ultrastructure , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Gene Knockdown Techniques , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Knockout , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Mutagenesis, Site-Directed , Myography , Protein Conformation, alpha-Helical/physiology , Receptors, Histamine H1/physiology , Stress, Mechanical
6.
Mol Pharmacol ; 96(1): 90-98, 2019 07.
Article in English | MEDLINE | ID: mdl-31171574

ABSTRACT

Although the overall structure of many classical transient receptor potential proteins (TRPC), including human and murine TRPC6, were recently resolved by cryoelectron microscopy analysis, structural changes during channel activation by 1-oleoyl-1-acetyl-sn-glycerol (OAG), the membrane-permeable analog of diacylglycerol, were not defined. Moreover, data on carboxyl- and amino-terminal interactions were not provided, as the amino-terminal regions of murine and human TRPC6 were not resolved. Therefore, we employed a Förster resonance energy transfer (FRET) approach using a small fluorescein arsenical hairpin (FlAsH) targeted to a short tetracysteine sequence at the unresolved amino-terminus and cerulean, a cyan fluorescent protein, as a tag at the carboxyl-terminus of the murine TRPC6 protein. After OAG as well as GSK-1702934A activation, FRET efficiency was simultaneously and significantly reduced, indicating a decreased interaction between the amino to carboxyl termini in the functional tagged murine TRPC6 tetramer (TRPC6 WT) heterologously expressed in human embryonic kidney 293T cells. There was a significant reduction in the FRET signal obtained from analysis of murine TRPC6 FRET constructs with homologous amino-terminal mutations (M131T, G108S) that had been identified in human patients with inherited focal segmental glomerulosclerosis, a condition that can lead to end-stage renal disease. A novel, designed loss-of-function TRPC6 mutation (N109A) in the amino-terminus in close proximity to the carboxyl-terminus produced similar FRET ratios. SIGNIFICANCE STATEMENT: Our data show for the first time that FlAsH-tagging of ion channels is a promising tool to study conformational changes after channel opening and may significantly advance the analysis of ion channel activation as well as their mutants involved in channelopathies.


Subject(s)
Diglycerides/pharmacology , Green Fluorescent Proteins/chemistry , TRPC6 Cation Channel/chemistry , TRPC6 Cation Channel/metabolism , Animals , Diglycerides/chemistry , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Mice , Mutation , Patch-Clamp Techniques , TRPC6 Cation Channel/genetics
7.
Cells ; 7(11)2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30463370

ABSTRACT

Transient receptor potential classical or canonical 4 (TRPC4) and TRPC5 channels are members of the classical or canonical transient receptor potential (TRPC) channel family of non-selective cation channels. TRPC4 and TRPC5 channels are widely accepted as receptor-operated cation channels that are activated in a phospholipase C-dependent manner, following the Gq/11 protein-coupled receptor activation. However, their precise activation mechanism has remained largely elusive for a long time, as the TRPC4 and TRPC5 channels were considered as being insensitive to the second messenger diacylglycerol (DAG) in contrast to the other TRPC channels. Recent findings indicate that the C-terminal interactions with the scaffolding proteins Na⁺/H⁺ exchanger regulatory factor 1 and 2 (NHERF1 and NHERF2) dynamically regulate the DAG sensitivity of the TRPC4 and TRPC5 channels. Interestingly, the C-terminal NHERF binding suppresses, while the dissociation of NHERF enables, the DAG sensitivity of the TRPC4 and TRPC5 channels. This leads to the assumption that all of the TRPC channels are DAG sensitive. The identification of the regulatory function of the NHERF proteins in the TRPC4/5-NHERF protein complex offers a new starting point to get deeper insights into the molecular basis of TRPC channel activation. Future studies will have to unravel the physiological and pathophysiological functions of this multi-protein channel complex.

8.
Cell Chem Biol ; 25(2): 215-223.e3, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29276045

ABSTRACT

Diacylglycerol-sensitive transient receptor potential (TRP) channels play crucial roles in a wide variety of biological processes and systems, but their activation mechanism is not well understood. We describe an optical toolkit by which activation and deactivation of these ion channels can be controlled with unprecedented speed and precision through light stimuli. We show that the photoswitchable diacylglycerols PhoDAG-1 and PhoDAG-3 enable rapid photoactivation of two DAG-sensitive TRP channels, Trpc2 and TRPC6, upon stimulation with UV-A light, whereas exposure to blue light terminates channel activation. PhoDAG photoconversion can be applied in heterologous expression systems, in native cells, and even in mammalian tissue slices. Combined laser scanning-controlled photoswitching and Ca2+ imaging enables both large-scale mapping of TRP channel-mediated neuronal activation and localized mapping in small cellular compartments. Light-switchable PhoDAGs provide an important advance to explore the pathophysiological relevance of DAG-sensitive TRP channels in the maintenance of body homeostasis.


Subject(s)
Diglycerides/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Diglycerides/chemistry , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Photochemical Processes , Transient Receptor Potential Channels/chemistry
9.
Cell ; 171(6): 1368-1382.e23, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195076

ABSTRACT

Blood platelets are critical for hemostasis and thrombosis and play diverse roles during immune responses. Despite these versatile tasks in mammalian biology, their skills on a cellular level are deemed limited, mainly consisting in rolling, adhesion, and aggregate formation. Here, we identify an unappreciated asset of platelets and show that adherent platelets use adhesion receptors to mechanically probe the adhesive substrate in their local microenvironment. When actomyosin-dependent traction forces overcome substrate resistance, platelets migrate and pile up the adhesive substrate together with any bound particulate material. They use this ability to act as cellular scavengers, scanning the vascular surface for potential invaders and collecting deposited bacteria. Microbe collection by migrating platelets boosts the activity of professional phagocytes, exacerbating inflammatory tissue injury in sepsis. This assigns platelets a central role in innate immune responses and identifies them as potential targets to dampen inflammatory tissue damage in clinical scenarios of severe systemic infection.


Subject(s)
Bacterial Infections/immunology , Blood Platelets/immunology , Animals , Bacteria/classification , Blood Platelets/cytology , Blood Vessels/injuries , Blood Vessels/pathology , Calcium/metabolism , Cell Movement , Cell Polarity , Humans , Inflammation/immunology , Integrins/metabolism , Mice , Myosins/metabolism , Neutrophils/cytology
11.
Pflugers Arch ; 469(5-6): 725-737, 2017 06.
Article in English | MEDLINE | ID: mdl-28386636

ABSTRACT

Analysis of G-protein-coupled receptor (GPCR) signaling, in particular of the second messenger cAMP that is tightly controlled by Gs- and Gi/o-proteins, is a central issue in biomedical research. The classical biochemical method to monitor increases in intracellular cAMP concentrations consists of a radioactive multicellular assay, which is well established, highly sensitive, and reproducible, but precludes continuous spatial and temporal assessment of cAMP levels in single living cells. For this purpose, Förster resonance energy transfer (FRET)-based Epac cAMP sensors are well suitable. So far, the latter sensors have been employed to monitor Gs-induced cAMP increases and it has remained elusive whether Epac sensors can reliably detect decreased intracellular cAMP levels as well. In this study, we systematically optimize experimental strategies employing FRET-based cAMP sensors to monitor Gi/o-mediated cAMP reductions. FRET experiments with adrenergic α2A or µ opioid receptors and a set of different Epac sensors allowed for time-resolved, valid, and reliable detection of cAMP level decreases upon Gi/o-coupled receptor activation in single living cells, and this effect can be reversed by selective receptor antagonists. Moreover, pre-treatment with forskolin or 3-isobutyl-1-methylxanthine (IBMX) to artificially increase basal cAMP levels was not required to monitor Gi/o-coupled receptor activation. Thus, using FRET-based cAMP sensors is of major advantage when compared to classical biochemical and multi-cellular assays.


Subject(s)
Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer/methods , Green Fluorescent Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans
12.
JACC Basic Transl Sci ; 2(6): 737-747, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29354781

ABSTRACT

Cardiovascular disease-related deaths frequently arise from arrhythmias, but treatment options are limited due to perilous side effects of commonly used antiarrhythmic drugs. Cardiac rhythmicity strongly depends on cardiomyocyte Ca2+ handling and prevalent cardiac diseases are causally associated with perturbations in intracellular Ca2+ handling. Therefore, intracellular Ca2+ transporters are lead candidate structures for novel and safer antiarrhythmic therapies. Mitochondria and mitochondrial Ca2+ transport proteins are important regulators of cardiac Ca2+ handling. Here we evaluated the potential of pharmacological activation of mitochondrial Ca2+ uptake for the treatment of cardiac arrhythmia. To this aim,we tested substances that enhance mitochondrial Ca2+ uptake for their ability to suppress arrhythmia in a murine model for ryanodine receptor 2 (RyR2)-mediated catecholaminergic polymorphic ventricular tachycardia (CPVT) in vitro and in vivo and in induced pluripotent stem cell-derived cardiomyocytes from a CPVT patient. In freshly isolated cardiomyocytes of RyR2R4496C/WT mice efsevin, a synthetic agonist of the voltage-dependent anion channel 2 (VDAC2) in the outer mitochondrial membrane, prevented the formation of diastolic Ca2+ waves and spontaneous action potentials. The antiarrhythmic effect of efsevin was abolished by blockade of the mitochondrial Ca2+ uniporter (MCU), but could be reproduced using the natural MCU activator kaempferol. Both mitochondrial Ca2+ uptake enhancers (MiCUps), efsevin and kaempferol, significantly reduced episodes of stress-induced ventricular tachycardia in RyR2R4496C/WT mice in vivo and abolished diastolic, arrhythmogenic Ca2+ events in human iPSC-derived cardiomyocytes.

13.
Proc Natl Acad Sci U S A ; 114(1): E37-E46, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27994151

ABSTRACT

The activation mechanism of the classical transient receptor potential channels TRPC4 and -5 via the Gq/11 protein-phospholipase C (PLC) signaling pathway has remained elusive so far. In contrast to all other TRPC channels, the PLC product diacylglycerol (DAG) is not sufficient for channel activation, whereas TRPC4/5 channel activity is potentiated by phosphatidylinositol 4,5-bisphosphate (PIP2) depletion. As a characteristic structural feature, TRPC4/5 channels contain a C-terminal PDZ-binding motif allowing for binding of the scaffolding proteins Na+/H+ exchanger regulatory factor (NHERF) 1 and 2. PKC inhibition or the exchange of threonine for alanine in the C-terminal PDZ-binding motif conferred DAG sensitivity to the channel. Altogether, we present a DAG-mediated activation mechanism for TRPC4/5 channels tightly regulated by NHERF1/2 interaction. PIP2 depletion evokes a C-terminal conformational change of TRPC5 proteins leading to dynamic dissociation of NHERF1/2 from the C terminus of TRPC5 as a prerequisite for DAG sensitivity. We show that NHERF proteins are direct regulators of ion channel activity and that DAG sensitivity is a distinctive hallmark of TRPC channels.


Subject(s)
Diglycerides/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , TRPC Cation Channels/metabolism , Type C Phospholipases/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Enzyme Activation/physiology , HEK293 Cells , Humans , Phosphoproteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Sodium-Hydrogen Exchangers/genetics
14.
Microcirculation ; 23(8): 621-625, 2016 11.
Article in English | MEDLINE | ID: mdl-27344060

ABSTRACT

Myogenic vasoconstriction (Bayliss effect) is mediated by vascular smooth muscle cells (VSMCs) of small resistance arteries sensing mechanical forces. During the last three decades, several proteins have been proposed as VSMC mechanosensors. Our previous studies highlighted agonist-independent mechanical activation of Gq/11 protein-coupled receptors (Gq/11 PCRs) in VSMCs of resistance arteries. In particular, angiotensin II AT1 receptors (AT1 Rs) emerged as mechanosensors mediating myogenic tone. Moreover, we found that the AT1B receptor isoform was more mechanosensitive than the AT1A receptor. Interestingly, cysteinyl leukotriene 1 receptors (CysLT1 Rs) were up-regulated in AT1 R-deficient arteries as an essential backup strategy to compensate for the loss of vasoconstrictor receptors. Up-regulation of CysLT1 Rs resulted in increased myogenic tone at low intraluminal pressures resulting in hyperactivity of AT1 R-deficient arteries. Only at high intraluminal pressures myogenic tone was reduced, thus reflecting the loss of AT1 Rs. Further, CysLT1 Rs were involved in myogenic vasoconstriction of wild-type arteries. Simultaneous blockade of AT1 Rs and CysLT1 Rs in wild-type arteries caused reduction in myogenic tone of more than 60% comparable to the application of the selective Gq/11 -protein inhibitor YM-254890. Our findings suggest that AT1 Rs and CysLT1 Rs are crucial mechanosensors in resistance arteries mediating 60% of myogenic vasoconstriction via the Gq/11 -protein pathway without involvement of endogenous agonists.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Muscle, Smooth, Vascular/physiology , Receptor, Angiotensin, Type 1/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Leukotriene/physiology , Vasoconstriction , Animals , Biomechanical Phenomena , Humans , Mechanoreceptors , Mechanotransduction, Cellular , Mice , Myocytes, Smooth Muscle
15.
J Am Soc Nephrol ; 27(3): 848-62, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26160898

ABSTRACT

Podocytes are specialized, highly differentiated epithelial cells in the kidney glomerulus that are exposed to glomerular capillary pressure and possible increases in mechanical load. The proteins sensing mechanical forces in podocytes are unconfirmed, but the classic transient receptor potential channel 6 (TRPC6) interacting with the MEC-2 homolog podocin may form a mechanosensitive ion channel complex in podocytes. Here, we observed that podocytes respond to mechanical stimulation with increased intracellular calcium concentrations and increased inward cation currents. However, TRPC6-deficient podocytes responded in a manner similar to that of control podocytes, and mechanically induced currents were unaffected by genetic inactivation of TRPC1/3/6 or administration of the broad-range TRPC blocker SKF-96365. Instead, mechanically induced currents were significantly decreased by the specific P2X purinoceptor 4 (P2X4) blocker 5-BDBD. Moreover, mechanical P2X4 channel activation depended on cholesterol and podocin and was inhibited by stabilization of the actin cytoskeleton. Because P2X4 channels are not intrinsically mechanosensitive, we investigated whether podocytes release ATP upon mechanical stimulation using a fluorometric approach. Indeed, mechanically induced ATP release from podocytes was observed. Furthermore, 5-BDBD attenuated mechanically induced reorganization of the actin cytoskeleton. Altogether, our findings reveal a TRPC channel-independent role of P2X4 channels as mechanotransducers in podocytes.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Mechanotransduction, Cellular , Podocytes/metabolism , Receptors, Purinergic P2X4/physiology , Adenosine Triphosphate/pharmacology , Animals , Benzodiazepinones/pharmacology , Cells, Cultured , Cholesterol/metabolism , Cytoskeleton/ultrastructure , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mechanotransduction, Cellular/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Stress, Mechanical , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics , TRPC6 Cation Channel
16.
Circ Res ; 117(4): 376-87, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26129975

ABSTRACT

RATIONALE: Platelets are known to play a crucial role in hemostasis. Sphingosine kinases (Sphk) 1 and 2 catalyze the conversion of sphingosine to the bioactive metabolite sphingosine 1-phosphate (S1P). Although platelets are able to secrete S1P on activation, little is known about a potential intrinsic effect of S1P on platelet function. OBJECTIVE: To investigate the role of Sphk1- and Sphk2-derived S1P in the regulation of platelet function. METHODS AND RESULTS: We found a 100-fold reduction in intracellular S1P levels in platelets derived from Sphk2(-/-) mutants compared with Sphk1(-/-) or wild-type mice, as analyzed by mass spectrometry. Sphk2(-/-) platelets also failed to secrete S1P on stimulation. Blood from Sphk2-deficient mice showed decreased aggregation after protease-activated receptor 4-peptide and adenosine diphosphate stimulation in vitro, as assessed by whole blood impedance aggregometry. We revealed that S1P controls platelet aggregation via the sphingosine 1-phosphate receptor 1 through modulation of protease-activated receptor 4-peptide and adenosine diphosphate-induced platelet activation. Finally, we show by intravital microscopy that defective platelet aggregation in Sphk2-deficient mice translates into reduced arterial thrombus stability in vivo. CONCLUSIONS: We demonstrate that Sphk2 is the major Sphk isoform responsible for the generation of S1P in platelets and plays a pivotal intrinsic role in the control of platelet activation. Correspondingly, Sphk2-deficient mice are protected from arterial thrombosis after vascular injury, but have normal bleeding times. Targeting this pathway could therefore present a new therapeutic strategy to prevent thrombosis.


Subject(s)
Blood Platelets/enzymology , Lysophospholipids/blood , Phosphotransferases (Alcohol Group Acceptor)/blood , Platelet Aggregation , Sphingosine/analogs & derivatives , Animals , Arachidonic Acid/blood , Blood Coagulation , Blood Coagulation Tests , Carotid Artery Injuries/blood , Carotid Artery Injuries/enzymology , Disease Models, Animal , Erythrocytes/enzymology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Platelet Adhesiveness , Platelet Function Tests , Receptors, Lysosphingolipid/blood , Signal Transduction , Sphingosine/blood , Sphingosine-1-Phosphate Receptors , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/prevention & control , Thromboxane A2/blood , Vascular System Injuries/blood , Vascular System Injuries/enzymology
17.
Hypertension ; 66(1): 108-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26034200

ABSTRACT

The protective effects of 5'-AMP-activated protein kinase (AMPK) on the metabolic syndrome may include direct effects on resistance artery vasomotor function. However, the precise actions of AMPK on microvessels and their potential interaction are largely unknown. Thus, we set to determine the effects of AMPK activation on vascular smooth muscle tone and the underlying mechanisms. Resistance arteries isolated from hamster and mouse exhibited a pronounced endothelium-independent dilation on direct pharmacological AMPK activation by 2 structurally unrelated compounds (PT1 and A769662). The dilation was associated with a decrease of intracellular-free calcium [Ca(2+)]i in vascular smooth muscle cell. AMPK stimulation induced activation of BKCa channels as assessed by patch clamp studies in freshly isolated hamster vascular smooth muscle cell and confirmed by direct proof of membrane hyperpolarization in intact arteries. The BKCa channel blocker iberiotoxin abolished the hyperpolarization but only partially reduced the dilation and did not affect the decrease of [Ca(2+)]i. By contrast, the sarcoplasmic/endoplasmic Ca(2+)-ATPase (SERCA) inhibitor thapsigargin largely reduced these effects, whereas combined inhibition of SERCA and BKCa channels virtually abolished them. AMPK stimulation significantly increased the phosphorylation of the SERCA modulator phospholamban at the regulatory T17 site. Stimulation of smooth muscle AMPK represents a new, potent vasodilator mechanism in resistance vessels. AMPK directly relaxes vascular smooth muscle cell by a decrease of [Ca(2+)]i. This is achieved by calcium sequestration via SERCA activation, as well as activation of BKCa channels. There is in part a mutual compensation of both calcium-lowering mechanisms. However, SERCA activation which involves an AMPK-dependent phosphorylation of phospholamban is the predominant mechanism in resistance vessels.


Subject(s)
AMP-Activated Protein Kinases/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Muscle, Smooth, Vascular/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Vascular Resistance/drug effects , Vasodilation/drug effects , Vasomotor System/drug effects , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cricetinae , Enzyme Activation/drug effects , Indoles/pharmacology , Membrane Potentials/drug effects , Mesocricetus , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/physiology , Peptides/pharmacology , RNA, Messenger/biosynthesis , Thapsigargin/pharmacology , Vascular Resistance/physiology , Vasodilation/physiology , Vasomotor System/physiology
18.
Arterioscler Thromb Vasc Biol ; 35(1): 121-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25395620

ABSTRACT

OBJECTIVE: Myogenic vasoconstriction is mediated by vascular smooth muscle cells of resistance arteries sensing mechanical stretch. Angiotensin II AT1 receptors and in particular AT1BRs in murine vascular smooth muscle cells have been characterized as mechanosensors that cannot fully account for myogenic vasoconstriction observed. Therefore, we aimed at uncovering novel vascular mechanosensors by expression profiling and functional characterization of candidate proteins. APPROACH AND RESULTS: Analyzing myogenic tone of isolated murine mesenteric arteries of AT1A and AT1B receptor double gene-deficient (AT1A/1B (-/-)) mice ex vivo, we observed a decreased myogenic tone at high intraluminal pressures and an unexpected hyper-reactivity at low intraluminal pressures because of upregulation of cysteinyl leukotriene 1 receptors (CysLT1Rs). Pharmacological blockade of CysLT1Rs with pranlukast significantly reduced myogenic tone not only in AT1A/1B (-/-) but also in wild-type arteries. In wild-type arteries, additional blockade of angiotensin II AT1 receptors with candesartan resulted in an additive reduction of myogenic tone. Furthermore, calcium imaging experiments were performed with fura-2-loaded human embryonic kidney 293 cells overexpressing CysLT1Rs and with isolated mesenteric vascular smooth muscle cells. Hypo-osmotically induced membrane stretch provoked calcium transients that were significantly reduced by pranlukast. Incubations of isolated mesenteric vascular smooth muscle cells with the 5-lipoxygenase inhibitor zileuton had no effect. Furthermore, the Gq/11-protein inhibitor YM 254890 profoundly reduced myogenic tone to the same extent as induced by the application of pranlukast plus candesartan. CONCLUSIONS: Here, we identify a novel, hitherto unappreciated role of CysLT1Rs in vascular regulation. We identified CysLT1Rs as novel mechanosensors in the vasculature involved in myogenic vasoconstriction. Moreover, our findings suggest that myogenic tone is determined by AT1 and CysLT1 receptors acting together as mechanosensors via Gq/11-protein activation.


