Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Elife ; 92020 10 07.
Article in English | MEDLINE | ID: mdl-33026343

ABSTRACT

Submucosal glands (SMGs) are a prominent structure that lines human cartilaginous airways. Although it has been assumed that SMGs contribute to respiratory defense, that hypothesis has gone without a direct test. Therefore, we studied pigs, which have lungs like humans, and disrupted the gene for ectodysplasin (EDA-KO), which initiates SMG development. EDA-KO pigs lacked SMGs throughout the airways. Their airway surface liquid had a reduced ability to kill bacteria, consistent with SMG production of antimicrobials. In wild-type pigs, SMGs secrete mucus that emerges onto the airway surface as strands. Lack of SMGs and mucus strands disrupted mucociliary transport in EDA-KO pigs. Consequently, EDA-KO pigs failed to eradicate a bacterial challenge in lung regions normally populated by SMGs. These in vivo and ex vivo results indicate that SMGs are required for normal antimicrobial activity and mucociliary transport, two key host defenses that protect the lung.


Subject(s)
Ectodysplasins/genetics , Exocrine Glands/immunology , Respiratory Mucosa/immunology , Staphylococcus aureus/physiology , Sus scrofa/immunology , Animals , Ectodysplasins/immunology , Female , Gene Knockout Techniques , Male , Sus scrofa/genetics
2.
Elife ; 62017 06 26.
Article in English | MEDLINE | ID: mdl-28650315

ABSTRACT

Attenuating the strength of fearful memories could benefit people disabled by memories of past trauma. Pavlovian conditioning experiments indicate that a retrieval cue can return a conditioned aversive memory to a labile state. However, means to enhance retrieval and render a memory more labile are unknown. We hypothesized that augmenting synaptic signaling during retrieval would increase memory lability. To enhance synaptic transmission, mice inhaled CO2 to induce an acidosis and activate acid sensing ion channels. Transient acidification increased the retrieval-induced lability of an aversive memory. The labile memory could then be weakened by an extinction protocol or strengthened by reconditioning. Coupling CO2 inhalation to retrieval increased activation of amygdala neurons bearing the memory trace and increased the synaptic exchange from Ca2+-impermeable to Ca2+-permeable AMPA receptors. The results suggest that transient acidosis during retrieval renders the memory of an aversive event more labile and suggest a strategy to modify debilitating memories.


Subject(s)
Acidosis , Fear , Memory , Amygdala/physiology , Animals , Carbon Dioxide/metabolism , Conditioning, Classical , Mice , Receptors, AMPA/metabolism
3.
PLoS One ; 11(11): e0166089, 2016.
Article in English | MEDLINE | ID: mdl-27820848

ABSTRACT

Neurons innervating the airways contribute to airway hyperreactivity (AHR), a hallmark feature of asthma. Several observations suggested that acid-sensing ion channels (ASICs), neuronal cation channels activated by protons, might contribute to AHR. For example, ASICs are found in vagal sensory neurons that innervate airways, and asthmatic airways can become acidic. Moreover, airway acidification activates ASIC currents and depolarizes neurons innervating airways. We found ASIC1a protein in vagal ganglia neurons, but not airway epithelium or smooth muscle. We induced AHR by sensitizing mice to ovalbumin and found that ASIC1a-/- mice failed to exhibit AHR despite a robust inflammatory response. Loss of ASIC1a also decreased bronchoalveolar lavage fluid levels of substance P, a sensory neuropeptide secreted from vagal sensory neurons that contributes to AHR. These findings suggest that ASIC1a is an important mediator of AHR and raise the possibility that inhibiting ASIC channels might be beneficial in asthma.


Subject(s)
Acid Sensing Ion Channels/metabolism , Respiratory Hypersensitivity/metabolism , Respiratory System/metabolism , Animals , Asthma/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Sensory Receptor Cells/metabolism , Substance P/metabolism , Vagus Nerve/metabolism
4.
Proc Natl Acad Sci U S A ; 111(24): 8961-6, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24889629

ABSTRACT

Stimulating presynaptic terminals can increase the proton concentration in synapses. Potential receptors for protons are acid-sensing ion channels (ASICs), Na(+)- and Ca(2+)-permeable channels that are activated by extracellular acidosis. Those observations suggest that protons might be a neurotransmitter. We found that presynaptic stimulation transiently reduced extracellular pH in the amygdala. The protons activated ASICs in lateral amygdala pyramidal neurons, generating excitatory postsynaptic currents. Moreover, both protons and ASICs were required for synaptic plasticity in lateral amygdala neurons. The results identify protons as a neurotransmitter, and they establish ASICs as the postsynaptic receptor. They also indicate that protons and ASICs are a neurotransmitter/receptor pair critical for amygdala-dependent learning and memory.


