Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cells ; 12(17)2023 09 03.
Article in English | MEDLINE | ID: mdl-37681933

ABSTRACT

Pharmacological adaptation is a common phenomenon observed during prolonged drug exposure and often leads to drug resistance. Understanding the cellular events involved in adaptation could provide new strategies to circumvent this resistance issue. We used the nematode Caenorhabditis elegans to analyze the adaptation to levamisole, an ionotropic acetylcholine receptor agonist, used for decades to treat nematode parasitic infections. Genetic screens in C. elegans identified "adapting mutants" that initially paralyze upon exposure to levamisole as the wild type (WT), but recover locomotion after a few hours whereas WT remain paralyzed. Here, we show that levamisole induces a sustained increase in cytosolic calcium concentration in the muscle cells of adapting mutants, lasting several hours and preceding a decrease in levamisole-sensitive acetylcholine receptors (L-AChR) at the muscle plasma membrane. This decrease correlated with a drop in calcium concentration, a relaxation of the animal's body and a resumption of locomotion. The decrease in calcium and L-AChR content depends on calcineurin activation in muscle cells. We also showed that levamisole adaptation triggers homeostatic mechanisms in muscle cells including mitochondria remodeling, lysosomal tubulation and an increase in autophagic activity. Levamisole adaptation thus provides a new experimental paradigm for studying how cells cope with calcium stress.


Subject(s)
Caenorhabditis elegans , Calcineurin , Animals , Calcium , Levamisole/pharmacology , Muscle Cells , Receptors, Cholinergic
2.
Nat Commun ; 14(1): 1939, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024493

ABSTRACT

Excitable cells can be stimulated or inhibited by optogenetics. Since optogenetic actuation regimes are often static, neurons and circuits can quickly adapt, allowing perturbation, but not true control. Hence, we established an optogenetic voltage-clamp (OVC). The voltage-indicator QuasAr2 provides information for fast, closed-loop optical feedback to the bidirectional optogenetic actuator BiPOLES. Voltage-dependent fluorescence is held within tight margins, thus clamping the cell to distinct potentials. We established the OVC in muscles and neurons of Caenorhabditis elegans, and transferred it to rat hippocampal neurons in slice culture. Fluorescence signals were calibrated to electrically measured potentials, and wavelengths to currents, enabling to determine optical I/V-relationships. The OVC reports on homeostatically altered cellular physiology in mutants and on Ca2+-channel properties, and can dynamically clamp spiking in C. elegans. Combining non-invasive imaging with control capabilities of electrophysiology, the OVC facilitates high-throughput, contact-less electrophysiology in individual cells and paves the way for true optogenetic control in behaving animals.


Subject(s)
Caenorhabditis elegans , Muscles , Animals , Rats , Caenorhabditis elegans/physiology , Action Potentials/physiology , Neurons/physiology , Optogenetics/methods
3.
Proc Natl Acad Sci U S A ; 119(29): e2113545119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858330

ABSTRACT

Biophysical properties of ligand-gated receptors can be profoundly modified by auxiliary subunits or by the lipid microenvironment of the membrane. Hence, it is sometimes challenging to relate the properties of receptors reconstituted in heterologous expression systems to those of their native counterparts. Here we show that the properties of Caenorhabditis elegans levamisole-sensitive acetylcholine receptors (L-AChRs), the ionotropic acetylcholine receptors targeted by the cholinergic anthelmintic levamisole at neuromuscular junctions, can be profoundly modified by their clustering machinery. We uncovered that L-AChRs exhibit a strong outward rectification in vivo, which was not previously described in heterologous systems. This unusual feature for an ionotropic AChR is abolished by disrupting the interaction of the receptors with the extracellular complex required for their synaptic clustering. When recorded at -60 mV, levamisole-induced currents are similar in the wild type and in L-AChR-clustering-defective mutants, while they are halved in these mutants at more depolarized physiological membrane potentials. Consequently, levamisole causes a strong muscle depolarization in the wild type, which leads to complete inactivation of the voltage-gated calcium channels and to an irreversible flaccid paralysis. In mutants defective for L-AChR clustering, the levamisole-induced depolarization is weaker, allowing voltage-gated calcium channels to remain partially active, which eventually leads to adaptation and survival of the worms. This explains why historical screens for C. elegans mutants resistant to levamisole identified the components of the L-AChR clustering machinery, in addition to proteins required for receptor biosynthesis or efficacy. This work further emphasizes the importance of pursuing ligand-gated channel characterization in their native environment.