Subject(s)
Mechanoreceptors/metabolism , Mechanotransduction, Cellular , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Leukotriene/metabolism , Vasoconstriction , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Arterial Pressure , Calcium Signaling , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Leukotriene Antagonists/pharmacology , Male , Mechanoreceptors/drug effects , Mechanotransduction, Cellular/drug effects , Mesenteric Arteries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/genetics , Receptors, Leukotriene/drug effects , Receptors, Leukotriene/genetics , Transfection , Vasoconstriction/drug effects
19.
Pflugers Arch ; 466(7): 1343-53, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24101294

ABSTRACT

Myogenic vasoconstriction is an inherent property of vascular smooth muscle cells (VSMCs) of resistance arteries harboring ill-defined mechanosensing and mechanotransducing elements. G protein-coupled receptors (GPCRs) are discussed as mechanosensors in VSMCs. In this study, we sought to identify and characterize the role and impact of GPCRs on myogenic vasoconstriction. Thus, we analyzed mRNA expression levels of GPCRs in resistance versus preceding conduit arteries revealing a significant enrichment of several GPCRs in resistance vessels. Selective pharmacological blockade of the highly expressed GPCRs in isolated murine mesenteric arteries ex vivo decreased myogenic vasoconstriction. In particular, candesartan and losartan most prominently suppressed myogenic tone, suggesting that AT1 receptors play a central role in myogenic vasoconstriction. Analyzing angiotensinogen(-/-) mice, a relevant contribution of locally produced angiotensin II to myogenic tone could be excluded. Investigation of AT1A (-/-) and AT1B (-/-) murine mesenteric arteries revealed that AT1B, but not AT1A, receptors are key components of myogenic regulation. This notion was supported by examining fura-2-loaded isolated AT1A (-/-) and AT1B (-/-) VSMCs. Our results indicate that in VSMCs, AT1B receptors are more mechanosensitive than AT1A receptors even at comparable receptor expression levels. Furthermore, we demonstrate that the mechanosensitivity of GPCRs is agonist-independent and positively correlates with receptor expression levels.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Receptor, Angiotensin, Type 1/metabolism , Vasoconstriction , Angiotensin II/metabolism , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Cells, Cultured , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiology , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/genetics
20.
Nat Commun ; 3: 649, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22337127

ABSTRACT

Lung ischaemia-reperfusion-induced oedema (LIRE) is a life-threatening condition that causes pulmonary oedema induced by endothelial dysfunction. Here we show that lungs from mice lacking nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox2(y/-)) or the classical transient receptor potential channel 6 (TRPC6(-/-)) are protected from LIR-induced oedema (LIRE). Generation of chimeric mice by bone marrow cell transplantation and endothelial-specific Nox2 deletion showed that endothelial Nox2, but not leukocytic Nox2 or TRPC6, are responsible for LIRE. Lung endothelial cells from Nox2- or TRPC6-deficient mice showed attenuated ischaemia-induced Ca(2+) influx, cellular shape changes and impaired barrier function. Production of reactive oxygen species was completely abolished in Nox2(y/-) cells. A novel mechanistic model comprising endothelial Nox2-derived production of superoxide, activation of phospholipase C-γ, inhibition of diacylglycerol (DAG) kinase, DAG-mediated activation of TRPC6 and ensuing LIRE is supported by pharmacological and molecular evidence. This mechanism highlights novel pharmacological targets for the treatment of LIRE.


Subject(s)
Edema/therapy , Lung/pathology , Reperfusion Injury , TRPC Cation Channels/genetics , Animals , Calcium/metabolism , Diacylglycerol Kinase/metabolism , Edema/pathology , Endothelial Cells/cytology , Gene Deletion , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , NADPH Oxidase 2 , NADPH Oxidases/genetics , Permeability , Phospholipase C gamma/metabolism , Reactive Oxygen Species , TRPC6 Cation Channel , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...