51
|
Kudryashova TV, Goncharov DA, Pena A, Ihida-Stansbury K, DeLisser H, Kawut SM, Goncharova EA. Profiling the role of mammalian target of rapamycin in the vascular smooth muscle metabolome in pulmonary arterial hypertension. Pulm Circ 2015; 5:667-80. [PMID: 26697174 DOI: 10.1086/683810] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Increased proliferation and resistance to apoptosis of pulmonary arterial vascular smooth muscle cells (PAVSMCs), coupled with metabolic reprogramming, are key components of pulmonary vascular remodeling, a major and currently irreversible pathophysiological feature of pulmonary arterial hypertension (PAH). We recently reported that activation of mammalian target of rapamycin (mTOR) plays a key role in increased energy generation and maintenance of the proliferative, apoptosis-resistant PAVSMC phenotype in human PAH, but the downstream effects of mTOR activation on PAH PAVSMC metabolism are not clear. Using liquid and gas chromatography-based mass spectrometry, we performed pilot metabolomic profiling of human microvascular PAVSMCs from idiopathic-PAH subjects before and after treatment with the selective adenosine triphosphate-competitive mTOR inhibitor PP242 and from nondiseased lungs. We have shown that PAH PAVSMCs have a distinct metabolomic signature of altered metabolites-components of fatty acid synthesis, deficiency of sugars, amino sugars, and nucleotide sugars-intermediates of protein and lipid glycosylation, and downregulation of key biochemicals involved in glutathione and nicotinamide adenine dinucleotide (NAD) metabolism. We also report that mTOR inhibition attenuated or reversed the majority of the PAH-specific abnormalities in lipogenesis, glycosylation, glutathione, and NAD metabolism without affecting altered polyunsaturated fatty acid metabolism. Collectively, our data demonstrate a critical role of mTOR in major PAH PAVSMC metabolic abnormalities and suggest the existence of de novo lipid synthesis in PAVSMCs in human PAH that may represent a new, important component of disease pathogenesis worthy of future investigation.
Collapse
Affiliation(s)
- Tatiana V Kudryashova
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dmitry A Goncharov
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andressa Pena
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kaori Ihida-Stansbury
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Horace DeLisser
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven M Kawut
- Pulmonary Vascular Disease Program and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elena A Goncharova
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
52
|
Nadler A, Yushchenko DA, Müller R, Stein F, Feng S, Mulle C, Carta M, Schultz C. Exclusive photorelease of signalling lipids at the plasma membrane. Nat Commun 2015; 6:10056. [PMID: 26686736 PMCID: PMC4703838 DOI: 10.1038/ncomms10056] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/28/2015] [Indexed: 12/17/2022] Open
Abstract
Photoactivation of caged biomolecules has become a powerful approach to study cellular signalling events. Here we report a method for anchoring and uncaging biomolecules exclusively at the outer leaflet of the plasma membrane by employing a photocleavable, sulfonated coumarin derivative. The novel caging group allows quantifying the reaction progress and efficiency of uncaging reactions in a live-cell microscopy setup, thereby greatly improving the control of uncaging experiments. We synthesized arachidonic acid derivatives bearing the new negatively charged or a neutral, membrane-permeant coumarin caging group to locally induce signalling either at the plasma membrane or on internal membranes in β-cells and brain slices derived from C57B1/6 mice. Uncaging at the plasma membrane triggers a strong enhancement of calcium oscillations in β-cells and a pronounced potentiation of synaptic transmission while uncaging inside cells blocks calcium oscillations in β-cells and causes a more transient effect on neuronal transmission, respectively. The precise subcellular site of arachidonic acid release is therefore crucial for signalling outcome in two independent systems. Caged signalling intermediates are powerful cell biological tools, however it can be challenging to precisely control where activation occurs. Nadler et al. develop a caging group that specifically targets the plasma membrane, and demonstrate spatially controlled activation of arachidonic acid signalling.
Collapse
Affiliation(s)
- André Nadler
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Dmytro A Yushchenko
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany.,Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo náměstí 2, 16610 Prague 6, Czech Republic
| | - Rainer Müller
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Frank Stein
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Suihan Feng
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Christophe Mulle
- Institut Interdisciplinaire de Neurosciences, CNRS UMR 5297 Université Bordeaux 2, 146, rue Léo-Saignat, 33077 Bordeaux, France
| | - Mario Carta
- Institut Interdisciplinaire de Neurosciences, CNRS UMR 5297 Université Bordeaux 2, 146, rue Léo-Saignat, 33077 Bordeaux, France
| | - Carsten Schultz
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
| |
Collapse
|
53
|
Bai J, Ding W, Kojima A, Seto T, Matsuura H. Putative binding sites for arachidonic acid on the human cardiac Kv 1.5 channel. Br J Pharmacol 2015; 172:5281-92. [PMID: 26292661 PMCID: PMC5341216 DOI: 10.1111/bph.13314] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/01/2015] [Accepted: 08/18/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE In human heart, the Kv 1.5 channel contributes to repolarization of atrial action potentials. This study examined the electrophysiological and molecular mechanisms underlying arachidonic acid (AA)-induced inhibition of the human Kv 1.5 (hKv 1.5) channel. EXPERIMENTAL APPROACH Site-directed mutagenesis was conducted to mutate amino acids that reside within the pore domain of the hKv 1.5 channel. Whole-cell patch-clamp method was used to record membrane currents through wild type and mutant hKv 1.5 channels heterologously expressed in CHO cells. Computer docking simulation was conducted to predict the putative binding site(s) of AA in an open-state model of the Kv 1.5 channel. KEY RESULTS The hKv 1.5 current was minimally affected at the onset of depolarization but was progressively reduced during depolarization by the presence of AA, suggesting that AA acts as an open-channel blocker. AA itself affected the channel at extracellular sites independently of its metabolites and signalling pathways. The blocking effect of AA was attenuated at pH 8.0 but not at pH 6.4. The blocking action of AA developed rather rapidly by co-expression of Kv β1.3. The AA-induced block was significantly attenuated in H463C, T480A, R487V, I502A, I508A, V512A and V516A, but not in T462C, A501V and L510A mutants of the hKv 1.5 channel. Docking simulation predicted that H463, T480, R487, I508, V512 and V516 are potentially accessible for interaction with AA. CONCLUSIONS AND IMPLICATIONS AA itself interacts with multiple amino acids located in the pore domain of the hKv 1.5 channel. These findings may provide useful information for future development of selective blockers of hKv 1.5 channels.
Collapse
Affiliation(s)
- Jia‐Yu Bai
- Department of PhysiologyShiga University of Medical ScienceOtsuJapan
| | - Wei‐Guang Ding
- Department of PhysiologyShiga University of Medical ScienceOtsuJapan
| | - Akiko Kojima
- Department of AnesthesiologyShiga University of Medical ScienceOtsuJapan
| | - Tomoyoshi Seto
- Department of AnesthesiologyShiga University of Medical ScienceOtsuJapan
| | - Hiroshi Matsuura
- Department of PhysiologyShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
54
|
Alter P, Glück T, Figiel JH, Koczulla AR, Vogelmeier CF, Rupp H. From Heart Failure to Highly Unsaturated Fatty Acid Deficiency and Vice Versa: Bidirectional Heart and Liver Interactions. Can J Cardiol 2015; 32:217-25. [PMID: 26277086 DOI: 10.1016/j.cjca.2015.05.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/15/2015] [Accepted: 05/15/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND In several trials, beneficial prognostic effects of highly unsaturated fatty acids (HUFAs) in heart failure were shown. Because other studies showed no incremental benefit in nearly preserved cardiac function, the question arises, whether the degree of cardiac dysfunction is involved. It is hypothesized that increased left ventricular (LV) wall stress affects the endogenous hepatic HUFA metabolism, which in turn exhibits adverse cardiac consequences. METHODS Cardiac magnetic resonance imaging was performed in 30 patients with suspected cardiomyopathy. The serum fatty acid profile was assessed using gas chromatography/mass spectrometry. RESULTS Docosahexaenoic acid (DHA; P = 0.002) and eicosapentaenoic acid (EPA; by trend) levels were decreased in patients with reduced LV ejection fraction (≤ 50%) or LV dilatation (≥ 90 mL/m(2)). Decreased DHA (P = 0.003) and EPA (P = 0.022) levels were associated with a reduced LV ejection fraction. Decreased DHA level was correlated with increased end-diastolic (P = 0.047) and end-systolic LV wall stress (P = 0.001). Pseudocholinesterase activity was inversely correlated with end-diastolic (P = 0.020) and end-systolic LV wall stress (P = 0.025). CONCLUSIONS DHA level was significantly reduced in heart failure. Similar, but less pronounced effects were found for EPA and arachidonic acid by trend. Increased LV wall stress was correlated with a reduced DHA level. Increased LV wall stress exhibits various adverse consequences (eg, increased oxygen consumption, favouring of arrhythmias, and an unfavourable remodelling). The increase of wall stress was paralleled by reduced HUFA level. Increased LV wall stress was correlated with reduced pseudocholinesterase, which is suggestive of hepatic congestion (ie, a cardiohepatic syndrome, involved in the altered fatty acid profile in heart failure) and has major consequences regarding the dose-efficacy of HUFA treatment.
Collapse
Affiliation(s)
- Peter Alter
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Marburg, Marburg, Germany.
| | - Tobias Glück
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Marburg, Marburg, Germany
| | - Jens H Figiel
- Department of Radiology, University of Marburg, Marburg, Germany
| | - A Rembert Koczulla
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Marburg, Marburg, Germany
| | - Claus F Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Marburg, Marburg, Germany
| | - Heinz Rupp
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Marburg, Marburg, Germany
| |
Collapse
|
55
|
Differential regulation of proton-sensitive ion channels by phospholipids: a comparative study between ASICs and TRPV1. PLoS One 2015; 10:e0122014. [PMID: 25781982 PMCID: PMC4362947 DOI: 10.1371/journal.pone.0122014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/05/2015] [Indexed: 12/31/2022] Open
Abstract
Protons are released in pain-generating pathological conditions such as inflammation, ischemic stroke, infection, and cancer. During normal synaptic activities, protons are thought to play a role in neurotransmission processes. Acid-sensing ion channels (ASICs) are typical proton sensors in the central nervous system (CNS) and the peripheral nervous system (PNS). In addition to ASICs, capsaicin- and heat-activated transient receptor potential vanilloid 1 (TRPV1) channels can also mediate proton-mediated pain signaling. In spite of their importance in perception of pH fluctuations, the regulatory mechanisms of these proton-sensitive ion channels still need to be further investigated. Here, we compared regulation of ASICs and TRPV1 by membrane phosphoinositides, which are general cofactors of many receptors and ion channels. We observed that ASICs do not require membrane phosphatidylinositol 4-phosphate (PI(4)P) or phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) for their function. However, TRPV1 currents were inhibited by simultaneous breakdown of PI(4)P and PI(4,5)P2. By using a novel chimeric protein, CF-PTEN, that can specifically dephosphorylate at the D3 position of phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P3), we also observed that neither ASICs nor TRPV1 activities were altered by depletion of PI(3,4,5)P3 in intact cells. Finally, we compared the effects of arachidonic acid (AA) on two proton-sensitive ion channels. We observed that AA potentiates the currents of both ASICs and TRPV1, but that they have different recovery aspects. In conclusion, ASICs and TRPV1 have different sensitivities toward membrane phospholipids, such as PI(4)P, PI(4,5)P2, and AA, although they have common roles as proton sensors. Further investigation about the complementary roles and respective contributions of ASICs and TRPV1 in proton-mediated signaling is necessary.
Collapse
|
56
|
Muslikhov ER, Sukhanova IF, Avdonin PV. Arachidonic acid activates release of calcium ions from reticulum via ryanodine receptor channels in C2C12 skeletal myotubes. BIOCHEMISTRY (MOSCOW) 2015; 79:435-9. [PMID: 24954594 DOI: 10.1134/s0006297914050071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Arachidonic acid causes an increase in free cytoplasmic calcium concentration ([Ca2+]i) in differentiated skeletal multinucleated myotubes C2C12 and does not induce calcium response in C2C12 myoblasts. The same reaction of myotubes to arachidonic acid is observed in Ca2+-free medium. This indicates that arachidonic acid induces release of calcium ions from intracellular stores. The blocker of ryanodine receptor channels of sarcoplasmic reticulum dantrolene (20 µM) inhibits this effect by 68.7 ± 6.3% (p < 0.001). The inhibitor of two-pore calcium channels of endolysosomal vesicles trans-NED19 (10 µM) decreases the response to arachidonic acid by 35.8 ± 5.4% (p < 0.05). The phospholipase C inhibitor U73122 (10 µM) has no effect. These data indicate the involvement of ryanodine receptor calcium channels of sarcoplasmic reticulum in [Ca2+]i elevation in skeletal myotubes caused by arachidonic acid and possible participation of two-pore calcium channels from endolysosomal vesicles in this process.
Collapse
Affiliation(s)
- E R Muslikhov
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
| | | | | |
Collapse
|
57
|
Yue Z, Xie J, Yu AS, Stock J, Du J, Yue L. Role of TRP channels in the cardiovascular system. Am J Physiol Heart Circ Physiol 2015; 308:H157-82. [PMID: 25416190 PMCID: PMC4312948 DOI: 10.1152/ajpheart.00457.2014] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/14/2014] [Indexed: 12/12/2022]
Abstract
The transient receptor potential (TRP) superfamily consists of a large number of nonselective cation channels with variable degree of Ca(2+)-permeability. The 28 mammalian TRP channel proteins can be grouped into six subfamilies: canonical, vanilloid, melastatin, ankyrin, polycystic, and mucolipin TRPs. The majority of these TRP channels are expressed in different cell types including both excitable and nonexcitable cells of the cardiovascular system. Unlike voltage-gated ion channels, TRP channels do not have a typical voltage sensor, but instead can sense a variety of other stimuli including pressure, shear stress, mechanical stretch, oxidative stress, lipid environment alterations, hypertrophic signals, and inflammation products. By integrating multiple stimuli and transducing their activity to downstream cellular signal pathways via Ca(2+) entry and/or membrane depolarization, TRP channels play an essential role in regulating fundamental cell functions such as contraction, relaxation, proliferation, differentiation, and cell death. With the use of targeted deletion and transgenic mouse models, recent studies have revealed that TRP channels are involved in numerous cellular functions and play an important role in the pathophysiology of many diseases in the cardiovascular system. Moreover, several TRP channels are involved in inherited diseases of the cardiovascular system. This review presents an overview of current knowledge concerning the physiological functions of TRP channels in the cardiovascular system and their contributions to cardiovascular diseases. Ultimately, TRP channels may become potential therapeutic targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Zhichao Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jia Xie
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Albert S Yu
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jonathan Stock
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jianyang Du
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
58
|
Swelling and eicosanoid metabolites differentially gate TRPV4 channels in retinal neurons and glia. J Neurosci 2015; 34:15689-700. [PMID: 25411497 DOI: 10.1523/jneurosci.2540-14.2014] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Activity-dependent shifts in ionic concentrations and water that accompany neuronal and glial activity can generate osmotic forces with biological consequences for brain physiology. Active regulation of osmotic gradients and cellular volume requires volume-sensitive ion channels. In the vertebrate retina, critical support to volume regulation is provided by Müller astroglia, but the identity of their osmosensor is unknown. Here, we identify TRPV4 channels as transducers of mouse Müller cell volume increases into physiological responses. Hypotonic stimuli induced sustained [Ca(2+)]i elevations that were inhibited by TRPV4 antagonists and absent in TRPV4(-/-) Müller cells. Glial TRPV4 signals were phospholipase A2- and cytochrome P450-dependent, characterized by slow-onset and Ca(2+) waves, and, in excess, were sufficient to induce reactive gliosis. In contrast, neurons responded to TRPV4 agonists and swelling with fast, inactivating Ca(2+) signals that were independent of phospholipase A2. Our results support a model whereby swelling and proinflammatory signals associated with arachidonic acid metabolites differentially gate TRPV4 in retinal neurons and glia, with potentially significant consequences for normal and pathological retinal function.
Collapse
|
59
|
Veldhuis NA, Poole DP, Grace M, McIntyre P, Bunnett NW. The G protein-coupled receptor-transient receptor potential channel axis: molecular insights for targeting disorders of sensation and inflammation. Pharmacol Rev 2015; 67:36-73. [PMID: 25361914 DOI: 10.1124/pr.114.009555] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Sensory nerves are equipped with receptors and ion channels that allow them to detect and respond to diverse chemical, mechanical, and thermal stimuli. These sensory proteins include G protein-coupled receptors (GPCRs) and transient receptor potential (TRP) ion channels. A subclass of peptidergic sensory nerves express GPCRs and TRP channels that detect noxious, irritant, and inflammatory stimuli. Activation of these nerves triggers protective mechanisms that lead to withdrawal from danger (pain), removal of irritants (itch, cough), and resolution of infection (neurogenic inflammation). The GPCR-TRP axis is central to these mechanisms. Signals that emanate from the GPCR superfamily converge on the small TRP family, leading to channel sensitization and activation, which amplify pain, itch, cough, and neurogenic inflammation. Herein we discuss how GPCRs and TRP channels function independently and synergistically to excite sensory nerves that mediate noxious and irritant responses and inflammation in the skin and the gastrointestinal and respiratory systems. We discuss the signaling mechanisms that underlie the GPCR-TRP axis and evaluate how new information about the structure of GPCRs and TRP channels provides insights into their functional interactions. We propose that a deeper understanding of the GPCR-TRP axis may facilitate the development of more selective and effective therapies to treat dysregulated processes that underlie chronic pain, itch, cough, and inflammation.
Collapse
Affiliation(s)
- Nicholas A Veldhuis
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Daniel P Poole
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Megan Grace
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Peter McIntyre
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| |
Collapse
|
60
|
Cavallo JS, Hamilton BN, Farley J. In vitro extinction learning in Hermissenda: involvement of conditioned inhibition molecules. Front Behav Neurosci 2014; 8:354. [PMID: 25374517 PMCID: PMC4204529 DOI: 10.3389/fnbeh.2014.00354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 09/23/2014] [Indexed: 11/30/2022] Open
Abstract
Extinction of a conditioned association is typically viewed as the establishment of new learning rather than the erasure of the original memory. However, recent research in the nudibranch, Hermissenda crassicornis (H.c.) demonstrated that extinction training (using repeated light-alone presentations) given 15 min, but not 23 h, after memory acquisition reversed both the cellular correlates of learning (enhanced Type B cell excitability) and the behavioral changes (reduced phototaxis) produced by associative conditioning (pairings of light, CS, and rotation, US). Here, we investigated the putative molecular signaling pathways that underlie this extinction in H.c. by using a novel in vitro protocol combined with pharmacological manipulations. After intact H.c. received either light-rotation pairings (Paired), random presentations of light and rotation (Random), or no stimulation (Untrained), B cells from isolated CNSs were recorded from during exposure to extinction training consisting of two series of 15 consecutive light-steps (LSs). When in vitro extinction was administered shortly (2 h, but not 24 h) after paired training, B cells from Paired animals showed progressive and robust declines in spike frequency by the 30th LS, while control cells (Random and Untrained) did not. We found that several molecules implicated in H.c. conditioned inhibitory (CI) learning, protein phosphatase 1 (PP1) and arachidonic acid (AA)/12-lipoxygenase (12-LOX) metabolites, also contributed to the spike frequency decreases produced by in vitro extinction. Protein phosphatase 2B (PP2B) also appeared to play a role. Calyculin A (PP1 inhibitor), cyclosporin A (PP2B inhibitor), and baicalein (a 12-LOX inhibitor) all blocked the spike frequency declines in Paired B cells produced by 30 LSs. Conversely, injection of catalytically-active PP1 (caPP1) or PP2B (caPP2B) into Untrained B cells partially mimicked the spike frequency declines observed in Paired cells, as did bath-applied AA, and occluded additional LS-produced reductions in spiking in Paired cells.
Collapse
Affiliation(s)
- Joel S Cavallo
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, USA
| | - Brittany N Hamilton
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, USA
| | - Joseph Farley
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, USA
| |
Collapse
|
61
|
Thompson MA, Prakash YS, Pabelick CM. Arachidonate-regulated Ca(2+) influx in human airway smooth muscle. Am J Respir Cell Mol Biol 2014; 51:68-76. [PMID: 24471656 DOI: 10.1165/rcmb.2013-0144oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Plasma membrane Ca(2+) influx, especially store-operated Ca(2+) entry triggered by sarcoplasmic reticulum (SR) Ca(2+) release, is a key component of intracellular calcium concentration ([Ca(2+)]i) regulation in airway smooth muscle (ASM). Agonist-induced Ca(2+) oscillations in ASM that involve both influx and SR mechanisms have been previously demonstrated. In nonexcitable cells, [Ca(2+)]i oscillations involve Ca(2+) influx via arachidonic acid (AA) -stimulated channels, which show similarities to store-operated Ca(2+) entry, although their molecular identity remains undetermined. Little is known about AA-regulated Ca(2+) channels or their regulation in ASM. In enzymatically dissociated human ASM cells loaded with the Ca(2+) indicator, fura-2, AA (1-10 μM) triggered [Ca(2+)]i oscillations that were inhibited by removal of extracellular Ca(2+). Other fatty acids, such as the diacylglycerol analog, 1-oleoyl-2-acetyl-SN-glycerol, oleic acid, and palmitic acid (10 μM each), failed to elicit similar [Ca(2+)]i responses. Preincubation with LaCl3 (1 μM or 1 mM) inhibited AA-induced oscillations. Inhibition of receptor-operated channels (SKF96,365 [10 μM]), lipoxygenase (zileuton [10 μM]), or cyclooxygenase (indomethacin [10 μM]) did not affect oscillation parameters. Inhibition of SR Ca(2+) release (ryanodine [10 μM] or inositol 1,4,5-trisphosphate receptor inhibitor, xestospongin C [1 μM]) decreased [Ca(2+)]i oscillation frequency and amplitude. Small interfering RNA against caveolin-1, stromal interaction molecule 1, or Orai3 (20 nM each) reduced the frequency and amplitude of AA-induced [Ca(2+)]i oscillations. In ASM cells derived from individuals with asthma, AA increased oscillation amplitude, but not frequency. These results are highly suggestive of a novel AA-mediated Ca(2+)-regulatory mechanism in human ASM, reminiscent of agonist-induced oscillations. Given the role of AA in ASM intracellular signaling, especially with inflammation, AA-regulated Ca(2+) channels could potentially contribute to increased [Ca(2+)]i in diseases such asthma.
Collapse
|
62
|
Olszewska A, Bednarczyk P, Siemen D, Szewczyk A. Modulation of the mitochondrial large-conductance calcium-regulated potassium channel by polyunsaturated fatty acids. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2014; 1837:1602-10. [PMID: 25046142 DOI: 10.1016/j.bbabio.2014.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 07/03/2014] [Accepted: 07/11/2014] [Indexed: 10/25/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) and their metabolites can modulate several biochemical processes in the cell and thus prevent various diseases. PUFAs have a number of cellular targets, including membrane proteins. They can interact with plasma membrane and intracellular potassium channels. The goal of this work was to verify the interaction between PUFAs and the most common and intensively studied mitochondrial large conductance Ca(2+)-regulated potassium channel (mitoBKCa). For this purpose human astrocytoma U87 MG cell lines were investigated using a patch-clamp technique. We analyzed the effects of arachidonic acid (AA); eicosatetraynoic acid (ETYA), which is a non-metabolizable analog of AA; docosahexaenoic acid (DHA); and eicosapentaenoic acid (EPA). The open probability (Po) of the channel did not change significantly after application of 10μM ETYA. Po increased, however, after adding 10μM AA. The application of 30μM DHA or 10μM EPA also increased the Po of the channel. Additionally, the number of open channels in the patch increased in the presence of 30μM EPA. Collectively, our results indicate that PUFAs regulate the BKCa channel from the inner mitochondrial membrane.
Collapse
Affiliation(s)
- Anna Olszewska
- Department of Biochemistry, Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland.
| | - Piotr Bednarczyk
- Department of Biochemistry, Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland; Department of Biophysics, Warsaw University of Life Sciences, Warsaw, Poland
| | - Detlef Siemen
- Department of Neurology, Otto-von-Guericke Universität Magdeburg, Germany
| | - Adam Szewczyk
- Department of Biochemistry, Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
63
|
Poveda J, Giudici A, Renart M, Molina M, Montoya E, Fernández-Carvajal A, Fernández-Ballester G, Encinar J, González-Ros J. Lipid modulation of ion channels through specific binding sites. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:1560-7. [DOI: 10.1016/j.bbamem.2013.10.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/24/2013] [Accepted: 10/30/2013] [Indexed: 01/08/2023]
|
64
|
Oz M, Al Kury L, Keun-Hang SY, Mahgoub M, Galadari S. Cellular approaches to the interaction between cannabinoid receptor ligands and nicotinic acetylcholine receptors. Eur J Pharmacol 2014; 731:100-5. [DOI: 10.1016/j.ejphar.2014.03.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 02/24/2014] [Accepted: 03/10/2014] [Indexed: 01/31/2023]
|
65
|
Beaulieu E, Ioffe J, Watson SN, Hermann PM, Wildering WC. Oxidative-stress induced increase in circulating fatty acids does not contribute to phospholipase A2-dependent appetitive long-term memory failure in the pond snail Lymnaea stagnalis. BMC Neurosci 2014; 15:56. [PMID: 24886155 PMCID: PMC4013061 DOI: 10.1186/1471-2202-15-56] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/23/2014] [Indexed: 11/23/2022] Open
Abstract
Background Reactive oxygen species (ROS) are essential for normal physiological functioning of the brain. However, uncompensated increase in ROS levels may results in oxidative stress. Phospholipase A2 (PLA2) is one of the key players activated by elevated ROS levels resulting in the hydrolysis of various products from the plasmamembrane such as peroxidized fatty acids. Free fatty acids (FFAs) and fatty acid metabolites are often implicated to the genesis of cognitive impairment. Previously we have shown that age-, and experimentally induced oxidative stress causes PLA2-dependent long-term memory (LTM) failure in an aversive operant conditioning model in Lymnaea stagnalis. In the present study, we investigate the effects of experimentally induced oxidative stress and the role of elevated levels of circulating FFAs on LTM function using a non-aversive appetitive classical conditioning paradigm. Results We show that intracoelomic injection of exogenous PLA2 or pro-oxidant induced PLA2 activation negatively affects LTM performance in our learning paradigm. In addition, we show that experimental induction of oxidative stress causes significant temporal changes in circulating FFA levels. Importantly, the time of training coincides with the peak of this change in lipid metabolism. However, intracoelomic injection with exogenous arachidonic acid, one of the main FFAs released by PLA2, does not affect LTM function. Moreover, sequestrating circulating FFAs with the aid of bovine serum albumin does not rescue pro-oxidant induced appetitive LTM failure. Conclusions Our data substantiates previous evidence linking lipid peroxidation and PLA2 activation to age- and oxidative stress-related cognitive impairment, neuronal dysfunction and disease. In addition however, our data indicate that lipid peroxidation induced increased levels of circulating (per)oxidized FFAs are not a factor in oxidative stress induced LTM impairment.
Collapse
Affiliation(s)
| | | | | | | | - Willem C Wildering
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
66
|
Chemin J, Cazade M, Lory P. Modulation of T-type calcium channels by bioactive lipids. Pflugers Arch 2014; 466:689-700. [PMID: 24531745 DOI: 10.1007/s00424-014-1467-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 01/24/2014] [Accepted: 01/29/2014] [Indexed: 01/10/2023]
Abstract
T-type calcium channels (T-channels/CaV3) have unique biophysical properties allowing a calcium influx at resting membrane potential of most cells. T-channels are ubiquitously expressed in many tissues and contribute to low-threshold spikes and burst firing in central neurons as well as to pacemaker activities in cardiac cells. They also emerged as potential targets to treat cancer and hypertension. Regulation of these channels appears complex, and several studies have indicated that CaV3.1, CaV3.2, and CaV3.3 currents are directly inhibited by multiple endogenous lipids independently of membrane receptors or intracellular pathways. These bioactive lipids include arachidonic acid and ω3 poly-unsaturated fatty acids; the endocannabinoid anandamide and other N-acylethanolamides; the lipoamino-acids and lipo-neurotransmitters; the P450 epoxygenase metabolite 5,6-epoxyeicosatrienoic acid; as well as similar molecules with 18-22 carbons in the alkyl chain. In this review, we summarize evidence for direct effects of these signaling molecules, the molecular mechanisms underlying the current inhibition, and the involved chemical features. The impact of this modulation in physiology and pathophysiology is discussed with a special emphasis on pain aspects and vasodilation. Overall, these data clearly indicate that T-current inhibition is an important mechanism by which bioactive lipids mediate their physiological functions.
Collapse
Affiliation(s)
- Jean Chemin
- Institut de Génomique Fonctionnelle, Universités Montpellier 1 & 2, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5203, 141, rue de la Cardonille, 34094, Montpellier cedex 05, France,
| | | | | |
Collapse
|
67
|
Carta M, Lanore F, Rebola N, Szabo Z, Da Silva SV, Lourenço J, Verraes A, Nadler A, Schultz C, Blanchet C, Mulle C. Membrane lipids tune synaptic transmission by direct modulation of presynaptic potassium channels. Neuron 2014; 81:787-99. [PMID: 24486086 DOI: 10.1016/j.neuron.2013.12.028] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2013] [Indexed: 12/20/2022]
Abstract
Voltage-gated potassium (Kv) channels are involved in action potential (AP) repolarization in excitable cells. Exogenous application of membrane-derived lipids, such as arachidonic acid (AA), regulates the gating of Kv channels. Whether membrane-derived lipids released under physiological conditions have an impact on neuronal coding through this mechanism is unknown. We show that AA released in an activity-dependent manner from postsynaptic hippocampal CA3 pyramidal cells acts as retrograde messenger, inducing a robust facilitation of mossy fiber (Mf) synaptic transmission over several minutes. AA acts by broadening presynaptic APs through the direct modulation of Kv channels. This form of short-term plasticity can be triggered when postsynaptic cell fires with physiologically relevant patterns and sets the threshold for the induction of the presynaptic form of long-term potentiation (LTP) at hippocampal Mf synapses. Hence, direct modulation of presynaptic Kv channels by activity-dependent release of lipids serves as a physiological mechanism for tuning synaptic transmission.
Collapse
Affiliation(s)
- Mario Carta
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - Frederic Lanore
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - Nelson Rebola
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - Zsolt Szabo
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - Silvia Viana Da Silva
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - Joana Lourenço
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - Agathe Verraes
- Institut Jacques Monod, UMR 7592, CNRS and INSERM ERL U950, University Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France
| | - André Nadler
- EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Carsten Schultz
- EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Christophe Blanchet
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - Christophe Mulle
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France.
| |
Collapse
|
68
|
Kweon HJ, Suh BC. Acid-sensing ion channels (ASICs): therapeutic targets for neurological diseases and their regulation. BMB Rep 2014; 46:295-304. [PMID: 23790972 PMCID: PMC4133903 DOI: 10.5483/bmbrep.2013.46.6.121] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Extracellular acidification occurs not only in pathological conditions such as inflammation and brain ischemia, but also in normal physiological conditions such as synaptic transmission. Acid-sensing ion channels (ASICs) can detect a broad range of physiological pH changes during pathological and synaptic cellular activities. ASICs are voltage-independent, proton-gated cation channels widely expressed throughout the central and peripheral nervous system. Activation of ASICs is involved in pain perception, synaptic plasticity, learning and memory, fear, ischemic neuronal injury, seizure termination, neuronal degeneration, and mechanosensation. Therefore, ASICs emerge as potential therapeutic targets for manipulating pain and neurological diseases. The activity of these channels can be regulated by many factors such as lactate, Zn2+, and Phe-Met-Arg-Phe amide (FMRFamide)-like neuropeptides by interacting with the channel’s large extracellular loop. ASICs are also modulated by G protein-coupled receptors such as CB1 cannabinoid receptors and 5-HT2. This review focuses on the physiological roles of ASICs and the molecular mechanisms by which these channels are regulated. [BMB Reports 2013; 46(6): 295-304]
Collapse
Affiliation(s)
- Hae-Jin Kweon
- Department of Brain Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Korea
| | | |
Collapse
|
69
|
Abstract
TRPV3 is a temperature-sensitive transient receptor potential (TRP) ion channel. The TRPV3 protein functions as a Ca(2+)-permeable nonselective cation channel with six transmembrane domains forming a tetrameric complex. TRPV3 is known to be activated by warm temperatures, synthetic small-molecule chemicals, and natural compounds from plants. Its function is regulated by a variety of physiological factors including extracellular divalent cations and acidic pH, intracellular adenosine triphosphate, membrane voltage, and arachidonic acid. TRPV3 shows a broad expression pattern in both neuronal and non-neuronal tissues including epidermal keratinocytes, epithelial cells in the gut, endothelial cells in blood vessels, and neurons in dorsal root ganglia and CNS. TRPV3 null mice exhibit abnormal hair morphogenesis and compromised skin barrier function. Recent advances suggest that TRPV3 may play critical roles in inflammatory skin disorders, itch, and pain sensation. Thus, identification of selective TRPV3 activators and inhibitors could potentially lead to beneficial pharmacological interventions in several diseases. The intent of this review is to summarize our current knowledge of the tissue expression, structure, function, and mechanisms of activation of TRPV3.
Collapse
Affiliation(s)
- Jialie Luo
- The Center for the Study of Itch, Washington University Pain Center, The Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Hongzhen Hu
- The Center for the Study of Itch, Washington University Pain Center, The Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
70
|
Martín P, Moncada M, Enrique N, Asuaje A, Valdez Capuccino JM, Gonzalez C, Milesi V. Arachidonic acid activation of BKCa (Slo1) channels associated to the β1-subunit in human vascular smooth muscle cells. Pflugers Arch 2013; 466:1779-92. [DOI: 10.1007/s00424-013-1422-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/03/2013] [Accepted: 12/05/2013] [Indexed: 01/10/2023]
|
71
|
Metabonomic profiling in studying anti-osteoporosis effects of strontium fructose 1,6-diphosphate on estrogen deficiency-induced osteoporosis in rats by GC/TOF-MS. Eur J Pharmacol 2013; 718:524-32. [DOI: 10.1016/j.ejphar.2013.06.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 06/05/2013] [Accepted: 06/21/2013] [Indexed: 01/28/2023]
|
72
|
Martins DA, Rocha F, Castanheira F, Mendes A, Pousão-Ferreira P, Bandarra N, Coutinho J, Morais S, Yúfera M, Conceição LEC, Martínez-Rodríguez G. Effects of dietary arachidonic acid on cortisol production and gene expression in stress response in Senegalese sole (Solea senegalensis) post-larvae. FISH PHYSIOLOGY AND BIOCHEMISTRY 2013; 39:1223-1238. [PMID: 23443720 DOI: 10.1007/s10695-013-9778-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 02/18/2013] [Indexed: 06/01/2023]
Abstract
Dietary fatty acids, particularly arachidonic acid (ARA), affect cortisol and may influence the expression of genes involved in stress response in fish. The involvement of ARA on stress, lipid, and eicosanoid metabolism genes, in Senegalese sole, was tested. Post-larvae were fed Artemia presenting graded ARA levels (0.1, 0.4, 0.8, 1.7, and 2.3%, dry matter basis), from 22 to 35 days after hatch. Whole-body cortisol levels were determined, before and 3 h after a 2 min air exposure, as well as the expression of phospholipase A2 (PLA 2 ), cyclooxygenase-2 (COX-2), steroidogenic acute regulatory protein (StAR), glucocorticoid receptors (GRs), phosphoenolpyruvate carboxykinase (PEPCK), and peroxisome proliferator-activated receptor alpha (PPARα). Relative growth rate (6.0-7.8% day(-1)) and survival at the end of the experiment (91-96%) and after stress (100%) were unaffected. Fish reflected dietary ARA content and post-stress cortisol increased with ARA supply up to 1.7%, whereas 2.3% ARA seemed to enhance basal cortisol slightly and alter the response to stress. Results suggested that elevating StAR transcription might not be necessary for a short-term response to acute stress. Basal cortisol and PLA 2 expression were strongly correlated, indicating a potential role for this enzyme in steroidogenesis. Under basal conditions, larval ARA was associated with GR1 expression, whereas the glucocorticoid responsive gene PEPCK was strongly related with cortisol but not GR1 mRNA levels, suggesting the latter might not reflect the amount of GR1 protein in sole. Furthermore, a possible role for PPARα in the expression of PEPCK following acute stress is proposed.
Collapse
Affiliation(s)
- Dulce Alves Martins
- Centro de Ciências do Mar do Algarve, Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Nassir F, Adewole OL, Brunt EM, Abumrad NA. CD36 deletion reduces VLDL secretion, modulates liver prostaglandins, and exacerbates hepatic steatosis in ob/ob mice. J Lipid Res 2013; 54:2988-97. [PMID: 23964120 DOI: 10.1194/jlr.m037812] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent findings described the role of CD36-mediated signaling in regulating cellular calcium and the release of various bioactive molecules, including the prostaglandins, neurotransmitters, cholecystokinin, and secretin. Here we document the role of CD36 in the secretion of hepatic VLDL. CD36 deletion resulted in 60% suppression of VLDL output in vivo, and VLDL secretion was reduced in vitro using incubated liver slices. The effect of CD36 deletion was mediated by enhancing formation of hepatic prostaglandins D2, F2, and E2. Treatment of CD36-deficient slices with inhibitors of cyclooxygenases reversed the reduction in triglyceride secretion. We also examined the effect of CD36 deletion on the obesity-associated spontaneous steatosis of the ob/ob mouse that is driven by enhanced de novo lipogenesis. Homozygous ob/ob mice lacking CD36 (ob-CD36⁻/⁻) were generated and studied for hepatic triglyceride accumulation and VLDL secretion. Livers of ob/ob mice were steatotic as expected and had 5-fold more CD36 on Kupffer cells and hepatocytes. CD36 deletion exacerbated the steatosis by impairing hepatic triglyceride and apoB secretion through increasing prostaglandin levels. These findings suggest an unappreciated role of CD36 in regulating VLDL secretion, which might have relevance to some forms of fatty liver. They provide insight into the association reported in humans between CD36 protein expression and serum levels of apoB and VLDL particle number.
Collapse
Affiliation(s)
- Fatiha Nassir
- Department of Medicine Washington University School of Medicine, St. Louis, MO 63110
| | | | | | | |
Collapse
|
74
|
Hermann PM, Park D, Beaulieu E, Wildering WC. Evidence for inflammation-mediated memory dysfunction in gastropods: putative PLA2 and COX inhibitors abolish long-term memory failure induced by systemic immune challenges. BMC Neurosci 2013; 14:83. [PMID: 23915010 PMCID: PMC3750374 DOI: 10.1186/1471-2202-14-83] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/02/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Previous studies associate lipid peroxidation with long-term memory (LTM) failure in a gastropod model (Lymnaea stagnalis) of associative learning and memory. This process involves activation of Phospholipase A2 (PLA2), an enzyme mediating the release of fatty acids such as arachidonic acid that form the precursor for a variety of pro-inflammatory lipid metabolites. This study investigated the effect of biologically realistic challenges of L. stagnalis host defense response system on LTM function and potential involvement of PLA2, COX and LOX therein. RESULTS Systemic immune challenges by means of β-glucan laminarin injections induced elevated H2O2 release from L. stagnalis circulatory immune cells within 3 hrs of treatment. This effect dissipated within 24 hrs after treatment. Laminarin exposure has no direct effect on neuronal activity. Laminarin injections disrupted LTM formation if training followed within 1 hr after injection but had no behavioural impact if training started 24 hrs after treatment. Intermediate term memory was not affected by laminarin injection. Chemosensory and motor functions underpinning the feeding response involved in this learning model were not affected by laminarin injection. Laminarin's suppression of LTM induction was reversed by treatment with aristolochic acid, a PLA2 inhibitor, or indomethacin, a putative COX inhibitor, but not by treatment with nordihydro-guaiaretic acid, a putative LOX inhibitor. CONCLUSIONS A systemic immune challenge administered shortly before behavioural training impairs associative LTM function in our model that can be countered with putative inhibitors of PLA2 and COX, but not LOX. As such, this study establishes a mechanistic link between the state of activity of this gastropod's innate immune system and higher order nervous system function. Our findings underwrite the rapidly expanding view of neuroinflammatory processes as a fundamental, evolutionary conserved cause of cognitive and other nervous system disorders.
Collapse
Affiliation(s)
- Petra M Hermann
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | | | | |
Collapse
|
75
|
Fokina NN, Ruokolainen TR, Nemova NN, Bakhmet IN. Changes of blue mussels Mytilus edulis L. lipid composition under cadmium and copper toxic effect. Biol Trace Elem Res 2013; 154:217-25. [PMID: 23793920 DOI: 10.1007/s12011-013-9727-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 06/04/2013] [Indexed: 02/04/2023]
Abstract
The lipid and fatty acid composition of the blue mussels Mytilus edulis L. gills and digestive glands was evaluated after 24 and 72 h of cadmium (Cd) and copper (Cu) exposure. Mussels were exposed to different cadmium (10, 100, and 500 μg/L) and copper (5, 50, and 250 μg/L) concentrations. Similar stress response of predominant membrane phospholipids level as well as polyenoic and non-methylene interrupted (NMI) fatty acids content was observed in mussel gills under both cadmium and copper effects. Increased NMI fatty acids level after 24 h, the metal ions treatment suggests that these acids contribute to the protective response to the membrane oxidative stress caused by accumulation of the metals. The content of cholesterol, some minor membrane phospholipids, and storage lipids (triacylglycerols, TAG) in the mussels' organs alter significantly under the cadmium and copper effect. A two-step response at the digestive glands TAG level depends on the duration of the cadmium and copper treatments (24 and 72 h) on the mussels. The results demonstrate that Cd and Cu impact has adverse effects on gills and digestive glands lipid and fatty acids composition. The type of observed effects varies with the nature and concentration of the metal ions and depends on the role of the metals in the mussels' life activity.
Collapse
Affiliation(s)
- Natalia N Fokina
- Institute of Biology Karelian Research Center of Russian Academy of Sciences, Pushkinskaja st., 11, Petrozavodsk 185910, Russia.
| | | | | | | |
Collapse
|
76
|
Okada Y, Miyazaki T, Fujiyama R, Toda K. Wing (Ib) cells in frog taste discs detect dietary unsaturated fatty acids. Comp Biochem Physiol A Mol Integr Physiol 2013; 166:434-40. [PMID: 23872318 DOI: 10.1016/j.cbpa.2013.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/11/2013] [Accepted: 07/10/2013] [Indexed: 11/17/2022]
Abstract
The effects of unsaturated fatty acids on membrane properties were studied using conventional whole-cell patch-clamp recording of isolated wing (Ib) cells in bullfrog (Lithobates catesbeianus) taste discs. Applying arachidonic acid to the bath induced monophasic inward currents in 60% of wing cells and biphasic inward and outward currents in the other cells. The intracellular dialysis of arachidonic acid did not induce an inward current; however, it enhanced a slowly developing Ba(2+)-sensitive outward current. The effects of various unsaturated fatty acids were explored under the condition of Cs(+) internal solution. Linoleic and α-linolenic acids induced large inward currents. Oleic, eicosapentaenoic and docosahexaenoic acids elicited the same inward currents as those of arachidonic acid. Wing cells, under the basal condition with Cs(+) internal solution, displayed a small inward current of -1.1±0.1pA/pF at -50mV (n=40), in which the peak existed at a membrane potential of -49mV. Removing external Ca(2+) further increased the inward current by -2.9±0.3pA/pF at -50mV (n=4) from the basal current and the peak was located at -55mV. External linoleic acid (50μM) also induced a similar inward current of -5.6±0.6pA/pF at -50mV (n=19) from the basal current and the peak was located at -61mV. External Ca(2+)-free saline and linoleic acid induced similar current/voltage (I/V) relationships elicited by a ramp voltage as well as voltage steps. Linoleic acid-induced currents were not influenced by replacing internal EGTA with BAPTA, whereas inward currents disappeared under the elimination of external Na(+) and addition of flufenamic acid. These results suggest that dietary unsaturated fatty acids may depolarize wing (Ib) cells, which affects the excitability of these cells.
Collapse
Affiliation(s)
- Yukio Okada
- Integrative Sensory Physiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki 852-8588, Japan.
| | | | | | | |
Collapse
|
77
|
Montserrat-de la Paz S, García-Giménez MD, Ángel-Martín M, Marín-Aguilar F, Fernández-Arche A. Dietary supplementation evening primrose oil improve symptoms of fibromyalgia syndrome. J Funct Foods 2013. [DOI: 10.1016/j.jff.2013.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
78
|
Kukkonen JP. Lipid signaling cascades of orexin/hypocretin receptors. Biochimie 2013; 96:158-65. [PMID: 23810911 DOI: 10.1016/j.biochi.2013.06.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 06/18/2013] [Indexed: 11/18/2022]
Abstract
Orexins - orexin-A and orexin-B - are neuropeptides with significant role in regulation of fundamental physiological processes such as sleep-wakefulness cycle. Orexins act via G-protein-coupled OX1 and OX2 receptors, which are found, in addition to the central nervous system, also in a number of peripheral organs. Orexin receptors show high degree of signaling promiscuity. One particularly prominent way of signaling for these receptors is via phospholipase cascades, including the phospholipase C, phospholipase D and phospholipase A2 cascades, and also diacylglycerol lipase and phosphoinositide-3-kinase pathways. Most analyses have been performed in recombinant cells; there are indications of some of these cascades in native cells while the significance of other cascades remains to be shown. In this review, I present these pathways, their activation mechanisms and their physiological significance.
Collapse
Key Words
- 2-AG
- 2-arachidonoylglycerol
- AA
- CNS
- DAG
- DAG lipase
- DAGL
- DOG
- ERK
- Endocannabinoid
- G-protein-coupled receptor
- GPCR
- GPL
- Hypocretin
- IP(3)
- Ion fluxes
- KB-R7943
- MAFP
- N-acyl-phosphatidylethanolamine
- N-arachidonoylethanolamine
- NAPE
- NSCC
- OX(1)
- OX(2)
- Orexin
- PA
- PC
- PC-PLC
- PC-specific PLC
- PDK1
- PI
- PI3K
- PIP
- PIP(2)
- PIP(3)
- PIs
- PKB, PKC and PKD
- PLA(1), PLA(2), PLB, PLC and PLD
- Phospholipase
- TRP (channel)
- U73122
- a NCX inhibitor
- a PLC inhibitor
- a cPLA(2)α/ζ inhibitor
- anandamide
- arachidonic acid
- cPLA(2) and iPLA(2)
- central nervous system
- cytosolic (Ca(2+)-dependent) and intracellular (Ca(2+)-independent) PLA(2), respectively
- diacylglycerol
- dioctanoylglycerol
- extracellular signal-regulated kinase
- glycerophospholipid
- inositol-1,4,5-trisphosphate
- lyso(glycero)phospholipid
- lysoGPL
- lysoPA
- lysophosphatidic acid
- methyl arachidonyl fluorophosphonate
- non-selective cation channel
- orexin 1 receptor
- orexin 2 receptor
- phosphatidic acid
- phosphatidylcholine
- phosphatidylinositol
- phosphatidylinositol-3,4,5-trisphosphate
- phosphatidylinositol-4,5-bisphosphate
- phosphatidylinositolmonophosphate
- phosphatidylinositols (including differentially phosphorylated species PI, PIP, PIP(2) and PIP(3))
- phosphoinositide-3-kinase
- phosphoinositide-dependent kinase 1
- phospholipase A(1), A(2), B, C and D, respectively
- protein kinase B, C and D, respectively
- pyrrophenone
- transient receptor potential (channel)
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Biochemistry and Cell Biology, Department of Veterinary Biosciences, POB 66, FIN-00014, University of Helsinki, Finland.
| |
Collapse
|
79
|
Zheng X, Zinkevich NS, Gebremedhin D, Gauthier KM, Nishijima Y, Fang J, Wilcox DA, Campbell WB, Gutterman DD, Zhang DX. Arachidonic acid-induced dilation in human coronary arterioles: convergence of signaling mechanisms on endothelial TRPV4-mediated Ca2+ entry. J Am Heart Assoc 2013; 2:e000080. [PMID: 23619744 PMCID: PMC3698766 DOI: 10.1161/jaha.113.000080] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Arachidonic acid (AA) and/or its enzymatic metabolites are important lipid mediators contributing to endothelium-derived hyperpolarizing factor (EDHF)-mediated dilation in multiple vascular beds, including human coronary arterioles (HCAs). However, the mechanisms of action of these lipid mediators in endothelial cells (ECs) remain incompletely defined. In this study, we investigated the role of the transient receptor potential vanilloid 4 (TRPV4) channel in AA-induced endothelial Ca(2+) response and dilation of HCAs. METHODS AND RESULTS AA induced concentration-dependent dilation in isolated HCAs. The dilation was largely abolished by the TRPV4 antagonist RN-1734 and by inhibition of endothelial Ca(2+)-activated K(+) channels. In native and TRPV4-overexpressing human coronary artery ECs (HCAECs), AA increased intracellular Ca(2+) concentration ([Ca(2+)]i), which was mediated by TRPV4-dependent Ca(2+) entry. The AA-induced [Ca(2+)]i increase was inhibited by cytochrome P450 (CYP) inhibitors. Surprisingly, the CYP metabolites of AA, epoxyeicosatrienoic acids (EETs), were much less potent activators of TRPV4, and CYP inhibitors did not affect EET production in HCAECs. Apart from its effect on [Ca(2+)]i, AA induced endothelial hyperpolarization, and this effect was required for Ca(2+) entry through TRPV4. AA-induced and TRPV4-mediated Ca(2+) entry was also inhibited by the protein kinase A inhibitor PKI. TRPV4 exhibited a basal level of phosphorylation, which was inhibited by PKI. Patch-clamp studies indicated that AA activated TRPV4 single-channel currents in cell-attached and inside-out patches of HCAECs. CONCLUSIONS AA dilates HCAs through a novel mechanism involving endothelial TRPV4 channel-dependent Ca(2+) entry that requires endothelial hyperpolarization, PKA-mediated basal phosphorylation of TRPV4, and direct activation of TRPV4 channels by AA.
Collapse
Affiliation(s)
- Xiaodong Zheng
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
Leucettamols, bifunctionalized sphingoid-like compounds obtained from a marine sponge Leucetta sp., act as non-electrophilic activators of the TRPA1 channel and potent inhibitors of the icilin-mediated activation of the TRPM8 channel, while they are inactive on CB₁, CB₂ and TRPV1 receptors. Leucettamols represent the first compounds of marine origin to target TRPA1 and the first class of natural products to inhibit TRPM8 channels. The preparation of a small series of semi-synthetic derivatives revealed interesting details on the structure-activity relationships within this new chemotype of simple acyclic TRP modulators.
Collapse
|
81
|
Gutla PVK, Boccaccio A, De Angeli A, Gambale F, Carpaneto A. Modulation of plant TPC channels by polyunsaturated fatty acids. JOURNAL OF EXPERIMENTAL BOTANY 2012; 63:6187-97. [PMID: 23105130 PMCID: PMC3481210 DOI: 10.1093/jxb/ers272] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) are powerful modulators of several animal ion channels. It is shown here that PUFAs strongly affect the activity of the Slow Vacuolar (SV) channel encoded by the plant TPC1 gene. The patch-clamp technique was applied to isolated vacuoles from carrot taproots and Arabidopsis thaliana mesophyll cells and arachidonic acid (AA) was chosen as a model molecule for PUFAs. Our study was extended to different PUFAs including the endogenous alpha-linolenic acid (ALA). The addition of micromolar concentrations of AA reversibly inhibited the SV channel decreasing the maximum open probability and shifting the half activation voltage to positive values. Comparing the effects of different PUFAs, it was found that the length of the lipophilic acyl chain, the number of double bonds and the polar head were critical for channel modulation.The experimental data can be reproduced by a simple three-state model, in which PUFAs do not interact directly with the voltage sensors but affect the voltage-independent transition that leads the channel from the open state to the closed configuration. The results indicate that lipids play an important role in co-ordinating ion channel activities similar to what is known from animal cells.
Collapse
Affiliation(s)
- Paul Vijay Kanth Gutla
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via De Marini 6, 16149 Genova, Italy
| | - Anna Boccaccio
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via De Marini 6, 16149 Genova, Italy
| | - Alexis De Angeli
- Institute of Plant Biology, University of Zurich, Zollikerstrasse 107, CH-8008, Zurich, Switzerland
| | - Franco Gambale
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via De Marini 6, 16149 Genova, Italy
| | - Armando Carpaneto
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via De Marini 6, 16149 Genova, Italy
| |
Collapse
|
82
|
Smithers N, Bolivar JH, Lee AG, East JM. Characterizing the fatty acid binding site in the cavity of potassium channel KcsA. Biochemistry 2012; 51:7996-8002. [PMID: 22971149 PMCID: PMC3466778 DOI: 10.1021/bi3009196] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
We show that interactions of fatty acids with the central
cavity of potassium channel KcsA can be characterized using the fluorescence
probe 11-dansylaminoundecanoic acid (Dauda). The fluorescence emission
spectrum of Dauda bound to KcsA in bilayers of dioleoylphosphatidylcholine
contains three components, which can be attributed to KcsA-bound and
lipid-bound Dauda together with unbound Dauda. The binding of Dauda
to KcsA was characterized by a dissociation constant of 0.47 ±
0.10 μM with 0.94 ± 0.06 binding site per KcsA tetramer.
Displacement of KcsA-bound Dauda by the tetrabutylammonium (TBA) ion
confirmed that the Dauda binding site was in the central cavity of
KcsA. Dissociation constants for a range of fatty acids were determined
by displacement of Dauda: binding of fatty acids increased in strength
with an increasing chain length from C14 to C20 but then decreased
in strength from C20 to C22. Increasing the number of double bonds
in the chain from one to four had little effect on binding, dissociation
constants for oleic acid and arachidonic acid, for example, being
2.9 ± 0.2 and 3.0 ± 0.4 μM, respectively. Binding
of TBA to KcsA was very slow, whereas binding of Dauda was fast, suggesting
that TBA can enter the cavity only through an open channel whereas
Dauda can bind to the closed channel, presumably entering the cavity
via the lipid bilayer.
Collapse
Affiliation(s)
- Natalie Smithers
- Centre for Biological Sciences, Life Sciences Building, University of Southampton , Southampton SO17 1BJ, UK
| | | | | | | |
Collapse
|
83
|
Ruparel S, Green D, Chen P, Hargreaves KM. The cytochrome P450 inhibitor, ketoconazole, inhibits oxidized linoleic acid metabolite-mediated peripheral inflammatory pain. Mol Pain 2012; 8:73. [PMID: 23006841 PMCID: PMC3488501 DOI: 10.1186/1744-8069-8-73] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 09/20/2012] [Indexed: 01/26/2023] Open
Abstract
Background Oxidized linoleic acid metabolites (OLAMs) are a class of endogenous agonists to the transient receptor potential V1 (TRPV1) receptor. Although TRPV1 mediates inflammatory heat hyperalgesia, it is not known if the OLAMs contribute to the peripheral activation of this receptor during tissue inflammation. In the present study, we evaluated whether the OLAM system is activated during inflammation and whether cytochrome P450 enzymes mediate OLAM contributions to heat hyperalgesia using the complete Freund’s adjuvant (CFA) model of inflammation. Results Our results demonstrate that the intraplantar (ipl) injection of anti-OLAM antibodies significantly reversed CFA-induced heat hyperalgesia. Moreover, application of lipid extracts from inflamed rat skin to cultured sensory neurons triggered a significant release of iCGRP that is blocked by co-treatment with I-RTX, a TRPV1 antagonist. To determine the role of CYP enzymes in mediating OLAM effects, we used a broad spectrum CYP inhibitor, ketoconazole. Pretreatment with ketoconazole inhibited the release of TRPV1 agonists in lipid extracts from inflamed skin and significantly reversed CFA-induced heat hyperalgesia by a peripheral mechanism of action. Moreover, the ipl injection of linoleic acid to rats 24 hr after CFA evoked spontaneous nocifensive behaviors that were significantly reduced by capsazepine, by knockout of the TRPV1 gene, or by pretreatment with either anti-OLAM antibodies or ketoconazole. Conclusions Taken together, our data suggests that OLAMs contribute to inflammatory nociception in the periphery and that cytochrome P450 enzymes play a crucial role in mediating OLAM contributions to inflammatory heat hyperalgesia.
Collapse
Affiliation(s)
- Shivani Ruparel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | | | | | | |
Collapse
|
84
|
Moreno C, Macias A, Prieto A, De La Cruz A, Valenzuela C. Polyunsaturated Fatty acids modify the gating of kv channels. Front Pharmacol 2012; 3:163. [PMID: 22973228 PMCID: PMC3437463 DOI: 10.3389/fphar.2012.00163] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/20/2012] [Indexed: 11/13/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) have been reported to exhibit antiarrhythmic properties, which are attributed to their capability to modulate ion channels. This PUFAs ability has been reported to be due to their effects on the gating properties of ion channels. In the present review, we will focus on the role of PUFAs on the gating of two Kv channels, Kv1.5 and Kv11.1. Kv1.5 channels are blocked by n-3 PUFAs of marine [docosahexaenoic acid (DHA) and eicosapentaenoic acid] and plant origin (alpha-linolenic acid, ALA) at physiological concentrations. The blockade of Kv1.5 channels by PUFAs steeply increased in the range of membrane potentials coinciding with those of Kv1.5 channel activation, suggesting that PUFAs-channel binding may derive a significant fraction of its voltage sensitivity through the coupling to channel gating. A similar shift in the activation voltage was noted for the effects of n-6 arachidonic acid (AA) and DHA on Kv1.1, Kv1.2, and Kv11.1 channels. PUFAs-Kv1.5 channel interaction is time-dependent, producing a fast decay of the current upon depolarization. Thus, Kv1.5 channel opening is a prerequisite for the PUFA-channel interaction. Similar to the Kv1.5 channels, the blockade of Kv11.1 channels by AA and DHA steeply increased in the range of membrane potentials that coincided with the range of Kv11.1 channel activation, suggesting that the PUFAs-Kv channel interactions are also coupled to channel gating. Furthermore, AA regulates the inactivation process in other Kv channels, introducing a fast voltage-dependent inactivation in non-inactivating Kv channels. These results have been explained within the framework that AA closes voltage-dependent potassium channels by inducing conformational changes in the selectivity filter, suggesting that Kv channel gating is lipid dependent.
Collapse
Affiliation(s)
- Cristina Moreno
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid Madrid, Spain
| | | | | | | | | |
Collapse
|
85
|
Wei HP, Yao YY, Zhang RW, Zhao XF, Du JL. Activity-Induced Long-Term Potentiation of Excitatory Synapses in Developing Zebrafish Retina In Vivo. Neuron 2012; 75:479-89. [DOI: 10.1016/j.neuron.2012.05.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2012] [Indexed: 11/16/2022]
|
86
|
Bolivar JH, Smithers N, East JM, Marsh D, Lee AG. Multiple binding sites for fatty acids on the potassium channel KcsA. Biochemistry 2012; 51:2889-98. [PMID: 22409348 PMCID: PMC3336937 DOI: 10.1021/bi300153v] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interactions of fatty acids with the potassium channel KcsA were studied using Trp fluorescence quenching and electron paramagnetic resonance (EPR) techniques. The brominated analogue of oleic acid was shown to bind to annular sites on KcsA and to the nonannular sites at each protein-protein interface in the homotetrameric structure with binding constants relative to dioleoylphosphatidylcholine of 0.67 ± 0.04 and 0.87 ± 0.08, respectively. Mutation of the two Arg residues close to the nonannular binding sites had no effect on fatty acid binding. EPR studies with a spin-labeled analogue of stearic acid detected a high-affinity binding site for the fatty acid with strong immobilization. Fluorescence quenching studies with the spin-labeled analogue showed that the binding site detected in the EPR experiments could not be one of the annular or nonannular binding sites. Instead, it is proposed that the EPR studies detect binding to the central hydrophobic cavity of the channel, with a binding constant in the range of ~0.1-1 μM.
Collapse
Affiliation(s)
- Juan H Bolivar
- Centre for Biological Sciences, Life Sciences Building, University of Southampton, Southampton SO17 1BJ, UK
| | | | | | | | | |
Collapse
|
87
|
Kantserova NP, Fokina NN, Lysenko LA, Nemova NN. Correlation between the activity of intracellular Ca2+-dependent proteinase and the content of membrane lipid components in mussel, Mytilus edulis, upon accumulation of heavy metals. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2012; 38:86-91. [DOI: 10.1134/s1068162012010116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
88
|
Jiang H, Zeng B, Chen GL, Bot D, Eastmond S, Elsenussi SE, Atkin SL, Boa AN, Xu SZ. Effect of non-steroidal anti-inflammatory drugs and new fenamate analogues on TRPC4 and TRPC5 channels. Biochem Pharmacol 2012; 83:923-31. [PMID: 22285229 DOI: 10.1016/j.bcp.2012.01.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 01/04/2012] [Accepted: 01/12/2012] [Indexed: 01/13/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used anti-inflammatory therapeutic agents, among which the fenamate analogues play important roles in regulating intracellular Ca²⁺ transient and ion channels. However, the effect of NSAIDs on TRPC4 and TRPC5 is still unknown. To understand the structure-activity of fenamate analogues on TRPC channels, we have synthesized a series of fenamate analogues and investigated their effects on TRPC4 and TRPC5 channels. Human TRPC4 and TRPC5 cDNAs in tetracycline-regulated vectors were transfected into HEK293 T-REx cells. The whole cell current and Ca²⁺ movement were recorded by patch clamp and calcium imaging, respectively. Flufenamic acid (FFA), mefenamic acid (MFA), niflumic acid (NFA) and diclofenac sodium (DFS) showed inhibition on TRPC4 and TRPC5 channels in a concentration-dependent manner. The potency was FFA>MFA>NFA>DFS. Modification of 2-phenylamino ring by substitution of the trifluoromethyl group in FFA with F, CH₃, OCH₃, OCH₂CH₃, COOH, and NO₂ led to the changes in their channel blocking activity. However, 2-(2'-methoxy-5'-methylphenyl)aminobenzoic acid stimulated TRPC4 and TRPC5 channels. Selective COX1-3 inhibitors (aspirin, celecoxib, acetaminophen, and indomethacin) had no effect on the channels. Longer perfusion (> 5 min) with FFA (100 μM) and MFA (100 μM) caused a potentiation of TRPC4 and TRPC5 currents after their initial blocking effects that appeared to be partially mediated by the mitochondrial Ca²⁺ release. Our results suggest that fenamate analogues are direct modulators of TRPC4 and TRPC5 channels. The substitution pattern and conformation of the 2-phenylamino ring could alter their blocking activity, which is important for understanding fenamate pharmacology and new drug development targeting the TRPC channels.
Collapse
Affiliation(s)
- Hongni Jiang
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Tse A, Lee AK, Tse FW. Ca2+ signaling and exocytosis in pituitary corticotropes. Cell Calcium 2012; 51:253-9. [PMID: 22225940 DOI: 10.1016/j.ceca.2011.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 12/08/2011] [Accepted: 12/10/2011] [Indexed: 10/14/2022]
Abstract
The secretion of adrenocorticotrophin (ACTH) from corticotropes is a key component in the endocrine response to stress. The resting potential of corticotropes is set by the basal activities of TWIK-related K(+) (TREK)-1 channel. Corticotrophin-releasing hormone (CRH), the major ACTH secretagogue, closes the background TREK-1 channels via the cAMP-dependent pathway, resulting in depolarization and a sustained rise in cytosolic [Ca(2+)] ([Ca(2+)](i)). By contrast, arginine vasopressin and norepinephrine evoke Ca(2+) release from the inositol trisphosphate (IP(3))-sensitive store, resulting in the activation of small conductance Ca(2+)-activated K(+) channels and hyperpolarization. Following [Ca(2+)](i) rise, cytosolic Ca(2+) is taken into the mitochondria via the uniporter. Mitochondrial inhibition slows the decay of the Ca(2+) signal and enhances the depolarization-triggered exocytotic response. Both voltage-gated Ca(2+) channel activation and intracellular Ca(2+) release generate spatial Ca(2+) gradients near the exocytic sites such that the local [Ca(2+)] is ~3-fold higher than the average [Ca(2+)](i). The stimulation of mitochondrial metabolism during the agonist-induced Ca(2+) signal and the robust endocytosis following stimulated exocytosis enable corticotropes to maintain sustained secretion during the diurnal ACTH surge. Arachidonic acid (AA) which is generated during CRH stimulation activates TREK-1 channels and causes hyperpolarization. Thus, corticotropes may regulate ACTH release via an autocrine feedback mechanism.
Collapse
Affiliation(s)
- Amy Tse
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada.
| | | | | |
Collapse
|
90
|
Yeung-Yam-Wah V, Lee AK, Tse A. Arachidonic acid mobilizes Ca2+ from the endoplasmic reticulum and an acidic store in rat pancreatic β cells. Cell Calcium 2011; 51:140-8. [PMID: 22197025 DOI: 10.1016/j.ceca.2011.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 11/30/2011] [Accepted: 11/30/2011] [Indexed: 11/17/2022]
Abstract
In rat pancreatic β cells, arachidonic acid (AA) triggered intracellular Ca(2+) release. This effect could be mimicked by eicosatetraynoic acid, indicating that AA metabolism is not required. The AA-mediated Ca(2+) signal was not affected by inhibition of ryanodine receptors or emptying of ryanodine-sensitive store but was reduced by ∼70% following the disruption of acidic stores (treatment with bafilomycin A1 or glycyl-phenylalanyl-β-naphthylamide (GPN)). The action of AA did not involve TRPM2 channels or NAADP receptors because intracellular dialysis of adenosine diphosphoribose (ADPR; an activator of TRPM2 channels) or NAADP did not affect the AA response. In contrast, stimulation of IP(3) receptors via intracellular dialysis of adenophostin A, or exogenous application of ATP largely abolished the AA-mediated Ca(2+) signal. Intracellular dialysis of heparin abolished the ATP-mediated Ca(2+) signal but not the AA response, suggesting that the action of AA did not involve the IP(3)-binding site. Treatment with the SERCA pump inhibitor, thapsigargin, reduced the amplitude of the AA-mediated Ca(2+) signal by ∼70%. Overall, our finding suggests that AA mobilizes Ca(2+) from the endoplasmic reticulum as well as an acidic store and both stores could be depleted by IP(3) receptor agonist. The possibility of secretory granules as targets of AA is discussed.
Collapse
|
91
|
Rupp H, Rupp TP, Alter P, Maisch B. Mechanisms involved in the differential reduction of omega-3 and omega-6 highly unsaturated fatty acids by structural heart disease resulting in "HUFA deficiency". Can J Physiol Pharmacol 2011; 90:55-73. [PMID: 22188440 DOI: 10.1139/y11-101] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The causes of reduced levels of omega-3 and omega-6 highly unsaturated fatty acids ("HUFA deficiency") in heart failure remain unresolved. HUFA profiles were examined in the serum of 331 patients with failing versus nonfailing heart disease. Arachidonic acid was positively correlated (P < 0.001) with eicosapentaenoic acid (EPA) (r = 0.40) and docosahexaenoic acid (DHA) (r = 0.53) and negatively with palmitic (r = 0.42), palmitoleic (r = 0.38), and oleic acid (r = 0.48). Delta-5 desaturase activity was reduced (P < 0.01) in heart failure patients with low ejection fraction, dilatation, increased wall stress, and reduced heart rate variability (SDNN). In these patients, the reduced (P < 0.01) HUFA and increased palmitic (P < 0.01) and oleic acid (P = 0.05) arose from separate influences involving reduced cardiac contractility (arachidonic acid and palmitic acid predicted by ejection fraction) and chamber dilatation (DHA and oleic acid predicted by end-diastolic diameter). A low DHA (0.2%-0.9% versus 1.4%-3.1%) was associated (P < 0.025) with atrial dilatation (44 ± 8 mm versus 40 ± 8 mm). Equidirectional but less pronounced effects on HUFA were induced by sympathetic activation and (or) insulin resistance (fat and sugar fed to deoxycorticosterone acetate (DOCA)-salt rats) but not by compensated cardiac overload alone (DOCA-salt or aortic constriction), or reduced fatty acid oxidation (CPT-1 inhibition). Based on administration of omega-3 HUFA (OMACOR), dilatation is identified as a target for 1-2 g omega-3 HUFA·day(-1). Interventions for reduced arachidonic acid remain to be explored.
Collapse
Affiliation(s)
- Heinz Rupp
- Experimental Cardiology Laboratory, Department of Internal Medicine - Cardiology, Philipps University Marburg, Baldingerstrasse 1, 35043 Marburg, Germany.
| | | | | | | |
Collapse
|
92
|
Teleost fish larvae adapt to dietary arachidonic acid supply through modulation of the expression of lipid metabolism and stress response genes. Br J Nutr 2011; 108:864-74. [PMID: 22172151 DOI: 10.1017/s0007114511006143] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Dietary fatty acid supply can affect stress response in fish during early development. Although knowledge on the mechanisms involved in fatty acid regulation of stress tolerance is scarce, it has often been hypothesised that eicosanoid profiles can influence cortisol production. Genomic cortisol actions are mediated by cytosolic receptors which may respond to cellular fatty acid signalling. An experiment was designed to test the effects of feeding gilthead sea-bream larvae with four microdiets, containing graded arachidonic acid (ARA) levels (0·4, 0·8, 1·5 and 3·0 %), on the expression of genes involved in stress response (steroidogenic acute regulatory protein, glucocorticoid receptor and phosphoenolpyruvate carboxykinase), lipid and, particularly, eicosanoid metabolism (hormone-sensitive lipase, PPARα, phospholipase A2, cyclo-oxygenase-2 and 5-lipoxygenase), as determined by real-time quantitative PCR. Fish fatty acid phenotypes reflected dietary fatty acid profiles. Growth performance, survival after acute stress and similar whole-body basal cortisol levels suggested that sea-bream larvae could tolerate a wide range of dietary ARA levels. Transcription of all genes analysed was significantly reduced at dietary ARA levels above 0·4 %. Nonetheless, despite practical suppression of phospholipase A2 transcription, higher leukotriene B4 levels were detected in larvae fed 3·0 % ARA, whereas a similar trend was observed regarding PGE2 production. The present study demonstrates that adaptation to a wide range of dietary ARA levels in gilthead sea-bream larvae involves the modulation of the expression of genes related to eicosanoid synthesis, lipid metabolism and stress response. The roles of ARA, other polyunsaturates and eicosanoids as signals in this process are discussed.
Collapse
|
93
|
Effects of diet and behavioral enrichment on free fatty acids in the aged canine brain. Neuroscience 2011; 202:326-33. [PMID: 22183056 DOI: 10.1016/j.neuroscience.2011.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 11/30/2011] [Accepted: 12/01/2011] [Indexed: 01/19/2023]
Abstract
Despite several recent studies suggesting that dysregulation of brain lipid metabolism might contribute to the mechanisms of aging and Alzheimer's disease (AD), lipid metabolism has not been evaluated extensively in the aging brain. Here, we use a lipidomic approach to demonstrate that antioxidants plus mitochondrial cofactors treatment, either alone or in combination with behavioral enrichment, attenuates lipid abnormalities in the frontal cortices of aged canine in a manner correlated with cognitive scores. Our analyses revealed that the levels of free palmitoleic acid and nervonic acid were decreased in frontal cortices of aged dogs (n=5-6/group) treated with antioxidant compared with the control group. The monounsaturated/saturated fatty acid ratio, also known as "desaturation index"-an ex-vivo indicator of stearoyl-CoA desaturase activity, was also reduced in the frontal cortex of dogs treated with antioxidants compared with control groups. Increased palmitoleic acid levels and desaturation index were positively correlated with increased reversal learning errors and decreased cognitive performance. In conclusion, our study indicates that the addition of antioxidants and mitochondrial cofactors to the regular diet alters the composition of free fatty acids in the aged brain. Together with data showing increased palmitoleic acid levels in AD patients, our data suggest that reducing palmitoleic acid levels and desaturation index in the brain may be associated with improved cognitive performance.