Subject(s)
Antinematodal Agents , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Calcium Channels , Cholinergic Agonists , Levamisole , Receptors, Cholinergic , Acetylcholine/metabolism , Animals , Antinematodal Agents/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Calcium Channels/metabolism , Cholinergic Agonists/pharmacology , Levamisole/pharmacology , Receptors, Cholinergic/metabolism
4.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34213535

ABSTRACT

The extracellular matrix has emerged as an active component of chemical synapses regulating synaptic formation, maintenance, and homeostasis. The heparan sulfate proteoglycan (HSPG) syndecans are known to regulate cellular and axonal migration in the brain. They are also enriched at synapses, but their synaptic functions remain more elusive. Here, we show that SDN-1, the sole orthologue of syndecan in C. elegans, is absolutely required for the synaptic clustering of homomeric α7-like acetylcholine receptors (AChRs) and regulates the synaptic content of heteromeric AChRs. SDN-1 is concentrated at neuromuscular junctions (NMJs) by the neurally secreted synaptic organizer Ce-Punctin/MADD-4, which also activates the transmembrane netrin receptor DCC. Those cooperatively recruit the FARP and CASK orthologues that localize α7-like-AChRs at cholinergic NMJs through physical interactions. Therefore, SDN-1 stands at the core of the cholinergic synapse organization by bridging the extracellular synaptic determinants to the intracellular synaptic scaffold that controls the postsynaptic receptor content.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Receptors, Cholinergic/metabolism , Synapses/metabolism , Syndecans/metabolism , Acetylcholine/metabolism , Angiopoietin-Like Protein 4/genetics , Angiopoietin-Like Protein 4/metabolism , Animals , Brain/cytology , Brain/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , DCC Receptor/genetics , DCC Receptor/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Gene Expression Regulation , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Nerve Tissue Proteins/genetics , Neuromuscular Junction/ultrastructure , Neurons/cytology , Neurons/metabolism , Receptors, Cholinergic/genetics , Synapses/ultrastructure , Synaptic Transmission/genetics , Syndecans/genetics
5.
Nat Commun ; 11(1): 2674, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471987

ABSTRACT

Increasing evidence indicates that guidance molecules used during development for cellular and axonal navigation also play roles in synapse maturation and homeostasis. In C. elegans the netrin receptor UNC-40/DCC controls the growth of dendritic-like muscle cell extensions towards motoneurons and is required to recruit type A GABA receptors (GABAARs) at inhibitory neuromuscular junctions. Here we show that activation of UNC-40 assembles an intracellular synaptic scaffold by physically interacting with FRM-3, a FERM protein orthologous to FARP1/2. FRM-3 then recruits LIN-2, the ortholog of CASK, that binds the synaptic adhesion molecule NLG-1/Neuroligin and physically connects GABAARs to prepositioned NLG-1 clusters. These processes are orchestrated by the synaptic organizer CePunctin/MADD-4, which controls the localization of GABAARs by positioning NLG-1/neuroligin at synapses and regulates the synaptic content of GABAARs through the UNC-40-dependent intracellular scaffold. Since DCC is detected at GABA synapses in mammals, DCC might also tune inhibitory neurotransmission in the mammalian brain.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Adhesion Molecules/metabolism , Receptors, Cell Surface/metabolism , Receptors, GABA-A/metabolism , Synaptic Transmission/physiology , Animals , Axon Guidance/physiology , Cell Adhesion Molecules, Neuronal/metabolism , Cytoskeletal Proteins/metabolism , Helminth Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Synapses/physiology
6.
Proc Natl Acad Sci U S A ; 115(29): E6890-E6899, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29959203

ABSTRACT

Neuropeptides are ubiquitous modulators of behavior and physiology. They are packaged in specialized secretory organelles called dense core vesicles (DCVs) that are released upon neural stimulation. Unlike synaptic vesicles, which can be recycled and refilled close to release sites, DCVs must be replenished by de novo synthesis in the cell body. Here, we dissect DCV cell biology in vivo in a Caenorhabditis elegans sensory neuron whose tonic activity we can control using a natural stimulus. We express fluorescently tagged neuropeptides in the neuron and define parameters that describe their subcellular distribution. We measure these parameters at high and low neural activity in 187 mutants defective in proteins implicated in membrane traffic, neuroendocrine secretion, and neuronal or synaptic activity. Using unsupervised hierarchical clustering methods, we analyze these data and identify 62 groups of genes with similar mutant phenotypes. We explore the function of a subset of these groups. We recapitulate many previous findings, validating our paradigm. We uncover a large battery of proteins involved in recycling DCV membrane proteins, something hitherto poorly explored. We show that the unfolded protein response promotes DCV production, which may contribute to intertissue communication of stress. We also find evidence that different mechanisms of priming and exocytosis may operate at high and low neural activity. Our work provides a defined framework to study DCV biology at different neural activity levels.


Subject(s)
Caenorhabditis elegans , Mutation , Neuropeptides , Secretory Vesicles , Sensory Receptor Cells/metabolism , Synaptic Vesicles , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Secretory Vesicles/genetics , Secretory Vesicles/metabolism , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism
7.
Neuron ; 86(6): 1407-19, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26028575

ABSTRACT

Positioning type A GABA receptors (GABA(A)Rs) in front of GABA release sites sets the strength of inhibitory synapses. The evolutionarily conserved Ce-Punctin/MADD-4 is an anterograde synaptic organizer that specifies GABAergic versus cholinergic identity of postsynaptic domains at the C. elegans neuromuscular junctions (NMJs). Here we show that the Ce-Punctin secreted by GABAergic motor neurons controls the clustering of GABA(A)Rs through the synaptic adhesion molecule neuroligin (NLG-1) and the netrin receptor UNC-40/DCC. The short isoform of Ce-Punctin binds and clusters NLG-1 postsynaptically at GABAergic NMJs. NLG-1 disruption causes a strong reduction of GABA(A)R content at GABAergic synapses. Ce-Punctin also binds and localizes UNC-40 receptors in the postsynaptic membrane of NMJs, which promotes the recruitment of GABA(A)Rs by NLG-1. Since the mammalian orthologs of these genes are expressed in the central nervous system and their mutations are implicated in neuropsychiatric diseases, this molecular pathway might have been evolutionarily conserved.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules/metabolism , Nerve Tissue Proteins/metabolism , Receptors, GABA-A/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules, Neuronal/genetics , Electric Stimulation , GABAergic Neurons/physiology , HEK293 Cells , Humans , Luminescent Proteins/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Muscle Cells/metabolism , Mutation/genetics , Nerve Tissue Proteins/genetics , Neuromuscular Junction/metabolism , Protein Binding/genetics , Receptors, GABA-A/genetics , Tumor Suppressor Proteins/genetics
8.
Nat Commun ; 5: 4974, 2014 09 18.
Article in English | MEDLINE | ID: mdl-25232639

ABSTRACT

Single-molecule (SM) fluorescence microscopy allows the imaging of biomolecules in cultured cells with a precision of a few nanometres but has yet to be implemented in living adult animals. Here we used split-GFP (green fluorescent protein) fusions and complementation-activated light microscopy (CALM) for subresolution imaging of individual membrane proteins in live Caenorhabditis elegans (C. elegans). In vivo tissue-specific SM tracking of transmembrane CD4 and voltage-dependent Ca(2+) channels (VDCC) was achieved with a precision of 30 nm within neuromuscular synapses and at the surface of muscle cells in normal and dystrophin-mutant worms. Through diffusion analyses, we reveal that dystrophin is involved in modulating the confinement of VDCC within sarcolemmal membrane nanodomains in response to varying tonus of C. elegans body-wall muscles. CALM expands the applications of SM imaging techniques beyond the petri dish and opens the possibility to explore the molecular basis of homeostatic and pathological cellular processes with subresolution precision, directly in live animals.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Dystrophin/genetics , Mutation , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cell Membrane/metabolism , Diffusion , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/metabolism , Homeostasis , Membrane Proteins/metabolism , Microscopy, Fluorescence , Recombinant Fusion Proteins/metabolism , Sarcolemma/metabolism
9.
J Exp Biol ; 217(Pt 21): 3805-14, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25214488

ABSTRACT

Several human diseases, including hypokalemic periodic paralysis and Timothy syndrome, are caused by mutations in voltage-gated calcium channels. The effects of these mutations are not always well understood, partially because of difficulties in expressing these channels in heterologous systems. The use of Caenorhabditis elegans could be an alternative approach to determine the effects of mutations on voltage-gated calcium channel function because all the main types of voltage-gated calcium channels are found in C. elegans, a large panel of mutations already exists and efficient genetic tools are available to engineer customized mutations in any gene. In this study, we characterize the effects of two gain-of-function mutations in egl-19, which encodes the L-type calcium channel α1 subunit. One of these mutations, ad695, leads to the replacement of a hydrophobic residue in the IIIS4 segment. The other mutation, n2368, changes a conserved glycine of IS6 segment; this mutation has been identified in patients with Timothy syndrome. We show that both egl-19 (gain-of-function) mutants have defects in locomotion and morphology that are linked to higher muscle tone. Using in situ electrophysiological approaches in striated muscle cells, we provide evidence that this high muscle tone is due to a shift of the voltage dependency towards negative potentials, associated with a decrease of the inactivation rate of the L-type Ca(2+) current. Moreover, we show that the maximal conductance of the Ca(2+) current is decreased in the strongest mutant egl-19(n2368), and that this decrease is correlated with a mislocalization of the channel.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Striated/metabolism , Protein Subunits/genetics , Animals , DNA Primers/genetics , Gene Transfer Techniques , Locomotion/genetics , Locomotion/physiology , Microscopy, Fluorescence , Patch-Clamp Techniques , Point Mutation/genetics
10.
J Biol Chem ; 286(42): 36180-7, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21878625

ABSTRACT

Voltage-gated calcium channels, which play key roles in many physiological processes, are composed of a pore-forming α1 subunit associated with up to three auxiliary subunits. In vertebrates, the role of auxiliary subunits has mostly been studied in heterologous systems, mainly because of the severe phenotypes of knock-out animals. The genetic model Caenorhabditis elegans has all main types of voltage-gated calcium channels and strong loss-of-function mutations in all pore-forming and auxiliary subunits; it is therefore a useful model to investigate the roles of auxiliary subunits in their native context. By recording calcium currents from channel and auxiliary subunit mutants, we molecularly dissected the voltage-dependent calcium currents in striated muscle of C. elegans. We show that EGL-19 is the only α1 subunit that carries calcium currents in muscle cells. We then demonstrate that the α2/δ subunit UNC-36 modulates the voltage dependence, the activation kinetics, and the conductance of calcium currents, whereas another α2/δ subunit TAG-180 has no effect. Finally, we characterize mutants of the two ß subunits, CCB-1 and CCB-2. CCB-1 is necessary for viability, and voltage-dependent calcium currents are abolished in the absence of CCB-1 whereas CCB-2 does not affect currents. Altogether these results show that EGL-19, UNC-36, and CCB-1 underlie voltage-dependent calcium currents in C. elegans striated muscle.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Calcium/metabolism , Muscle Proteins/metabolism , Muscle, Striated/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Subunits/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Muscle Proteins/genetics , Mutation , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/metabolism , Potassium Channels, Voltage-Gated/genetics , Protein Subunits/genetics
11.
PLoS Biol ; 7(12): e1000265, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20027209

ABSTRACT

In the nematode Caenorhabditis elegans, cholinergic motor neurons stimulate muscle contraction as well as activate GABAergic motor neurons that inhibit contraction of the contralateral muscles. Here, we describe the composition of an ionotropic acetylcholine receptor that is required to maintain excitation of the cholinergic motor neurons. We identified a gain-of-function mutation that leads to spontaneous muscle convulsions. The mutation is in the pore domain of the ACR-2 acetylcholine receptor subunit and is identical to a hyperactivating mutation in the muscle receptor of patients with myasthenia gravis. Screens for suppressors of the convulsion phenotype led to the identification of other receptor subunits. Cell-specific rescue experiments indicate that these subunits function in the cholinergic motor neurons. Expression of these subunits in Xenopus oocytes demonstrates that the functional receptor is comprised of three alpha-subunits, UNC-38, UNC-63 and ACR-12, and two non-alpha-subunits, ACR-2 and ACR-3. Although this receptor exhibits a partially overlapping subunit composition with the C. elegans muscle acetylcholine receptor, it shows distinct pharmacology. Recordings from intact animals demonstrate that loss-of-function mutations in acr-2 reduce the excitability of the cholinergic motor neurons. By contrast, the acr-2(gf) mutation leads to a hyperactivation of cholinergic motor neurons and an inactivation of downstream GABAergic motor neurons in a calcium dependent manner. Presumably, this imbalance between excitatory and inhibitory input into muscles leads to convulsions. These data indicate that the ACR-2 receptor is important for the coordinated excitation and inhibition of body muscles underlying sinusoidal movement.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Locomotion , Motor Neurons/metabolism , Muscle Contraction , Receptors, Nicotinic/metabolism , Acetylcholine/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Molecular Sequence Data , Mutation , Receptors, Nicotinic/genetics , Synaptic Transmission , Xenopus , gamma-Aminobutyric Acid/metabolism
12.
Curr Biol ; 17(18): 1595-600, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17825559

ABSTRACT

Synaptojanin is a lipid phosphatase required to degrade phosphatidylinositol 4,5 bisphosphate (PIP(2)) at cell membranes during synaptic vesicle recycling. Synaptojanin mutants in C. elegans are severely uncoordinated and are depleted of synaptic vesicles, possibly because of accumulation of PIP(2). To identify proteins that act downstream of PIP(2) during endocytosis, we screened for suppressors of synaptojanin mutants in the nematode C. elegans. A class of uncoordinated mutants called "fainters" partially suppress the locomotory, vesicle depletion, and electrophysiological defects in synaptojanin mutants. These suppressor loci include the genes for the NCA ion channels, which are homologs of the vertebrate cation leak channel NALCN, and a novel gene called unc-80. We demonstrate that unc-80 encodes a novel, but highly conserved, neuronal protein required for the proper localization of the NCA-1 and NCA-2 ion channel subunits. These data suggest that activation of the NCA ion channel in synaptojanin mutants leads to defects in recycling of synaptic vesicles.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Endocytosis/physiology , Ion Channels/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Phosphoric Monoester Hydrolases/genetics , Animals , Axons/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/genetics , Endocytosis/genetics , Green Fluorescent Proteins/analysis , Ion Channels/analysis , Models, Genetic , Mutation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synaptic Transmission/genetics , Synaptic Vesicles/metabolism
13.
J Mol Biol ; 358(2): 387-95, 2006 Apr 28.
Article in English | MEDLINE | ID: mdl-16527307

ABSTRACT

The Caenorhabditis elegans SLO-1 channel belongs to the family of calcium-activated large conductance BK potassium channels. SLO-1 has been shown to be involved in neurotransmitter release and ethanol response. Here, we report that SLO-1 also has a critical role in muscles. Inactivation of the slo-1 gene in muscles leads to phenotypes similar to those caused by mutations of the dystrophin homologue dys-1. Notably, slo-1 mutations result in a progressive muscle degeneration when put into a sensitized genetic background. slo-1 localization was observed by gfp reporter gene in both the M-line and the dense bodies (Z line) of the C.elegans body-wall muscles. Using the inside-out configuration of the patch clamp technique on body-wall muscle cells of acutely dissected wild-type worms, we characterized a Ca2+-activated K+ channel that was identified unambiguously as SLO-1. Since neither the abundance nor the conductance of SLO-1 was changed significantly in dys-1 mutants compared to wild-type animals, it is likely that the inactivation of dys-1 causes a misregulation of SLO-1. All in all, these results indicate that SLO-1 function in C.elegans muscles is related to the dystrophin homologue DYS-1.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Dystrophin/metabolism , Large-Conductance Calcium-Activated Potassium Channels/physiology , Muscles/metabolism , Muscular Dystrophy, Animal/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans Proteins/metabolism , Muscles/pathology , Muscular Dystrophy, Animal/pathology , Mutation/genetics , Phenotype , Potassium Channels, Calcium-Activated/metabolism
14.
J Physiol ; 559(Pt 3): 715-20, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15254157

ABSTRACT

About 30 genes are predicted to encode degenerin/epithelial sodium channels (DEG/ENaCs) in Caenorhabditis elegans but the gating mode of these channels has not been determined. Using the whole-cell configuration of the patch-clamp technique in acutely dissected C. elegans, we investigated the effects of H+ as a potential activating factor of DEG/ENaCs on electrical properties of body wall muscle cells. Under current-clamp conditions, decreasing external pH from 7.2 to 6.1 led to a reversible depolarization of muscle cells associated with a decrease in input resistance which was partially inhibited by amiloride. Under voltage-clamp conditions, extracellular acidification activated an inward desensitizing current at -60 mV. In the absence of external Ca2+, H+ -gated channels were found to be slightly more permeable to Na+ than to K+ and were blocked by amiloride with a K0.5 of 31 microm at -60 mV. An inward current could be also activated by protons in a GABA receptor null mutant in the presence of D-tubocurare and in an unc-105 null mutant. These results demonstrate that ion channels sharing common properties with mammalian acid-sensing ion channels (ASICs) are functional in C. elegans muscle which should prove useful for understanding proton sensing in animals.


Subject(s)
Amiloride/pharmacology , Caenorhabditis elegans Proteins/physiology , Muscle Fibers, Skeletal/physiology , Protons , Sodium Channels/physiology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/agonists , Dose-Response Relationship, Drug , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins , Muscle Fibers, Skeletal/drug effects , Sodium Channel Agonists
15.
J Physiol ; 557(Pt 2): 379-88, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15020702

ABSTRACT

Degenerins have emerged from genetic studies in Caenorhabditis elegans as candidate mechanically gated amiloride-sensitive ion channels for transducing mechanical stimuli into cellular responses. In C. elegans muscle, the existence of a genetic interaction between the unc-105 degenerin gene and let-2, a gene encoding an alpha2(IV) collagen, raised the possibility that UNC-105 may function as a mechanically gated channel in a stretch receptor complex. However, to date, ion channel activity of UNC-105 has only been recorded in a gain-of-function mutant form in heterologous expression systems. In this study we investigated the in situ properties of UNC-105 using the whole cell configuration of the patch clamp technique on body wall muscle cells from acutely dissected C. elegans. Amiloride was found to be without effect on membrane potential of wild-type muscle cells, suggesting that the UNC-105 degenerin is electrically silent in resting muscle. Hypo-osmotic shocks induced a reversible depolarization of muscle cells but which was not affected by amiloride. Deformation of the cells by applying tension to the filamentous complex on which muscle cells remained attached or by ejecting external solution under pressure failed to induce any change of membrane potential. In gain-of-function unc-105(n506) mutant cells, an amiloride-sensitive inward Na(+) current was found to be constitutively active, leading to maintained muscle depolarization. An associated mutation in the alpha2(IV) collagen LET-2 led to the closure of the mutant UNC-105(n506) channel while a collagenase treatment of these double mutant cells caused it to re-open, giving evidence for a functional interaction between LET-2 collagen and mutant UNC-105 channel.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Collagen Type IV/physiology , Sodium Channels/physiology , Amiloride/pharmacology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Collagen Type IV/chemistry , Collagen Type IV/genetics , Collagenases/pharmacology , DNA, Helminth , Ion Channels/drug effects , Ion Channels/physiology , Mechanotransduction, Cellular , Membrane Potentials/drug effects , Membrane Proteins , Muscle Cells/drug effects , Muscle Cells/physiology , Muscle Spindles/physiology , Patch-Clamp Techniques , Sodium Channels/genetics
16.
J Physiol ; 544(2): 373-84, 2002 10 15.
Article in English | MEDLINE | ID: mdl-12381812

ABSTRACT

The properties of K(+) channels in body wall muscle cells acutely dissected from the nematode Caenorhabditis elegans were investigated at the macroscopic and unitary level using an in situ patch clamp technique. In the whole-cell configuration, depolarizations to potentials positive to -40 mV gave rise to outward currents resulting from the activation of two kinetically distinct voltage-dependent K(+) currents: a fast activating and inactivating 4-aminopyridine-sensitive component and a slowly activating and maintained tetraethylammonium-sensitive component. In cell-attached patches, voltage-dependent K(+) channels, with unitary conductances of 34 and 80 pS in the presence of 5 and 140 mM external K(+), respectively, activated at membrane potentials positive to -40 mV. Excision revealed that these channels corresponded to Ca(2+)-activated K(+) channels exhibiting an unusual sensitivity to internal Cl(-) and whose activity progressively decreased in inside-out conditions. After complete run-down of these channels, one third of inside-out patches displayed activity of another Ca(2+)-activated K(+) channel of smaller unitary conductance (6 pS at 0 mV in the presence of 5 mM external K(+)). In providing a detailed description of native K(+) currents in body wall muscle cells of C. elegans, this work lays the basis for further comparisons with mutants to assess the function of K(+) channels in this model organism that is highly amenable to molecular and classical genetics.


Subject(s)
Caenorhabditis elegans/physiology , Muscle Cells/metabolism , Potassium Channels/physiology , 4-Aminopyridine/pharmacology , Animals , Electric Conductivity , Electrophysiology , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels, Calcium-Activated/classification , Potassium Channels, Calcium-Activated/physiology , Tetraethylammonium/pharmacology
17.
J Cell Biol ; 159(2): 337-48, 2002 Oct 28.
Article in English | MEDLINE | ID: mdl-12391025

ABSTRACT

Caenorhabditis elegans is a powerful model system widely used to investigate the relationships between genes and complex behaviors like locomotion. However, physiological studies at the cellular level have been restricted by the difficulty to dissect this microscopic animal. Thus, little is known about the properties of body wall muscle cells used for locomotion. Using in situ patch clamp technique, we show that body wall muscle cells generate spontaneous spike potentials and develop graded action potentials in response to injection of positive current of increasing amplitude. In the presence of K+ channel blockers, membrane depolarization elicited Ca2+ currents inhibited by nifedipine and exhibiting Ca2+-dependent inactivation. Our results give evidence that the Ca2+ channel involved belongs to the L-type class and corresponds to EGL-19, a putative Ca2+ channel originally thought to be a member of this class on the basis of genomic data. Using Ca2+ fluorescence imaging on patch-clamped muscle cells, we demonstrate that the Ca2+ transients elicited by membrane depolarization are under the control of Ca2+ entry through L-type Ca2+ channels. In reduction of function egl-19 mutant muscle cells, Ca2+ currents displayed slower activation kinetics and provided a significantly smaller Ca2+ entry, whereas the threshold for Ca2+ transients was shifted toward positive membrane potentials.


Subject(s)
Caenorhabditis elegans Proteins , Calcium Channels/metabolism , Locomotion/physiology , Muscle Proteins/metabolism , Muscles/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Caenorhabditis elegans , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Dihydropyridines/pharmacology , Muscle Proteins/genetics , Mutation/physiology , Patch-Clamp Techniques , Sodium Chloride
SELECTION OF CITATIONS
SEARCH DETAIL
...