51
|
Uchitel OD, González Inchauspe C, Weissmann C. Synaptic signals mediated by protons and acid-sensing ion channels. Synapse 2019; 73:e22120. [PMID: 31180161 DOI: 10.1002/syn.22120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 01/04/2023]
Abstract
Extracellular pH changes may constitute significant signals for neuronal communication. During synaptic transmission, changes in pH in the synaptic cleft take place. Its role in the regulation of presynaptic Ca2+ currents through multivesicular release in ribbon-type synapses is a proven phenomenon. In recent years, protons have been recognized as neurotransmitters that participate in neuronal communication in synapses of several regions of the CNS such as amygdala, nucleus accumbens, and brainstem. Protons are released by nerve stimulation and activate postsynaptic acid-sensing ion channels (ASICs). Several types of ASIC channels are expressed in the peripheral and central nervous system. The influx of Ca2+ through some subtypes of ASICs, as a result of synaptic transmission, agrees with the participation of ASICs in synaptic plasticity. Pharmacological and genetical inhibition of ASIC1a results in alterations in learning, memory, and phenomena like fear and cocaine-seeking behavior. The recognition of endogenous molecules, such as arachidonic acid, cytokines, histamine, spermine, lactate, and neuropeptides, capable of inhibiting or potentiating ASICs suggests the existence of mechanisms of synaptic modulation that have not yet been fully identified and that could be tuned by new emerging pharmacological compounds with potential therapeutic benefits.
Collapse
Affiliation(s)
- Osvaldo D Uchitel
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlota González Inchauspe
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carina Weissmann
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
52
|
González-Inchauspe C, Gobetto MN, Uchitel OD. Modulation of acid sensing ion channel dependent protonergic neurotransmission at the mouse calyx of Held. Neuroscience 2019; 439:195-210. [PMID: 31022462 DOI: 10.1016/j.neuroscience.2019.04.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 11/18/2022]
Abstract
Acid-sensing ion channels (ASICs) regulate synaptic activities and play important roles in neurodegenerative diseases. It has been reported that homomeric ASIC-1a channels are expressed in neurons of the medial nucleus of the trapezoid body (MNTB) of the auditory system in the CNS. During synaptic transmission, acidification of the synaptic cleft presumably due to the co-release of neurotransmitter and H+ from synaptic vesicles activates postsynaptic ASIC-1a channels in mice up to 3 weeks old. This generates synaptic currents (ASIC1a-SCs) that add to the glutamatergic excitatory postsynaptic currents (EPSCs). Here we report that neuromodulators like histamine and natural products like lactate and spermine potentiate ASIC1a-SCs in an additive form such that excitatory ASIC synaptic currents as well as the associated calcium influx become significantly large and physiologically relevant. We show that ASIC1a-SCs enhanced by endogenous neuromodulators are capable of supporting synaptic transmission in the absence of glutamatergic EPSCs. Furthermore, at high frequency stimulation (HFS), ASIC1a-SCs contribute to diminish short term depression (STD) and their contribution is even more relevant at early stages of development. Since ASIC channels are present in almost all types of neurons and synaptic vesicles content is acid, the participation of protons in synaptic transmission and its potentiation by endogenous substances could be a general phenomenon across the central nervous system. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.
Collapse
Affiliation(s)
- Carlota González-Inchauspe
- Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET. Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Universitaria. (C1428EGA) Ciudad Autónoma de Buenos Aires, Argentina.
| | - María Natalia Gobetto
- Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET. Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Universitaria. (C1428EGA) Ciudad Autónoma de Buenos Aires, Argentina
| | - Osvaldo D Uchitel
- Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET. Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Universitaria. (C1428EGA) Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
53
|
Zuo L, Zhu Y, Hu L, Liu Y, Wang Y, Hu Y, Wang H, Pan X, Li K, Du N, Huang Y. PI3-kinase/Akt pathway-regulated membrane transportation of acid-sensing ion channel 1a/Calcium ion influx/endoplasmic reticulum stress activation on PDGF-induced HSC Activation. J Cell Mol Med 2019; 23:3940-3950. [PMID: 30938088 PMCID: PMC6533492 DOI: 10.1111/jcmm.14275] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/14/2019] [Accepted: 01/27/2019] [Indexed: 12/15/2022] Open
Abstract
Acid-sensing ion channel 1a (ASIC1a) allows Na+ and Ca2+ flow into cells. It is expressed during inflammation, in tumour and ischaemic tissue, in the central nervous system and non-neuronal injury environments. Endoplasmic reticulum stress (ERS) is caused by the accumulation of misfolded proteins that interferes with intracellular calcium homoeostasis. Our recent reports showed ASIC1a and ERS are involved in liver fibrosis progression, particularly in hepatic stellate cell (HSC) activation. In this study, we investigated the roles of ASIC1a and ERS in activated HSC. We found that ASIC1a and ERS-related proteins were up-regulated in carbon tetrachloride (CCl4 )-induced fibrotic mouse liver tissues, and in patient liver tissues with hepatocellular carcinoma with severe liver fibrosis. The results show silencing ASIC1a reduced the expression of ERS-related biomarkers GRP78, Caspase12 and IREI-XBP1. And, ERS inhibition by 4-PBA down-regulated the high expression of ASIC1a induced by PDGF, suggesting an interactive relationship. In PDGF-induced HSCs, ASIC1a was activated and migrated to the cell membrane, leading to extracellular calcium influx and ERS, which was mediated by PI3K/AKT pathway. Our work shows PDGF-activated ASIC1a via the PI3K/AKT pathway, induced ERS and promoted liver fibrosis progression.
Collapse
Affiliation(s)
- Longquan Zuo
- Department of Pharmacy, Hospital of Armed Police of Anhui Province, Hefei, China
| | - Yueqin Zhu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Lili Hu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Yanyi Liu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Yinghong Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yamin Hu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Huan Wang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Xuesheng Pan
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Kuayue Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Na Du
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Yan Huang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| |
Collapse
|
54
|
Gao WF, Xu YY, Ge JF, Chen FH. Inhibition of acid‑sensing ion channel 1a attenuates acid‑induced activation of autophagy via a calcium signaling pathway in articular chondrocytes. Int J Mol Med 2019; 43:1778-1788. [PMID: 30720055 PMCID: PMC6414154 DOI: 10.3892/ijmm.2019.4085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 01/30/2019] [Indexed: 12/16/2022] Open
Abstract
Acid-sensing ion channel 1a (ASIC1a), member of the degenerin/epithelial sodium channel protein superfamily, serves a critical role in various physiological and pathological processes. The aim of the present study was to examine the role of ASIC1a in the autophagy of rat articular chondrocytes. Autophagy was induced by acidic stimulation in rat articular chondrocytes and the extent of autophagy was evaluated via the expression levels of microtubule-associated protein 1 light chain 3II, Beclin1 and uncoordinated-51 like kinase1. Suppression of ASIC1a was achieved using small interfering RNA technology and/or inhibitor psalmotoxin-1. The expression levels of autophagy markers were measured by western blot analysis and reverse transcription-quantitative polymerase chain reaction methods. Intracellular calcium ([Ca2+]i) was analyzed using a Ca2+-imaging method. Additionally, protein expression levels of the Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ)/5′-monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway were measured by western blot analysis. The results showed that autophagy was increased in a pH-and time-dependent manner with exposure to an acidic environment. In addition, silencing ASIC1a significantly decreased the expression levels of autophagy makers, accompanied by abrogation of the acid-induced [Ca2+]i increase. Furthermore, silencing of ASIC1a downregulated the levels of CaMKKβ/β-actin and phosphorylated (p-) AMPK/AMPK, and upregulated the levels of p-mTOR/mTOR. These results indicated that ASIC1a is a potent regulator of autophagy in chondrocytes, which may be associated with decreased Ca2+ influx and the CaMKKβ/AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Wen-Fan Gao
- Department of Pharmacy, Anhui Mental Health Center, Hefei, Anhui 230000, P.R. China
| | - Ya-Yun Xu
- Department of Pharmacy, Anhui Mental Health Center, Hefei, Anhui 230000, P.R. China
| | - Jin-Fang Ge
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Fei-Hu Chen
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
55
|
Palma-Cerda F, Papageorgiou G, Barbour B, Auger C, Ogden D. Photolysis of a Caged, Fast-Equilibrating Glutamate Receptor Antagonist, MNI-Caged γ-D-Glutamyl-Glycine, to Investigate Transmitter Dynamics and Receptor Properties at Glutamatergic Synapses. Front Cell Neurosci 2019; 12:465. [PMID: 30618624 PMCID: PMC6300705 DOI: 10.3389/fncel.2018.00465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/15/2018] [Indexed: 11/29/2022] Open
Abstract
Fast uncaging of low affinity competitive receptor antagonists can in principle measure the timing and concentration dependence of transmitter action at receptors during synaptic transmission. Here, we describe the development, synthesis and characterization of MNI-caged γ-D-glutamyl-glycine (γ-DGG), which combines the fast photolysis and hydrolytic stability of nitroindoline cages with the well-characterized fast-equilibrating competitive glutamate receptor antagonist γ-DGG. At climbing fiber-Purkinje cell (CF-PC) synapses MNI-caged-γ-DGG was applied at concentrations up to 5 mM without affecting CF-PC transmission, permitting release of up to 1.5 mM γ-DGG in 1 ms in wide-field flashlamp photolysis. In steady-state conditions, photoreleased γ-DGG at 0.55–1.7 mM inhibited the CF first and second paired EPSCs by on average 30% and 60%, respectively, similar to reported values for bath applied γ-DGG. Photolysis of the L-isomer MNI-caged γ-L-glutamyl-glycine was ineffective. The time-course of receptor activation by synaptically released glutamate was investigated by timed photolysis of MNI-caged-γ-DGG at defined intervals following CF stimulation in the second EPSCs. Photorelease of γ-DGG prior to the stimulus and up to 3 ms after showed strong inhibition similar to steady-state inhibition; in contrast γ-DGG applied by a flash at 3–4 ms post-stimulus produced weaker and variable block, suggesting transmitter-receptor interaction occurs mainly in this time window. The data also show a small and lasting component of inhibition when γ-DGG was released at 4–7 ms post stimulus, near the peak of the CF-PC EPSC, or at 10–11 ms. This indicates that competition for binding and activation of AMPA receptors occurs also during the late phase of the EPSC, due to either delayed transmitter release or persistence of glutamate in the synaptic region. The results presented here first show that MNI-caged-γ-DGG has properties suitable for use as a synaptic probe at high concentration and that its photolysis can resolve timing and extent of transmitter activation of receptors in glutamatergic transmission.
Collapse
Affiliation(s)
| | | | - Boris Barbour
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL University, Paris, France
| | - Céline Auger
- Brain Physiology Lab, UMR8118 Université Paris Descartes, Paris, France
| | - David Ogden
- Brain Physiology Lab, UMR8118 Université Paris Descartes, Paris, France.,The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
56
|
Abstract
A large series of different ion channels have been identified and investigated as potential targets for new medicines for the treatment of a variety of human diseases, including pain. Among these channels, the voltage gated calcium channels (VGCC) are inhibited by drugs for the treatment of migraine, neuropathic pain or intractable pain. Transient receptor potential (TRP) channels are emerging as important pain transducers as they sense low pH media or oxidative stress and other mediators and are abundantly found at sites of inflammation or tissue injury. Low pH may also activate acid sensing ion channels (ASIC) and mechanical forces stimulate the PIEZO channels. While potent agonists of TRP channels due to their desensitizing action on pain transmission are used as topical applications, the potential of TRP antagonists as pain therapeutics remains an exciting field of investigation. The study of ASIC or PIEZO channels in pain signaling is in an early stage, whereas antagonism of the purinergic P2X3 channels has been reported to provide beneficial effects in chronic intractable cough. The present chapter covers these intriguing channels in great detail, highlighting their diverse mechanisms and broad potential for therapeutic utility.
Collapse
Affiliation(s)
- Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| |
Collapse
|
57
|
Abstract
Many odors activate the intranasal chemosensory trigeminal system where they produce cooling and other somatic sensations such as tingling, burning, or stinging. Specific trigeminal receptors are involved in the mediation of these sensations. Importantly, the trigeminal system also mediates sensitivity to airflow. The intranasal trigeminal and the olfactory system are closely connected. With regard to central nervous processing, it is most interesting that trigeminal stimuli can activate the piriform cortex, which is typically viewed as the primary olfactory cortex. This suggests that interactions between the two systems may form at a relatively early stage of processing. For example, there is evidence showing that acquired olfactory loss leads to reduced trigeminal sensitivity, probably on account of the lack of interaction in the central nervous system. Decreased trigeminal sensitivity may also be responsible for changes in airflow perception, leading to the impression of congested nasal airways.
Collapse
Affiliation(s)
- Thomas Hummel
- Department of Otorhinolaryngology, Smell and Taste Clinic, Technische Universität Dresden, Dresden, Germany.
| | - Johannes Frasnelli
- Université du Québec à Trois-Rivières, Department of Anatomy, Trois-Rivières, QC, Canada
| |
Collapse
|
58
|
Vyvers A, Schmidt A, Wiemuth D, Gründer S. Screening of 109 neuropeptides on ASICs reveals no direct agonists and dynorphin A, YFMRFamide and endomorphin-1 as modulators. Sci Rep 2018; 8:18000. [PMID: 30573735 PMCID: PMC6301962 DOI: 10.1038/s41598-018-36125-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/14/2018] [Indexed: 01/01/2023] Open
Abstract
Acid-sensing ion channels (ASICs) belong to the DEG/ENaC gene family. While ASIC1a, ASIC1b and ASIC3 are activated by extracellular protons, ASIC4 and the closely related bile acid-sensitive ion channel (BASIC or ASIC5) are orphan receptors. Neuropeptides are important modulators of ASICs. Moreover, related DEG/ENaCs are directly activated by neuropeptides, rendering neuropeptides interesting ligands of ASICs. Here, we performed an unbiased screen of 109 short neuropeptides (<20 amino acids) on five homomeric ASICs: ASIC1a, ASIC1b, ASIC3, ASIC4 and BASIC. This screen revealed no direct agonist of any ASIC but three modulators. First, dynorphin A as a modulator of ASIC1a, which increased currents of partially desensitized channels; second, YFMRFamide as a modulator of ASIC1b and ASIC3, which decreased currents of ASIC1b and slowed desensitization of ASIC1b and ASIC3; and, third, endomorphin-1 as a modulator of ASIC3, which also slowed desensitization. With the exception of YFMRFamide, which, however, is not a mammalian neuropeptide, we identified no new modulator of ASICs. In summary, our screen confirmed some known peptide modulators of ASICs but identified no new peptide ligands of ASICs, suggesting that most short peptides acting as ligands of ASICs are already known.
Collapse
Affiliation(s)
- Anna Vyvers
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany
| | - Axel Schmidt
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany.
| | - Dominik Wiemuth
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany.
| |
Collapse
|
59
|
Peverini L, Beudez J, Dunning K, Chataigneau T, Grutter T. New Insights Into Permeation of Large Cations Through ATP-Gated P2X Receptors. Front Mol Neurosci 2018; 11:265. [PMID: 30108481 PMCID: PMC6080412 DOI: 10.3389/fnmol.2018.00265] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/13/2018] [Indexed: 11/28/2022] Open
Abstract
The permeability of large cations through the P2X pore has remained arguably the most controversial and complicated topic in P2X-related research, with the emergence of conflicting studies on the existence, mechanism and physiological relevance of a so-called “dilated” state. Due to the important role of several “dilating” P2X subtypes in numerous diseases, a clear and detailed understanding of this phenomenon represents a research priority. Recent advances, however, have challenged the existence of a progressive, ATP-induced pore dilation, by demonstrating that this phenomenon is an artifact of the method employed. Here, we discuss briefly the history of this controversial and enigmatic dilated state, from its initial discovery to its recent reconsideration. We will discuss the literature in which mechanistic pathways to a large cation-permeable state are proposed, as well as important advances in the methodology employed to study this elusive state. Considering recent literature, we will also open the discussion as to whether an intrinsically dilating P2X pore exists, as well as the physiological relevance of such a large cation-permeable pore and its potential use as therapeutic pathway.
Collapse
Affiliation(s)
- Laurie Peverini
- CNRS, CAMB UMR 7199, Équipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, Strasbourg, France
| | - Juline Beudez
- CNRS, CAMB UMR 7199, Équipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, Strasbourg, France
| | - Kate Dunning
- CNRS, CAMB UMR 7199, Équipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, Strasbourg, France
| | - Thierry Chataigneau
- CNRS, CAMB UMR 7199, Équipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, Strasbourg, France
| | - Thomas Grutter
- CNRS, CAMB UMR 7199, Équipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
60
|
Dual actions of Psalmotoxin at ASIC1a and ASIC2a heteromeric channels (ASIC1a/2a). Sci Rep 2018; 8:7179. [PMID: 29739981 PMCID: PMC5940917 DOI: 10.1038/s41598-018-25386-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023] Open
Abstract
Acid-Sensing Ion Channels (ASICs) are gated by extracellular protons and play important roles in physiological and pathological states, such as pain and stroke. ASIC1a and ASIC2a, two of the most highly expressed subunits in the brain, form functional homo- and hetero-meric (ASIC1a/2a) channels. The function of ASIC1a has been widely studied using psalmotoxin (PcTx1), a venom-derived peptide, as an ASIC1a-selective antagonist. Here, using whole-cell patch clamp, we show that PcTx1 has dual actions at ASIC1a/2a. It can either inhibit or potentiate the heteromeric channel, depending on the conditioning and stimulating pHs. Potent inhibition occurs only at conditioning pHs that begin to desensitize the channel (IC50 = 2.9 nM at pH7.0, a threshold pH for desensitization of ASIC1a/2a). By contrast, potent potentiation can occur at the physiological pH in both CHO cells (EC50 = 56.1 nM) and cortical neurons (threshold concentration < 10 nM). PcTx1 potentiates ASIC1a/2a by increasing the apparent affinity of channel activation for protons. As such, potentiation is the strongest at moderate pHs, diminishing with increasing proton concentrations. Our findings identify PcTx1 as a valuable tool for studying ASIC1a/2a function and contribute significantly to the understanding of the diverse and complex pharmacology of PcTx1.
Collapse
|
61
|
Stephan G, Huang L, Tang Y, Vilotti S, Fabbretti E, Yu Y, Nörenberg W, Franke H, Gölöncsér F, Sperlágh B, Dopychai A, Hausmann R, Schmalzing G, Rubini P, Illes P. The ASIC3/P2X3 cognate receptor is a pain-relevant and ligand-gated cationic channel. Nat Commun 2018; 9:1354. [PMID: 29636447 PMCID: PMC5893604 DOI: 10.1038/s41467-018-03728-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 03/09/2018] [Indexed: 12/21/2022] Open
Abstract
Two subclasses of acid-sensing ion channels (ASIC3) and of ATP-sensitive P2X receptors (P2X3Rs) show a partially overlapping expression in sensory neurons. Here we report that both recombinant and native receptors interact with each other in multiple ways. Current measurements with the patch-clamp technique prove that ASIC3 stimulation strongly inhibits the P2X3R current partly by a Ca2+-dependent mechanism. The proton-binding site is critical for this effect and the two receptor channels appear to switch their ionic permeabilities during activation. Co-immunoprecipation proves the close association of the two protein structures. BN-PAGE and SDS-PAGE analysis is also best reconciled with the view that ASIC3 and P2X3Rs form a multiprotein structure. Finally, in vivo measurements in rats reveal the summation of pH and purinergically induced pain. In conclusion, the receptor subunits do not appear to form a heteromeric channel, but tightly associate with each other to form a protein complex, mediating unidirectional inhibition. Two subclasses of ligand-gated ion channels (ASIC3 and P2X3) are both present at sensory neurons and might be therefore subject to receptor crosstalk. Here authors use electrophysiology, biochemistry and co-immunoprecipitation to show that the two ion channels interact and affect P2X3 currents.
Collapse
Affiliation(s)
- Gabriele Stephan
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Lumei Huang
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany.,Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Sandra Vilotti
- Neurobiology Sector, International School for Advanced Studies, Trieste, 34136, Italy
| | - Elsa Fabbretti
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Ye Yu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai, 100025, China
| | - Wolfgang Nörenberg
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Heike Franke
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Flóra Gölöncsér
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1043, Hungary.,János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Budapest, 1043, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1043, Hungary
| | - Anke Dopychai
- Molecular Pharmacology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, 52072, Germany
| | - Ralf Hausmann
- Molecular Pharmacology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, 52072, Germany
| | - Günther Schmalzing
- Molecular Pharmacology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, 52072, Germany
| | - Patrizia Rubini
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany.
| |
Collapse
|
62
|
Abstract
Metastatic bone pain is a complex, poorly understood process. Understanding the unique mechanisms causing cancer-induced bone pain may lead to potential therapeutic targets. This article discusses the effects of osteoclast overstimulation within the tumor microenvironment; the role of inflammatory factors at the tumor-nociceptor interface; the development of structural instability, causing mechanical nerve damage; and, ultimately, the neuroplastic changes in the setting of sustained pain. Several adjuvant therapies are available to attenuate metastatic bone pain. This article discusses the role of pharmacologic therapies, surgery, kyphoplasty, vertebroplasty, and radiofrequency ablation.
Collapse
Affiliation(s)
- Nicholas Figura
- Department of Radiation Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | - Joshua Smith
- Department of Supportive Care Medicine, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Hsiang-Hsuan Michael Yu
- Department of Radiation Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
63
|
Gonzales EB, Sumien N. Acidity and Acid-Sensing Ion Channels in the Normal and Alzheimer's Disease Brain. J Alzheimers Dis 2018; 57:1137-1144. [PMID: 28211811 DOI: 10.3233/jad-161131] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease prevalence has reached epidemic proportion with very few treatment options, which are associated with a multitude of side effects. A potential avenue of research for new therapies are protons, and their associated receptor: acid-sensing ion channels (ASIC). Protons are often overlooked neurotransmitters, and proton-gated currents have been identified in the brain. Furthermore, ASICs have been determined to be crucial for proper brain function. While there is more work to be done, this review is intended to highlight protons as neurotransmitters and their role along with the role of ASICs within physiological functioning of the brain. We will also cover the pathophysiological associations between ASICs and modulators of ASICs. Finally, this review will sum up how the studies of protons, ASICs and their modulators may generate new therapeutic molecules for Alzheimer's disease and other neurodegenerative diseases.
Collapse
|
64
|
Osmakov DI, Koshelev SG, Andreev YA, Dubinnyi MA, Kublitski VS, Efremov RG, Sobolevsky AI, Kozlov SA. Proton-independent activation of acid-sensing ion channel 3 by an alkaloid, lindoldhamine, from Laurus nobilis. Br J Pharmacol 2018; 175:924-937. [PMID: 29277899 DOI: 10.1111/bph.14134] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/05/2017] [Accepted: 12/07/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Acid-sensing ion channels (ASICs) play an important role in synaptic plasticity and learning, as well as in nociception and mechanosensation. ASICs are involved in pain and in neurological and psychiatric diseases, but their therapeutic potential is limited by the lack of ligands activating them at physiological pH. EXPERIMENTAL APPROACH We extracted, purified and determined the structure of a bisbenzylisoquinoline alkaloid, lindoldhamine, (LIN) from laurel leaves. Its effect on ASIC3 channels were characterized, using two-electrode voltage-clamp electrophysiological recordings from Xenopus laevis oocytes. KEY RESULTS At pH 7.4 or higher, LIN activated a sustained, proton-independent, current through rat and human ASIC3 channels, but not rat ASIC1a or ASIC2a channels. LIN also potentiated proton-induced transient currents and promoted recovery from desensitization in human, but not rat, ASIC3 channels. CONCLUSIONS AND IMPLICATIONS We describe a novel ASIC subtype-specific agonist LIN, which induced proton-independent activation of human and rat ASIC3 channels at physiological pH. LIN also acts as a positive allosteric modulator of human, but not rat, ASIC3 channels. This unique, species-selective, ligand of ASIC3, opens new avenues in studies of ASIC structure and function, as well as providing new approaches to drug design.
Collapse
Affiliation(s)
- Dmitry I Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sergey G Koshelev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yaroslav A Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maxim A Dubinnyi
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vadim S Kublitski
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Sergey A Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
65
|
Cherezova AL, Negulyaev JA, Zenin VV, Semenova SB. Extracellular pH Regulates the Entry of Calcium into Jurkat T-cells. CELL AND TISSUE BIOLOGY 2018; 12:41-47. [DOI: 10.1134/s1990519x18010042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Indexed: 04/08/2025]
|
66
|
Schuhmacher LN, Callejo G, Srivats S, Smith ESJ. Naked mole-rat acid-sensing ion channel 3 forms nonfunctional homomers, but functional heteromers. J Biol Chem 2017; 293:1756-1766. [PMID: 29237731 PMCID: PMC5798305 DOI: 10.1074/jbc.m117.807859] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/05/2017] [Indexed: 12/16/2022] Open
Abstract
Acid-sensing ion channels (ASICs) form both homotrimeric and heterotrimeric ion channels that are activated by extracellular protons and are involved in a wide range of physiological and pathophysiological processes, including pain and anxiety. ASIC proteins can form both homotrimeric and heterotrimeric ion channels. The ASIC3 subunit has been shown to be of particular importance in the peripheral nervous system with pharmacological and genetic manipulations demonstrating a role in pain. Naked mole-rats, despite having functional ASICs, are insensitive to acid as a noxious stimulus and show diminished avoidance of acidic fumes, ammonia, and carbon dioxide. Here we cloned naked mole-rat ASIC3 (nmrASIC3) and used a cell-surface biotinylation assay to demonstrate that it traffics to the plasma membrane, but using whole-cell patch clamp electrophysiology we observed that nmrASIC3 is insensitive to both protons and the non-proton ASIC3 agonist 2-guanidine-4-methylquinazoline. However, in line with previous reports of ASIC3 mRNA expression in dorsal root ganglia neurons, we found that the ASIC3 antagonist APETx2 reversibly inhibits ASIC-like currents in naked mole-rat dorsal root ganglia neurons. We further show that like the proton-insensitive ASIC2b and ASIC4, nmrASIC3 forms functional, proton-sensitive heteromers with other ASIC subunits. An amino acid alignment of ASIC3s between 9 relevant rodent species and human identified unique sequence differences that might underlie the proton insensitivity of nmrASIC3. However, introducing nmrASIC3 differences into rat ASIC3 (rASIC3) produced only minor differences in channel function, and replacing the nmrASIC3 sequence with that of rASIC3 did not produce a proton-sensitive ion channel. Our observation that nmrASIC3 forms nonfunctional homomers may reflect a further adaptation of the naked mole-rat to living in an environment with high-carbon dioxide levels.
Collapse
Affiliation(s)
- Laura-Nadine Schuhmacher
- From the Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Gerard Callejo
- From the Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Shyam Srivats
- From the Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Ewan St John Smith
- From the Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| |
Collapse
|
67
|
Wei J, Li M, Wang D, Zhu H, Kong X, Wang S, Zhou YL, Ju Z, Xu GY, Jiang GQ. Overexpression of suppressor of cytokine signaling 3 in dorsal root ganglion attenuates cancer-induced pain in rats. Mol Pain 2017; 13:1744806916688901. [PMID: 28326931 PMCID: PMC5302175 DOI: 10.1177/1744806916688901] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Cancer-induced pain (CIP) is one of the most severe types of chronic pain with which clinical treatment remains challenging and the involved mechanisms are largely unknown. Suppressor of cytokine signaling 3 (SOCS3) is an important intracellular protein and provides a classical negative feedback loop, thus involving in a wide variety of processes including inflammation and nociception. However, the role of SOCS3 pathway in CIP is poorly understood. The present study was designed to investigate the role of SOCS3 in dorsal root ganglion (DRG) in the development of CIP. Method CIP was established by injection of Walker 256 mammary gland tumor cells into the rat tibia canal. Whole-cell patch clamping and Western blotting were performed. Results Following the development of bone cancer, SOCS3 expression was significantly downregulated in rat DRGs at L2-L5 segments. Overexpression of SOCS3, using lentiviral-mediated production of SOCS3 at spinal cord level, drastically attenuated mechanical allodynia and body weight-bearing difference, but not thermal hyperalgesia in bone cancer rats. In addition, overexpression of SOCS3 reversed the hyperexcitability of DRG neurons innervating the tibia, and reduced abnormal expression of toll-like receptors 4 in the DRGs. Conclusions These results suggest that SOCS3 might be a key molecular involved in the development of complicated cancer pain and that overexpression of SOCS3 might be an important strategy for treatment for mechanical allodynia associated with bone cancer.
Collapse
Affiliation(s)
- Jinrong Wei
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China
| | - Meng Li
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China
| | - Dieyu Wang
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China
| | - Hongyan Zhu
- 2 Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, P.R. China
| | - Xiangpeng Kong
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China
| | - Shusheng Wang
- 2 Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, P.R. China
| | - You-Lang Zhou
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China
| | - Zhong Ju
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China
| | - Guang-Yin Xu
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China.,2 Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, P.R. China
| | - Guo-Qin Jiang
- 1 Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, P.R. China
| |
Collapse
|
68
|
Yu H, Yang L, Liu L, Zhao X, Huang X. Molecular dynamics simulations investigate the mechanism of Psalmotoxin 1 regulating gating process of an acid-sensing ion channel 1a at pH 5.5. J Biomol Struct Dyn 2017; 36:2558-2566. [PMID: 28768463 DOI: 10.1080/07391102.2017.1363087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Acid-sensing ion channel 1a (ASIC1a) is a cation channel activated by protons and causes neuronal death through central nervous system. Psalmotoxin1 (PcTx1) is a gating modifier for ASIC1a. The process of PcTx1 regulating the channel gating from the extracellular domain to the transmembrane domain is unclear. Here we used molecular dynamics (MD) simulations method to investigate how PcTx1 regulates the gating of the ASIC1a. Our results indicated that PcTx1can mainly regulate ASIC1a gating process through hydrogen bonds, which can affect their relative positions of several key domains in ASIC1a, further, a long-range conformational changes path was determined, which is composed of β1, β2, β10, α6, α7, β11, and β12 in ASIC1a.
Collapse
Affiliation(s)
- Hui Yu
- a Chemistry Teaching Center, College of Chemistry and Biology , Beihua University , Jilin , People's Republic of China
| | - Lianjuan Yang
- b The Fungal Reference Laboratory of Shanghai Dermatology Hospital , Shanghai , China
| | - Lu Liu
- c Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , People's Republic of China
| | - Xi Zhao
- c Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , People's Republic of China
| | - Xuri Huang
- c Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , People's Republic of China
| |
Collapse
|
69
|
Drummond HA, Xiang L, Chade AR, Hester R. Enhanced maximal exercise capacity, vasodilation to electrical muscle contraction, and hind limb vascular density in ASIC1a null mice. Physiol Rep 2017; 5:e13368. [PMID: 28784852 PMCID: PMC5555894 DOI: 10.14814/phy2.13368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 12/15/2022] Open
Abstract
Acid-sensing ion channel (ASIC) proteins form extracellular proton-gated, cation-selective channels in neurons and vascular smooth muscle cells and are proposed to act as extracellular proton sensors. However, their importance to vascular responses under conditions associated with extracellular acidosis, such as strenuous exercise, is unclear. Therefore, the purpose of this study was to determine if one ASIC protein, ASIC1a, contributes to extracellular proton-gated vascular responses and exercise tolerance. To determine if ASIC1a contributes to exercise tolerance, we determined peak oxygen (O2) uptake in conscious ASIC1a-/- mice during exhaustive treadmill running. Loss of ASIC1a was associated with a greater peak running speed (60 ± 2 vs. 53 ± 3 m·min-1, P = 0.049) and peak oxygen (O2) uptake during exhaustive treadmill running (9563 ± 120 vs. 8836 ± 276 mL·kg-1·h-1, n = 6-7, P = 0.0082). There were no differences in absolute or relative lean body mass, as determined by EchoMRI. To determine if ASIC1a contributes to vascular responses during muscle contraction, we measured femoral vascular conductance (FVC) during a stepwise electrical stimulation (0.5-5.0 Hz at 3 V for 60 sec) of the left major hind limb muscles. FVC increased to a greater extent in ASIC1a-/- versus ASIC1a+/+ mice (0.44 ± 0.03 vs. 0.30 ± 0.04 mL·min-1·100 g hind limb mass-1 · mmHg-1, n = 5 each, P = 0.0009). Vasodilation following local application of external protons in the spinotrapezius muscle increased the duration, but not the magnitude, of the vasodilatory response in ASIC1a-/- mice. Finally, we examined hind limb vascular density using micro-CT and found increased density of 0-80 μm vessels (P < 0.05). Our findings suggest an increased vascular density and an enhanced vasodilatory response to local protons, to a lesser degree, may contribute to the enhanced vascular conductance and increased peak exercise capacity in ASIC1a-/- mice.
Collapse
Affiliation(s)
- Heather A Drummond
- Department of Physiology and Biophysics and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lusha Xiang
- Department of Physiology and Biophysics and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Alejandro R Chade
- Department of Physiology and Biophysics and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Robert Hester
- Department of Physiology and Biophysics and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
70
|
Effect of an Acid-sensing Ion Channels Inhibitor on Pain-related Behavior by Nucleus Pulposus Applied on the Nerve Root in Rats. Spine (Phila Pa 1976) 2017; 42:E633-E641. [PMID: 27879566 DOI: 10.1097/brs.0000000000001918] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Controlled, interventional animal study. OBJECTIVE To examine the effect of an inhibitor of acid-sensing ion channel 3 (ASIC3) on pain-related behavior induced by application of the nucleus pulposus (NP) onto the dorsal root ganglion (DRG) in rats. SUMMARY OF BACKGROUND DATA ASIC3 is associated with acidosis pain in inflamed or ischemic tissues and is expressed in sensory neurons and NP cells. The ASIC3 inhibitor, APETx2, increases the mechanical threshold of pain in models of knee osteoarthritis or postoperative pain. However, the efficacy of APETx2 for pain relief in the NP application model remains unknown. METHODS Autologous NP was applied to the left L5 nerve root of 183 adult female Sprague-Dawley rats. The DRGs were treated with NP plus one of the following four treatments: saline solution (SM), low (0.01 μg: LD), medium (0.1 μg: MD), or high dose (1.0 μg: HD) of APETx2. Behavioral testing was performed to investigate the mechanical withdrawal threshold using von Frey hairs. Expression of nerve growth factor, hypoxia-inducible factor-1α (HIF1α), activating transcription factor-3, and ionized calcium-binding adaptor molecule-1 was evaluated using immunohistochemistry. Statistical differences among multiple groups were assessed using the Steel test, the Tukey-Kramer test, and the Dunnett test. P < 0.05 were considered significant. RESULTS The thresholds in the HD group were higher than those in the SM group at Days 14 and 21 (P < 0.05). In the MD group, the threshold was higher than in the SM group at Day 14 (P < 0.05). High doses of APETx2 reduced the expression of HIF1α after Day 14 compared with the SM group (P < 0.05). CONCLUSION APETx2 significantly improved pain-related behavior in a dose-dependent manner. APETx2 may inhibit ASIC3 and partly inhibit Nav1.8 channels. This ASIC3 channel inhibitor may be a potential therapeutic agent in early-stage lumbar disc herniation. LEVEL OF EVIDENCE N/A.
Collapse
|
71
|
Hoshikawa M, Kato A, Hojo H, Shibata Y, Kumamoto N, Watanabe M, Ugawa S. Distribution of ASIC4 transcripts in the adult wild-type mouse brain. Neurosci Lett 2017; 651:57-64. [PMID: 28461138 DOI: 10.1016/j.neulet.2017.03.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 01/27/2023]
Abstract
Acid-sensing ion channel 4 (ASIC4) belongs to the ASIC gene family of neuronal proton-gated cation channels, and is the least understood subtype among the members. Previous studies of ASIC4 expression in the mammalian central nervous system have shown that ASIC4 is abundantly expressed in the spinal cord and in various brain regions, such as the cerebral cortex, the hippocampus, and the cerebellum. However, the detailed distribution of ASIC4 transcripts in mammalian brains still remains to be elucidated. In the present study, radioactive in situ hybridization histochemistry with an ASIC4-specific cRNA probe was performed on wild-type mouse brains, followed by X-gal staining experiments with Asic4-lacZ reporter mice Asic4tm1a(KOMP)Mbp. It was found that ASIC4 mRNAs were widely expressed throughout the wild-type brain, but preferentially concentrated in the olfactory bulb, the piriform cortex, the caudate putamen, the preoptic area, the paraventricular nucleus, the medial habenular nucleus, the pretectal area, the lateral geniculate nucleus, the amygdaloid complex, the superior colliculus, the interpeduncular nucleus, and the granule cell layer of the ventral hippocampus, and these results were in agreement with the X-gal-positive reactions observed in the mutant brain. In addition, X-gal staining combined with immunohistochemistry identified intense signals for ASIC4 transcriptional activity in most of the choline acetyltransferase (ChAT)-positive principal neurons located in the basal forebrain cholinergic nuclei. Our data provide useful information to speculate possible roles of ASIC4 in diverse brain functions.
Collapse
Affiliation(s)
- M Hoshikawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - A Kato
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - H Hojo
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Y Shibata
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - N Kumamoto
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - M Watanabe
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - S Ugawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan.
| |
Collapse
|
72
|
Acid-sensing ion channel (ASIC) structure and function: Insights from spider, snake and sea anemone venoms. Neuropharmacology 2017; 127:173-184. [PMID: 28457973 DOI: 10.1016/j.neuropharm.2017.04.042] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/07/2017] [Accepted: 04/27/2017] [Indexed: 01/14/2023]
Abstract
Acid-sensing ion channels (ASICs) are proton-activated cation channels that are expressed in a variety of neuronal and non-neuronal tissues. As proton-gated channels, they have been implicated in many pathophysiological conditions where pH is perturbed. Venom derived compounds represent the most potent and selective modulators of ASICs described to date, and thus have been invaluable as pharmacological tools to study ASIC structure, function, and biological roles. There are now ten ASIC modulators described from animal venoms, with those from snakes and spiders favouring ASIC1, while the sea anemones preferentially target ASIC3. Some modulators, such as the prototypical ASIC1 modulator PcTx1 have been studied in great detail, while some of the newer members of the club remain largely unstudied. Here we review the current state of knowledge on venom derived ASIC modulators, with a particular focus on their molecular interaction with ASICs, what they have taught us about channel structure, and what they might still reveal about ASIC function and pathophysiological roles. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
|
73
|
Rash LD. Acid-Sensing Ion Channel Pharmacology, Past, Present, and Future …. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 79:35-66. [PMID: 28528673 DOI: 10.1016/bs.apha.2017.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
pH is one of the most strictly controlled parameters in mammalian physiology. An extracellular pH of ~7.4 is crucial for normal physiological processes, and perturbations to this have profound effects on cell function. Acidic microenvironments occur in many physiological and pathological conditions, including inflammation, bone remodeling, ischemia, trauma, and intense synaptic activity. Cells exposed to these conditions respond in different ways, from tumor cells that thrive to neurons that are either suppressed or hyperactivated, often fatally. Acid-sensing ion channels (ASICs) are primary pH sensors in mammals and are expressed widely in neuronal and nonneuronal cells. There are six main subtypes of ASICs in rodents that can form homo- or heteromeric channels resulting in many potential combinations. ASICs are present and activated under all of the conditions mentioned earlier, suggesting that they play an important role in how cells respond to acidosis. Compared to many other ion channel families, ASICs were relatively recently discovered-1997-and there is a substantial lack of potent, subtype-selective ligands that can be used to elucidate their structural and functional properties. In this chapter I cover the history of ASIC channel pharmacology, which began before the proteins were even identified, and describe the current arsenal of tools available, their limitations, and take a glance into the future to predict from where new tools are likely to emerge.
Collapse
Affiliation(s)
- Lachlan D Rash
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
74
|
Fló M, Margenat M, Pellizza L, Graña M, Durán R, Báez A, Salceda E, Soto E, Alvarez B, Fernández C. Functional diversity of secreted cestode Kunitz proteins: Inhibition of serine peptidases and blockade of cation channels. PLoS Pathog 2017; 13:e1006169. [PMID: 28192542 PMCID: PMC5325619 DOI: 10.1371/journal.ppat.1006169] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 02/24/2017] [Accepted: 01/06/2017] [Indexed: 01/01/2023] Open
Abstract
We previously reported a multigene family of monodomain Kunitz proteins from Echinococcus granulosus (EgKU-1-EgKU-8), and provided evidence that some EgKUs are secreted by larval worms to the host interface. In addition, functional studies and homology modeling suggested that, similar to monodomain Kunitz families present in animal venoms, the E. granulosus family could include peptidase inhibitors as well as channel blockers. Using enzyme kinetics and whole-cell patch-clamp, we now demonstrate that the EgKUs are indeed functionally diverse. In fact, most of them behaved as high affinity inhibitors of either chymotrypsin (EgKU-2-EgKU-3) or trypsin (EgKU-5-EgKU-8). In contrast, the close paralogs EgKU-1 and EgKU-4 blocked voltage-dependent potassium channels (Kv); and also pH-dependent sodium channels (ASICs), while showing null (EgKU-1) or marginal (EgKU-4) peptidase inhibitory activity. We also confirmed the presence of EgKUs in secretions from other parasite stages, notably from adult worms and metacestodes. Interestingly, data from genome projects reveal that at least eight additional monodomain Kunitz proteins are encoded in the genome; that particular EgKUs are up-regulated in various stages; and that analogous Kunitz families exist in other medically important cestodes, but not in trematodes. Members of this expanded family of secreted cestode proteins thus have the potential to block, through high affinity interactions, the function of host counterparts (either peptidases or cation channels) and contribute to the establishment and persistence of infection. From a more general perspective, our results confirm that multigene families of Kunitz inhibitors from parasite secretions and animal venoms display a similar functional diversity and thus, that host-parasite co-evolution may also drive the emergence of a new function associated with the Kunitz scaffold.
Collapse
Affiliation(s)
- Martín Fló
- Cátedra de Inmunología, Facultad de Química, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Mariana Margenat
- Cátedra de Inmunología, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Leonardo Pellizza
- Cátedra de Inmunología, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Martín Graña
- Unidad de Bioinformática, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Rosario Durán
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo and Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Adriana Báez
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Emilio Salceda
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Enrique Soto
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Beatriz Alvarez
- Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Fernández
- Cátedra de Inmunología, Facultad de Química, Universidad de la República, Montevideo, Uruguay
- * E-mail:
| |
Collapse
|
75
|
Acid-Sensing Ion Channels Activated by Evoked Released Protons Modulate Synaptic Transmission at the Mouse Calyx of Held Synapse. J Neurosci 2017; 37:2589-2599. [PMID: 28159907 DOI: 10.1523/jneurosci.2566-16.2017] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 01/10/2017] [Accepted: 01/13/2017] [Indexed: 12/30/2022] Open
Abstract
Acid-sensing ion channels (ASICs) regulate synaptic activities and play important roles in neurodegenerative diseases. We found that these channels can be activated in neurons of the medial nucleus of the trapezoid body (MNTB) of the auditory system in the CNS. A drop in extracellular pH induces transient inward ASIC currents (IASICs) in postsynaptic MNTB neurons from wild-type mice. The inhibition of IASICs by psalmotoxin-1 (PcTx1) and the absence of these currents in knock-out mice for ASIC-1a subunit (ASIC1a-/-) suggest that homomeric ASIC-1as are mediating these currents in MNTB neurons. Furthermore, we detect ASIC1a-dependent currents during synaptic transmission, suggesting an acidification of the synaptic cleft due to the corelease of neurotransmitter and H+ from synaptic vesicles. These currents are capable of eliciting action potentials in the absence of glutamatergic currents. A significant characteristic of these homomeric ASIC-1as is their permeability to Ca2+ Activation of ASIC-1a in MNTB neurons by exogenous H+ induces an increase in intracellular Ca2+ Furthermore, the activation of postsynaptic ASIC-1as during high-frequency stimulation (HFS) of the presynaptic nerve terminal leads to a PcTx1-sensitive increase in intracellular Ca2+ in MNTB neurons, which is independent of glutamate receptors and is absent in neurons from ASIC1a-/- mice. During HFS, the lack of functional ASICs in synaptic transmission results in an enhanced short-term depression of glutamatergic EPSCs. These results strongly support the hypothesis of protons as neurotransmitters and demonstrate that presynaptic released protons modulate synaptic transmission by activating ASIC-1as at the calyx of Held-MNTB synapse.SIGNIFICANCE STATEMENT The manuscript demonstrates that postsynaptic neurons of the medial nucleus of the trapezoid body at the mouse calyx of Held synapse express functional homomeric Acid-sensing ion channel-1a (ASIC-1as) that can be activated by protons (coreleased with neurotransmitter from acidified synaptic vesicles). These ASIC-1as contribute to the generation of postsynaptic currents and, more relevant, to calcium influx, which could be involved in the modulation of presynaptic transmitter release. Inhibition or deletion of ASIC-1a leads to enhanced short-term depression, demonstrating that they are concerned with short-term plasticity of the synapse. ASICs represent a widespread communication system with unique properties. We expect that our experiments will have an impact in the neurobiology field and will spread in areas related to neuronal plasticity.
Collapse
|
76
|
Jiang N, Wu J, Leng T, Yang T, Zhou Y, Jiang Q, Wang B, Hu Y, Ji YH, Simon RP, Chu XP, Xiong ZG, Zha XM. Region specific contribution of ASIC2 to acidosis-and ischemia-induced neuronal injury. J Cereb Blood Flow Metab 2017; 37:528-540. [PMID: 26861816 PMCID: PMC5381448 DOI: 10.1177/0271678x16630558] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acidosis in the brain plays a critical role in neuronal injury in neurological diseases, including brain ischemia. One key mediator of acidosis-induced neuronal injury is the acid-sensing ion channels (ASICs). Current literature has focused on ASIC1a when studying acid signaling. The importance of ASIC2, which is also widely expressed in the brain, has not been appreciated. We found here a region-specific effect of ASIC2 on acid-mediated responses. Deleting ASIC2 reduced acid-activated current in cortical and striatal neurons, but had no significant effect in cerebellar granule neurons. In addition, we demonstrated that ASIC2 was important for ASIC1a expression, and that ASIC2a but not 2b facilitated ASIC1a surface trafficking in the brain. Further, we showed that ASIC2 deletion attenuated acidosis/ischemia-induced neuronal injury in organotypic hippocampal slices but had no effect in organotypic cerebellar slices. Consistent with an injurious role of ASIC2, we showed that ASIC2 deletion significantly protected the mouse brain from ischemic damage in vivo. These data suggest a critical region-specific contribution of ASIC2 to neuronal injury and reveal an important functional difference between ASIC2a and 2b in the brain.
Collapse
Affiliation(s)
- Nan Jiang
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA.,2 School of Life Sciences, Shanghai University, Shanghai, China
| | - Junjun Wu
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA.,3 China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Tiandong Leng
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Tao Yang
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Yufan Zhou
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA
| | - Qian Jiang
- 5 Department of Basic Medical Science, University of Missouri-Kansas City, Kansas City, USA
| | - Bin Wang
- 6 Department of Mathematics and Statistics, University of South Alabama, Mobile, USA
| | - Youjia Hu
- 3 China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Yong-Hua Ji
- 2 School of Life Sciences, Shanghai University, Shanghai, China
| | - Roger P Simon
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Xiang-Ping Chu
- 5 Department of Basic Medical Science, University of Missouri-Kansas City, Kansas City, USA
| | - Zhi-Gang Xiong
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Xiang-Ming Zha
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA
| |
Collapse
|
77
|
Schuhmacher LN, Smith ESJ. Expression of acid-sensing ion channels and selection of reference genes in mouse and naked mole rat. Mol Brain 2016; 9:97. [PMID: 27964758 PMCID: PMC5154015 DOI: 10.1186/s13041-016-0279-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/02/2016] [Indexed: 12/19/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are a family of ion channels comprised of six subunits encoded by four genes and they are expressed throughout the peripheral and central nervous systems. ASICs have been implicated in a wide range of physiological and pathophysiological processes: pain, breathing, synaptic plasticity and excitotoxicity. Unlike mice and humans, naked mole-rats do not perceive acid as a noxious stimulus, even though their sensory neurons express functional ASICs, likely an adaptation to living in a hypercapnic subterranean environment. Previous studies of ASIC expression in the mammalian nervous system have often not examined all subunits, or have failed to adequately quantify expression between tissues; to date there has been no attempt to determine ASIC expression in the central nervous system of the naked mole-rat. Here we perform a geNorm study to identify reliable housekeeping genes in both mouse and naked mole-rat and then use quantitative real-time PCR to estimate the relative amounts of ASIC transcripts in different tissues of both species. We identify RPL13A (ribosomal protein L13A) and CANX (calnexin), and β-ACTIN and EIF4A (eukaryotic initiation factor 4a) as being the most stably expressed housekeeping genes in mouse and naked mole-rat, respectively. In both species, ASIC3 was most highly expressed in dorsal root ganglia (DRG), and ASIC1a, ASIC2b and ASIC3 were more highly expressed across all brain regions compared to the other subunits. We also show that ASIC4, a proton-insensitive subunit of relatively unknown function, was highly expressed in all mouse tissues apart from DRG and hippocampus, but was by contrast the lowliest expressed ASIC in all naked mole-rat tissues.
Collapse
Affiliation(s)
- Laura-Nadine Schuhmacher
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK.,Department of Cell & Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK.
| |
Collapse
|
78
|
Acid-sensing ion channels are expressed in the ventrolateral medulla and contribute to central chemoreception. Sci Rep 2016; 6:38777. [PMID: 27934921 PMCID: PMC5146928 DOI: 10.1038/srep38777] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 11/14/2016] [Indexed: 12/30/2022] Open
Abstract
The role of acid-sensing ion channels (ASICs) in the ventrolateral medulla (VLM) remains uncertain. Here, we found that ASIC1a and ASIC2 are widely expressed in rat medulla, and the expression level is higher at neonatal stage as compared to adult stage. The two ASIC subunits co-localized in medualla neurons. Furthermore, pH reduction triggered typical ASIC-type currents in the medulla, including the VLM. These currents showed a pH50 value of 6.6 and were blocked by amiloride. Based on their sensitivity to psalmotoxin 1 (PcTx1) and zinc, homomeric ASIC1a and heteromeric ASIC1a/2 channels were likely responsible for acid-mediated currents in the mouse medulla. ASIC currents triggered by pH 5 disappeared in the VLM neurons from ASIC1−/−, but not ASIC2−/− mice. Activation of ASICs in the medulla also triggered neuronal excitation. Moreover, microinjection of artificial cerebrospinal fluid at a pH of 6.5 into the VLM increased integrated phrenic nerve discharge, inspiratory time and respiratory drive in rats. Both amiloride and PcTx1 inhibited the acid-induced stimulating effect on respiration. Collectively, our data suggest that ASICs are highly expressed in the medulla including the VLM, and activation of ASICs in the VLM contributes to central chemoreception.
Collapse
|
79
|
Lei Z, Sami Shaikh A, Zheng W, Yu X, Yu J, Li J. Non-proton ligand-sensing domain of acid-sensing ion channel 3 is required for itch sensation. J Neurochem 2016; 139:1093-1101. [PMID: 27770439 DOI: 10.1111/jnc.13869] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/30/2016] [Accepted: 10/04/2016] [Indexed: 02/05/2023]
Abstract
Itch, the unpleasant sensation that evokes a desire to scratch, accompanies numerous skin and nervous system disorders. However, the molecular mechanisms of itch are unclear. Acid-sensing ion channel 3 (ASIC3) is a sensor of acidic and primary inflammatory pain. The whole-cell patch clamp technique was used to determine the effect of chloroquine (CQ) on ASICs currents in primary sensory neurons or the Chinese hamster ovary cells transfected with rat ASIC1a or ASIC3. Site-directed mutagenesis of plasmid was performed. Scratching behavior was evaluated by measuring the number of bouts during 30 min after injection. CQ, an anti-malarial drug defined as a histamine-independent pruritogen, selectively enhanced the sustained phase of ASIC3 current in a concentration-dependent manner either in ASIC3-transfected Chinese hamster ovary cells or in primary cultured rat dorsal root ganglion neurons. Further studies revealed that the effect of CQ on ASIC3 channels depends on the newly identified non-proton ligand-sensing domain. Importantly, CQ-evoked scratching behavior was largely alleviated by APETx2, a selective ASIC3 channel blocker. Like CQ, other compounds such as amiloride, 2-guanidine-4-methylquinazoline and neuropeptide FF, which have been previously reported to be non-proton ligands that activate ASIC3, undoubtedly evoked the scratching response. In conclusion, ASIC3, a proton-gated ion channel critical for pain sensation, also functions as an essential component of itch transduction.
Collapse
Affiliation(s)
- Zhen Lei
- Department of Physiology, School of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China.,Department of Anesthesiology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Abdul Sami Shaikh
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenshuai Zheng
- Department of Physiology, School of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xiao Yu
- Department of Physiology, School of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Jingui Yu
- Department of Anesthesiology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Jingxin Li
- Department of Physiology, School of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
80
|
Abdelhamid RE, Sluka KA. ASICs Mediate Pain and Inflammation in Musculoskeletal Diseases. Physiology (Bethesda) 2016; 30:449-59. [PMID: 26525344 DOI: 10.1152/physiol.00030.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Chronic musculoskeletal pain is debilitating and affects ∼ 20% of adults. Tissue acidosis is present in painful musculoskeletal diseases like rheumatoid arthritis. ASICs are located on skeletal muscle and joint nociceptors as well as on nonneuronal cells in the muscles and joints, where they mediate nociception. This review discusses the properties of different types of ASICs, factors affecting their pH sensitivity, and their role in musculoskeletal hyperalgesia and inflammation.
Collapse
Affiliation(s)
- Ramy E Abdelhamid
- Department of Physical Therapy and Rehabilitation Science, Neuroscience Graduate Program, Pain Research Program, University of Iowa, Iowa City, Iowa
| | - Kathleen A Sluka
- Department of Physical Therapy and Rehabilitation Science, Neuroscience Graduate Program, Pain Research Program, University of Iowa, Iowa City, Iowa
| |
Collapse
|
81
|
Ergonul Z, Yang L, Palmer LG. Properties of acid-induced currents in mouse dorsal root ganglia neurons. Physiol Rep 2016; 4:4/9/e12795. [PMID: 27173673 PMCID: PMC4873640 DOI: 10.14814/phy2.12795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 04/15/2016] [Indexed: 11/29/2022] Open
Abstract
Acid‐sensing ion channels (ASICs) are cation channels that are activated by protons (H+). They are expressed in neurons throughout the nervous system and may play important roles in several neurologic disorders including inflammation, cerebral ischemia, seizures, neurodegeneration, anxiety, depression, and migraine. ASICs generally produce transient currents that desensitize in response to a decrease in extracellular pH. Under certain conditions, the inactivation of ASICs can be incomplete and allow them to produce sustained currents. Here, we characterize the properties of both transient and sustained acid‐induced currents in cultured mouse dorsal root ganglia (DRG) neurons. At pH levels between 7.3 and 7.1 they include “window currents” through ASICs. With stronger acid signals sustained currents are maintained in the absence of extracellular Na+ or the presence of the ASIC blockers amiloride and Psalmotoxin‐1(PcTx1). These sustained responses may have several different origins in these cells, including acid‐induced stimulation of inward Cl− currents, block of outward K+ currents, and augmentation of inward H+ currents, properties that distinguish these novel sustained currents from the well‐characterized transient currents.
Collapse
Affiliation(s)
- Zuhal Ergonul
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York Department of Pediatrics, NewYork-Presbyterian/Weill Cornell Medical Center, New York, New York
| | - Lei Yang
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York Department of Physiology, Harbin Medical University, Harbin, China
| | - Lawrence G Palmer
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| |
Collapse
|
82
|
Boscardin E, Alijevic O, Hummler E, Frateschi S, Kellenberger S. The function and regulation of acid-sensing ion channels (ASICs) and the epithelial Na(+) channel (ENaC): IUPHAR Review 19. Br J Pharmacol 2016; 173:2671-701. [PMID: 27278329 DOI: 10.1111/bph.13533] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/19/2016] [Accepted: 06/02/2016] [Indexed: 12/30/2022] Open
Abstract
Acid-sensing ion channels (ASICs) and the epithelial Na(+) channel (ENaC) are both members of the ENaC/degenerin family of amiloride-sensitive Na(+) channels. ASICs act as proton sensors in the nervous system where they contribute, besides other roles, to fear behaviour, learning and pain sensation. ENaC mediates Na(+) reabsorption across epithelia of the distal kidney and colon and of the airways. ENaC is a clinically used drug target in the context of hypertension and cystic fibrosis, while ASIC is an interesting potential target. Following a brief introduction, here we will review selected aspects of ASIC and ENaC function. We discuss the origin and nature of pH changes in the brain and the involvement of ASICs in synaptic signalling. We expose how in the peripheral nervous system, ASICs cover together with other ion channels a wide pH range as proton sensors. We introduce the mechanisms of aldosterone-dependent ENaC regulation and the evidence for an aldosterone-independent control of ENaC activity, such as regulation by dietary K(+) . We then provide an overview of the regulation of ENaC by proteases, a topic of increasing interest over the past few years. In spite of the profound differences in the physiological and pathological roles of ASICs and ENaC, these channels share many basic functional and structural properties. It is likely that further research will identify physiological contexts in which ASICs and ENaC have similar or overlapping roles.
Collapse
Affiliation(s)
- Emilie Boscardin
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Omar Alijevic
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Edith Hummler
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|
83
|
Acid-Sensing Ion Channel 2a (ASIC2a) Promotes Surface Trafficking of ASIC2b via Heteromeric Assembly. Sci Rep 2016; 6:30684. [PMID: 27477936 PMCID: PMC4967927 DOI: 10.1038/srep30684] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/08/2016] [Indexed: 12/27/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-activated cation channels that play important roles as typical proton sensors during pathophysiological conditions and normal synaptic activities. Among the ASIC subunits, ASIC2a and ASIC2b are alternative splicing products from the same gene, ACCN1. It has been shown that ASIC2 isoforms have differential subcellular distribution: ASIC2a targets the cell surface by itself, while ASIC2b resides in the ER. However, the underlying mechanism for this differential subcellular localization remained to be further elucidated. By constructing ASIC2 chimeras, we found that the first transmembrane (TM1) domain and the proximal post-TM1 domain (17 amino acids) of ASIC2a are critical for membrane targeting of the proteins. We also observed that replacement of corresponding residues in ASIC2b by those of ASIC2a conferred proton-sensitivity as well as surface expression to ASIC2b. We finally confirmed that ASIC2b is delivered to the cell surface from the ER by forming heteromers with ASIC2a, and that the N-terminal region of ASIC2a is additionally required for the ASIC2a-dependent membrane targeting of ASIC2b. Together, our study supports an important role of ASIC2a in membrane targeting of ASIC2b.
Collapse
|
84
|
Joeres N, Augustinowski K, Neuhof A, Assmann M, Gründer S. Functional and pharmacological characterization of two different ASIC1a/2a heteromers reveals their sensitivity to the spider toxin PcTx1. Sci Rep 2016; 6:27647. [PMID: 27277303 PMCID: PMC4899722 DOI: 10.1038/srep27647] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 05/24/2016] [Indexed: 12/23/2022] Open
Abstract
Acid Sensing Ion Channels (ASICs) detect extracellular proton signals and are involved in synaptic transmission and pain sensation. ASIC subunits assemble into homo- and heteromeric channels composed of three subunits. Single molecule imaging revealed that heteromers composed of ASIC1a and ASIC2a, which are widely expressed in the central nervous system, have a flexible 2:1/1:2 stoichiometry. It was hitherto not possible, however, to functionally differentiate these two heteromers. To have a homogenous population of ASIC1a/2a heteromers with either 2:1 or 1:2 stoichiometry, we covalently linked subunits in the desired configuration and characterized their functional properties in Xenopus oocytes. We show that the two heteromers have slightly different proton affinity, with an additional ASIC1a subunit increasing apparent affinity. Moreover, we found that zinc, which potentiates ASIC2a-containing ASICs but not homomeric ASIC1a, potentiates both heteromers. Finally, we show that PcTx1, which binds at subunit-subunit interfaces of homomeric ASIC1a, inhibits both heteromers suggesting that ASIC2a can also contribute to a PcTx1 binding site. Using this functional fingerprint, we show that rat cortical neurons predominantly express the ASIC1a/2a heteromer with a 2:1 stoichiometry. Collectively, our results reveal the contribution of individual subunits to the functional properties of ASIC1a/2a heteromers.
Collapse
Affiliation(s)
- Niko Joeres
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Katrin Augustinowski
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Andreas Neuhof
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Marc Assmann
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| |
Collapse
|
85
|
Leng TD, Si HF, Li J, Yang T, Zhu M, Wang B, Simon RP, Xiong ZG. Amiloride Analogs as ASIC1a Inhibitors. CNS Neurosci Ther 2016; 22:468-76. [PMID: 26890278 DOI: 10.1111/cns.12524] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 12/30/2015] [Accepted: 01/16/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND ASIC1a, the predominant acid-sensing ion channels (ASICs), is implicated in neurological disorders including stroke, traumatic spinal cord injury, and ALS. Potent ASIC1a inhibitors should have promising therapeutic potential for ASIC1a-related diseases. AIMS We examined the inhibitory effects of a number of amiloride analogs on ASIC1a currents, aimed at understanding the structure-activity relationship and identifying potent ASIC1a inhibitors for stroke intervention. METHODS Whole-cell patch-clamp techniques and a mouse model of middle cerebral artery occlusion (MCAO)-induced focal ischemia were used. Surflex-Dock was used to dock the analogs into the pocket with default parameters. RESULTS Amiloride and its analogs inhibit ASIC1a currents expressed in Chinese hamster ovary cells with a potency rank order of benzamil > phenamil > 5-(N,N-dimethyl)amiloride (DMA) > amiloride > 5-(N,N-hexamethylene)amiloride (HMA) ≥ 5-(N-methyl-N-isopropyl)amiloride (MIA) > 5-(N-ethyl-N-isopropyl)amiloride (EIPA). In addition, amiloride and its analogs inhibit ASIC currents in cortical neurons with the same potency rank order. In mice, benzamil and EIPA decreased MCAO-induced infarct volume. Similar to its effect on the ASIC current, benzamil showed a much higher potency than EIPA. CONCLUSION Addition of a benzyl group to the terminal guanidinyl group resulted in enhanced inhibitory activity on ASIC1a. On the other hand, the bulky groups added to the 5-amino residues slightly decreased the activity. Among the tested amiloride analogs, benzamil is the most potent ASIC1a inhibitor.
Collapse
Affiliation(s)
- Tian-Dong Leng
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Hong-Fang Si
- Anhui Medical University, School of Pharmacy, Hefei, Anhui, China
| | - Jun Li
- Anhui Medical University, School of Pharmacy, Hefei, Anhui, China
| | - Tao Yang
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Mengyuan Zhu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Roger P Simon
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
86
|
Syntabulin regulates the trafficking of PICK1-containing vesicles in neurons. Sci Rep 2016; 6:20924. [PMID: 26868290 PMCID: PMC4751430 DOI: 10.1038/srep20924] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/08/2016] [Indexed: 11/08/2022] Open
Abstract
PICK1 (protein interacting with C-kinase 1) is a peripheral membrane protein that interacts with diverse membrane proteins. PICK1 has been shown to regulate the clustering and membrane localization of synaptic receptors such as AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors, metabotropic glutamate receptor 7, and ASICs (acid-sensing ion channels). Moreover, recent evidence suggests that PICK1 can mediate the trafficking of various vesicles out from the Golgi complex in several cell systems, including neurons. However, how PICK1 affects vesicle-trafficking dynamics remains unexplored. Here, we show that PICK1 mediates vesicle trafficking by interacting with syntabulin, a kinesin-binding protein that mediates the trafficking of both synaptic vesicles and mitochondria in axons. Syntabulin recruits PICK1 onto microtubule structures and mediates the trafficking of PICK1-containing vesicles along microtubules. In neurons, syntabulin alters PICK1 expression by recruiting PICK1 into axons and regulates the trafficking dynamics of PICK1-containing vesicles. Furthermore, we show that syntabulin forms a complex with PICK1 and ASICs, regulates ASIC protein expression in neurons, and participates in ASIC-induced acidotoxicity.
Collapse
|
87
|
Wu J, Xu Y, Jiang YQ, Xu J, Hu Y, Zha XM. ASIC subunit ratio and differential surface trafficking in the brain. Mol Brain 2016; 9:4. [PMID: 26746198 PMCID: PMC4706662 DOI: 10.1186/s13041-016-0185-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Background Acid-sensing ion channels (ASICs) are key mediators of acidosis-induced responses in neurons. However, little is known about the relative abundance of different ASIC subunits in the brain. Such data are fundamental for interpreting the relative contribution of ASIC1a homomers and 1a/2 heteromers to acid signaling, and essential for designing therapeutic interventions to target these channels. We used a simple biochemical approach and semi-quantitatively determined the molar ratio of ASIC1a and 2 subunits in mouse brain. Further, we investigated differential surface trafficking of ASIC1a, ASIC2a, and ASIC2b. Results and conclusions ASIC1a subunits outnumber the sum of ASIC2a and ASIC2b. There is a region-specific variation in ASIC2a and 2b expression, with cerebellum and striatum expressing predominantly 2b and 2a, respectively. Further, we performed surface biotinylation and found that surface ASIC1a and ASIC2a ratio correlates with their total expression. In contrast, ASIC2b exhibits little surface presence in the brain. This result is consistent with increased co-localization of ASIC2b with an ER marker in 3T3 cells. Our data are the first semi-quantitative determination of relative subunit ratio of various ASICs in the brain. The differential surface trafficking of ASICs suggests that the main functional ASICs in the brain are ASIC1a homomers and 1a/2a heteromers. This finding provides important insights into the relative contribution of various ASIC complexes to acid signaling in neurons.
Collapse
Affiliation(s)
- Junjun Wu
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, 5851 USA Dr N, MSB3074, Mobile, AL, 36688, USA. .,China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai, 201203, China.
| | - Yuanyuan Xu
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, 5851 USA Dr N, MSB3074, Mobile, AL, 36688, USA. .,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Yu-Qing Jiang
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, 5851 USA Dr N, MSB3074, Mobile, AL, 36688, USA. .,Department of Urology, The Third Hospital of Hebei Medical University, Shijiazhuang, HeBei, China.
| | - Jiangping Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Youjia Hu
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai, 201203, China.
| | - Xiang-ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, 5851 USA Dr N, MSB3074, Mobile, AL, 36688, USA.
| |
Collapse
|
88
|
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
|
89
|
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]
|
90
|
Guo Y, Chen J, Li J, Cheng L, Lin N. Unique roles played by Acid-sensing ion channel 2. Channels (Austin) 2015:0. [PMID: 26552578 DOI: 10.1080/19336950.2015.1106653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The discovery of Acid-sensing ion channels (ASICs) provided us the theoretical basis to understand the pathological acidic environment. They belong to the degenerin/epithelial Na+ channel family and function once extracellular pH decreases to a certain level, and this characteristic make them spotlights in the regulation or response of pH change. As a regulatory system, keeping the intra- and extra-balance seems to be significant for ASICs, in which ASIC2 plays an important role. We surprisingly noticed that ASIC2 owns some distinctive properties, including its inter-system regulation, specific distribution and transporting patterns, influence on cell migration and the unique role in mechanosensitivity. Therefore, to conclude the functions and characterisitics of ASIC2 indeed assist the understanding of interaction among ASICs subunits and the regulation from extracellular environment to ASICs.
Collapse
Affiliation(s)
- Yingjun Guo
- a Dept. of Orthopedic Surgery , Qilu Hospital, Shandong University . West Wenhua Road, No. 107, Ji'nan, Shandong Province , P.R. China . Zip code: 250012
| | - Jingying Chen
- b Dept. of Gynaecology and Obstetrics , Qilu Hospital, Shandong University . West Wenhua Road, No. 107, Ji'nan, Shandong Province , P.R. China. Zip code: 250012
| | - Jingkun Li
- a Dept. of Orthopedic Surgery , Qilu Hospital, Shandong University . West Wenhua Road, No. 107, Ji'nan, Shandong Province , P.R. China . Zip code: 250012
| | - Lei Cheng
- a Dept. of Orthopedic Surgery , Qilu Hospital, Shandong University . West Wenhua Road, No. 107, Ji'nan, Shandong Province , P.R. China . Zip code: 250012
| | - Nie Lin
- a Dept. of Orthopedic Surgery , Qilu Hospital, Shandong University . West Wenhua Road, No. 107, Ji'nan, Shandong Province , P.R. China . Zip code: 250012
| |
Collapse
|
91
|
Abstract
Sensory nerve fibers innervating the ocular anterior surface detect external stimuli producing innocuous and painful sensations. Protons are among the first mediators released by damaged cells during inflammation, tissue injury, or other chronic ophthalmic conditions. We studied whether acid-sensing ion channels (ASICs) are expressed in corneal sensory neurons and their roles in the response to moderate acidifications of the ocular surface and in pathologies producing ocular surface inflammation. Moderate acidic pH (6.6) activated ASIC-like currents in corneal sensory neurons, which were blocked by ASIC1- or ASIC3-specific toxins. Acidic pH depolarizes corneal sensory neurons to fire action potentials, an effect blocked by the ASIC3 inhibitor APETx2. 2-Guanidino-4-methylquinazoline, an ASIC3 agonist, activated a population of corneal polymodal sensory nerve fibers and significantly increased the blinking and tearing rate. The nocifensive behaviors produced by application of either a moderate acidic stimulus or ophthalmic drugs formulated in acidic solution were abolished by ASIC blockers. In a model of allergic keratoconjunctivitis, nocifensive behavior was greatly reduced by ASIC3 blockade, presumably by reducing nociceptor sensitization during the inflammatory process. Our results show that, in addition to the established role of TRPV1, ASICs play a significant role in the detection of acidic insults at the ocular surface. The identification of ASICs in corneal neurons and their alterations during different diseases is critical for the understanding of sensory ocular pathophysiology. They are likely to mediate some of the discomfort sensations accompanying several ophthalmic formulations and may represent novel targets for the development of new therapeutics for ocular pathologies.
Collapse
|
92
|
Osmakov DI, Andreev YA, Kozlov SA. Acid-sensing ion channels and their modulators. BIOCHEMISTRY (MOSCOW) 2015; 79:1528-45. [PMID: 25749163 DOI: 10.1134/s0006297914130069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
According to a modern look acid-sensing ion channels (ASICs) are one of the most important receptors that perceive pH change in the body. ASICs represent proton-gated Na+-selective channels, which are expressed in neurons of the central and peripheral nervous system. These channels are attracting attention of researchers around the world, as they are involved in various physiological processes in the body. Drop of pH may occur in tissues in norm (e.g. the accumulation of lactic acid, the release of protons upon ATP hydrolysis) and pathology (inflammation, ischemic stroke, tissue damage and seizure). These processes are accompanied by unpleasant pain sensations, which may be short-lived or can lead to chronic inflammatory diseases. Modulators of ASIC channels activity are potential candidates for new effective analgesic and neuroprotection drugs. This review summarizes available information about structure, function, and physiological role of ASIC channels. In addition a description of all known ligands of these channels and their practical relevance is provided.
Collapse
Affiliation(s)
- D I Osmakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| | | | | |
Collapse
|
93
|
Physical basis of apparent pore dilation of ATP-activated P2X receptor channels. Nat Neurosci 2015; 18:1577-83. [PMID: 26389841 DOI: 10.1038/nn.4120] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/25/2015] [Indexed: 12/11/2022]
Abstract
The selectivity of ion channels is fundamental for their roles in electrical and chemical signaling and in ion homeostasis. Although most ion channels exhibit stable ion selectivity, the prevailing view of purinergic P2X receptor channels, transient receptor potential V1 (TRPV1) channels and acid-sensing ion channels (ASICs) is that their ion conduction pores dilate upon prolonged activation. We investigated this mechanism in P2X receptors and found that the hallmark shift in equilibrium potential observed with prolonged channel activation does not result from pore dilation, but from time-dependent alterations in the concentration of intracellular ions. We derived a physical model to calculate ion concentration changes during patch-clamp recordings, which validated our experimental findings and provides a quantitative guideline for effectively controlling ion concentration. Our results have fundamental implications for understanding ion permeation and gating in P2X receptor channels, as well as more broadly for using patch-clamp techniques to study ion channels and neuronal excitability.
Collapse
|
94
|
Slosky LM, Largent-Milnes TM, Vanderah TW. Use of Animal Models in Understanding Cancer-induced Bone Pain. CANCER GROWTH AND METASTASIS 2015; 8:47-62. [PMID: 26339191 PMCID: PMC4552039 DOI: 10.4137/cgm.s21215] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/14/2015] [Accepted: 06/16/2015] [Indexed: 12/13/2022]
Abstract
Many common cancers have a propensity to metastasize to bone. Although malignancies often go undetected in their native tissues, bone metastases produce excruciating pain that severely compromises patient quality of life. Cancer-induced bone pain (CIBP) is poorly managed with existing medications, and its multifaceted etiology remains to be fully elucidated. Novel analgesic targets arise as more is learned about this complex and distinct pain state. Over the past two decades, multiple animal models have been developed to study CIBP’s unique pathology and identify therapeutic targets. Here, we review animal models of CIBP and the mechanistic insights gained as these models evolve. Findings from immunocompromised and immunocompetent host systems are discussed separately to highlight the effect of model choice on outcome. Gaining an understanding of the unique neuromolecular profile of cancer pain through the use of appropriate animal models will aid in the development of more effective therapeutics for CIBP.
Collapse
Affiliation(s)
- Lauren M Slosky
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Tally M Largent-Milnes
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Todd W Vanderah
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
95
|
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
|
96
|
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
|
97
|
Duzhyy DE, Viatchenko-Karpinski VY, Khomula EV, Voitenko NV, Belan PV. Upregulation of T-type Ca2+ channels in long-term diabetes determines increased excitability of a specific type of capsaicin-insensitive DRG neurons. Mol Pain 2015; 11:29. [PMID: 25986602 PMCID: PMC4490764 DOI: 10.1186/s12990-015-0028-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/13/2015] [Indexed: 01/15/2023] Open
Abstract
Background Previous studies have shown that increased excitability of capsaicin-sensitive DRG neurons and thermal hyperalgesia in rats with short-term (2–4 weeks) streptozotocin-induced diabetes is mediated by upregulation of T-type Ca2+ current. In longer–term diabetes (after the 8th week) thermal hyperalgesia is changed to hypoalgesia that is accompanied by downregulation of T-type current in capsaicin-sensitive small-sized nociceptors. At the same time pain symptoms of diabetic neuropathy other than thermal persist in STZ-diabetic animals and patients during progression of diabetes into later stages suggesting that other types of DRG neurons may be sensitized and contribute to pain. In this study, we examined functional expression of T-type Ca2+ channels in capsaicin-insensitive DRG neurons and excitability of these neurons in longer-term diabetic rats and in thermally hypoalgesic diabetic rats. Results Here we have demonstrated that in STZ-diabetes T-type current was upregulated in capsaicin-insensitive low-pH-sensitive small-sized nociceptive DRG neurons of longer-term diabetic rats and thermally hypoalgesic diabetic rats. This upregulation was not accompanied by significant changes in biophysical properties of T-type channels suggesting that a density of functionally active channels was increased. Sensitivity of T-type current to amiloride (1 mM) and low concentration of Ni2+ (50 μM) implicates prevalence of Cav3.2 subtype of T-type channels in the capsaicin-insensitive low-pH-sensitive neurons of both naïve and diabetic rats. The upregulation of T-type channels resulted in the increased neuronal excitability of these nociceptive neurons revealed by a lower threshold for action potential initiation, prominent afterdepolarizing potentials and burst firing. Sodium current was not significantly changed in these neurons during long-term diabetes and could not contribute to the diabetes-induced increase of neuronal excitability. Conclusions Capsaicin-insensitive low-pH-sensitive type of DRG neurons shows diabetes-induced upregulation of Cav3.2 subtype of T-type channels. This upregulation results in the increased excitability of these neurons and may contribute to nonthermal nociception at a later-stage diabetes.
Collapse
Affiliation(s)
- Dmytro E Duzhyy
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Viacheslav Y Viatchenko-Karpinski
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Eugen V Khomula
- International Center of Molecular Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Nana V Voitenko
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Pavel V Belan
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| |
Collapse
|
98
|
Mata X, Ducasse A, Vaiman A, Diribarne M, Fraud AS, Guérin G. Genomic structure, polymorphism and expression of ACCN1 and ACCN3 genes in the horse. Anim Genet 2015; 41 Suppl 2:138-44. [PMID: 21070287 DOI: 10.1111/j.1365-2052.2010.02123.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A category of cation gate proteins was shown to be present in sensory neurons and act as receptors of protons present in tissues such as muscles. The Amiloride-sensitive Cation Channel, Neuronal (ACCN) gene family is known to play a role in the transmission of pain through specialized pH sensitive neurons. Muscles from horses submitted to strenuous exercises produce lactic acid, which may induce variable pain through ACCN differential properties. The sequences of the equine cDNAs were determined to be 2.6 kb in length with an open reading frame of 1539 bp for ACCN1 and 2.1 kb in length with an open reading frame of 1602 bp for ACCN3. The ACCN1 gene is 990 kb long and contains 10 exons, and the ACCN3 gene is 4.2 kb long and contains 11 exons. The equine ACCN1 and ACCN3 genes have an ubiquitous expression but ACCN1 is more highly expressed in the spinal cord. We identified one alternative ACCN3 splicing variant present in various equine tissues. These mRNA variants may encode two different protein isoforms 533 and 509 amino acids long. Ten single nucleotide polymorphisms (SNPs) were detected for ACCN1; five in the coding and five in the non-coding region, with no amino acid change, while the three SNPs identified in the coding region of the ACCN3 gene introduce amino acid changes. The equine in silico promoter sequence reveals a structure similar to those of other mammalian species, especially for the ACCN1 gene.
Collapse
Affiliation(s)
- X Mata
- INRA, UMR1313, Unité de Génétique Animale et Biologie Intégrative, Centre de Recherche de Jouy, 78350 Jouy-en-Josas, France.
| | | | | | | | | | | |
Collapse
|
99
|
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
|
100
|
de Ceglia R, Chaabane L, Biffi E, Bergamaschi A, Ferrigno G, Amadio S, Del Carro U, Mazzocchi N, Comi G, Bianchi V, Taverna S, Forti L, D'Adamo P, Martino G, Menegon A, Muzio L. Down-sizing of neuronal network activity and density of presynaptic terminals by pathological acidosis are efficiently prevented by Diminazene Aceturate. Brain Behav Immun 2015; 45:263-76. [PMID: 25499583 DOI: 10.1016/j.bbi.2014.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 12/02/2014] [Accepted: 12/02/2014] [Indexed: 11/26/2022] Open
Abstract
Local acidosis is associated with neuro-inflammation and can have significant effects in several neurological disorders, including multiple sclerosis, brain ischemia, spinal cord injury and epilepsy. Despite local acidosis has been implicated in numerous pathological functions, very little is known about the modulatory effects of pathological acidosis on the activity of neuronal networks and on synaptic structural properties. Using non-invasive MRI spectroscopy we revealed protracted extracellular acidosis in the CNS of Experimental Autoimmune Encephalomyelitis (EAE) affected mice. By multi-unit recording in cortical neurons, we established that acidosis affects network activity, down-sizing firing and bursting behaviors as well as amplitudes. Furthermore, a protracted acidosis reduced the number of presynaptic terminals, while it did not affect the postsynaptic compartment. Application of the diarylamidine Diminazene Aceturate (DA) during acidosis significantly reverted both the loss of neuronal firing and bursting and the reduction of presynaptic terminals. Finally, in vivo DA delivery ameliorated the clinical disease course of EAE mice, reducing demyelination and axonal damage. DA is known to block acid-sensing ion channels (ASICs), which are proton-gated, voltage-insensitive, Na(+) permeable channels principally expressed by peripheral and central nervous system neurons. Our data suggest that ASICs activation during acidosis modulates network electrical activity and exacerbates neuro-degeneration in EAE mice. Therefore pharmacological modulation of ASICs in neuroinflammatory diseases could represent a new promising strategy for future therapies aimed at neuro-protection.
Collapse
Affiliation(s)
- Roberta de Ceglia
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Linda Chaabane
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy; Department of Neurology, Institute of Experimental Neurology (INSPE), Vita Salute San Raffaele University, Milan, Italy
| | - Emilia Biffi
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy; Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Andrea Bergamaschi
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Giancarlo Ferrigno
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy
| | - Stefano Amadio
- Neurophysiology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Ubaldo Del Carro
- Neurophysiology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Nausicaa Mazzocchi
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giancarlo Comi
- Department of Neurology, Institute of Experimental Neurology (INSPE), Vita Salute San Raffaele University, Milan, Italy
| | - Veronica Bianchi
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy
| | - Stefano Taverna
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Lia Forti
- Center for Neuroscience and Dept. of Theoretical and Applied Sciences, Biomedical Division, University of Insubria, 21052 Busto Arsizio, Italy
| | - Patrizia D'Adamo
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy
| | - Gianvito Martino
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy.
| | - Andrea Menegon
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luca Muzio
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy.
| |
Collapse
|