Subject(s)
Acid Sensing Ion Channels/genetics , Amygdala/physiology , Neuronal Plasticity , Neurotransmitter Agents/metabolism , Protons , Synapses/physiology , Acid Sensing Ion Channel Blockers/chemistry , Acidosis , Amygdala/metabolism , Animals , Brain/metabolism , Electrodes , Excitatory Postsynaptic Potentials , Hydrogen-Ion Concentration , Ion Channels/chemistry , Learning , Long-Term Potentiation , Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Peptides/chemistry , Spider Venoms/chemistry
5.
PLoS One ; 7(4): e35225, 2012.
Article in English | MEDLINE | ID: mdl-22506072

ABSTRACT

Three observations have suggested that acid-sensing ion channels (ASICs) might be mammalian cutaneous mechanoreceptors; they are structurally related to Caenorhabditis elegans mechanoreceptors, they are localized in specialized cutaneous mechanosensory structures, and mechanical displacement generates an ASIC-dependent depolarization in some neurons. However, previous studies of mice bearing a single disrupted ASIC gene showed only subtle or no alterations in cutaneous mechanosensitivity. Because functional redundancy of ASIC subunits might explain limited phenotypic alterations, we hypothesized that disrupting multiple ASIC genes would markedly impair cutaneous mechanosensation. We found the opposite. In behavioral studies, mice with simultaneous disruptions of ASIC1a, -2 and -3 genes (triple-knockouts, TKOs) showed increased paw withdrawal frequencies when mechanically stimulated with von Frey filaments. Moreover, in single-fiber nerve recordings of cutaneous afferents, mechanical stimulation generated enhanced activity in A-mechanonociceptors of ASIC TKOs compared to wild-type mice. Responses of all other fiber types did not differ between the two genotypes. These data indicate that ASIC subunits influence cutaneous mechanosensitivity. However, it is unlikely that ASICs directly transduce mechanical stimuli. We speculate that physical and/or functional association of ASICs with other components of the mechanosensory transduction apparatus contributes to normal cutaneous mechanosensation.


Subject(s)
Behavior, Animal/physiology , Mechanoreceptors/physiology , Mechanotransduction, Cellular/physiology , Nerve Tissue Proteins/genetics , Sodium Channels/genetics , Acid Sensing Ion Channels , Acids/metabolism , Animals , Female , Male , Mechanoreceptors/metabolism , Mechanotransduction, Cellular/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Stress, Mechanical
6.
Trustee ; 64(9): 6-7, 1, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22132666

ABSTRACT

After ending inpatient care, a hospital recommits to its community.


Subject(s)
Community-Institutional Relations , Health Facility Closure , Health Services Accessibility , Kansas , Organizational Case Studies , Professional Role , Trustees
7.
Circ Res ; 105(3): 279-86, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19590043

ABSTRACT

RATIONALE: Acid-sensing ion channels (ASICs) are Na+ channels that are activated by acidic pH. Their expression in cardiac afferents and remarkable sensitivity to small pH changes has made them leading candidates to sense cardiac ischemia. OBJECTIVE: Four genes encode six different ASIC subunits, however it is not yet clear which of the ASIC subunits contribute to the composition of ASICs in cardiac afferents. METHODS AND RESULTS: Here, we labeled cardiac afferents using a retrograde tracer dye in mice, which allowed for patch-clamp studies of murine cardiac afferents. We found that a higher percentage of cardiac sensory neurons from the dorsal root ganglia respond to acidic pH and generated larger currents compared to those from the nodose ganglia. The ASIC-like current properties of the cardiac dorsal root ganglia neurons from wild-type mice most closely matched the properties of ASIC2a/3 heteromeric channels. This was supported by studies in ASIC-null mice: acid-evoked currents from ASIC3(-/-) cardiac afferents matched the properties of ASIC2a channels, and currents from ASIC2(-/-) cardiac afferents matched the properties of ASIC3 channels. CONCLUSIONS: We conclude that ASIC2a and -3 are the major ASIC subunits in cardiac dorsal root ganglia neurons and provide potential molecular targets to attenuate chest pain and deleterious reflexes associated with cardiac disease.


Subject(s)
Ganglia, Spinal/metabolism , Heart/innervation , Nerve Tissue Proteins/metabolism , Protein Multimerization/physiology , Sodium Channels/metabolism , Acid Sensing Ion Channels , Adenosine Triphosphate/pharmacology , Animals , Capsaicin/pharmacology , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Protons , Sodium Channels/genetics
8.
J Neurosci ; 29(17): 5381-8, 2009 Apr 29.
Article in English | MEDLINE | ID: mdl-19403806

ABSTRACT

No animal models replicate the complexity of human depression. However, a number of behavioral tests in rodents are sensitive to antidepressants and may thus tap important underlying biological factors. Such models may also offer the best opportunity to discover novel treatments. Here, we used several of these models to test the hypothesis that the acid-sensing ion channel-1a (ASIC1a) might be targeted to reduce depression. Genetically disrupting ASIC1a in mice produced antidepressant-like effects in the forced swim test, the tail suspension test, and following unpredictable mild stress. Pharmacologically inhibiting ASIC1a also had antidepressant-like effects in the forced swim test. The effects of ASIC1a disruption in the forced swim test were independent of and additive to those of several commonly used antidepressants. Furthermore, ASIC1a disruption interfered with an important biochemical marker of depression, the ability of stress to reduce BDNF in the hippocampus. Restoring ASIC1a to the amygdala of ASIC1a(-/-) mice with a viral vector reversed the forced swim test effects, suggesting that the amygdala is a key site of ASIC1a action in depression-related behavior. These data are consistent with clinical studies emphasizing the importance of the amygdala in mood regulation, and suggest that ASIC1a antagonists may effectively combat depression.


Subject(s)
Amygdala/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Drug Delivery Systems/methods , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Acid Sensing Ion Channels , Amygdala/drug effects , Animals , Antidepressive Agents/administration & dosage , Depressive Disorder/psychology , Female , Isoquinolines/administration & dosage , Male , Mice , Mice, Transgenic , Naphthalenes/administration & dosage , Nerve Tissue Proteins/deficiency , Sodium Channels/deficiency , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Stress, Psychological/psychology
9.
Proc Natl Acad Sci U S A ; 105(8): 3140-4, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18287010

ABSTRACT

Acid-sensing ion channels (ASICs) are neuronal non-voltage-gated cation channels that are activated when extracellular pH falls. They contribute to sensory function and nociception in the peripheral nervous system, and in the brain they contribute to synaptic plasticity and fear responses. Some of the physiologic consequences of disrupting ASIC genes in mice suggested that ASIC channels might modulate neuronal function by mechanisms in addition to their H(+)-evoked opening. Within ASIC channel's large extracellular domain, we identified sequence resembling that in scorpion toxins that inhibit K(+) channels. Therefore, we tested the hypothesis that ASIC channels might inhibit K(+) channel function by coexpressing ASIC1a and the high-conductance Ca(2+)- and voltage-activated K(+) (BK) channel. We found that ASIC1a associated with BK channels and inhibited their current. Reducing extracellular pH disrupted the association and relieved the inhibition. BK channels, in turn, altered the kinetics of ASIC1a current. In addition to BK, ASIC1a inhibited voltage-gated Kv1.3 channels. Other ASIC channels also inhibited BK, although acidosis-dependent relief of inhibition varied. These results reveal a mechanism of ion channel interaction and reciprocal regulation. Finding that a reduced pH activated ASIC1a and relieved BK inhibition suggests that extracellular protons may enhance the activity of channels with opposing effects on membrane voltage. The wide and varied expression patterns of ASICs, BK, and related K(+) channels suggest broad opportunities for this signaling system to alter neuronal function.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Signal Transduction/physiology , Sodium Channels/genetics , Sodium Channels/metabolism , Acid Sensing Ion Channels , Amino Acid Sequence , Cell Line , DNA, Complementary/genetics , Electrophysiology , Humans , Hydrogen-Ion Concentration , Kinetics , Large-Conductance Calcium-Activated Potassium Channels/genetics , Molecular Sequence Data , Neurons/metabolism , Protein Structure, Tertiary , Scorpion Venoms/genetics
10.
Gastroenterology ; 133(1): 184-94, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17553498

ABSTRACT

BACKGROUND & AIMS: Using a mouse model that reproduces major features of irritable bowel syndrome (long-lasting colon hypersensitivity without inflammation), we examined the contributions of 2 proteins, transient receptor potential vanilloid 1 (TRPV1) and acid-sensing ion channel 3 (ASIC3), on development of behavioral hypersensitivity and assessed the function of colon mechanoreceptors of hypersensitive mice. METHODS: Visceral nociceptive behavior was measured as the visceromotor response (VMR) to colorectal distention (CRD) before and after intracolonic treatment with zymosan or saline. Colon pathology was assessed in parallel experiments by quantifying myeloperoxidase activity, intralumenal pH, and tissue histology. Electrophysiologic experiments were performed on naïve and zymosan-treated hypersensitive mice using an in vitro colon-pelvic nerve preparation. RESULTS: Zymosan, but not saline, produced significant and persistent increases in the VMRs of control mice; zymosan produced nonsignificant increases in the VMRs in TRPV1 and ASIC3 knockout mice. Colon myeloperoxidase activity and pH were unaffected by either CRD or intracolonic treatments. Pelvic nerve mechanoreceptors recorded from zymosan-treated or naïve mice had similar sensitivity to stretch of the colon. When applied acutely, zymosan sensitized muscular/mucosal mechanoreceptors in both naïve and hypersensitive mice. CONCLUSIONS: Zymosan produced sensitization of colon mechanoreceptors acutely in vitro and chronic (>or=7 weeks) behavioral hypersensitivity in the absence of inflammation. The behavioral hypersensitivity was partially dependent on both TRPV1 and ASIC3 because deletions of either of these genes blunted zymosan's effect, suggesting that these proteins may be important peripheral mediators for development of functional (ie, noninflammatory) visceral hypersensitivity.


Subject(s)
Irritable Bowel Syndrome/physiopathology , Mechanoreceptors/physiopathology , Sodium Channels/genetics , TRPV Cation Channels/genetics , Acid Sensing Ion Channels , Animals , Colon/innervation , Colon/physiopathology , Disease Models, Animal , Electromyography , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Irritable Bowel Syndrome/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/physiology , Sodium Channels/metabolism , TRPV Cation Channels/metabolism , Zymosan/pharmacology
11.
Pain ; 129(1-2): 102-12, 2007 May.
Article in English | MEDLINE | ID: mdl-17134831

ABSTRACT

Peripheral initiators of muscle pain are virtually unknown, but likely key to development of chronic pain after muscle insult. The current study tested the hypothesis that ASIC3 in muscle is necessary for development of cutaneous mechanical, but not heat, hyperalgesia induced by muscle inflammation. Using mechanical and heat stimuli, we assessed behavioral responses in ASIC3-/- and ASIC3+/+ mice after induction of carrageenan muscle inflammation. ASIC3-/- mice did not develop cutaneous mechanical hyperalgesia after muscle inflammation when compared to ASIC3+/+ mice; heat hyperalgesia developed similarly between groups. We then tested if the phenotype could be rescued in ASIC3-/- mice by using a recombinant herpes virus vector to express ASIC3 in skin (where testing occurred) or muscle (where inflammation occurred). Infection of mouse DRG neurons with ASIC3-encoding virus resulted in functional expression of ASICs. Injection of ASIC3-encoding virus into muscle or skin of ASIC3-/- mice resulted in ASIC3 mRNA in DRG and protein expression in DRG and the peripheral injection site. Injection of ASIC3-encoding virus into muscle, but not skin, resulted in development of mechanical hyperalgesia similar to that observed in ASIC3+/+ mice. Thus, ASIC3 in primary afferent fibers innervating muscle is critical to development of hyperalgesia that results from muscle insult.


Subject(s)
Hyperalgesia/etiology , Hyperalgesia/pathology , Inflammation/complications , Muscle, Skeletal/metabolism , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Behavior, Animal , Cells, Cultured , Enzyme-Linked Immunosorbent Assay/methods , Ganglia, Spinal/cytology , Immunohistochemistry/methods , Inflammation/etiology , Inflammation/pathology , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/physiopathology , Muscle, Skeletal/virology , Neurons/physiology , Pain Threshold , Patch-Clamp Techniques/methods , Physical Stimulation , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Simplexvirus , Skin/metabolism , Skin/physiopathology , Sodium Channels/deficiency
12.
Trends Neurosci ; 29(10): 578-86, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16891000

ABSTRACT

Extracellular acid can have important effects on neuron function. In central and peripheral neurons, acid-sensing ion channels (ASICs) have emerged as key receptors for extracellular protons, and recent studies suggest diverse roles for these channels in the pathophysiology of pain, ischemic stroke and psychiatric disease. ASICs have also been implicated in mechanosensation in the peripheral nervous system and in neurotransmission in the central nervous system. Here, we briefly review advances in our understanding of ASICs, their potential contributions to disease, and the possibility for their therapeutic modification.


Subject(s)
Central Nervous System/physiology , Mechanotransduction, Cellular/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Peripheral Nervous System/physiology , Sodium Channels/physiology , Acid Sensing Ion Channels , Animals , Extracellular Fluid/chemistry , Humans , Membrane Proteins/chemistry , Mental Disorders/physiopathology , Nerve Tissue Proteins/chemistry , Pain/physiopathology , Sodium Channels/chemistry , Stroke/physiopathology
13.
J Neurosci ; 24(45): 10167-75, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537887

ABSTRACT

Ion channels in the degenerin-epithelial sodium channel (DEG-ENaC) family perform diverse functions, including mechanosensation. Here we explored the role of the vertebrate DEG-ENaC protein, acid-sensing ion channel 2 (ASIC2), in auditory transduction. Contributions of ASIC2 to hearing were examined by comparing hearing threshold and noise sensitivity of wild-type and ASIC2 null mice. ASIC2 null mice showed no significant hearing loss, indicating that the ASIC2 was not directly involved in the mechanotransduction of the mammalian cochlea. However, we found that (1) ASIC2 was present in the spiral ganglion (SG) neurons in the adult cochlea and that externally applied protons induced amiloride-sensitive sodium currents and action potentials in SG neurons in vitro, (2) proton-induced responses were greatly reduced in SG neurons obtained from ASIC2 null mice, indicating that activations of ASIC2 contributed a major portion of the proton-induced excitatory response in SG neurons, and (3) ASIC2 null mice were considerably more resistant to noise-induced temporary, but not permanent, threshold shifts. Together, these data suggest that ASIC2 contributes to suprathreshold functions of the cochlea. The presence of ASIC2 in SG neurons could provide sensors to directly convert local acidosis to excitatory responses, therefore offering a cellular mechanism linking hearing losses caused by many enigmatic causes (e.g., ischemia or inflammation of the inner ear) to excitotoxicity.


Subject(s)
Cochlea/physiopathology , Hearing Loss, Sensorineural/physiopathology , Hearing/physiology , Hydrogen-Ion Concentration , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons, Afferent/physiology , Noise/adverse effects , Sodium Channels/physiology , Spiral Ganglion/physiology , Acid Sensing Ion Channels , Action Potentials/drug effects , Amiloride/pharmacology , Animals , Auditory Threshold , Cells, Cultured/drug effects , Cells, Cultured/physiology , Choline/pharmacology , Elapid Venoms/pharmacology , Extracellular Fluid/chemistry , Hearing Loss, Sensorineural/etiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons, Afferent/drug effects , Patch-Clamp Techniques , Protons , Sodium Channels/deficiency , Sodium Channels/genetics , Spiral Ganglion/cytology , Spiral Ganglion/drug effects
14.
J Neurosci ; 24(40): 8678-89, 2004 Oct 06.
Article in English | MEDLINE | ID: mdl-15470133

ABSTRACT

Acid-sensing ion channels (ASICs), a novel class of ligand-gated cation channels activated by protons, are highly expressed in peripheral sensory and central neurons. Activation of ASICs may play an important role in physiological processes such as nociception, mechanosensation, and learning-memory, and in the pathology of neurological conditions such as brain ischemia. Modulation of the activities of ASICs is expected to have a significant influence on the roles that these channels can play in both physiological and/or pathological processes. Here we show that the divalent cation Zn2+, an endogenous trace element, dose-dependently inhibits ASIC currents in cultured mouse cortical neurons at nanomolar concentrations. With ASICs expressed in Chinese hamster ovary cells, Zn2+ inhibits currents mediated by homomeric ASIC1a and heteromeric ASIC1a-ASIC2a channels, without affecting currents mediated by homomeric ASIC1beta, ASIC2a, or ASIC3. Consistent with ASIC1a-specific modulation, high-affinity Zn2+ inhibition is absent in neurons from ASIC1a knock-out mice. Current-clamp recordings and Ca2+-imaging experiments demonstrated that Zn2+ inhibits acid-induced membrane depolarization and the increase of intracellular Ca2+. Mutation of lysine-133 in the extracellular domain of the ASIC1a subunit abolishes the high-affinity Zn2+ inhibition. Our studies suggest that Zn2+ may play an important role in a negative feedback system for preventing overexcitation of neurons during normal synaptic transmission and ASIC1a-mediated excitotoxicity in pathological conditions.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/physiology , Zinc/pharmacology , Acid Sensing Ion Channels , Amino Acid Sequence , Animals , CHO Cells , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chelating Agents/pharmacology , Cricetinae , Cricetulus , Electric Conductivity , Hydrogen-Ion Concentration , Lysine/genetics , Membrane Potentials , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/drug effects , Patch-Clamp Techniques , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Sodium Channels/chemistry , Sodium Channels/genetics
15.
J Biol Chem ; 279(51): 53886-91, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15471860

ABSTRACT

Acid-sensing ion channels (ASICs) are H(+)-gated members of the degenerin/epithelial Na(+) channel (DEG/ENaC) family in vertebrate neurons. Several ASICs are expressed in sensory neurons, where they play a role in responses to nociceptive, taste, and mechanical stimuli; others are expressed in central neurons, where they participate in synaptic plasticity and some forms of learning. Stomatin is an integral membrane protein found in lipid/protein-rich microdomains, and it is believed to regulate the function of ion channels and transporters. In Caenorhabditis elegans, stomatin homologs interact with DEG/ENaC channels, which together are necessary for normal mechanosensation in the worm. Therefore, we asked whether stomatin interacts with and modulates the function of ASICs. We found that stomatin co-immunoprecipitated and co-localized with ASIC proteins in heterologous cells. Moreover, stomatin altered the function of ASIC channels. Stomatin potently reduced acid-evoked currents generated by ASIC3 without changing steady state protein levels or the amount of ASIC3 expressed at the cell surface. In contrast, stomatin accelerated the desensitization rate of ASIC2 and heteromeric ASICs, whereas current amplitude was unaffected. These data suggest that stomatin binds to and alters the gating of ASICs. Our findings indicate that modulation of DEG/ENaC channels by stomatin-like proteins is evolutionarily conserved and may have important implications for mammalian nociception and mechanosensation.


Subject(s)
Blood Proteins/physiology , Membrane Proteins/chemistry , Membrane Proteins/physiology , Nerve Tissue Proteins/chemistry , Sodium Channels/chemistry , Acid Sensing Ion Channels , Animals , Biotinylation , CHO Cells , COS Cells , Caenorhabditis elegans , Cell Membrane/metabolism , Conserved Sequence , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Evolution, Molecular , Humans , Hydrogen-Ion Concentration , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Fluorescence , Neurons/metabolism , Patch-Clamp Techniques , Time Factors , Transfection
16.
Cell ; 118(6): 687-98, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15369669

ABSTRACT

Ca2+ toxicity remains the central focus of ischemic brain injury. The mechanism by which toxic Ca2+ loading of cells occurs in the ischemic brain has become less clear as multiple human trials of glutamate antagonists have failed to show effective neuroprotection in stroke. Acidosis is a common feature of ischemia and is assumed to play a critical role in brain injury; however, the mechanism(s) remain ill defined. Here, we show that acidosis activates Ca2+ -permeable acid-sensing ion channels (ASICs), inducing glutamate receptor-independent, Ca2+ -dependent, neuronal injury inhibited by ASIC blockers. Cells lacking endogenous ASICs are resistant to acid injury, while transfection of Ca2+ -permeable ASIC1a establishes sensitivity. In focal ischemia, intracerebroventricular injection of ASIC1a blockers or knockout of the ASIC1a gene protects the brain from ischemic injury and does so more potently than glutamate antagonism. Thus, acidosis injures the brain via membrane receptor-based mechanisms with resultant toxicity of [Ca2+]i, disclosing new potential therapeutic targets for stroke.


Subject(s)
Acidosis/metabolism , Brain Ischemia/metabolism , Calcium/metabolism , Glutamic Acid/metabolism , Membrane Proteins/metabolism , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Acid Sensing Ion Channels , Acidosis/complications , Acidosis/drug therapy , Animals , Brain Ischemia/drug therapy , COS Cells , Calcium/toxicity , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cells, Cultured , Disease Models, Animal , Drug Design , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/toxicity , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neuroprotective Agents/pharmacology , Rats , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/genetics
17.
J Biol Chem ; 279(45): 46962-8, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15317815

ABSTRACT

The acid-sensing ion channel-3 (ASIC3) is a degenerin/epithelial sodium channel expressed in the peripheral nervous system. Previous studies indicate that it participates in the response to mechanical and painful stimuli, perhaps contributing to mechanoreceptor and/or H+ -gated nociceptor function. ASIC3 subunits contain intracellular N and C termini that may control channel localization and function. We found that a PDZ-binding motif at the ASIC3 C terminus interacts with four different proteins that contain PDZ domains: PSD-95, Lin-7b, MAGI-1b, and PIST. ASIC3 and these interacting proteins were expressed in dorsal root ganglia and spinal cord, and PSD-95 co-precipitated ASIC3 from spinal cord. When expressed in heterologous cells, PSD-95 reduced the amplitude of ASIC3 acid-evoked currents, whereas Lin-7b increased current amplitude. PSD-95 and Lin-7b altered current density by decreasing or increasing, respectively, the amount of ASIC3 on the cell surface. The finding that multiple PDZ-containing proteins bind ASIC3 and can influence its presence in the plasma membrane suggests that they may play an important role in the contribution of ASIC3 to nociception and mechanosensation.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Protons , Sodium Channels/chemistry , Acid Sensing Ion Channels , Amino Acid Motifs , Animals , Biotinylation , CHO Cells , COS Cells , Cell Membrane/metabolism , Cricetinae , DNA/metabolism , Disks Large Homolog 4 Protein , Electrophysiology , Ganglia, Spinal/metabolism , Green Fluorescent Proteins/metabolism , Guanylate Kinases , Humans , Hydrogen-Ion Concentration , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Microscopy, Fluorescence , Nerve Tissue Proteins/metabolism , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/metabolism , Tissue Distribution , Transfection , Two-Hybrid System Techniques
18.
J Physiol ; 556(Pt 3): 691-710, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14990679

ABSTRACT

The molecular basis of mechanosensory transduction by primary sensory neurones remains poorly understood. Amongst candidate transducer molecules are members of the acid-sensing ion channel (ASIC) family; nerve fibre recordings have shown ASIC2 and ASIC3 null mutants have aberrant responses to suprathreshold mechanical stimuli. Using the neuronal cell body as a model of the sensory terminal we investigated if ASIC2 or 3 contributed to mechanically activated currents in dorsal root ganglion (DRG) neurones. We cultured neurones from ASIC2 and ASIC3 null mutants and compared response properties with those of wild-type controls. Neuronal subpopulations [categorized by cell size, action potential duration and isolectin B4 (IB4) binding] generated distinct responses to mechanical stimulation consistent with their predicted in vivo phenotypes. In particular, there was a striking relationship between action potential duration and mechanosensitivity as has been observed in vivo. Putative low threshold mechanoreceptors exhibited rapidly adapting mechanically activated currents. Conversely, when nociceptors responded they displayed slowly or intermediately adapting currents that were smaller in amplitude than responses of low threshold mechanoreceptor neurones. No differences in current amplitude or kinetics were found between ASIC2 and/or ASIC3 null mutants and controls. Ruthenium red (5 microm) blocked mechanically activated currents in a voltage-dependent manner, with equal efficacy in wild-type and knockout animals. Analysis of proton-gated currents revealed that in wild-type and ASIC2/3 double knockout mice the majority of putative low threshold mechanoreceptors did not exhibit ASIC-like currents but exhibited a persistent current in response to low pH. Our findings are consistent with another ion channel type being important in DRG mechanotransduction.


Subject(s)
Mechanotransduction, Cellular/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons, Afferent/physiology , Sodium Channels/physiology , Acid Sensing Ion Channels , Action Potentials/drug effects , Action Potentials/physiology , Animals , Brain Chemistry , Capsaicin/pharmacology , Cells, Cultured , Ganglia, Spinal/chemistry , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Gene Expression/genetics , Hydrogen-Ion Concentration , Kinetics , Mechanotransduction, Cellular/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Neurons, Afferent/chemistry , Neurons, Afferent/drug effects , Nociceptors/physiology , Patch-Clamp Techniques , Physical Stimulation , Reverse Transcriptase Polymerase Chain Reaction , Ruthenium Red/pharmacology , Sodium Channels/genetics , Spinal Cord/chemistry , Stress, Mechanical , Tetrodotoxin/pharmacology
19.
J Biol Chem ; 279(18): 18296-305, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14960591

ABSTRACT

Hippocampal neurons express subunits of the acid-sensing ion channel (ASIC1 and ASIC2) and exhibit large cation currents that are transiently activated by acidic extracellular solutions. Earlier work indicated that ASIC1 contributed to the current in these neurons and suggested its importance for normal behavior. However, the specific contribution of ASIC1 and ASIC2 subunits to acid-evoked currents in hippocampal neurons remained uncertain. To decipher the individual role of the ASIC subunits, we studied H(+)-gated currents in neurons from both ASIC1 and ASIC2 null mice. We found that much of the current was produced by ASIC1a/2a heteromultimeric channels, and individual subunits made distinct contributions. The ASIC1a subunit was key in establishing current amplitude. The ASIC2a subunit had little effect on amplitude but influenced desensitization, recovery from desensitization, pH sensitivity, and the response to modulatory agents. We also found heterogeneity in the contribution of ASIC2 throughout the neuronal population, with individual neurons expressing both ASIC1a homomultimeric and ASIC1a/2a heteromultimeric channels. Studies of neurons heterozygous for disrupted ASIC alleles indicated that the properties of H(+)-gated currents are dependent on the proportion of the individual subunits. These findings indicate that the absolute and relative amounts of ASIC subunits determine the amplitude and properties of hippocampal H(+)-gated currents and therefore may contribute to normal physiology and pathophysiology.


Subject(s)
Hippocampus/cytology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Sodium Channels/physiology , Acid Sensing Ion Channels , Animals , Benzamides/pharmacology , Cells, Cultured , Electrophysiology , FMRFamide/pharmacology , Hydrogen-Ion Concentration , Membrane Proteins/chemistry , Mice , Mice, Knockout , Nerve Tissue Proteins/chemistry , Neurons/chemistry , Neurons/physiology , Protein Subunits/chemistry , Protein Subunits/physiology , Protons , Sodium Channels/chemistry , Zinc/pharmacology
20.
Pain ; 106(3): 229-239, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14659506

ABSTRACT

Clinically, chronic pain and hyperalgesia induced by muscle injury are disabling and difficult to treat. Cellular and molecular mechanisms underlying chronic muscle-induced hyperalgesia are not well understood. For this reason, we developed an animal model where repeated injections of acidic saline into one gastrocnemius muscle produce bilateral, long-lasting mechanical hypersensitivity of the paw (i.e. hyperalgesia) without associated tissue damage. Since acid sensing ion channels (ASICs) are found on primary afferent fibers and respond to decreases in pH, we tested the hypothesis that ASICs on primary afferent fibers innervating muscle are critical to development of hyperalgesia and central sensitization in response to repeated intramuscular acid. Dorsal root ganglion neurons innervating muscle express ASIC3 and respond to acidic pH with fast, transient inward and sustained currents that resemble those of ASICs. Mechanical hyperalgesia produced by repeated intramuscular acid injections is prevented by prior treatment of the muscle with the non-selective ASIC antagonist, amiloride, suggesting ASICs might be involved. ASIC3 knockouts do not develop mechanical hyperalgesia to repeated intramuscular acid injection when compared to wildtype littermates. In contrast, ASIC1 knockouts develop hyperalgesia similar to their wildtype littermates. Extracellular recordings of spinal wide dynamic range (WDR) neurons from wildtype mice show an expansion of the receptive field to include the contralateral paw, an increased response to von Frey filaments applied to the paw both ipsilaterally and contralaterally, and increased response to noxious pinch contralaterally after the second intramuscular acid injection. These changes in WDR neurons do not occur in ASIC3 knockouts. Thus, activation of ASIC3s on muscle afferents is required for development of mechanical hyperalgesia and central sensitization that normally occurs in response to repeated intramuscular acid. Therefore, interfering with ASIC3 might be of benefit in treatment or prevention of chronic hyperalgesia.


Subject(s)
Hyperalgesia/metabolism , Membrane Proteins/deficiency , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/deficiency , Sodium Channels/deficiency , Sodium Chloride/toxicity , Acid Sensing Ion Channels , Action Potentials/drug effects , Action Potentials/physiology , Animals , Chronic Disease , Hydrogen-Ion Concentration , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Injections, Intramuscular , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Nerve Tissue Proteins/genetics , Pain Measurement/drug effects , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Sodium Channels/genetics , Sodium Chloride/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL