Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(30): e2407461121, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39018191

RESUMEN

The Shaker family of voltage-gated K+ channels has been thought of as an animal-specific ion channel family that diversified in concert with nervous systems. It comprises four functionally independent gene subfamilies (Kv1-4) that encode diverse neuronal K+ currents. Comparison of animal genomes predicts that only the Kv1 subfamily was present in the animal common ancestor. Here, we show that some choanoflagellates, the closest protozoan sister lineage to animals, also have Shaker family K+ channels. Choanoflagellate Shaker family channels are surprisingly most closely related to the animal Kv2-4 subfamilies which were believed to have evolved only after the divergence of ctenophores and sponges from cnidarians and bilaterians. Structural modeling predicts that the choanoflagellate channels share a T1 Zn2+ binding site with Kv2-4 channels that is absent in Kv1 channels. We functionally expressed three Shakers from Salpingoeca helianthica (SheliKvT1.1-3) in Xenopus oocytes. SheliKvT1.1-3 function only in two heteromultimeric combinations (SheliKvT1.1/1.2 and SheliKvT1.1/1.3) and encode fast N-type inactivating K+ channels with distinct voltage dependence that are most similar to the widespread animal Kv1-encoded A-type Shakers. Structural modeling of the T1 assembly domain supports a preference for heteromeric assembly in a 2:2 stoichiometry. These results push the origin of the Shaker family back into a common ancestor of metazoans and choanoflagellates. They also suggest that the animal common ancestor had at least two distinct molecular lineages of Shaker channels, a Kv1 subfamily lineage predicted from comparison of animal genomes and a Kv2-4 lineage predicted from comparison of animals and choanoflagellates.


Asunto(s)
Coanoflagelados , Evolución Molecular , Canales de Potasio de la Superfamilia Shaker , Animales , Coanoflagelados/genética , Coanoflagelados/metabolismo , Canales de Potasio de la Superfamilia Shaker/genética , Canales de Potasio de la Superfamilia Shaker/metabolismo , Filogenia , Secuencia de Aminoácidos
2.
J Biol Chem ; 298(12): 102621, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36272643

RESUMEN

Cav3 T-type calcium channels from great pond snail Lymnaea stagnalis have a selectivity-filter ring of five acidic residues, EE(D)DD. Splice variants with exons 12b or 12a spanning the extracellular loop between the outer helix IIS5 and membrane-descending pore helix IIP1 (IIS5-P1) in Domain II of the pore module possess calcium selectivity or dominant sodium permeability, respectively. Here, we use AlphaFold2 neural network software to predict that a lysine residue in exon 12a is salt-bridged to the aspartate residue immediately C terminal to the second-domain glutamate in the selectivity filter. Exon 12b has a similar folding but with an alanine residue in place of lysine in exon 12a. We express LCav3 channels with mutated exons Ala-12b-Lys and Lys-12a-Ala and demonstrate that they switch the ion preference to high sodium permeability and calcium selectivity, respectively. We propose that in the calcium-selective variants, a calcium ion chelated between Domain II selectivity-filter glutamate and aspartate is knocked-out by the incoming calcium ion in the process of calcium permeation, whereas sodium ions are repelled. The aspartate is neutralized by the lysine residue in the sodium-permeant variants, allowing for sodium permeation through the selectivity-filter ring of four negatively charged residues akin to the prokaryotic sodium channels with four glutamates in the selectivity filter. The evolutionary adaptation in invertebrate LCav3 channels highlight the involvement of a key, ubiquitous aspartate, "a calcium beacon" of sorts in the outer pore of Domain II, as determinative for the calcium ion preference over sodium ions through eukaryotic Cav1, Cav2, and Cav3 channels.


Asunto(s)
Canales de Calcio Tipo T , Calcio , Lisina , Sodio , Ácido Aspártico , Calcio/química , Ácido Glutámico , Iones , Lisina/química , Sodio/química , Lymnaea , Animales , Canales de Calcio Tipo T/química
3.
J Biol Chem ; 292(49): 20010-20031, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-28972185

RESUMEN

Calcium (Cav1 and Cav2) and sodium channels possess homologous CaM-binding motifs, known as IQ motifs in their C termini, which associate with calmodulin (CaM), a universal calcium sensor. Cav3 T-type channels, which serve as pacemakers of the mammalian brain and heart, lack a C-terminal IQ motif. We illustrate that T-type channels associate with CaM using co-immunoprecipitation experiments and single particle cryo-electron microscopy. We demonstrate that protostome invertebrate (LCav3) and human Cav3.1, Cav3.2, and Cav3.3 T-type channels specifically associate with CaM at helix 2 of the gating brake in the I-II linker of the channels. Isothermal titration calorimetry results revealed that the gating brake and CaM bind each other with high-nanomolar affinity. We show that the gating brake assumes a helical conformation upon binding CaM, with associated conformational changes to both CaM lobes as indicated by amide chemical shifts of the amino acids of CaM in 1H-15N HSQC NMR spectra. Intact Ca2+-binding sites on CaM and an intact gating brake sequence (first 39 amino acids of the I-II linker) were required in Cav3.2 channels to prevent the runaway gating phenotype, a hyperpolarizing shift in voltage sensitivities and faster gating kinetics. We conclude that the presence of high-nanomolar affinity binding sites for CaM at its universal gating brake and its unique form of regulation via the tuning of the voltage range of activity could influence the participation of Cav3 T-type channels in heart and brain rhythms. Our findings may have implications for arrhythmia disorders arising from mutations in the gating brake or CaM.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Calmodulina/fisiología , Caveolina 3/metabolismo , Activación del Canal Iónico , Animales , Sitios de Unión , Encéfalo/fisiología , Calcio/metabolismo , Calmodulina/metabolismo , Corazón/fisiología , Humanos , Invertebrados , Periodicidad
4.
J Biol Chem ; 289(17): 11952-11969, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24596098

RESUMEN

T-type (Cav3) channels are categorized as calcium channels, but invertebrate ones can be highly sodium-selective channels. We illustrate that the snail LCav3 T-type channel becomes highly sodium-permeable through exon splicing of an extracellular turret and descending helix in domain II of the four-domain Cav3 channel. Highly sodium-permeable T-type channels are generated without altering the invariant ring of charged residues in the selectivity filter that governs calcium selectivity in calcium channels. The highly sodium-permeant T-type channel expresses in the brain and is the only splice isoform expressed in the snail heart. This unique splicing of turret residues offers T-type channels a capacity to serve as a pacemaking sodium current in the primitive heart and brain in lieu of Nav1-type sodium channels and to substitute for voltage-gated sodium channels lacking in many invertebrates. T-type channels would also contribute substantially to sodium leak conductances at rest in invertebrates because of their large window currents.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Sodio/metabolismo , Animales , Secuencia de Bases , Canales de Calcio Tipo T/genética , Cationes , Células Cultivadas , Clonación Molecular , Cartilla de ADN , Técnicas de Placa-Clamp , Permeabilidad , Reacción en Cadena de la Polimerasa , Empalme del ARN , Caracoles
5.
Channels (Austin) ; 17(1): 2191773, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37075164

RESUMEN

One of nature's exceptions was discovered when a Cav3 T-type channel was observed to switch phenotype from a calcium channel into a sodium channel by neutralizing an aspartate residue in the high field strength (HFS) +1 position within the ion selectivity filter. The HFS+1 site is dubbed a "beacon" for its location at the entryway just above the constricted, minimum radius of the HFS site's electronegative ring. A classification is proposed based on the occupancy of the HFS+1 "beacon" which correlates with the calcium- or sodium-selectivity phenotype. If the beacon is a glycine, or neutral, non-glycine residue, then the cation channel is calcium-selective or sodium-permeable, respectively (Class I). Occupancy of a beacon aspartate are calcium-selective channels (Class II) or possessing a strong calcium block (Class III). A residue lacking in position of the sequence alignment for the beacon are sodium channels (Class IV). The extent to which animal channels are sodium-selective is dictated in the occupancy of the HFS site with a lysine residue (Class III/IV). Governance involving the beacon solves the quandary the HFS site as a basis for ion selectivity, where an electronegative ring of glutamates at the HFS site generates a sodium-selective channel in one-domain channels but generates a calcium-selective channel in four-domain channels. Discovery of a splice variant in an exceptional channel revealed nature's exploits, highlighting the "beacon" as a principal determinant for calcium and sodium selectivity, encompassing known ion channels composed of one and four domains, from bacteria to animals.


Asunto(s)
Ácido Aspártico , Calcio , Animales , Calcio/metabolismo , Secuencia de Aminoácidos , Canales de Calcio/química , Canales de Sodio/metabolismo , Sodio/metabolismo
6.
J Biol Chem ; 285(10): 7447-58, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20056611

RESUMEN

Here we describe features of the first non-mammalian T-type calcium channel (LCa(v)3) expressed in vitro. This molluscan channel possesses combined biophysical properties that are reminiscent of all mammalian T-type channels. It exhibits T-type features such as "transient" kinetics, but the "tiny" label, usually associated with Ba(2+) conductance, is hard to reconcile with the "bigness" of this channel in many respects. LCa(v)3 is 25% larger than any voltage-gated ion channel expressed to date. It codes for a massive, 322-kDa protein that conducts large macroscopic currents in vitro. LCa(v)3 is also the most abundant Ca(2+) channel transcript in the snail nervous system. A window current at typical resting potentials appears to be at least as large as that reported for mammalian channels. This distant gene provides a unique perspective to analyze the structural, functional, drug binding, and evolutionary aspects of T-type channels.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Sistema Nervioso Central/metabolismo , Activación del Canal Iónico/fisiología , Lymnaea , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Bloqueadores de los Canales de Calcio/metabolismo , Canales de Calcio Tipo T/química , Canales de Calcio Tipo T/clasificación , Canales de Calcio Tipo T/genética , Línea Celular , Humanos , Lymnaea/anatomía & histología , Lymnaea/metabolismo , Mibefradil/metabolismo , Datos de Secuencia Molecular , Níquel/metabolismo , Técnicas de Placa-Clamp , Filogenia , Conformación Proteica , Alineación de Secuencia , Homología de Secuencia de Aminoácido
7.
Brain Sci ; 11(6)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070393

RESUMEN

A 400-level undergraduate oral presentation and discussion course in Systems Neuroscience was delivered asynchronously online during the COVID-19 pandemic. Enrolled students banked their narrated oral presentations in video format online then engaged in peer evaluation in virtual classrooms through the course website. Student delivered their oral presentation and responded to peer questions at their leisure and convenience, without the stress and anxiety associated with a "live" performance delivery in front of their peers. A remote and asynchronously delivered course facilitated much more peer contact than "live" versions of the course, which included a total of 62 uploaded presentations, 301 video responses uploaded to 1985 questions posed by peers, a total of 1159 feedback questionnaires submitted, 1066 rankings submitted of viewed oral presentations, and 1091 scores submitted evaluating the quality of questions posed by reviewers of oral presentations. A major drawback in the remote, asynchronous deliver was the enormity of peer engagement through the course website portal, which was mostly blind to the instructor because of the inability to effectively cross-index data linked between the student entries in the LEARN course website and the uploaded videos stored within BONGO Video Assignment tool. Nonetheless, a consistent engagement of students, and the positive feedback from enrolled students, indicate that a future version of this oral/written discussion course will be delivered, in part, remotely and asynchronously, even without a mandated delivery of the course by a remote and asynchronous method due to the COVID-19 pandemic restrictions in 2020-2021.

8.
Sci Rep ; 10(1): 12404, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32710088

RESUMEN

Invertebrate LCaV3 shares the quintessential features of vertebrate CaV3 T-type channels, with a low threshold of channel activation, rapid activation and inactivation kinetics and slow deactivation kinetics compared to other known Ca2+ channels, the CaV1 and CaV2 channels. Unlike the vertebrates though, CaV3 T-type channels in non-cnidarian invertebrates possess an alternative exon 12 spanning the D2L5 extracellular loop, which alters the invertebrate LCaV3 channel into a higher Na+ and lower Ca2+ current passing channel, more resembling a classical NaV1 Na+ channel. Cnidarian CaV3 T-type channels can possess genes with alternative cysteine-rich, D4L6 extracellular loops in a manner reminiscent of the alternative cysteine-rich, D2L5 extracellular loops of non-cnidarian invertebrates. We illustrate here that the preferences for greater Na+ or Ca2+ ion current passing through CaV3 T-type channels are contributed by paired cysteines within D2L5 and D4L6 extracellular loops looming above the pore selectivity filter. Swapping of invertebrate tri- and tetra-cysteine containing extracellular loops, generates higher Na+ current passing channels in human CaV3.2 channels, while corresponding mono- and di-cysteine loop pairs in human CaV3.2 generates greater Ca2+ current passing, invertebrate LCaV3 channels. Alanine substitutions of unique D2L5 loop cysteines of LCaV3 channels increases relative monovalent ion current sizes and increases the potency of Zn2+ and Ni2+ block by ~ 50× and ~ 10× in loop cysteine mutated channels respectively, acquiring characteristics of the high affinity block of CaV3.2 channels, including the loss of the slowing of inactivation kinetics during Zn2+ block. Charge neutralization of a ubiquitous aspartate residue of calcium passing CaV1, CaV2 and CaV3 channels, in the outer pore of the selectivity filter residues in Domain II generates higher Na+ current passing channels in a manner that may resemble how the unique D2L5 extracellular loops of invertebrate CaV3 channels may confer a relatively higher peak current size for Na+ ions over Ca2+ The extracellular loops of CaV3 channels are not engaged with accessory subunit binding, as the other Na+ (NaV1) and Ca2+ (CaV1/CaV2) channels, enabling diversity and expansion of cysteine-bonded extracellular loops, which appears to serve, amongst other possibilities, to alter to the preferences for passage of Ca2+ or Na+ ions through invertebrate CaV3 channels.


Asunto(s)
Bloqueadores de los Canales de Calcio/química , Bloqueadores de los Canales de Calcio/farmacología , Caveolina 3/antagonistas & inhibidores , Caveolina 3/química , Cisteína , Espacio Extracelular/metabolismo , Secuencia de Aminoácidos , Calcio/metabolismo , Caveolina 3/metabolismo , Humanos
9.
Biochim Biophys Acta Biomembr ; 1862(11): 183439, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32814116

RESUMEN

Eukaryote voltage-gated Ca2+ channels of the CaV2 channel family are hetero-oligomers formed by the pore-forming CaVα1 protein assembled with auxiliary CaVα2δ and CaVß subunits. CaVß subunits are formed by a Src homology 3 (SH3) domain and a guanylate kinase (GK) domain connected through a HOOK domain. The GK domain binds a conserved cytoplasmic region of the pore-forming CaVα1 subunit referred as the "AID". Herein we explored the phylogenetic and functional relationship between CaV channel subunits in distant eukaryotic organisms by investigating the function of a MAGUK protein (XM_004990081) cloned from the choanoflagellate Salpingoeca rosetta (Sro). This MAGUK protein (Sroß) features SH3 and GK structural domains with a 25% primary sequence identity to mammalian CaVß. Recombinant expression of its cDNA with mammalian high-voltage activated Ca2+ channel CaV2.3 in mammalian HEK cells produced robust voltage-gated inward Ca2+ currents with typical activation and inactivation properties. Like CaVß, Sroß prevents fast degradation of total CaV2.3 proteins in cycloheximide assays. The three-dimensional homology model predicts an interaction between the GK domain of Sroß and the AID motif of the pore-forming CaVα1 protein. Substitution of AID residues Trp (W386A) and Tyr (Y383A) significantly impaired co-immunoprecipitation of CaV2.3 with Sroß and functional upregulation of CaV2.3 currents. Likewise, a 6-residue deletion within the GK domain of Sroß, similar to the locus found in mammalian CaVß, significantly reduced peak current density. Altogether our data demonstrate that an ancestor MAGUK protein reconstitutes the biophysical and molecular features responsible for channel upregulation by mammalian CaVß through a minimally conserved molecular interface.


Asunto(s)
Canales de Calcio Tipo R/química , Proteínas de Transporte de Catión/química , Guanilato-Quinasas/química , Proteínas Protozoarias/química , Sustitución de Aminoácidos , Canales de Calcio Tipo R/genética , Canales de Calcio Tipo R/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Células HEK293 , Humanos , Mutación Missense , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
10.
J Neurosci ; 28(10): 2601-12, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18322103

RESUMEN

Activation of protein kinase C (PKC) potentiates secretion in Aplysia peptidergic neurons, in part by inducing new sites for peptide release at growth cone terminals. The mechanisms by which ion channels are trafficked to such sites are, however, not well understood. We now show that PKC activation rapidly recruits new Ca(V)2 subunits to the plasma membrane, and that recruitment is blocked by latrunculin B, an inhibitor of actin polymerization. In contrast, inhibition of microtubule polymerization selectively prevents the appearance of Ca(V)2 subunits only at the distal edge of the growth cone. In resting neurons, Ca(V)2-containing organelles reside in the central region of growth cones, but are absent from distal lamellipodia. After activation of PKC, these organelles are transported on microtubules to the lamellipodium. The ability to traffic to the most distal sites of channel insertion inside the lamellipodium does, therefore, not require intact actin but requires intact microtubules. Only after activation of PKC do Ca(V)2 channels associate with actin and undergo insertion into the plasma membrane.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Canales de Calcio/metabolismo , Membrana Celular/metabolismo , Líquido Intracelular/fisiología , Proteína Quinasa C/fisiología , Subunidades de Proteína/metabolismo , Animales , Aplysia , Células Cultivadas , Activación Enzimática/fisiología , Transporte de Proteínas/fisiología
11.
Biochem Biophys Res Commun ; 368(3): 827-31, 2008 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-18279668

RESUMEN

Rem2 belongs to the RGK family of small GTPases whose members are known to interact with the voltage gated calcium channel beta subunit, and to inhibit or abolish calcium currents. To identify the underlying functional domains of Rem2, we created several N- or C-terminally truncated Rem2 proteins and examined their abilities to interact with the Ca(v) beta subunit and to regulate the activities of Ca(v)2.2 N-type calcium channels. Confocal imaging of Rem2 in tsA-201 cells revealed that it contains a membrane-targeting signal in its C-terminus, consistent with previous studies. Co-precipitation assays showed that Ca(v) beta(3) interaction depends on Rem2 residues 1-123. Only Rem2 proteins that targeted the cell membrane as well as bound the beta subunit were able to reduce whole cell calcium currents.


Asunto(s)
Canales de Calcio Tipo N/química , Canales de Calcio Tipo N/metabolismo , Calcio/metabolismo , Activación del Canal Iónico/fisiología , Riñón/metabolismo , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/metabolismo , Animales , Línea Celular , Humanos , Ratones , Relación Estructura-Actividad
12.
Front Physiol ; 9: 1406, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30519187

RESUMEN

The appearance of voltage-gated, sodium-selective channels with rapid gating kinetics was a limiting factor in the evolution of nervous systems. Two rounds of domain duplications generated a common 24 transmembrane segment (4 × 6 TM) template that is shared amongst voltage-gated sodium (Nav1 and Nav2) and calcium channels (Cav1, Cav2, and Cav3) and leak channel (NALCN) plus homologs from yeast, different single-cell protists (heterokont and unikont) and algae (green and brown). A shared architecture in 4 × 6 TM channels include an asymmetrical arrangement of extended extracellular L5/L6 turrets containing a 4-0-2-2 pattern of cysteines, glycosylated residues, a universally short III-IV cytoplasmic linker and often a recognizable, C-terminal PDZ binding motif. Six intron splice junctions are conserved in the first domain, including a rare U12-type of the minor spliceosome provides support for a shared heritage for sodium and calcium channels, and a separate lineage for NALCN. The asymmetrically arranged pores of 4x6 TM channels allows for a changeable ion selectivity by means of a single lysine residue change in the high field strength site of the ion selectivity filter in Domains II or III. Multicellularity and the appearance of systems was an impetus for Nav1 channels to adapt to sodium ion selectivity and fast ion gating. A non-selective, and slowly gating Nav2 channel homolog in single cell eukaryotes, predate the diversification of Nav1 channels from a basal homolog in a common ancestor to extant cnidarians to the nine vertebrate Nav1.x channel genes plus Nax. A close kinship between Nav2 and Nav1 homologs is evident in the sharing of most (twenty) intron splice junctions. Different metazoan groups have lost their Nav1 channel genes altogether, while vertebrates rapidly expanded their gene numbers. The expansion in vertebrate Nav1 channel genes fills unique functional niches and generates overlapping properties contributing to redundancies. Specific nervous system adaptations include cytoplasmic linkers with phosphorylation sites and tethered elements to protein assemblies in First Initial Segments and nodes of Ranvier. Analogous accessory beta subunit appeared alongside Nav1 channels within different animal sub-phyla. Nav1 channels contribute to pace-making as persistent or resurgent currents, the former which is widespread across animals, while the latter is a likely vertebrate adaptation.

13.
Curr Opin Neurobiol ; 13(3): 308-14, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12850215

RESUMEN

In vertebrates, the physical coupling between presynaptic calcium channels and synaptic vesicle release proteins enhances the efficiency of neurotransmission. Recent evidence indicates that these synaptic proteins may feedback directly on synaptic release by negatively regulating calcium entry, and indirectly through pathways involving second messenger molecules. Studies of individual neurons from both vertebrates and invertebrates have provided novel insights into the roles of scaffolding proteins in calcium channel targeting and neurotransmitter release. These studies require us to expand current models of synaptic transmission.


Asunto(s)
Canales de Calcio/química , Canales de Calcio/metabolismo , Neurotransmisores/metabolismo , Terminales Presinápticos/química , Terminales Presinápticos/metabolismo , Animales , Canales de Calcio/fisiología , Humanos , Neurotransmisores/fisiología , Terminales Presinápticos/fisiología
14.
Front Physiol ; 6: 153, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26042044

RESUMEN

How nature discriminates sodium from calcium ions in eukaryotic channels has been difficult to resolve because they contain four homologous, but markedly different repeat domains. We glean clues from analyzing the changing pore region in sodium, calcium and NALCN channels, from single-cell eukaryotes to mammals. Alternative splicing in invertebrate homologs provides insights into different structural features underlying calcium and sodium selectivity. NALCN generates alternative ion selectivity with splicing that changes the high field strength (HFS) site at the narrowest level of the hourglass shaped pore where the selectivity filter is located. Alternative splicing creates NALCN isoforms, in which the HFS site has a ring of glutamates contributed by all four repeat domains (EEEE), or three glutamates and a lysine residue in the third (EEKE) or second (EKEE) position. Alternative splicing provides sodium and/or calcium selectivity in T-type channels with extracellular loops between S5 and P-helices (S5P) of different lengths that contain three or five cysteines. All eukaryotic channels have a set of eight core cysteines in extracellular regions, but the T-type channels have an infusion of 4-12 extra cysteines in extracellular regions. The pattern of conservation suggests a possible pairing of long loops in Domains I and III, which are bridged with core cysteines in NALCN, Cav, and Nav channels, and pairing of shorter loops in Domains II and IV in T-type channel through disulfide bonds involving T-type specific cysteines. Extracellular turrets of increasing lengths in potassium channels (Kir2.2, hERG, and K2P1) contribute to a changing landscape above the pore selectivity filter that can limit drug access and serve as an ion pre-filter before ions reach the pore selectivity filter below. Pairing of extended loops likely contributes to the large extracellular appendage as seen in single particle electron cryo-microscopy images of the eel Nav1 channel.

15.
PLoS One ; 9(6): e98808, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24945283

RESUMEN

The properties of leaky patch currents in whole cell recording of HEK-293T cells were examined as a means to separate these control currents from expressed sodium and calcium leak channel currents from snail NALCN leak channels possessing both sodium (EKEE) and calcium (EEEE) selectivity filters. Leak currents were generated by the weakening of gigaohm patch seals by artificial membrane rupture using the ZAP function on the patch clamp amplifier. Surprisingly, we found that leak currents generated from the weakened membrane/glass seal can be surprisingly stable and exhibit behavior that is consistent with a sodium leak current derived from an expressible channel. Leaky patch currents differing by 10 fold in size were similarly reduced in size when external sodium ions were replaced with the large monovalent ion NMDG+. Leaky patch currents increased when external Ca2+ (1.2 mM) was lowered to 0.1 mM and were inhibited (>40% to >90%) with 10 µM Gd3+, 100 µM La3+, 1 mM Co2+ or 1 mM Cd2+. Leaky patch currents were relatively insensitive (<30%) to 1 mM Ni2+ and exhibited a variable amount of block with 1 mM verapamil and were insensitive to 100 µM mibefradil or 100 µM nifedipine. We hypothesize that the rapid changes in leak current size in response to changing external cations or drugs relates to their influences on the membrane seal adherence and the electro-osmotic flow of mobile cations channeling in crevices of a particular pore size in the interface between the negatively charged patch electrode and the lipid membrane. Observed sodium leak conductance currents in weak patch seals are reproducible between the electrode glass interface with cell membranes, artificial lipid or Sylgard rubber.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Canales de Sodio/biosíntesis , Sodio/metabolismo , Canales de Calcio/biosíntesis , Canales de Calcio/metabolismo , Membrana Celular/química , Células HEK293 , Humanos , Canales Iónicos , Proteínas de la Membrana , Nifedipino/farmacología , Técnicas de Placa-Clamp , Canales de Sodio/metabolismo , Verapamilo/administración & dosificación
16.
PLoS One ; 9(4): e92941, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24690951

RESUMEN

The accessory beta subunit (Ca(v)ß) of calcium channels first appear in the same genome as Ca(v)1 L-type calcium channels in single-celled coanoflagellates. The complexity of this relationship expanded in vertebrates to include four different possible Ca(v)ß subunits (ß1, ß2, ß3, ß4) which associate with four Ca(v)1 channel isoforms (Ca(v)1.1 to Ca(v)1.4) and three Ca(v)2 channel isoforms (Ca(v)2.1 to Ca(v)2.3). Here we assess the fundamentally-shared features of the Ca(v)ß subunit in an invertebrate model (pond snail Lymnaea stagnalis) that bears only three homologous genes: (LCa(v)1, LCa(v)2, and LCa(v)ß). Invertebrate Ca(v)ß subunits (in flatworms, snails, squid and honeybees) slow the inactivation kinetics of Ca(v)2 channels, and they do so with variable N-termini and lacking the canonical palmitoylation residues of the vertebrate ß2a subunit. Alternative splicing of exon 7 of the HOOK domain is a primary determinant of a slow inactivation kinetics imparted by the invertebrate LCa(v)ß subunit. LCa(v)ß will also slow the inactivation kinetics of LCa(v)3 T-type channels, but this is likely not physiologically relevant in vivo. Variable N-termini have little influence on the voltage-dependent inactivation kinetics of differing invertebrate Ca(v)ß subunits, but the expression pattern of N-terminal splice isoforms appears to be highly tissue specific. Molluscan LCa(v)ß subunits have an N-terminal "A" isoform (coded by exons: 1a and 1b) that structurally resembles the muscle specific variant of vertebrate ß1a subunit, and has a broad mRNA expression profile in brain, heart, muscle and glands. A more variable "B" N-terminus (exon 2) in the exon position of mammalian ß3 and has a more brain-centric mRNA expression pattern. Lastly, we suggest that the facilitation of closed-state inactivation (e.g. observed in Ca(v)2.2 and Ca(v)ß3 subunit combinations) is a specialization in vertebrates, because neither snail subunit (LCa(v)2 nor LCa(v)ß) appears to be compatible with this observed property.


Asunto(s)
Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Lymnaea/genética , Subunidades de Proteína/química , Subunidades de Proteína/genética , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Canales de Calcio Tipo L/metabolismo , Secuencia Conservada , Exones/genética , Perfilación de la Expresión Génica , Humanos , Intrones/genética , Activación del Canal Iónico , Cinética , Datos de Secuencia Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia
17.
Channels (Austin) ; 7(2): 60-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23442378

RESUMEN

NALCN is an intriguing, orphan ion channel among the 4x6TM family of related voltage-gated cation channels, sharing a common architecture of four homologous domains consisting of six transmembrane helices, separated by three cytoplasmic linkers and delimited by N and C-terminal ends. NALCN is one of the shortest 4x6TM family members, lacking much of the variation that provides the diverse palate of gating features, and tissue specific adaptations of sodium and calcium channels. NALCN's most distinctive feature is that that it possesses a highly adaptable pore with a calcium-like EEEE selectivity filter in radially symmetrical animals and a more sodium-like EEKE or EKEE selectivity filter in bilaterally symmetrical animals including vertebrates. Two lineages of animals evolved alternative calcium-like EEEE and sodium-like EEKE / EKEE pores, spliced to regulate NALCN functions in differing cellular environments, such as muscle (heart and skeletal) and secretory tissue (brain and glands), respectively. A highly adaptable pore in an otherwise conserved ion channel in the 4x6TM channel family is not consistent with a role for NALCN in directly gating a significant ion conductance that can be either sodium ions or calcium ions. NALCN was proposed to be an expressible Gd ( 3+) -sensitive, NMDG (+) -impermeant, non-selective and ohmic leak conductance in HEK-293T cells, but we were unable to distinguish these reported currents from leaky patch currents (ILP) in control HEK-293T cells. We suggest that NALCN functions as a sensor for the much larger UNC80/UNC79 complex, in a manner consistent with the coupling mechanism known for other weakly or non-conducting 4x6TM channel sensor proteins such as Nax or Cav 1.1. We propose that NALCN serves as a variable sensor that responds to calcium or sodium ion flux, depending on whether the total cellular current density is generated more from calcium-selective or sodium-selective channels.


Asunto(s)
Canales de Calcio/química , Canales de Sodio/química , Adaptación Fisiológica/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas Portadoras/metabolismo , Evolución Molecular , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Sodio/metabolismo , Canales de Sodio/genética , Canales de Sodio/metabolismo
18.
PLoS One ; 8(1): e55088, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383067

RESUMEN

NALCN is a member of the family of ion channels with four homologous, repeat domains that include voltage-gated calcium and sodium channels. NALCN is a highly conserved gene from simple, extant multicellular organisms without nervous systems such as sponges and placozoans and mostly remains a single gene compared to the calcium and sodium channels which diversified into twenty genes in humans. The single NALCN gene has alternatively-spliced exons at exons 15 or exon 31 that splices in novel selectivity filter residues that resemble calcium channels (EEEE) or sodium channels (EKEE or EEKE). NALCN channels with alternative calcium, (EEEE) and sodium, (EKEE or EEKE) -selective pores are conserved in simple bilaterally symmetrical animals like flatworms to non-chordate deuterostomes. The single NALCN gene is limited as a sodium channel with a lysine (K)-containing pore in vertebrates, but originally NALCN was a calcium-like channel, and evolved to operate as both a calcium channel and sodium channel for different roles in many invertebrates. Expression patterns of NALCN-EKEE in pond snail, Lymnaea stagnalis suggest roles for NALCN in secretion, with an abundant expression in brain, and an up-regulation in secretory organs of sexually-mature adults such as albumen gland and prostate. NALCN-EEEE is equally abundant as NALCN-EKEE in snails, but is greater expressed in heart and other muscle tissue, and 50% less expressed in the brain than NALCN-EKEE. Transfected snail NALCN-EEEE and NALCN-EKEE channel isoforms express in HEK-293T cells. We were not able to distinguish potential NALCN currents from background, non-selective leak conductances in HEK293T cells. Native leak currents without expressing NALCN genes in HEK-293T cells are NMDG(+) impermeant and blockable with 10 µM Gd(3+) ions and are indistinguishable from the hallmark currents ascribed to mammalian NALCN currents expressed in vitro by Lu et al. in Cell. 2007 Apr 20;129(2):371-83.


Asunto(s)
Empalme Alternativo , Canales de Calcio/química , Canales de Sodio/química , Canales de Sodio/genética , Secuencia de Aminoácidos , Animales , Canales de Calcio/genética , Secuencia Conservada , Evolución Molecular , Regulación de la Expresión Génica , Humanos , Canales Iónicos , Proteínas de la Membrana , Datos de Secuencia Molecular , Filogenia , Porosidad , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Caracoles , Canales de Sodio/metabolismo
19.
PLoS One ; 8(4): e61765, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23626724

RESUMEN

NSCaTE is a short linear motif of (xWxxx(I or L)xxxx), composed of residues with a high helix-forming propensity within a mostly disordered N-terminus that is conserved in L-type calcium channels from protostome invertebrates to humans. NSCaTE is an optional, lower affinity and calcium-sensitive binding site for calmodulin (CaM) which competes for CaM binding with a more ancient, C-terminal IQ domain on L-type channels. CaM bound to N- and C- terminal tails serve as dual detectors to changing intracellular Ca(2+) concentrations, promoting calcium-dependent inactivation of L-type calcium channels. NSCaTE is absent in some arthropod species, and is also lacking in vertebrate L-type isoforms, Cav1.1 and Cav1.4 channels. The pervasiveness of a methionine just downstream from NSCaTE suggests that L-type channels could generate alternative N-termini lacking NSCaTE through the choice of translational start sites. Long N-terminus with an NSCaTE motif in L-type calcium channel homolog LCav1 from pond snail Lymnaea stagnalis has a faster calcium-dependent inactivation than a shortened N-termini lacking NSCaTE. NSCaTE effects are present in low concentrations of internal buffer (0.5 mM EGTA), but disappears in high buffer conditions (10 mM EGTA). Snail and mammalian NSCaTE have an alpha-helical propensity upon binding Ca(2+)-CaM and can saturate both CaM N-terminal and C-terminal domains in the absence of a competing IQ motif. NSCaTE evolved in ancestors of the first animals with internal organs for promoting a more rapid, calcium-sensitive inactivation of L-type channels.


Asunto(s)
Canales de Calcio Tipo L/química , Calmodulina/metabolismo , Caracoles/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Evolución Biológica , Canales de Calcio Tipo L/clasificación , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Calmodulina/química , Calmodulina/genética , Secuencia Conservada , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Filogenia , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Homología de Secuencia de Aminoácido , Caracoles/genética
20.
PLoS One ; 7(6): e37409, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22719839

RESUMEN

T-type calcium channels operate within tightly regulated biophysical constraints for supporting rhythmic firing in the brain, heart and secretory organs of invertebrates and vertebrates. The snail T-type gene, LCa(v)3 from Lymnaea stagnalis, possesses alternative, tandem donor splice sites enabling a choice of a large exon 8b (201 aa) or a short exon 25c (9 aa) in cytoplasmic linkers, similar to mammalian homologs. Inclusion of optional 25c exons in the III-IV linker of T-type channels speeds up kinetics and causes hyperpolarizing shifts in both activation and steady-state inactivation of macroscopic currents. The abundant variant lacking exon 25c is the workhorse of embryonic Ca(v)3 channels, whose high density and right-shifted activation and availability curves are expected to increase pace-making and allow the channels to contribute more significantly to cellular excitation in prenatal tissue. Presence of brain-enriched, optional exon 8b conserved with mammalian Ca(v)3.1 and encompassing the proximal half of the I-II linker, imparts a ~50% reduction in total and surface-expressed LCa(v)3 channel protein, which accounts for reduced whole-cell calcium currents of +8b variants in HEK cells. Evolutionarily conserved optional exons in cytoplasmic linkers of Ca(v)3 channels regulate expression (exon 8b) and a battery of biophysical properties (exon 25c) for tuning specialized firing patterns in different tissues and throughout development.


Asunto(s)
Canales de Calcio Tipo T/genética , Regulación de la Expresión Génica , Activación del Canal Iónico , Empalme del ARN , Transcripción Genética , Secuencia de Aminoácidos , Animales , Canales de Calcio Tipo T/química , Exones , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA