351
|
Insights into the channel gating of P2X receptors from structures, dynamics and small molecules. Acta Pharmacol Sin 2016; 37:44-55. [PMID: 26725734 DOI: 10.1038/aps.2015.127] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/02/2015] [Indexed: 12/16/2022]
Abstract
P2X receptors, as ATP-gated non-selective trimeric ion channels, are permeable to Na(+), K(+) and Ca(2+). Comparing with other ligand-gated ion channel families, P2X receptors are distinct in their unique gating properties and pathophysiological roles, and have attracted attention as promising drug targets for a variety of diseases, such as neuropathic pain, multiple sclerosis, rheumatoid arthritis and thrombus. Several small molecule inhibitors for distinct P2X subtypes have entered into clinical trials. However, many questions regarding the gating mechanism of P2X remain unsolved. The structural determinations of P2X receptors at the resting and ATP-bound open states revealed that P2X receptor gating is a cooperative allosteric process involving multiple domains, which marks the beginning of the post-structure era of P2X research at atomic level. Here, we review the current knowledge on the structure-function relationship of P2X receptors, depict the whole picture of allosteric changes during the channel gating, and summarize the active sites that may contribute to new strategies for developing novel allosteric drugs targeting P2X receptors.
Collapse
|
352
|
Giblin JP, Comes N, Strauss O, Gasull X. Ion Channels in the Eye: Involvement in Ocular Pathologies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 104:157-231. [PMID: 27038375 DOI: 10.1016/bs.apcsb.2015.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The eye is the sensory organ of vision. There, the retina transforms photons into electrical signals that are sent to higher brain areas to produce visual sensations. In the light path to the retina, different types of cells and tissues are involved in maintaining the transparency of avascular structures like the cornea or lens, while others, like the retinal pigment epithelium, have a critical role in the maintenance of photoreceptor function by regenerating the visual pigment. Here, we have reviewed the roles of different ion channels expressed in ocular tissues (cornea, conjunctiva and neurons innervating the ocular surface, lens, retina, retinal pigment epithelium, and the inflow and outflow systems of the aqueous humor) that are involved in ocular disease pathophysiologies and those whose deletion or pharmacological modulation leads to specific diseases of the eye. These include pathologies such as retinitis pigmentosa, macular degeneration, achromatopsia, glaucoma, cataracts, dry eye, or keratoconjunctivitis among others. Several disease-associated ion channels are potential targets for pharmacological intervention or other therapeutic approaches, thus highlighting the importance of these channels in ocular physiology and pathophysiology.
Collapse
Affiliation(s)
- Jonathan P Giblin
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Comes
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Xavier Gasull
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
353
|
Deplazes E, Davies J, Bonvin AMJJ, King GF, Mark AE. Combination of Ambiguous and Unambiguous Data in the Restraint-driven Docking of Flexible Peptides with HADDOCK: The Binding of the Spider Toxin PcTx1 to the Acid Sensing Ion Channel (ASIC) 1a. J Chem Inf Model 2015; 56:127-38. [PMID: 26642380 DOI: 10.1021/acs.jcim.5b00529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Peptides that bind to ion channels have attracted much interest as potential lead molecules for the development of new drugs and insecticides. However, the structure determination of large peptide-channel complexes using experimental methods is challenging. Thus structural models are often derived from combining experimental information with restraint-driven docking approaches. Using the complex formed by the venom peptide PcTx1 and the acid sensing ion channel (ASIC) 1a as a case study, we have examined the effect of different combinations of restraints and input structures on the statistical likelihood of (a) correctly predicting the structure of the binding interface and (b) the ability to predict which residues are involved in specific pairwise peptide-channel interactions. For this, we have analyzed over 200,000 water-refined docked structures obtained with various amounts and types of restraints of the peptide-channel complex predicted using the docking program HADDOCK. We found that increasing the number of restraints or even the use of pairwise interaction data resulted in only a modest improvement in the likelihood of finding a structure within a given accuracy. This suggests that shape complementarity and the force field make a large contribution to the accuracy of the predicted structure. The results also showed that there are large variations in the accuracy of the predicted structure depending on the precise combination of residues used as restraints. Finally, we reflect on the limitations of relying on geometric criteria such as root-mean square deviations to assess the accuracy of docking procedures. We propose that in addition to currently used measures, the likelihood of finding a structure within a given level of accuracy should be also used to evaluate docking methods.
Collapse
Affiliation(s)
- Evelyne Deplazes
- Institute for Molecular Bioscience, The University of Queensland , St. Lucia, Queensland 4072, Australia.,School of Chemistry & Molecular Biosciences, The University of Queensland , St. Lucia, Queensland 4072, Australia
| | - Josephine Davies
- School of Chemistry & Molecular Biosciences, The University of Queensland , St. Lucia, Queensland 4072, Australia
| | - Alexandre M J J Bonvin
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University , 3584 CH Utrecht, The Netherlands
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland , St. Lucia, Queensland 4072, Australia
| | - Alan E Mark
- School of Chemistry & Molecular Biosciences, The University of Queensland , St. Lucia, Queensland 4072, Australia
| |
Collapse
|
354
|
Mourier G, Salinas M, Kessler P, Stura EA, Leblanc M, Tepshi L, Besson T, Diochot S, Baron A, Douguet D, Lingueglia E, Servent D. Mambalgin-1 Pain-relieving Peptide, Stepwise Solid-phase Synthesis, Crystal Structure, and Functional Domain for Acid-sensing Ion Channel 1a Inhibition. J Biol Chem 2015; 291:2616-29. [PMID: 26680001 DOI: 10.1074/jbc.m115.702373] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Indexed: 01/04/2023] Open
Abstract
Mambalgins are peptides isolated from mamba venom that specifically inhibit a set of acid-sensing ion channels (ASICs) to relieve pain. We show here the first full stepwise solid phase peptide synthesis of mambalgin-1 and confirm the biological activity of the synthetic toxin both in vitro and in vivo. We also report the determination of its three-dimensional crystal structure showing differences with previously described NMR structures. Finally, the functional domain by which the toxin inhibits ASIC1a channels was identified in its loop II and more precisely in the face containing Phe-27, Leu-32, and Leu-34 residues. Moreover, proximity between Leu-32 in mambalgin-1 and Phe-350 in rASIC1a was proposed from double mutant cycle analysis. These data provide information on the structure and on the pharmacophore for ASIC channel inhibition by mambalgins that could have therapeutic value against pain and probably other neurological disorders.
Collapse
Affiliation(s)
- Gilles Mourier
- From the Commissariat à l'Energie Atomique, iBiTecS, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif-sur-Yvette
| | - Miguel Salinas
- the CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, the Université de Nice Sophia Antipolis, and the LabEx Ion Channel Science and Therapeutics, UMR 7275, 06560 Valbonne, France
| | - Pascal Kessler
- From the Commissariat à l'Energie Atomique, iBiTecS, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif-sur-Yvette
| | - Enrico A Stura
- From the Commissariat à l'Energie Atomique, iBiTecS, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif-sur-Yvette
| | - Mathieu Leblanc
- From the Commissariat à l'Energie Atomique, iBiTecS, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif-sur-Yvette
| | - Livia Tepshi
- From the Commissariat à l'Energie Atomique, iBiTecS, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif-sur-Yvette
| | - Thomas Besson
- the CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, the Université de Nice Sophia Antipolis, and the LabEx Ion Channel Science and Therapeutics, UMR 7275, 06560 Valbonne, France
| | - Sylvie Diochot
- the CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, the Université de Nice Sophia Antipolis, and the LabEx Ion Channel Science and Therapeutics, UMR 7275, 06560 Valbonne, France
| | - Anne Baron
- the CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, the Université de Nice Sophia Antipolis, and the LabEx Ion Channel Science and Therapeutics, UMR 7275, 06560 Valbonne, France
| | - Dominique Douguet
- the CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, the Université de Nice Sophia Antipolis, and
| | - Eric Lingueglia
- the CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, the Université de Nice Sophia Antipolis, and the LabEx Ion Channel Science and Therapeutics, UMR 7275, 06560 Valbonne, France
| | - Denis Servent
- From the Commissariat à l'Energie Atomique, iBiTecS, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif-sur-Yvette,
| |
Collapse
|
355
|
Gao M, Long H, Ma W, Liao L, Yang X, Zhou Y, Shan D, Huang R, Jian F, Wang Y, Lai W. The role of periodontal ASIC3 in orofacial pain induced by experimental tooth movement in rats. Eur J Orthod 2015; 38:577-583. [DOI: 10.1093/ejo/cjv082] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
356
|
Acid-sensing ion channel (ASIC) 4 predominantly localizes to an early endosome-related organelle upon heterologous expression. Sci Rep 2015; 5:18242. [PMID: 26667795 PMCID: PMC4678866 DOI: 10.1038/srep18242] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 10/23/2015] [Indexed: 12/25/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are voltage-independent proton-gated amiloride sensitive sodium channels, belonging to the DEG/ENaC gene family. Six different ASICs have been identified (ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3, ASIC4) that are activated by a drop in extracellular pH, either as homo- or heteromers. An exception is ASIC4, which is not activated by protons as a homomer and which does not contribute to functional heteromeric ASICs. Insensitivity of ASIC4 to protons and its comparatively low sequence identity to other ASICs (45%) raises the question whether ASIC4 may have different functions than other ASICs. In this study, we therefore investigated the subcellular localization of ASIC4 in heterologous cell lines, which revealed a surprising accumulation of the channel in early endosome-related vacuoles. Moreover, we identified an unique amino-terminal motif as important for forward-trafficking from the ER/Golgi to the early endosome-related compartment. Collectively, our results show that heterologously expressed ASIC4 predominantly resides in an intracellular endosomal compartment.
Collapse
|
357
|
Munro G, Christensen JK, Erichsen HK, Dyhring T, Demnitz J, Dam E, Ahring PK. NS383 Selectively Inhibits Acid-Sensing Ion Channels Containing 1a and 3 Subunits to Reverse Inflammatory and Neuropathic Hyperalgesia in Rats. CNS Neurosci Ther 2015; 22:135-45. [PMID: 26663905 DOI: 10.1111/cns.12487] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 12/22/2022] Open
Abstract
AIMS Here, we investigate the pharmacology of NS383, a novel small molecule inhibitor of acid-sensing ion channels (ASICs). METHODS ASIC inhibition by NS383 was characterized in patch-clamp electrophysiological studies. Analgesic properties were evaluated in four rat behavioral models of pain. RESULTS NS383 inhibited H(+)-activated currents recorded from rat homomeric ASIC1a, ASIC3, and heteromeric ASIC1a+3 with IC50 values ranging from 0.61 to 2.2 μM. However, NS383 was completely inactive at homomeric ASIC2a. Heteromeric receptors containing AISC2a, such as ASIC1a+2a and ASIC2a+3, were only partially inhibited, presumably as a result of stoichiometry-dependent binding. NS383 (10-60 mg/kg, i.p.), amiloride (50-200 mg/kg, i.p.), acetaminophen (100-400 mg/kg, i.p.), and morphine (3-10 mg/kg, i.p.) all dose-dependently attenuated nocifensive behaviors in the rat formalin test, reversed pathological inflammatory hyperalgesia in complete Freund's adjuvant-inflamed rats, and reversed mechanical hypersensitivity in the chronic constriction injury model of neuropathic pain. However, in contrast to acetaminophen and morphine, motor function was unaffected by NS383 at doses at least 8-fold greater than those that were effective in pain models, whilst analgesic doses of amiloride were deemed to be toxic. CONCLUSIONS NS383 is a potent and uniquely selective inhibitor of rat ASICs containing 1a and/or 3 subunits. It is well tolerated and capable of reversing pathological painlike behaviors, presumably via peripheral actions, but possibly also via actions within central pain circuits.
Collapse
Affiliation(s)
| | | | | | - Tino Dyhring
- NeuroSearch A/S, Ballerup, Denmark.,Saniona A/S, Ballerup, Denmark
| | | | - Eva Dam
- NeuroSearch A/S, Ballerup, Denmark
| | - Philip K Ahring
- NeuroSearch A/S, Ballerup, Denmark.,Saniona A/S, Ballerup, Denmark.,Faculty of Pharmacy, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
358
|
Muley MM, Krustev E, McDougall JJ. Preclinical Assessment of Inflammatory Pain. CNS Neurosci Ther 2015; 22:88-101. [PMID: 26663896 DOI: 10.1111/cns.12486] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
While acute inflammation is a natural physiological response to tissue injury or infection, chronic inflammation is maladaptive and engenders a considerable amount of adverse pain. The chemical mediators responsible for tissue inflammation act on nociceptive nerve endings to lower neuronal excitation threshold and sensitize afferent firing rate leading to the development of allodynia and hyperalgesia, respectively. Animal models have aided in our understanding of the pathophysiological mechanisms responsible for the generation of chronic inflammatory pain and allowed us to identify and validate numerous analgesic drug candidates. Here we review some of the commonly used models of skin, joint, and gut inflammatory pain along with their relative benefits and limitations. In addition, we describe and discuss several behavioral and electrophysiological approaches used to assess the inflammatory pain in these preclinical models. Despite significant advances having been made in this area, a gap still exists between fundamental research and the implementation of these findings into a clinical setting. As such we need to characterize inherent pathophysiological pathways and develop new endpoints in these animal models to improve their predictive value of human inflammatory diseases in order to design safer and more effective analgesics.
Collapse
Affiliation(s)
- Milind M Muley
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Eugene Krustev
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
359
|
Qu ZW, Liu TT, Ren C, Gan X, Qiu CY, Ren P, Rao Z, Hu WP. 17β-Estradiol Enhances ASIC Activity in Primary Sensory Neurons to Produce Sex Difference in Acidosis-Induced Nociception. Endocrinology 2015; 156:4660-71. [PMID: 26441237 DOI: 10.1210/en.2015-1557] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sex differences have been reported in a number of pain conditions. Women are more sensitive to most types of painful stimuli than men, and estrogen plays a key role in the sex differences in pain perception. However, it is unclear whether there is a sex difference in acidosis-evoked pain. We report here that both male and female rats exhibit nociceptive behaviors in response to acetic acid, with females being more sensitive than males. Local application of exogenous 17β-estradiol (E2) exacerbated acidosis-evoked nociceptive response in male rats. E2 and estrogen receptor (ER)-α agonist 1,3,5-Tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole, but not ERβ agonist 2,3-bis(4-hydroxyphenyl)-propionitrile, replacement also reversed attenuation of the acetic acid-induced nociceptive response in ovariectomized females. Moreover, E2 can exert a rapid potentiating effect on the functional activity of acid-sensing ion channels (ASICs), which mediated the acidosis-induced events. E2 dose dependently increased the amplitude of ASIC currents with a 42.8 ± 1.6 nM of EC50. E2 shifted the concentration-response curve for proton upward with a 50.1% ± 6.2% increase of the maximal current response to proton. E2 potentiated ASIC currents via an ERα and ERK1/2 signaling pathway. E2 also altered acidosis-evoked membrane excitability of dorsal root ganglia neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acidic stimuli. E2 potentiation of the functional activity of ASICs revealed a peripheral mechanism underlying this sex difference in acetic acid-induced nociception.
Collapse
Affiliation(s)
- Zu-Wei Qu
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| | - Ting-Ting Liu
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| | - Cuixia Ren
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| | - Xiong Gan
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| | - Chun-Yu Qiu
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| | - Ping Ren
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| | - Zhiguo Rao
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| | - Wang-Ping Hu
- Institute of Ion Channels (Z.-W.Q., T.-T.L., C.R., X.G., C.-Y.Q., P.R., W.-P.H.), Department of Pharmacology, Hubei University of Science and Technology, Xianning 437100, Hubei, People's Republic of China; and Department of Oncology (Z.R.), Wuhan of General Hospital of Guangzhou, People's Liberation Army, Wuhan 430070, People's Republic of China
| |
Collapse
|
360
|
Mazzocchi N, De Ceglia R, Mazza D, Forti L, Muzio L, Menegon A. Fluorescence-Based Automated Screening Assay for the Study of the pH-Sensitive Channel ASIC1a. ACTA ACUST UNITED AC 2015; 21:372-80. [DOI: 10.1177/1087057115617455] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/26/2015] [Indexed: 11/16/2022]
Abstract
Acid-sensing ion channel 1a (ASIC1a) is involved in several pathologies, including neurodegenerative and neuroinflammatory disorders, stroke, epilepsy, and inflammatory pain. ASIC1a has been the subject of intense drug discovery programs devoted to the development of new pharmacological tools for its modulation. However, these efforts to generate new compounds have faced the lack of an efficient screening procedure. In the past decades, improvements in screening technologies and fluorescent sensors for the study of ion channels have provided new opportunities in this field. Unfortunately, ASIC1a is mainly a Na+ permeable channel and undergoes desensitization after its activation, two features that make the use of the available screening procedures problematic. We propose here a novel screening approach for the study of ASIC1a activity in full automation. Our method is based on the stimulation of ASIC1a-expressing cells by protons and the use of electrochromic fluorescent voltage sensors as a readout of ion channel activation. This method will prove to be useful for drug screening programs aimed at ASIC1a modulation.
Collapse
Affiliation(s)
- Nausicaa Mazzocchi
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, Milan, Italy
| | - Roberta De Ceglia
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Davide Mazza
- Advanced Fluorescence Microscopy and Nanoscopy Research Unit, Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Lia Forti
- Center for Neuroscience and Dept. of Theoretical and Applied Sciences (DiSTA), Biomedical Division, University of Insubria, Busto Arsizio (VA), Italy
| | - Luca Muzio
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Menegon
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
361
|
The RS685012 Polymorphism of ACCN2, the Human Ortholog of Murine Acid-Sensing Ion Channel (ASIC1) Gene, is Highly Represented in Patients with Panic Disorder. Neuromolecular Med 2015; 18:91-8. [PMID: 26589317 DOI: 10.1007/s12017-015-8380-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/11/2015] [Indexed: 10/22/2022]
Abstract
Panic disorder (PD) is a disabling anxiety disorder that is characterized by unexpected, recurrent panic attacks, associated with fear of dying and worrying about possible future attacks or other behavioral changes as a consequence of the attacks. The acid-sensing ion channels (ASICs) are a family of proton-sensing channels expressed throughout the nervous system. Their activity is linked to a variety of behaviors including fear, anxiety, pain, depression, learning, and memory. The human analog of ASIC1a is the amiloride-sensitive cation channel 2 (ACCN2). Adenosine A2A receptors are suggested to play an important role in different brain circuits and pathways involved in anxiety reactions. In this work we aimed to evaluate the distribution of ACCN2 rs685012 and ADORA2A rs2298383 polymorphisms in PD patients compared with healthy subjects. We found no association between ADORA2A polymorphism and PD. Instead, the C mutated allele for ACCN2 rs685012 polymorphism was significantly more frequent in patients than in controls. On the contrary, the TT homozygous wild-type genotype and also the ACCN2 TT/ADORA2A CT diplotype were significantly more represented in controls. These results are suggestive for a role of ACCN2 rs685012 polymorphism in PD development in Caucasian people.
Collapse
|
362
|
Prager EM, Bergstrom HC, Wynn GH, Braga MFM. The basolateral amygdala γ-aminobutyric acidergic system in health and disease. J Neurosci Res 2015; 94:548-67. [PMID: 26586374 DOI: 10.1002/jnr.23690] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/01/2015] [Accepted: 10/18/2015] [Indexed: 01/13/2023]
Abstract
The brain comprises an excitatory/inhibitory neuronal network that maintains a finely tuned balance of activity critical for normal functioning. Excitatory activity in the basolateral amygdala (BLA), a brain region that plays a central role in emotion and motivational processing, is tightly regulated by a relatively small population of γ-aminobutyric acid (GABA) inhibitory neurons. Disruption in GABAergic inhibition in the BLA can occur when there is a loss of local GABAergic interneurons, an alteration in GABAA receptor activation, or a dysregulation of mechanisms that modulate BLA GABAergic inhibition. Disruptions in GABAergic control of the BLA emerge during development, in aging populations, or after trauma, ultimately resulting in hyperexcitability. BLA hyperexcitability manifests behaviorally as an increase in anxiety, emotional dysregulation, or development of seizure activity. This Review discusses the anatomy, development, and physiology of the GABAergic system in the BLA and circuits that modulate GABAergic inhibition, including the dopaminergic, serotonergic, noradrenergic, and cholinergic systems. We highlight how alterations in various neurotransmitter receptors, including the acid-sensing ion channel 1a, cannabinoid receptor 1, and glutamate receptor subtypes, expressed on BLA interneurons, modulate GABAergic transmission and how defects of these systems affect inhibitory tonus within the BLA. Finally, we discuss alterations in the BLA GABAergic system in neurodevelopmental (autism/fragile X syndrome) and neurodegenerative (Alzheimer's disease) diseases and after the development of epilepsy, anxiety, and traumatic brain injury. A more complete understanding of the intrinsic excitatory/inhibitory circuit balance of the amygdala and how imbalances in inhibitory control contribute to excessive BLA excitability will guide the development of novel therapeutic approaches in neuropsychiatric diseases.
Collapse
Affiliation(s)
- Eric M Prager
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services, University of the Health Sciences, Bethesda, Maryland
| | | | - Gary H Wynn
- Center for the Study of Traumatic Stress, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Program in Neuroscience, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services, University of the Health Sciences, Bethesda, Maryland.,Center for the Study of Traumatic Stress, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Program in Neuroscience, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
363
|
Wang Y, O’Bryant Z, Wang H, Huang Y. Regulating Factors in Acid-Sensing Ion Channel 1a Function. Neurochem Res 2015; 41:631-45. [DOI: 10.1007/s11064-015-1768-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/04/2015] [Accepted: 11/08/2015] [Indexed: 12/11/2022]
|
364
|
Ren C, Gan X, Wu J, Qiu CY, Hu WP. Enhancement of acid-sensing ion channel activity by metabotropic P2Y UTP receptors in primary sensory neurons. Purinergic Signal 2015; 12:69-78. [PMID: 26538146 DOI: 10.1007/s11302-015-9479-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/23/2015] [Indexed: 12/16/2022] Open
Abstract
Peripheral purinergic signaling plays an important role in nociception. Increasing evidence suggests that metabotropic P2Y receptors are also involved, but little is known about the underlying mechanism. Herein, we report that selective P2Y receptor agonist uridine 5'-triphosphate (UTP) can exert an enhancing effect on the functional activity of acid-sensing ion channels (ASICs), key sensors for extracellular protons, in rat dorsal root ganglia (DRG) neurons. First, UTP dose-dependently increased the amplitude of ASIC currents. UTP also shifted the concentration-response curve for proton upwards, with a 56.6 ± 6.4% increase of the maximal current response to proton. Second, UTP potentiation of proton-gated currents can be mimicked by adenosine 5'-triphosphate (ATP), but not by P2Y1 receptor agonist ADP. Potentiation of UTP was blocked by P2Y receptor antagonist suramin and by inhibition of intracellular G protein, phospholipase C (PLC), protein kinase C (PKC), or protein interacting with C-kinase 1 (PICK1) signaling. Third, UTP altered acidosis-evoked membrane excitability of DRG neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acid stimuli. Finally, UTP dose-dependently exacerbated nociceptive responses to injection of acetic acid in rats. These results suggest that UTP enhanced ASIC-mediated currents and nociceptive responses, which reveal a novel peripheral mechanism underlying UTP-sensitive P2Y2 receptor involvement in hyperalgesia by sensitizing ASICs in primary sensory neurons.
Collapse
Affiliation(s)
- Cuixia Ren
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Xiong Gan
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Jing Wu
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Chun-Yu Qiu
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Wang-Ping Hu
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China.
| |
Collapse
|
365
|
Wang YZ, Wang JJ, Huang Y, Liu F, Zeng WZ, Li Y, Xiong ZG, Zhu MX, Xu TL. Tissue acidosis induces neuronal necroptosis via ASIC1a channel independent of its ionic conduction. eLife 2015; 4. [PMID: 26523449 PMCID: PMC4629285 DOI: 10.7554/elife.05682] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 09/23/2015] [Indexed: 12/15/2022] Open
Abstract
Acidotoxicity is common among neurological disorders, such as ischemic stroke. Traditionally, Ca2+ influx via homomeric acid-sensing ion channel 1a (ASIC1a) was considered to be the leading cause of ischemic acidotoxicity. Here we show that extracellular protons trigger a novel form of neuronal necroptosis via ASIC1a, but independent of its ion-conducting function. We identified serine/threonine kinase receptor interaction protein 1 (RIP1) as a critical component of this form of neuronal necroptosis. Acid stimulation recruits RIP1 to the ASIC1a C-terminus, causing RIP1 phosphorylation and subsequent neuronal death. In a mouse model of focal ischemia, middle cerebral artery occlusion causes ASIC1a-RIP1 association and RIP1 phosphorylation in affected brain areas. Deletion of the Asic1a gene significantly prevents RIP1 phosphorylation and brain damage, suggesting ASIC1a-mediated RIP1 activation has an important role in ischemic neuronal injury. Our findings indicate that extracellular protons function as a novel endogenous ligand that triggers neuronal necroptosis during ischemia via ASIC1a independent of its channel function. DOI:http://dx.doi.org/10.7554/eLife.05682.001 What happens in the minutes and hours after a stroke can determine how much brain damage occurs. In some types of stroke, a blood clot cuts off the blood supply to part of the brain, depriving the brain cells of oxygen and other nutrients, including glucose. One of the consequences is that the blood-starved brain becomes more acidic, which triggers cell death. Protecting brain cells from acidity-induced death could therefore reduce the damage caused by a stroke, and may also be an effective treatment for other brain disorders that involve increased brain acidity, like multiple sclerosis and Huntington's disease. To create such treatments, researchers must first understand how increased acidity in the brain triggers cell death. A protein called the acid-sensing ion channel 1a (ASIC1a) is thought to contribute to acid-induced cell death by allowing calcium to flow into cells. However, this increased flow of calcium occurs only briefly (for seconds) in response to increased acidity, which cannot explain why the severity of cell death strongly depends on the length of increased brain acidity that lasts for hours during stroke. Wang, Wang et al. now show that while ASIC1a is essential for acid-induced brain cell death, this is not because it allows calcium to enter cells. Instead, when acid levels increase, a protein called RIP1 comes to bind to one end of the ASIC1a protein. This causes the addition of a phosphate tag to RIP1, an important cellular process well known to cause the cell to die. Wang, Wang et al. found that in mice genetically engineered to lack ASIC1a, the phosphate tag is not added to RIP1, and the brain cells survive the increased acidity caused by stroke. This suggests that preventing ASIC1a and RIP1 from interacting could be a new way to protect brain cells from the increased acidity caused by brain diseases. DOI:http://dx.doi.org/10.7554/eLife.05682.002
Collapse
Affiliation(s)
- Yi-Zhi Wang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Jing Wang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Huang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Liu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Zheng Zeng
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Li
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, United States
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, United States
| | - Tian-Le Xu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
366
|
Abstract
Chronic widespread pain is a serious medical problem, yet the mechanisms of nociception and pain are poorly understood. Using a reserpine-induced pain model originally reported as a putative animal model for fibromyalgia, this study was undertaken to examine the following: (1) expression of several ion channels responsible for pain, mechanotransduction, and generation/propagation of action potentials in the dorsal root ganglion (DRG), (2) activities of peripheral nociceptive afferents, and (3) alterations in spinal microglial cells. A significant increase in mRNA expression of the acid-sensing ion channel (ASIC)-3 was detected in the DRG, and the behavioral mechanical hyperalgesia was significantly reversed by subcutaneous injection of APETx2, a selective blocker of ASIC3. Single-fiber recordings in vitro revealed facilitated mechanical responses of mechanoresponsive C-fibers both in the skin and muscle although the proportion of mechanoresponsive C-nociceptors was paradoxically decreased. In the spinal dorsal horn, microglial cells labeled with Iba1 immunoreactivity was activated, especially in laminae I-II where the nociceptive input is mainly processed compared with the other laminae. The activated microglia and behavioral hyperalgesia were significantly tranquilized by intraperitoneal injection of minocycline. These results suggest that the increase in ASIC3 in the DRG facilitated mechanical response of the remaining C-nociceptors and that activated spinal microglia may direct to intensify pain in this model. Pain may be further amplified by reserpine-induced dysfunction of the descending pain inhibitory system and by the decrease in peripheral drive to this system resulting from a reduced proportion of mechanoresponsive C-nociceptors.
Collapse
|
367
|
Niculescu AB, Levey DF, Phalen PL, Le-Niculescu H, Dainton HD, Jain N, Belanger E, James A, George S, Weber H, Graham DL, Schweitzer R, Ladd TB, Learman R, Niculescu EM, Vanipenta NP, Khan FN, Mullen J, Shankar G, Cook S, Humbert C, Ballew A, Yard M, Gelbart T, Shekhar A, Schork NJ, Kurian SM, Sandusky GE, Salomon DR. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry 2015; 20:1266-85. [PMID: 26283638 PMCID: PMC4759104 DOI: 10.1038/mp.2015.112] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/25/2015] [Accepted: 06/29/2015] [Indexed: 12/26/2022]
Abstract
Worldwide, one person dies every 40 seconds by suicide, a potentially preventable tragedy. A limiting step in our ability to intervene is the lack of objective, reliable predictors. We have previously provided proof of principle for the use of blood gene expression biomarkers to predict future hospitalizations due to suicidality, in male bipolar disorder participants. We now generalize the discovery, prioritization, validation, and testing of such markers across major psychiatric disorders (bipolar disorder, major depressive disorder, schizoaffective disorder, and schizophrenia) in male participants, to understand commonalities and differences. We used a powerful within-participant discovery approach to identify genes that change in expression between no suicidal ideation and high suicidal ideation states (n=37 participants out of a cohort of 217 psychiatric participants followed longitudinally). We then used a convergent functional genomics (CFG) approach with existing prior evidence in the field to prioritize the candidate biomarkers identified in the discovery step. Next, we validated the top biomarkers from the prioritization step for relevance to suicidal behavior, in a demographically matched cohort of suicide completers from the coroner's office (n=26). The biomarkers for suicidal ideation only are enriched for genes involved in neuronal connectivity and schizophrenia, the biomarkers also validated for suicidal behavior are enriched for genes involved in neuronal activity and mood. The 76 biomarkers that survived Bonferroni correction after validation for suicidal behavior map to biological pathways involved in immune and inflammatory response, mTOR signaling and growth factor regulation. mTOR signaling is necessary for the effects of the rapid-acting antidepressant agent ketamine, providing a novel biological rationale for its possible use in treating acute suicidality. Similarly, MAOB, a target of antidepressant inhibitors, was one of the increased biomarkers for suicidality. We also identified other potential therapeutic targets or biomarkers for drugs known to mitigate suicidality, such as omega-3 fatty acids, lithium and clozapine. Overall, 14% of the top candidate biomarkers also had evidence for involvement in psychological stress response, and 19% for involvement in programmed cell death/cellular suicide (apoptosis). It may be that in the face of adversity (stress), death mechanisms are turned on at a cellular (apoptosis) and organismal level. Finally, we tested the top increased and decreased biomarkers from the discovery for suicidal ideation (CADM1, CLIP4, DTNA, KIF2C), prioritization with CFG for prior evidence (SAT1, SKA2, SLC4A4), and validation for behavior in suicide completers (IL6, MBP, JUN, KLHDC3) steps in a completely independent test cohort of psychiatric participants for prediction of suicidal ideation (n=108), and in a future follow-up cohort of psychiatric participants (n=157) for prediction of psychiatric hospitalizations due to suicidality. The best individual biomarker across psychiatric diagnoses for predicting suicidal ideation was SLC4A4, with a receiver operating characteristic (ROC) area under the curve (AUC) of 72%. For bipolar disorder in particular, SLC4A4 predicted suicidal ideation with an AUC of 93%, and future hospitalizations with an AUC of 70%. SLC4A4 is involved in brain extracellular space pH regulation. Brain pH has been implicated in the pathophysiology of acute panic attacks. We also describe two new clinical information apps, one for affective state (simplified affective state scale, SASS) and one for suicide risk factors (Convergent Functional Information for Suicide, CFI-S), and how well they predict suicidal ideation across psychiatric diagnoses (AUC of 85% for SASS, AUC of 89% for CFI-S). We hypothesized a priori, based on our previous work, that the integration of the top biomarkers and the clinical information into a universal predictive measure (UP-Suicide) would show broad-spectrum predictive ability across psychiatric diagnoses. Indeed, the UP-Suicide was able to predict suicidal ideation across psychiatric diagnoses with an AUC of 92%. For bipolar disorder, it predicted suicidal ideation with an AUC of 98%, and future hospitalizations with an AUC of 94%. Of note, both types of tests we developed (blood biomarkers and clinical information apps) do not require asking the individual assessed if they have thoughts of suicide, as individuals who are truly suicidal often do not share that information with clinicians. We propose that the widespread use of such risk prediction tests as part of routine or targeted healthcare assessments will lead to early disease interception followed by preventive lifestyle modifications and proactive treatment.
Collapse
Affiliation(s)
- A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - D F Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - P L Phalen
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H D Dainton
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N Jain
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E Belanger
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - A James
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - S George
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - H Weber
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - D L Graham
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Schweitzer
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - T B Ladd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Learman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E M Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N P Vanipenta
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - F N Khan
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J Mullen
- Advanced Biomedical IT Core, Indiana University School of Medicine, Indianapolis, IN, USA
| | - G Shankar
- Advanced Biomedical IT Core, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Cook
- Marion County Coroner's Office, Indianapolis, IN, USA
| | - C Humbert
- Marion County Coroner's Office, Indianapolis, IN, USA
| | - A Ballew
- Marion County Coroner's Office, Indianapolis, IN, USA
| | - M Yard
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - T Gelbart
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N J Schork
- J. Craig Venter Institute, La Jolla, CA, USA
| | - S M Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - G E Sandusky
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D R Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
368
|
Liang JJ, Huang LF, Chen XM, Pan SQ, Lu ZN, Xiao ZM. Amiloride suppresses pilocarpine-induced seizures via ASICs other than NHE in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:14507-14513. [PMID: 26823770 PMCID: PMC4713556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/20/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND AND PURPOSE Although recent studies have indicated that acid-sensing ion channels (ASICs) may play an important role in suppressing status epilepticus (SE) in rats, the precise mechanism is unclear. We attempted to investigate the antiepileptic effect of amiloride in SE rats and its mechanism. METHODS Rats with seizures induced by Li-pilocarpine were randomly divided into four groups, phosphate buffer saline (PBS) group, amiloride group, levetiracetam group and acidic liquid group, respectively. The electroencephalogram (EEG) of each group was recorded. Then rats treated with different drugs (2 h after amiloride or PBS injection or 1 h after PBS injection) and a normal control group was selected for reverse transcription-polymerase chain reaction (RT-PCR). The expression of ASIC1a, ASIC3 and sodium-hydrogen exchanger (NHE) in each group was detected. RESULTS Amiloride reduced the frequency of discharge in 60~90 min after injection significantly. In acidic liquid group, the epileptic discharge was increased in 0~30 min. Moreover, the expression of ASIC1a, ASIC3 and NHE was obviously increased in the SE groups. Compared with SE groups, the expression of ASIC1a and ASIC3 mRNA in amiloride group decreased significantly. While NHE mRNA expression in the SE groups showed no significant difference. CONCLUSION Amiloride inhibited pilocarpine-induced SE and the anti-epileptic mechanism was associated with deactivation of the ASIC1a and ASIC3 instead of NHE in rats.
Collapse
Affiliation(s)
- Jing-Jing Liang
- Department of Neurology, Renmin Hospital of Wuhan University Wuhan 430060, Hubei Province, P. R. C
| | - Li-Fang Huang
- Department of Neurology, Renmin Hospital of Wuhan University Wuhan 430060, Hubei Province, P. R. C
| | - Xu-Ming Chen
- Department of Neurology, Renmin Hospital of Wuhan University Wuhan 430060, Hubei Province, P. R. C
| | - Song-Qing Pan
- Department of Neurology, Renmin Hospital of Wuhan University Wuhan 430060, Hubei Province, P. R. C
| | - Zu-Neng Lu
- Department of Neurology, Renmin Hospital of Wuhan University Wuhan 430060, Hubei Province, P. R. C
| | - Zhe-Man Xiao
- Department of Neurology, Renmin Hospital of Wuhan University Wuhan 430060, Hubei Province, P. R. C
| |
Collapse
|
369
|
Quantifying Preferences and Responsiveness of Marine Zooplankton to Changing Environmental Conditions using Microfluidics. PLoS One 2015; 10:e0140553. [PMID: 26517120 PMCID: PMC4627805 DOI: 10.1371/journal.pone.0140553] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 09/28/2015] [Indexed: 01/31/2023] Open
Abstract
Global environmental change significantly affects marine species composition. However, analyzing the impact of these changes on marine zooplankton communities was so far mostly limited to assessing lethal doses through mortality assays and hence did not allow a direct assessment of the preferred conditions, or preferendum. Here, we use a microfluidic device to characterize individual behavior of actively swimming zooplankton, and to quantitatively determine their ecological preferendum. For the annelid zooplankton model Platynereis dumerilii we observe a broader pH preferendum than for the copepod Euterpina acutifrons, and reveal previously unrecognized sub-populations with different pH preferenda. For Platynereis, the minimum concentration difference required to elicit a response (responsiveness) is ~1 μM for H+ and ~13.7 mM for NaCl. Furthermore, using laser ablations we show that olfactomedin-expressing sensory cells mediate chemical responsiveness in the Platynereis foregut. Taken together, our microfluidic approach allows precise assessment and functional understanding of environmental perception on planktonic behaviour.
Collapse
|
370
|
Mari Y, Katnik C, Cuevas J. σ-1 Receptor Inhibition of ASIC1a Channels is Dependent on a Pertussis Toxin-Sensitive G-Protein and an AKAP150/Calcineurin Complex. Neurochem Res 2015; 40:2055-67. [PMID: 24925261 DOI: 10.1007/s11064-014-1324-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/17/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
ASIC1a channels play a major role in various pathophysiological conditions including depression, anxiety, epilepsy, and neurodegeneration following ischemic stroke. Sigma-1 (σ-1) receptor stimulation depresses the activity of ASIC1a channels in cortical neurons, but the mechanism(s) by which σ-1 receptors exert their influence on ASIC1a remains unknown. Experiments were undertaken to elucidate the signaling cascade linking σ-1 receptors to ASIC1a channels. Immunohistochemical studies showed that σ-1 receptors, ASIC1a and A-kinase anchoring peptide 150 colocalize in the plasma membrane of the cell body and processes of cortical neurons. Fluorometric Ca(2+) imaging experiments showed that disruption of the macromolecular complexes containing AKAP150 diminished the effects of the σ-1 on ASIC1a, as did application of the calcineurin inhibitors, cyclosporin A and FK-506. Moreover, whole-cell patch clamp experiments showed that σ-1 receptors were less effective at decreasing ASIC1a-mediated currents in the presence of the VIVIT peptide, which binds to calcineurin and prevents cellular effects dependent on AKAP150/calcineurin interaction. The coupling of σ-1 to ASIC1a was also disrupted by preincubation of the neurons in the G-protein inhibitor, pertussis toxin (PTX). Taken together, our data reveal that σ-1 receptor block of ASIC1a function is dependent on activation of a PTX-sensitive G-protein and stimulation of AKAP150 bound calcineurin.
Collapse
Affiliation(s)
- Yelenis Mari
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC-9, Tampa, FL, 33612-4799, USA
| | - Christopher Katnik
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC-9, Tampa, FL, 33612-4799, USA
| | - Javier Cuevas
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC-9, Tampa, FL, 33612-4799, USA.
| |
Collapse
|
371
|
Ovalle-Magallanes B, Déciga-Campos M, Mata R. Antinociceptive and hypoglycaemic evaluation of Conyza filaginoides (D.C.) Hieron Asteraceae. J Pharm Pharmacol 2015; 67:1733-43. [DOI: 10.1111/jphp.12477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/19/2015] [Indexed: 12/31/2022]
Abstract
Abstract
Objectives
This work was undertaken to assess the antinociceptive and hypoglycaemic properties of a quantified extract of Conyza filaginoides (CFOE), as well as the antinociceptive potential of rutin, the main active compound of the plant, in normoglycaemic and/or hyperglycaemic mice (nicotinamide-streptozotocin, NA-STZ).
Methods
The antinociceptive effect of CFOE was evaluated using the writhing, hotplate and formalin tests in mice. Rutin was also examined with the formalin test. In addition, the antihyperalgesic effect of CFOE was evaluated in hyperglycaemic mice. The hypoglycaemic effect of CFOE was tested using an acute hypoglycaemic assay, and oral glucose and sucrose tests in normoglycaemic and hyperglycaemic mice.
Key findings
CFOE showed antinociceptive effect when tested in normoglycaemic mice in the writhing and hotplate tests (31.6–316 mg/kg). CFOE was also active in both normoglycaemic and hyperglycaemic mice in the formalin test (10–100 μg/paw) revealing its antihyperalgesic property. Rutin reduced the nociceptive behaviour in the formalin test; its mechanism of action seems to involve GABAergic and opioid pathways. CFOE possessed noted hypoglycaemic and antihyperglycaemic effects in normoglycaemic and hyperglycaemic mice (31.6–316 mg/kg).
Conclusions
The antinociceptive, antihyperalgesic and hypoglycaemic effects of C. filaginoides found in this study support the contemporary uses of the plant in Mexican folk medicine.
Collapse
Affiliation(s)
| | - Myrna Déciga-Campos
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México D.F., México
| | - Rachel Mata
- Facultad de Química, Universidad Nacional Autónoma de México, México D.F., México
| |
Collapse
|
372
|
Gibbons DD, Kutschke WJ, Weiss RM, Benson CJ. Heart failure induces changes in acid-sensing ion channels in sensory neurons innervating skeletal muscle. J Physiol 2015; 593:4575-87. [PMID: 26314284 DOI: 10.1113/jp270690] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/13/2015] [Indexed: 12/19/2022] Open
Abstract
Heart failure is associated with diminished exercise capacity, which is driven, in part, by alterations in exercise-induced autonomic reflexes triggered by skeletal muscle sensory neurons (afferents). These overactive reflexes may also contribute to the chronic state of sympathetic excitation, which is a major contributor to the morbidity and mortality of heart failure. Acid-sensing ion channels (ASICs) are highly expressed in muscle afferents where they sense metabolic changes associated with ischaemia and exercise, and contribute to the metabolic component of these reflexes. Therefore, we tested if ASICs within muscle afferents are altered in heart failure. We used whole-cell patch clamp to study the electrophysiological properties of acid-evoked currents in isolated, labelled muscle afferent neurons from control and heart failure (induced by myocardial infarction) mice. We found that the percentage of muscle afferents that displayed ASIC-like currents, the current amplitudes, and the pH dose-response relationships were not altered in mice with heart failure. On the other hand, the biophysical properties of ASIC-like currents were significantly different in a subpopulation of cells (40%) from heart failure mice. This population displayed diminished pH sensitivity, altered desensitization kinetics, and very fast recovery from desensitization. These unique properties define these channels within this subpopulation of muscle afferents as being heteromeric channels composed of ASIC2a and -3 subunits. Heart failure induced a shift in the subunit composition of ASICs within muscle afferents, which significantly altered their pH sensing characteristics. These results might, in part, contribute to the changes in exercise-mediated reflexes that are associated with heart failure.
Collapse
Affiliation(s)
- David D Gibbons
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,The Department of Veterans Medical Center, Iowa City, IA, 52242, USA
| | - William J Kutschke
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Robert M Weiss
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Christopher J Benson
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,The Department of Veterans Medical Center, Iowa City, IA, 52242, USA
| |
Collapse
|
373
|
Saez NJ, Deplazes E, Cristofori-Armstrong B, Chassagnon IR, Lin X, Mobli M, Mark AE, Rash LD, King GF. Molecular dynamics and functional studies define a hot spot of crystal contacts essential for PcTx1 inhibition of acid-sensing ion channel 1a. Br J Pharmacol 2015; 172:4985-95. [PMID: 26248594 DOI: 10.1111/bph.13267] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/29/2015] [Accepted: 07/13/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The spider-venom peptide PcTx1 is the most potent and selective inhibitor of acid-sensing ion channel (ASIC) 1a. It has centrally acting analgesic activity and is neuroprotective in rodent models of ischaemic stroke. Understanding the molecular details of the PcTx1 : ASIC1a interaction should facilitate development of therapeutically useful ASIC1a modulators. Previously, we showed that several key pharmacophore residues of PcTx1 reside in a dynamic β-hairpin loop; conclusions confirmed by recent crystal structures of the complex formed between PcTx1 and chicken ASIC1 (cASIC1). Numerous peptide : channel contacts were observed in these crystal structures, but it remains unclear which of these are functionally important. EXPERIMENTAL APPROACH We combined molecular dynamics (MD) simulations of the PcTx1 : cASIC1 complex with mutagenesis of PcTx1 and rat ASIC1a. KEY RESULTS Crystal structures of the PcTx1 : cASIC1 complex indicated that 15 PcTx1 residues form a total of 57 pairwise intermolecular contacts (<5 Å) with 32 channel residues. MD simulations, however, suggested that about half of these interactions do not persist in solution. Mutation to alanine of only eight of 15 PcTx1 contact residues substantially altered ASIC1a inhibition by PcTx1. Our data reveal that many of the peptide-channel interactions observed in the PcTx1 : cASIC1 crystal structures are not important for PcTx1 inhibition of rat ASIC1a. CONCLUSIONS AND IMPLICATIONS We identified the atomic interactions that are critical for PcTx1 inhibition of ASIC1a. Our data highlight the value of combining structural information, MD and functional experiments to obtain detailed insight into the molecular basis of protein : protein interactions.
Collapse
Affiliation(s)
- Natalie J Saez
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Evelyne Deplazes
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Ben Cristofori-Armstrong
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Irène R Chassagnon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | | | - Mehdi Mobli
- Centre for Advanced Imaging, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Alan E Mark
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Lachlan D Rash
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
374
|
Zeng WZ, Liu DS, Liu L, She L, Wu LJ, Xu TL. Activation of acid-sensing ion channels by localized proton transient reveals their role in proton signaling. Sci Rep 2015; 5:14125. [PMID: 26370138 PMCID: PMC4569896 DOI: 10.1038/srep14125] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/18/2015] [Indexed: 12/31/2022] Open
Abstract
Extracellular transients of pH alterations likely mediate signal transduction in the nervous system. Neuronal acid-sensing ion channels (ASICs) act as sensors for extracellular protons, but the mechanism underlying ASIC activation remains largely unknown. Here, we show that, following activation of a light-activated proton pump, Archaerhodopsin-3 (Arch), proton transients induced ASIC currents in both neurons and HEK293T cells co-expressing ASIC1a channels. Using chimera proteins that bridge Arch and ASIC1a by a glycine/serine linker, we found that successful coupling occurred within 15 nm distance. Furthermore, two-cell sniffer patch recording revealed that regulated release of protons through either Arch or voltage-gated proton channel Hv1 activated neighbouring cells expressing ASIC1a channels. Finally, computational modelling predicted the peak proton concentration at the intercellular interface to be at pH 6.7, which is acidic enough to activate ASICs in vivo. Our results highlight the pathophysiological role of proton signalling in the nervous system.
Collapse
Affiliation(s)
- Wei-Zheng Zeng
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Di-Shi Liu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lu Liu
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang She
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Tian-Le Xu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
375
|
Luo J, Feng J, Liu S, Walters ET, Hu H. Molecular and cellular mechanisms that initiate pain and itch. Cell Mol Life Sci 2015; 72:3201-23. [PMID: 25894692 PMCID: PMC4534341 DOI: 10.1007/s00018-015-1904-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/20/2015] [Accepted: 04/07/2015] [Indexed: 12/17/2022]
Abstract
Somatosensory neurons mediate our sense of touch. They are critically involved in transducing pain and itch sensations under physiological and pathological conditions, along with other skin-resident cells. Tissue damage and inflammation can produce a localized or systemic sensitization of our senses of pain and itch, which can facilitate our detection of threats in the environment. Although acute pain and itch protect us from further damage, persistent pain and itch are debilitating. Recent exciting discoveries have significantly advanced our knowledge of the roles of membrane-bound G protein-coupled receptors and ion channels in the encoding of information leading to pain and itch sensations. This review focuses on molecular and cellular events that are important in early stages of the biological processing that culminates in our senses of pain and itch.
Collapse
Affiliation(s)
- Jialie Luo
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA
| | | | | | | | | |
Collapse
|
376
|
Quintana P, Soto D, Poirot O, Zonouzi M, Kellenberger S, Muller D, Chrast R, Cull-Candy SG. Acid-sensing ion channel 1a drives AMPA receptor plasticity following ischaemia and acidosis in hippocampal CA1 neurons. J Physiol 2015; 593:4373-86. [PMID: 26174503 PMCID: PMC4594240 DOI: 10.1113/jp270701] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 07/08/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The hippocampal CA1 region is highly vulnerable to ischaemic stroke. Two forms of AMPA receptor (AMPAR) plasticity - an anoxic form of long-term potentiation and a delayed increase in Ca(2+) -permeable (CP) AMPARs - contribute to this susceptibility by increasing excitotoxicity. In CA1, the acid-sensing ion channel 1a (ASIC1a) is known to facilitate LTP and contribute to ischaemic acidotoxicity. We have examined the role of ASIC1a in AMPAR ischaemic plasticity in organotypic hippocampal slice cultures exposed to oxygen glucose deprivation (a model of ischaemic stroke), and in hippocampal pyramidal neuron cultures exposed to acidosis. We find that ASIC1a activation promotes both forms of AMPAR plasticity and that neuroprotection, by inhibiting ASIC1a, circumvents any further benefit of blocking CP-AMPARs. Our observations establish a new interaction between acidotoxicity and excitotoxicity, and provide insight into the role of ASIC1a and CP-AMPARs in neurodegeneration. Specifically, we propose that ASIC1a activation drives certain post-ischaemic forms of CP-AMPAR plasticity. ABSTRACT The CA1 region of the hippocampus is particularly vulnerable to ischaemic damage. While NMDA receptors play a major role in excitotoxicity, it is thought to be exacerbated in this region by two forms of post-ischaemic AMPA receptor (AMPAR) plasticity - namely, anoxic long-term potentiation (a-LTP), and a delayed increase in the prevalence of Ca(2+) -permeable GluA2-lacking AMPARs (CP-AMPARs). The acid-sensing ion channel 1a (ASIC1a), which is expressed in CA1 pyramidal neurons, is also known to contribute to post-ischaemic neuronal death and to physiologically induced LTP. This raises the question does ASIC1a activation drive the post-ischaemic forms of AMPAR plasticity in CA1 pyramidal neurons? We have tested this by examining organotypic hippocampal slice cultures (OHSCs) exposed to oxygen glucose deprivation (OGD), and dissociated cultures of hippocampal pyramidal neurons (HPNs) exposed to low pH (acidosis). We find that both a-LTP and the delayed increase in the prevalence of CP-AMPARs are dependent on ASIC1a activation during ischaemia. Indeed, acidosis alone is sufficient to induce the increase in CP-AMPARs. We also find that inhibition of ASIC1a channels circumvents any potential neuroprotective benefit arising from block of CP-AMPARs. By demonstrating that ASIC1a activation contributes to post-ischaemic AMPAR plasticity, our results identify a functional interaction between acidotoxicity and excitotoxicity in hippocampal CA1 cells, and provide insight into the role of ASIC1a and CP-AMPARs as potential drug targets for neuroprotection. We thus propose that ASIC1a activation can drive certain forms of CP-AMPAR plasticity, and that inhibiting ASIC1a affords neuroprotection.
Collapse
Affiliation(s)
- Patrice Quintana
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva 4, Switzerland.,Department of Medical Genetics, University of Lausanne, 1005, Lausanne, Switzerland
| | - David Soto
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Olivier Poirot
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, WC1E 6BT, UK.,Department of Pharmacology and Toxicology, University of Lausanne, 1005, Lausanne, Switzerland.,Department of Medical Genetics, University of Lausanne, 1005, Lausanne, Switzerland
| | - Marzieh Zonouzi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Stephan Kellenberger
- Department of Pharmacology and Toxicology, University of Lausanne, 1005, Lausanne, Switzerland
| | - Dominique Muller
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva 4, Switzerland
| | - Roman Chrast
- Department of Medical Genetics, University of Lausanne, 1005, Lausanne, Switzerland
| | - Stuart G Cull-Candy
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
377
|
Li Y, Xu G, Huang K, Wang J, Zhang J, Liu J, Wang Z, Chen G. Alteration of ASIC1 expression in clear cell renal cell carcinoma. Onco Targets Ther 2015; 8:2121-7. [PMID: 26316781 PMCID: PMC4542551 DOI: 10.2147/ott.s86927] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background Acidic extracellular pH is a major feature of tumor tissue. Acid-sensing ion channels (ASICs) represent an H+-gated subgroup of the degenerin/epithelial Na+ channel family and are activated by acidic microenvironment. Little is known about the expression and clinical significance of ASICs in solid tumors. The purpose of this study was to examine the expression of ASIC1 in human clear cell renal cell carcinoma (CCRCC) and to determine if the expression of ASIC1 is associated with clinicopathological features. Methods The expression of ASIC1 in CCRCC tissues at the mRNA and protein levels was determined by real-time quantitative polymerase chain reaction and Western blot analysis, respectively. A tissue microarray was used to assess the expression of ASIC1 protein in tumor tissue and matched adjacent normal tissues from 75 patients with CCRCC. Results ASIC1 expression was detected in normal renal and CCRCC samples. The expressions of ASIC1 protein and mRNA were significantly decreased in the CCRCC tissues compared with matched normal renal tissues (P<0.05). The staining density measurement showed that the expression of ASIC1 was significantly decreased in stage I (P=0.037), stage II (P=0.026), and stage III (P=0.026), grades I–II CCRCC (P=0.004), and CCRCC from male patients (P=0.00002). However, no significant difference was observed for ASIC1 expression between CCRCC and normal tissue in patients with stage IV CCRCC (P=0.236), patients with grades III–IV CCRCC (P=0.314), and female patients (P=0.095). Spearman correlations demonstrated that ASIC1 expression did not correlate to tumor stage (correlation coefficient [CC =0.168], P=0.149) and the age of patients (CC −0.147, P=0.688) but showed a positive correlation to higher tumor grades (CC =0.270, P=0.018). Conclusion ASIC1 is downregulated in CCRCC. ASIC1 expression may be potentially used as a novel biomarker and even a CCRCC therapeutic target. Further efforts will be made to clarify the mechanism of ASIC1 in occurrence, progression, and metastasis of CCRCC.
Collapse
Affiliation(s)
- Yan Li
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Guoxiong Xu
- Central Laboratory, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Kai Huang
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Jun Wang
- Department of Urology, Shanghai First People's Hospital, Medical College of Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jihong Zhang
- Central Laboratory, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jikai Liu
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Zhanyu Wang
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Gang Chen
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
378
|
van Bemmelen MX, Huser D, Gautschi I, Schild L. The Human Acid-Sensing Ion Channel ASIC1a: Evidence for a Homotetrameric Assembly State at the Cell Surface. PLoS One 2015; 10:e0135191. [PMID: 26252376 PMCID: PMC4529235 DOI: 10.1371/journal.pone.0135191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/18/2015] [Indexed: 12/23/2022] Open
Abstract
The chicken acid-sensing ion channel ASIC1 has been crystallized as a homotrimer. We address here the oligomeric state of the functional ASIC1 in situ at the cell surface. The oligomeric states of functional ASIC1a and mutants with additional cysteines introduced in the extracellular pore vestibule were resolved on SDS-PAGE. The functional ASIC1 complexes were stabilized at the cell surface of Xenopus laevis oocytes or CHO cells either using the sulfhydryl crosslinker BMOE, or sodium tetrathionate (NaTT). Under these different crosslinking conditions ASIC1a migrates as four distinct oligomeric states that correspond by mass to multiples of a single ASIC1a subunit. The relative importance of each of the four ASIC1a oligomers was critically dependent on the availability of cysteines in the transmembrane domain for crosslinking, consistent with the presence of ASIC1a homo-oligomers. The expression of ASIC1a monomers, trimeric or tetrameric concatemeric cDNA constructs resulted in functional channels. The resulting ASIC1a complexes are resolved as a predominant tetramer over the other oligomeric forms, after stabilization with BMOE or NaTT and SDS-PAGE/western blot analysis. Our data identify a major ASIC1a homotetramer at the surface membrane of the cell expressing functional ASIC1a channel.
Collapse
Affiliation(s)
- Miguel Xavier van Bemmelen
- Department of Pharmacology & Toxicology, Faculty of Biology and Medicine, Lausanne University, Lausanne, Switzerland
| | - Delphine Huser
- Department of Pharmacology & Toxicology, Faculty of Biology and Medicine, Lausanne University, Lausanne, Switzerland
| | - Ivan Gautschi
- Department of Pharmacology & Toxicology, Faculty of Biology and Medicine, Lausanne University, Lausanne, Switzerland
| | - Laurent Schild
- Department of Pharmacology & Toxicology, Faculty of Biology and Medicine, Lausanne University, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
379
|
Liu TT, Qu ZW, Ren C, Gan X, Qiu CY, Hu WP. Prolactin potentiates the activity of acid-sensing ion channels in female rat primary sensory neurons. Neuropharmacology 2015; 103:174-82. [PMID: 26188144 DOI: 10.1016/j.neuropharm.2015.07.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 02/06/2023]
Abstract
Prolactin (PRL) is a polypeptide hormone produced and released from the pituitary and extrapituitary tissues. It regulates activity of nociceptors and causes hyperalgesia in pain conditions, but little is known the molecular mechanism. We report here that PRL can exert a potentiating effect on the functional activity of acid-sensing ion channels (ASICs), key sensors for extracellular protons. First, PRL dose-dependently increased the amplitude of ASIC currents with an EC50 of (5.89 ± 0.28) × 10(-8) M. PRL potentiation of ASIC currents was also pH dependent. Second, PRL potentiation of ASIC currents was blocked by Δ1-9-G129R-hPRL, a PRL receptor antagonist, and removed by intracellular dialysis of either protein kinase C inhibitor GF109203X, protein interacting with C-kinase 1(PICK1) inhibitor FSC-231, or PI3K inhibitor AS605240. Third, PRL altered acidosis-evoked membrane excitability of DRG neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acid stimuli. Four, PRL exacerbated nociceptive responses to injection of acetic acid in female rats. Finally, PRL displayed a stronger effect on ASIC mediated-currents and nociceptive behavior in intact female rats than OVX female and male rats and thus modulation of PRL may be gender-dependent. These results suggest that PRL up-regulates the activity of ASICs and enhances ASIC mediated nociceptive responses in female rats, which reveal a novel peripheral mechanism underlying PRL involvement in hyperalgesia.
Collapse
Affiliation(s)
- Ting-Ting Liu
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Zu-Wei Qu
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Cuixia Ren
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Xiong Gan
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Chun-Yu Qiu
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China.
| | - Wang-Ping Hu
- Institute of Ion Channels, Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China.
| |
Collapse
|
380
|
Noh S, Lee SR, Jeong YJ, Ko KS, Rhee BD, Kim N, Han J. The direct modulatory activity of zinc toward ion channels. Integr Med Res 2015; 4:142-146. [PMID: 28664120 PMCID: PMC5481804 DOI: 10.1016/j.imr.2015.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/08/2015] [Indexed: 12/19/2022] Open
Abstract
The divalent zinc ion is a cation that plays an indispensable role as a structural constituent of numerous proteins, including enzymes and transcription factors. Recently, it has been suggested that zinc also plays a dynamic role in extracellular and intracellular signaling as well. Ion channels are pore-forming proteins that control the flow of specific ions across the membrane, which is important to maintain ion gradients. In this review, we outline the modulatory effect of zinc on the activities of several ion channels through direct binding of zinc into histidine, cysteine, aspartate, and glutamate moieties of channel proteins. The binding of zinc to ion channels results in the activation or inhibition of the channel due to conformational changes. These novel aspects of ion-channel activity modulation by zinc provide new insights into the physiological regulation of ion channels.
Collapse
Affiliation(s)
- Sujin Noh
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Sung Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Yu Jeong Jeong
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| |
Collapse
|
381
|
Dussor G. ASICs as therapeutic targets for migraine. Neuropharmacology 2015; 94:64-71. [PMID: 25582295 PMCID: PMC4458434 DOI: 10.1016/j.neuropharm.2014.12.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/26/2014] [Accepted: 12/04/2014] [Indexed: 01/05/2023]
Abstract
Migraine is the most common neurological disorder and one of the most common chronic pain conditions. Despite its prevalence, the pathophysiology leading to migraine is poorly understood and the identification of new therapeutic targets has been slow. Several processes are currently thought to contribute to migraine including altered activity in the hypothalamus, cortical-spreading depression (CSD), and afferent sensory input from the cranial meninges. Decreased extracellular pH and subsequent activation of acid-sensing ion channels (ASICs) may contribute to each of these processes and may thus play a role in migraine pathophysiology. Although few studies have directly examined a role of ASICs in migraine, studies directly examining a connection have generated promising results including efficacy of ASIC blockers in both preclinical migraine models and in human migraine patients. The purpose of this review is to discuss the pathophysiology thought to contribute to migraine and findings that implicate decreased pH and/or ASICs in these events, as well as propose issues to be resolved in future studies of ASICs and migraine. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Greg Dussor
- The University of Texas at Dallas, School of Behavioral and Brain Sciences, GR-41, 800 West Campbell Road, Richardson, TX, 75080, USA.
| |
Collapse
|
382
|
Abstract
During synaptic vesicle (SV) recycling, the vacuolar-type H(+) ATPase creates a proton electrochemical gradient (ΔμH(+)) that drives neurotransmitter loading into SVs. Given the low estimates of free luminal protons, it has been envisioned that the influx of a limited number of protons suffices to establish ΔμH(+). Consistent with this, the time constant of SV re-acidification was reported to be <5 s, much faster than glutamate loading (τ of ∼ 15 s) and thus unlikely to be rate limiting for neurotransmitter loading. However, such estimates have relied on pHluorin-based probes that lack sensitivity in the lower luminal pH range. Here, we reexamined re-acidification kinetics using the mOrange2-based probe that should report the SV pH more accurately. In recordings from cultured mouse hippocampal neurons, we found that re-acidification took substantially longer (τ of ∼ 15 s) than estimated previously. In addition, we found that the SV lumen exhibited a large buffering capacity (∼ 57 mm/pH), corresponding to an accumulation of ∼ 1200 protons during re-acidification. Together, our results uncover hitherto unrecognized robust proton influx and storage in SVs that can restrict the rate of neurotransmitter refilling.
Collapse
|
383
|
Gwiazda K, Bonifacio G, Vullo S, Kellenberger S. Extracellular Subunit Interactions Control Transitions between Functional States of Acid-sensing Ion Channel 1a. J Biol Chem 2015; 290:17956-17966. [PMID: 26070563 DOI: 10.1074/jbc.m115.641688] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Indexed: 12/16/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are neuronal, voltage-independent Na(+) channels that are transiently activated by extracellular acidification. They are involved in pain sensation, the expression of fear, and in neurodegeneration after ischemic stroke. Our study investigates the role of extracellular subunit interactions in ASIC1a function. We identified two regions involved in critical intersubunit interactions. First, formation of an engineered disulfide bond between the palm and thumb domains leads to partial channel closure. Second, linking Glu-235 of a finger loop to either one of two different residues of the knuckle of a neighboring subunit opens the channel at physiological pH or disrupts its activity. This suggests that one finger-knuckle disulfide bond (E235C/K393C) sets the channel in an open state, whereas the other (E235C/Y389C) switches the channel to a non-conducting state. Voltage-clamp fluorometry experiments indicate that both the finger loop and the knuckle move away from the β-ball residue Trp-233 during acidification and subsequent desensitization. Together, these observations reveal that ASIC1a opening is accompanied by a distance increase between adjacent thumb and palm domains as well as a movement of Glu-235 relative to the knuckle helix. Our study identifies subunit interactions in the extracellular loop and shows that dynamic changes of these interactions are critical for normal ASIC function.
Collapse
Affiliation(s)
- Karolina Gwiazda
- Department of Pharmacology and Toxicology, University of Lausanne, Rue du Bugnon 27, CH-1005 Lausanne, Switzerland
| | - Gaetano Bonifacio
- Department of Pharmacology and Toxicology, University of Lausanne, Rue du Bugnon 27, CH-1005 Lausanne, Switzerland
| | - Sabrina Vullo
- Department of Pharmacology and Toxicology, University of Lausanne, Rue du Bugnon 27, CH-1005 Lausanne, Switzerland
| | - Stephan Kellenberger
- Department of Pharmacology and Toxicology, University of Lausanne, Rue du Bugnon 27, CH-1005 Lausanne, Switzerland.
| |
Collapse
|
384
|
Ru F, jr BP, Kollarik M. Acid sensitivity of the spinal dorsal root ganglia C-fiber nociceptors innervating the guinea pig esophagus. Neurogastroenterol Motil 2015; 27:865-74. [PMID: 25846134 PMCID: PMC4446164 DOI: 10.1111/nmo.12561] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 03/05/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gastroesophageal reflux can cause high acidity in the esophagus and trigger heartburn and pain. However, because of the esophageal mucosal barrier, the acidity at the nerve terminals of pain-mediating C-fibers in esophageal mucosa is predicted to be substantially lower. We hypothesized that the esophageal dorsal root ganglia (DRG) C-fibers are activated by mild acid (compared to acidic reflux), and express receptors and ion channels highly sensitive to acid. METHODS Extracellular single unit recordings of activity originating in esophageal DRG C-fiber nerve terminals were performed in the innervated esophagus preparation ex vivo. Acid was delivered in a manner that bypassed the esophageal mucosal barrier. The expression of mRNA for selected receptors in esophagus-specific DRG neurons was evaluated using single cell RT-PCR. KEY RESULTS Mild acid (pH = 6.5-5.5) activated esophageal DRG C-fibers in a pH-dependent manner. The response to mild acid at pH = 6 was not affected by the TRPV1 selective antagonist iodo-resiniferatoxin. The majority (70-95%) of esophageal DRG C-fiber neurons (TRPV1-positive) expressed mRNA for acid sensing ion channels (ASIC1a, ASIC1b, ASIC2b, and/or ASIC3), two-pore-domain (K2P) potassium channel TASK1, and the proton-sensing G-protein coupled receptor OGR1. Other evaluated targets (PKD2L1, TRPV4, TASK3, TALK1, G2A, GPR4, and TDAG8) were expressed rarely. CONCLUSIONS & INFERENCES Guinea pig esophageal DRG C-fibers are activated by mild acid via a TRPV1-independent mechanism, and express mRNA for several receptors and ion channels highly sensitive to acid. The high acid sensitivity of esophageal C-fibers may contribute to heartburn and pain in conditions of reduced mucosal barrier function.
Collapse
Affiliation(s)
- F Ru
- Medicine, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Banovcin P jr
- Pathophysiology, Comenius University in Bratislava, Jessenius Faculty of Medicine, Martin, Slovakia,Gastroenterology, Comenius University in Bratislava, Jessenius Faculty of Medicine, Martin, Slovakia
| | - M Kollarik
- Medicine, The Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
385
|
Cho CH, Lho YM, Ha E, Hwang I, Song KS, Min BW, Bae KC, Kim DH. Up-regulation of acid-sensing ion channels in the capsule of the joint in frozen shoulder. Bone Joint J 2015; 97-B:824-9. [DOI: 10.1302/0301-620x.97b6.35254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The purpose of this study was to evaluate the expression of acid-sensing ion channels (ASICs) in the capsule and synovial fluid of patients with frozen shoulder. Capsular tissue and synovial fluid were obtained from 18 patients with idiopathic frozen shoulder (FS group) and 18 patients with instability of the shoulder (control group). The expressions of ASIC1, ASIC2, and ASIC3 in the capsule were determined using the reverse transcriptase-polymerase chain reaction, immunoblot analysis, and immunohistochemistry (IHC). The concentrations in synovial fluid were evaluated using an enzyme-linked immunosorbent assay. The mRNA expression of ASIC1, ASIC2 and ASIC3 in the capsule were significantly increased in the FS group compared with the control group. The protein levels of these three ASICs were also increased. The increased expressions were confirmed by IHC. Of the ASICs, ASIC3 showed the greatest increase in both mRNA and levels of expression compared with the control group. The levels of ASIC1 and ASIC3 in synovial fluid were significantly increased in the FS group. This study suggests that ASICs may play a role as mediators of inflammatory pain and be involved in the pathogenesis of frozen shoulder. Cite this article: Bone Joint J 2015;97-B:824–9.
Collapse
Affiliation(s)
- C. H. Cho
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| | - Y. M. Lho
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| | - E. Ha
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| | - I. Hwang
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| | - K. S. Song
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| | - B. W. Min
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| | - K. C. Bae
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| | - D. H. Kim
- Keimyung University Dongsan Medical Center, 56
Dalseong-ro, Jung-gu, Daegu
700-712, South Korea
| |
Collapse
|
386
|
Liu S, Cheng XY, Wang F, Liu CF. Acid-sensing ion channels: potential therapeutic targets for neurologic diseases. Transl Neurodegener 2015; 4:10. [PMID: 26029363 PMCID: PMC4449961 DOI: 10.1186/s40035-015-0031-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 05/14/2015] [Indexed: 01/14/2023] Open
Abstract
Maintaining the physiological pH of interstitial fluid is crucial for normal cellular functions. In disease states, tissue acidosis is a common pathologic change causing abnormal activation of acid-sensing ion channels (ASICs), which according to cumulative evidence, significantly contributes to inflammation, mitochondrial dysfunction, and other pathologic mechanisms (i.e., pain, stroke, and psychiatric conditions). Thus, it has become increasingly clear that ASICs are critical in the progression of neurologic diseases. This review is focused on the importance of ASICs as potential therapeutic targets in combating neurologic diseases.
Collapse
Affiliation(s)
- Sha Liu
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
| | - Xiao-Yu Cheng
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
| | - Fen Wang
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
- />Institute of Neuroscience, Soochow University, Suzhou, 215123 China
| | - Chun-Feng Liu
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
- />Institute of Neuroscience, Soochow University, Suzhou, 215123 China
- />Beijing Key Laboratory for Parkinson’s Disease, Beijing, 100053 China
| |
Collapse
|
387
|
Buta A, Maximyuk O, Kovalskyy D, Sukach V, Vovk M, Ievglevskyi O, Isaeva E, Isaev D, Savotchenko A, Krishtal O. Novel Potent Orthosteric Antagonist of ASIC1a Prevents NMDAR-Dependent LTP Induction. J Med Chem 2015; 58:4449-61. [PMID: 25974655 DOI: 10.1021/jm5017329] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acid sensing ion channels 1a (ASIC1a) are of crucial importance in numerous physiological and pathological processes in the brain. Here we demonstrate that novel 2-oxo-2H-chromene-3-carboxamidine derivative 5b, designed with molecular modeling approach, inhibits ASIC1a currents with an apparent IC50 of 27 nM when measured at pH 6.7. Acidification to 5.0 decreases the inhibition efficacy by up to 3 orders of magnitude. The 5b molecule not only shifts pH dependence of ASIC1a activation but also inhibits its maximal evoked response. These findings suggest that compound 5b binds to pH sensor of ASIC1a acting as orthosteric noncompetitive antagonist. At 100 nM, compound 5b completely inhibits induction of long-term potentiation (LTP) in CA3-CA1 but not in MF-CA3 synapses. These findings support the knockout data indicating the crucial modulatory role of ASIC1a channels in the NMDAR-dependent LTP and introduce a novel type of ASIC1a antagonists.
Collapse
Affiliation(s)
- Andriy Buta
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine.,§State Key Laboratory for Molecular and Cellular Biology, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Oleksandr Maximyuk
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine.,§State Key Laboratory for Molecular and Cellular Biology, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Dmytro Kovalskyy
- ∥ChemBio Center, Taras Shevchenko University of Kyiv, 67 Chervonotkatska Str., 02094 Kyiv, Ukraine
| | - Volodymyr Sukach
- ‡Institute of Organic Chemistry of NAS Ukraine, 5 Murmanska Str., 02660 Kyiv, Ukraine
| | - Mykhailo Vovk
- ‡Institute of Organic Chemistry of NAS Ukraine, 5 Murmanska Str., 02660 Kyiv, Ukraine
| | - Oleksandr Ievglevskyi
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Elena Isaeva
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Dmytro Isaev
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Alina Savotchenko
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Oleg Krishtal
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine.,§State Key Laboratory for Molecular and Cellular Biology, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| |
Collapse
|
388
|
Proton-induced currents in substantia gelatinosa neurons of the rat trigeminal subnucleus caudalis. Eur J Pharmacol 2015; 762:18-25. [PMID: 25962663 DOI: 10.1016/j.ejphar.2015.04.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 04/30/2015] [Accepted: 04/30/2015] [Indexed: 11/20/2022]
Abstract
Acid-sensing ion channels (ASICs) are widely expressed in both the peripheral and central nervous system, and contribute to the modulation of central nociceptive transmission under both physiological and pathophysiological conditions. In this study, we characterized the proton-induced membrane currents in acutely isolated rat substantia gelatinosa (SG) neurons of the trigeminal subnucleus caudalis using the whole cell patch-clamp technique. Exposure to acidic conditions (pH<6.5) induced the inward currents in a pH-dependent manner. Amiloride, a general ASIC antagonist, significantly blocked the proton-induced currents in a non-competitive manner. The pH 6.0-induced membrane current (IpH6.0) was greatly attenuated in the Na(+)-free external solution, and the reversal potential of the proton-induced currents was similar to the theoretical Na(+) equilibrium potential. The IpH6.0 was reciprocally potentiated by a lower extracellular Ca(2+) concentration. The modulation of IpH6.0 by divalent cations and other modulators suggests that the proton-induced currents are mediated by multiple types of ASIC subunits, including ASIC1a and ASIC2a. Multi-cell RT-PCR analysis revealed that SG neurons express these subunits. Exposure to a pH 6.0 solution directly depolarized the membrane potential, and generated a burst of action potentials in a current-clamp mode. This acidic pH-induced depolarization was significantly blocked by amiloride. The present results suggest that ASICs expressed on SG neurons play important roles in the regulation of nociceptive transmission from the orofacial tissues.
Collapse
|
389
|
Acidic microenvironment and bone pain in cancer-colonized bone. BONEKEY REPORTS 2015; 4:690. [PMID: 25987988 DOI: 10.1038/bonekey.2015.58] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/20/2015] [Indexed: 12/15/2022]
Abstract
Solid cancers and hematologic cancers frequently colonize bone and induce skeletal-related complications. Bone pain is one of the most common complications associated with cancer colonization in bone and a major cause of increased morbidity and diminished quality of life, leading to poor survival in cancer patients. Although the mechanisms responsible for cancer-associated bone pain (CABP) are poorly understood, it is likely that complex interactions among cancer cells, bone cells and peripheral nerve cells contribute to the pathophysiology of CABP. Clinical observations that specific inhibitors of osteoclasts reduce CABP indicate a critical role of osteoclasts. Osteoclasts are proton-secreting cells and acidify extracellular bone microenvironment. Cancer cell-colonized bone also releases proton/lactate to avoid intracellular acidification resulting from increased aerobic glycolysis known as the Warburg effect. Thus, extracellular microenvironment of cancer-colonized bone is acidic. Acidosis is algogenic for nociceptive sensory neurons. The bone is densely innervated by the sensory neurons that express acid-sensing nociceptors. Collectively, CABP is evoked by the activation of these nociceptors on the sensory neurons innervating bone by the acidic extracellular microenvironment created by bone-resorbing osteoclasts and bone-colonizing cancer cells. As current treatments do not satisfactorily control CABP and can elicit serious side effects, new therapeutic interventions are needed to manage CABP. Understanding of the cellular and molecular mechanism by which the acidic extracellular microenvironment is created in cancer-colonized bone and by which the expression and function of the acid-sensing nociceptors on the sensory neurons are regulated would facilitate to develop novel therapeutic approaches for the management of CABP.
Collapse
|
390
|
Vollmer LL, Strawn JR, Sah R. Acid-base dysregulation and chemosensory mechanisms in panic disorder: a translational update. Transl Psychiatry 2015; 5:e572. [PMID: 26080089 PMCID: PMC4471296 DOI: 10.1038/tp.2015.67] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 02/19/2015] [Accepted: 04/06/2015] [Indexed: 12/13/2022] Open
Abstract
Panic disorder (PD), a complex anxiety disorder characterized by recurrent panic attacks, represents a poorly understood psychiatric condition which is associated with significant morbidity and an increased risk of suicide attempts and completed suicide. Recently however, neuroimaging and panic provocation challenge studies have provided insights into the pathoetiology of panic phenomena and have begun to elucidate potential neural mechanisms that may underlie panic attacks. In this regard, accumulating evidence suggests that acidosis may be a contributing factor in induction of panic. Challenge studies in patients with PD reveal that panic attacks may be reliably provoked by agents that lead to acid-base dysbalance such as CO2 inhalation and sodium lactate infusion. Chemosensory mechanisms that translate pH into panic-relevant fear, autonomic, and respiratory responses are therefore of high relevance to the understanding of panic pathophysiology. Herein, we provide a current update on clinical and preclinical studies supporting how acid-base imbalance and diverse chemosensory mechanisms may be associated with PD and discuss future implications of these findings.
Collapse
Affiliation(s)
- L L Vollmer
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - J R Strawn
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, USA,Cincinnati Children's Hospital Medical Center, Department of Psychiatry, Cincinnati, OH, USA
| | - R Sah
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, USA,Veterens' Affairs Medical Center, Cincinnati, OH, USA,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, 2170 East Galbraith Road, Cincinnati, OH 45237, USA. E-mail:
| |
Collapse
|
391
|
Schuhmacher LN, Srivats S, Smith ESJ. Structural domains underlying the activation of acid-sensing ion channel 2a. Mol Pharmacol 2015; 87:561-71. [PMID: 25583083 PMCID: PMC4747086 DOI: 10.1124/mol.114.096909] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/12/2015] [Indexed: 02/06/2023] Open
Abstract
The acid-sensing ion channels (ASICs) are a family of ion channels expressed throughout the mammalian nervous system. The principal activator of ASICs is extracellular protons, and ASICs have been demonstrated to play a significant role in many physiologic and pathophysiologic processes, including synaptic transmission, nociception, and fear. However, not all ASICs are proton-sensitive: ASIC2a is activated by acid, whereas its splice variant ASIC2b is not. We made a series of chimeric ASIC2 proteins, and using whole-cell electrophysiology we have identified the minimal region of the ASIC2a extracellular domain that is required for ASIC2 proton activation: the first 87 amino acids after transmembrane domain 1. We next examined the function of different domains within the ASIC2b N-terminus and identified a region proximal to the first transmembrane domain that confers tachyphylaxis upon ASIC2a. We have thus identified domains of ASIC2 that are crucial to channel function and may be important for the function of other members of the ASIC family.
Collapse
Affiliation(s)
| | - Shyam Srivats
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
392
|
Making sense of pain: are pluripotent stem cell-derived sensory neurons a new tool for studying pain mechanisms? Mol Ther 2015; 22:1403-1405. [PMID: 25082088 DOI: 10.1038/mt.2014.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
393
|
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
|
394
|
|
395
|
Yoneda T, Hiasa M, Nagata Y, Okui T, White F. Contribution of acidic extracellular microenvironment of cancer-colonized bone to bone pain. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2677-84. [PMID: 25687976 DOI: 10.1016/j.bbamem.2015.02.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 02/01/2015] [Accepted: 02/05/2015] [Indexed: 12/31/2022]
Abstract
Solid and hematologic cancer colonized bone produces a number of pathologies. One of the most common complications is bone pain. Cancer-associated bone pain (CABP) is a major cause of increased morbidity and diminishes the quality of life and affects survival. Current treatments do not satisfactorily control CABP and can elicit adverse effects. Thus, new therapeutic interventions are needed to manage CABP. However, the mechanisms responsible for CABP are poorly understood. The observation that specific osteoclast inhibitors can reduce CABP in patients indicates a critical role of osteoclasts in the pathophysiology of CABP. Osteoclasts create an acidic extracellular microenvironment by secretion of protons via vacuolar proton pumps during bone resorption. In addition, bone-colonized cancer cells also release protons and lactate via plasma membrane pH regulators to avoid intracellular acidification resulting from increased aerobic glycolysis known as the Warburg effect. Since acidosis is algogenic for sensory neurons and bone is densely innervated by sensory neurons that express acid-sensing nociceptors, the acidic bone microenvironments can evoke CABP. Understanding of the mechanism by which the acidic extracellular microenvironment is created in cancer-colonized bone and the expression and function of the acid-sensing nociceptors are regulated should facilitate the development of novel approaches for management of CABP. Here, the contribution of the acidic microenvironment created in cancer-colonized bone to elicitation of CABP and potential therapeutic implications of blocking the development and recognition of acidic microenvironment will be described. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Toshiyuki Yoneda
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Masahiro Hiasa
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Yuki Nagata
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Tatsuo Okui
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Fletcher White
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indiana University, 320 West 15th Street, Indianapolis, IN 46202, USA.
| |
Collapse
|
396
|
Affiliation(s)
- Lonny R. Levin
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065; ,
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065; ,
| |
Collapse
|
397
|
Burgos-Vega C, Moy J, Dussor G. Meningeal afferent signaling and the pathophysiology of migraine. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:537-64. [PMID: 25744685 DOI: 10.1016/bs.pmbts.2015.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Migraine is the most common neurological disorder. Attacks are complex and consist of multiple phases but are most commonly characterized by intense, unilateral, throbbing headache. The pathophysiology contributing to migraine is poorly understood and the disorder is not well managed with currently available therapeutics, often rendering patients disabled during attacks. The mechanisms most likely to contribute to the pain phase of migraine require activation of trigeminal afferent signaling from the cranial meninges and subsequent relay of nociceptive information into the central nervous system in a region of the dorsal brainstem known as the trigeminal nucleus caudalis. Events leading to activation of meningeal afferents are unclear, but nerve endings within this tissue are mechanosensitive and also express a variety of ion channels including acid-sensing ion channels and transient receptor-potential channels. These properties may provide clues into the pathophysiology of migraine by suggesting that decreased extracellular pH and environmental irritant exposure in the meninges contributes to headache. Neuroplasticity is also likely to play a role in migraine given that attacks are triggered by routine events that are typically nonnoxious in healthy patients and clear evidence of sensitization occurs during an attack. Where and how plasticity develops is also not clear but may include events directly on the afferents and/or within the TNC. Among the mediators potentially contributing to plasticity, calcitonin gene-related peptide has received the most attention within the migraine field but other mechanisms may also contribute. Ultimately, greater understanding of the molecules and mechanisms contributing to migraine will undoubtedly lead to better therapeutics and relief for the large number of patients across the globe who suffer from this highly disabling neurological disorder.
Collapse
Affiliation(s)
- Carolina Burgos-Vega
- Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Jamie Moy
- Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Gregory Dussor
- Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA.
| |
Collapse
|
398
|
Qiu F, Qiu CY, Cai H, Liu TT, Qu ZW, Yang Z, Li JD, Zhou QY, Hu WP. Oxytocin inhibits the activity of acid-sensing ion channels through the vasopressin, V1A receptor in primary sensory neurons. Br J Pharmacol 2015; 171:3065-76. [PMID: 24641084 DOI: 10.1111/bph.12635] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/01/2014] [Accepted: 02/04/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE A growing number of studies have demonstrated that oxytocin (OT) plays an analgesic role in modulation of nociception and pain. Most work to date has focused on the central mechanisms of OT analgesia, but little is known about whether peripheral mechanisms are also involved. Acid-sensing ion channels (ASICs) are distributed in peripheral sensory neurons and participate in nociception. Here, we investigated the effects of OT on the activity of ASICs in dorsal root ganglion (DRG) neurons. EXPERIMENTAL APPROACH Electrophysiological experiments were performed on neurons from rat DRG. Nociceptive behaviour was induced by acetic acid in rats and mice lacking vasopressin, V1A receptors. KEY RESULTS OT inhibited the functional activity of native ASICs. Firstly, OT dose-dependently decreased the amplitude of ASIC currents in DRG neurons. Secondly, OT inhibition of ASIC currents was mimicked by arginine vasopressin (AVP) and completely blocked by the V1A receptor antagonist SR49059, but not by the OT receptor antagonist L-368899. Thirdly, OT altered acidosis-evoked membrane excitability of DRG neurons and significantly decreased the amplitude of the depolarization and number of action potentials induced by acid stimuli. Finally, peripherally administered OT or AVP inhibited nociceptive responses to intraplantar injection of acetic acid in rats. Both OT and AVP also induced an analgesic effect on acidosis-evoked pain in wild-type mice, but not in V1A receptor knockout mice. CONCLUSIONS AND IMPLICATIONS These results reveal a novel peripheral mechanism for the analgesic effect of OT involving the modulation of native ASICs in primary sensory neurons mediated by V1A receptors.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Pharmacology, Hubei University of Science and Technology, Xianning, China; College of Life Sciences, Hubei University, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
399
|
Jernigan NL. Smooth muscle acid-sensing ion channel 1: pathophysiological implication in hypoxic pulmonary hypertension. Exp Physiol 2015; 100:111-20. [DOI: 10.1113/expphysiol.2014.081612] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/04/2014] [Indexed: 01/20/2023]
Affiliation(s)
- Nikki L. Jernigan
- Vascular Physiology Group; Department of Cell Biology and Physiology; University of New Mexico Health Sciences Center; Albuquerque, NM 87131-0001 USA
| |
Collapse
|
400
|
Huang Y, Jiang N, Li J, Ji YH, Xiong ZG, Zha XM. Two aspects of ASIC function: Synaptic plasticity and neuronal injury. Neuropharmacology 2015; 94:42-8. [PMID: 25582290 DOI: 10.1016/j.neuropharm.2014.12.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 12/06/2014] [Accepted: 12/09/2014] [Indexed: 12/17/2022]
Abstract
Extracellular brain pH fluctuates in both physiological and disease conditions. The main postsynaptic proton receptor is the acid-sensing ion channels (ASICs). During the past decade, much progress has been made on protons, ASICs, and neurological disease. This review summarizes the recent progress on synaptic role of protons and our current understanding of how ASICs contribute to various types of neuronal injury in the brain. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Yan Huang
- School of Pharmacy, Anhui Medical University, Hefei, China; Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310, USA
| | - Nan Jiang
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Lab of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yong-Hua Ji
- Lab of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, China
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310, USA.
| | - Xiang-ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA.
| |
Collapse
|