Collapse
|
94
|
Arachidonic acid modulates Na+ currents by non-metabolic and metabolic pathways in rat cerebellar granule cells. Biochem J 2011; 438:203-15. [PMID: 21564022 DOI: 10.1042/bj20110569] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AA (arachidonic acid), which possesses both neurotoxic and neurotrophic activities, has been implicated as a messenger in both physiological and pathophysiological processes. In the present study, we investigated the effects of both extracellular and intracellular application of AA on the activity of Na(V) (voltage-gated Na(+) channels) in rat cerebellar GCs (granule cells). The extracellular application of AA inhibited the resultant I(Na) (Na(V) current), wherein the current-voltage curve shifted to a negative voltage direction. Because this effect could be reproduced by treating the GCs with ETYA (eicosa-5,8,11,14-tetraynoic acid) or a membrane-impermeable analogue of AA, AA-CoA (arachidonoyl coenzyme A), we inferred that AA itself exerted the observed modulatory effects on I(Na). In contrast, intracellular AA significantly augmented the elicited I(Na) peak when the same protocol that was used for extracellular AA was followed. The observed I(Na) increase that was induced by intracellular AA was mimicked by the AA cyclo-oxygenase metabolite PGE(2) (prostaglandin E(2)), but not by ETYA. Furthermore, cyclo-oxygenase inhibitors decreased I(Na) and quenched AA-induced channel activation, indicating that the effect of intracellular AA on Na(V) was possibly mediated through AA metabolites. In addition, the PGE2-induced activation of I(Na) was mimicked by cAMP and quenched by a PKA (protein kinase A) inhibitor, a G(s) inhibitor and EP (E-series of prostaglandin) receptor antagonists. The results of the present study suggest that extracellular AA modulates Na(V) channel activity in rat cerebellar GCs without metabolic conversion, whereas intracellular AA augments the I(Na) by PGE(2)-mediated activation of cAMP/PKA pathways. These observations may explain the dual character of AA in neuronal pathogenesis.
Collapse
|
95
|
Oleic acid inhibits store-operated calcium entry in human colorectal adenocarcinoma cells. Eur J Nutr 2011; 51:677-84. [DOI: 10.1007/s00394-011-0246-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 09/07/2011] [Indexed: 11/26/2022]
|
96
|
Fiorio Pla A, Avanzato D, Munaron L, Ambudkar IS. Ion channels and transporters in cancer. 6. Vascularizing the tumor: TRP channels as molecular targets. Am J Physiol Cell Physiol 2011; 302:C9-15. [PMID: 21832241 DOI: 10.1152/ajpcell.00280.2011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumor vascularization is a critical process that determines tumor growth and metastasis. In the last decade new experimental evidence obtained from in vitro and in vivo studies have challenged the classical angiogenesis model forcing us to consider new scenarios for tumor neovascularization. In particular, the genetic stability of tumor-derived endothelial cells (TECs) has been recently questioned in several studies, which show that TECs, as well as pericytes, differ significantly from their normal counterparts at genetic and functional levels. In addition to such an epigenetic action of tumor microenvironment on endothelial cells (ECs) commitment, the distinct characteristics of TECs could be due to differences in their origin compared with preexisting differentiated ECs. Intracellular Ca(2+) signals are involved at different critical phases in the regulation of the complex process of angiogenesis and tumor progression. These signals are generated by a wide variety of intrinsic and extrinsic factors. Several key components of Ca(2+) signaling including Ca(2+) channels in the plasma membrane, endoplasmic reticulum, calcium pumps, and mitochondria contribute to the generation, amplitude, and frequency of these Ca(2+) change. In particular, several members of the transient receptor potential (TRP) family of calcium-permeable channels have profound effects on the function of ECs. Because of its multifaceted role in the control of cell function, proliferation, and motility, TRP channels have been suggested as a potential molecular target for control of tumor neovascularization. Since plasma membrane Ca(2+) channels are easily and directly accessible via the bloodstream, they are potential targets for a number of pharmacological and antibody-targeted therapeutic strategies, with specificity being the main limitation. In this review we discuss recent advances in understanding the role of Ca(2+) channels, with specific reference to TRP channels, in tumor vascularization process.
Collapse
|
97
|
Kukkonen JP. A ménage à trois made in heaven: G-protein-coupled receptors, lipids and TRP channels. Cell Calcium 2011; 50:9-26. [DOI: 10.1016/j.ceca.2011.04.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/21/2011] [Accepted: 04/22/2011] [Indexed: 12/15/2022]
|
98
|
Pavlov TS, Ilatovskaya DV, Levchenko V, Mattson DL, Roman RJ, Staruschenko A. Effects of cytochrome P-450 metabolites of arachidonic acid on the epithelial sodium channel (ENaC). Am J Physiol Renal Physiol 2011; 301:F672-81. [PMID: 21697242 DOI: 10.1152/ajprenal.00597.2010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sodium reabsorption via the epithelial Na(+) channel (ENaC) in the aldosterone-sensitive distal nephron plays a central role in the regulation of body fluid volume. Previous studies have indicated that arachidonic acid (AA) and its metabolite 11,12-EET but not other regioisomers of EETs inhibit ENaC activity in the collecting duct. The goal of this study was to investigate the endogenous metabolism of AA in cultured mpkCCD(c14) principal cells and the effects of these metabolites on ENaC activity. Liquid chromatography/mass spectrometry analysis of the mpkCCD(c14) cells indicated that these cells produce prostaglandins, 8,9-EET, 11,12-EET, 14,15-EET, 5-HETE, 12/8-HETE, and 15-HETE, but not 20-HETE. Single-channel patch-clamp experiments revealed that 8,9-EET, 14,15-EET, and 11,12-EET all decrease ENaC activity. Neither 5-, 12-, nor 15-HETE had any effect on ENaC activity. Diclofenac and ibuprofen, inhibitors of cyclooxygenase, decreased transepithelial Na(+) transport in the mpkCCD(c14) cells. Inhibition of cytochrome P-450 (CYP450) with MS-PPOH activated ENaC-mediated sodium transport when cells were pretreated with AA and diclofenac. Coexpression of CYP2C8, but not CYP4A10, with ENaC in Chinese hamster ovary cells significantly decreased ENaC activity in whole-cell experiments, whereas 11,12-EET mimicked this effect. Thus both endogenously formed EETs and their exogenous application decrease ENaC activity. Downregulation of ENaC activity by overexpression of CYP2C8 was PKA dependent and was prevented by myristoylated PKI treatment. Biotinylation experiments and single-channel analysis revealed that long-term treatment with 11,12-EET and overexpression of CYP2C8 decreased the number of channels in the membrane. In contrast, the acute inhibitory effects are mediated by a decrease in the open probability of the ENaC. We conclude that 11,12-EET, 8,9-EET, and 14,15-EET are endogenously formed eicosanoids that modulate ENaC activity in the collecting duct.
Collapse
Affiliation(s)
- Tengis S Pavlov
- Dept. of Physiology, Medical College of Wisconsin, Milwaukee, 53226, USA
| | | | | | | | | | | |
Collapse
|
99
|
Fiorio Pla A, Ong HL, Cheng KT, Brossa A, Bussolati B, Lockwich T, Paria B, Munaron L, Ambudkar IS. TRPV4 mediates tumor-derived endothelial cell migration via arachidonic acid-activated actin remodeling. Oncogene 2011; 31:200-12. [PMID: 21685934 DOI: 10.1038/onc.2011.231] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Changes in intracellular calcium [Ca(2+)](i) levels control critical cytosolic and nuclear events that are involved in the initiation and progression of tumor angiogenesis in endothelial cells (ECs). Therefore, the mechanism(s) involved in agonist-induced Ca(2+)(i) signaling is a potentially important molecular target for controlling angiogenesis and tumor growth. Several studies have shown that blood vessels in tumors differ from normal vessels in their morphology, blood flow and permeability. We had previously reported a key role for arachidonic acid (AA)-mediated Ca(2+) entry in the initial stages of tumor angiogenesis in vitro. In this study we assessed the mechanism involved in AA-induced EC migration. We report that TRPV4, an AA-activated channel, is differentially expressed in EC derived from human breast carcinomas (BTEC) as compared with 'normal' EC (HMVEC). BTEC display a significant increase in TRPV4 expression, which was correlated with greater Ca(2+) entry, induced by AA or 4αPDD (a selective TRPV4 agonist) in the tumor-derived ECs. Wound-healing assays revealed a key role of TRPV4 in regulating cell migration of BTEC but not HMVEC. Knockdown of TRPV4 expression completely abolished AA-induced BTEC migration, suggesting that TRPV4 mediates the pro-angiogenic effects promoted by AA. Furthermore, pre-incubation of BTEC with AA induced actin remodeling and a subsequent increase in the surface expression of TRPV4. This was consistent with the increased plasma membrane localization of TRPV4 and higher AA-stimulated Ca(2+) entry in the migrating cells. Together, the data presented herein demonstrate that: (1) TRPV4 is differentially expressed in tumor-derived versus 'normal' EC; (2) TRPV4 has a critical role in the migration of tumor-derived but not 'normal' EC migration; and (3) AA induces actin remodeling in BTEC, resulting in a corresponding increase of TRPV4 expression in the plasma membrane. We suggest that the latter is critical for migration of EC and thus in promoting angiogenesis and tumor growth.
Collapse
Affiliation(s)
- A Fiorio Pla
- Department of Animal and Human Biology, University of Torino, Torino, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Hayama K, Suzuki Y, Inoue T, Ochiai T, Terui T, Ra C. Gold activates mast cells via calcium influx through multiple H2O2-sensitive pathways including L-type calcium channels. Free Radic Biol Med 2011; 50:1417-28. [PMID: 21376117 DOI: 10.1016/j.freeradbiomed.2011.02.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 02/17/2011] [Accepted: 02/23/2011] [Indexed: 11/23/2022]
Abstract
Heavy metals, including gold, induce severe contact hypersensitivity and autoimmune disorders, which develop through an initial Th2-independent process followed by a Th2-dependent process. It has been shown that mast cell activation plays a role in the Th2-independent process and that gold stimulates histamine release in vitro. However, the mechanisms of the gold-induced mast cell activation remain largely unclear. Here we report that gold directly activates mast cells in a Ca2+-dependent manner. HAuCl4 [Au(III)] at nontoxic concentrations (≤50 μM) induced substantial degranulation and leukotriene C4 secretion in an extracellular Ca2+-dependent manner. Au(III) induced a robust Ca2+ influx but not Ca2+ mobilization from internal stores. Au(III) also stimulated intracellular production of reactive oxygen species, including H2O2, and blockade of the production abolished the mediator release and Ca2+ influx. Au(III) induced Ca2+ influx through multiple store-independent Ca2+ channels, including Cav1.2 L-type Ca2+ channels (LTCCs) and 2-aminoethoxydiphenyl borate (2-APB)-sensitive Ca2+ channels. The 2-APB-sensitive channel seemed to mediate Au(III)-induced degranulation. Our results indicate that gold stimulates Ca2+ influx and mediator release in mast cells through multiple H2O2-sensitive Ca2+ channels including LTCCs and 2-APB-sensitive Ca2+ channels. These findings provide insight into the roles of these Ca2+ channels in the Th2-independent process of gold-induced immunological disorders.
Collapse
Affiliation(s)
- Koremasa Hayama
- Division of Molecular Cell Immunology and Allergology, Graduate School of Medical Science, Nihon University, and Department of Dermatology, Nihon University Surugadai Hospital, Tokyo 173-8610, Japan
| | | | | | | | | | | |
Collapse
|