Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Int J Mol Sci ; 23(7)2022 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-35409240

RESUMEN

The activity of the epithelial Na+ Channel (ENaC) is strongly dependent on the membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2). PIP2 binds two distinct cationic clusters within the N termini of ß- and γ-ENaC subunits (ßN1 and γN2). The affinities of these sites were previously determined using short synthetic peptides, yet their role in sensitizing ENaC to changes in PIP2 levels in the cellular system is not well established. We addressed this question by comparing the effects of PIP2 depletion and recovery on ENaC channel activity and intracellular Na+ levels [Na+]i. We tested effects on ENaC activity with mutations to the PIP2 binding sites using the optogenetic system CIBN/CRY2-OCRL to selectively deplete PIP2. We monitored changes of [Na+]i by measuring the fluorescent Na+ indicator, CoroNa Green AM, and changes in channel activity by performing patch clamp electrophysiology. Whole cell patch clamp measurements showed a complete lack of response to PIP2 depletion and recovery in ENaC with mutations to ßN1 or γN2 or both sites, compared to wild type ENaC. Whereas mutant ßN1 also had no change in CoroNa Green fluorescence in response to PIP2 depletion, γN2 did have reduced [Na+]i, which was explained by having shorter CoroNa Green uptake and half-life. These results suggest that CoroNa Green measurements should be interpreted with caution. Importantly, the electrophysiology results show that the ßN1 and γN2 sites on ENaC are each necessary to permit maximal ENaC activity in the presence of PIP2.


Asunto(s)
Canales Epiteliales de Sodio , Fosfatidilinositol 4,5-Difosfato , Sitios de Unión , Canales Epiteliales de Sodio/metabolismo , Optogenética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositoles/metabolismo , Sodio/metabolismo
2.
J Neurosci ; 39(9): 1566-1587, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30593498

RESUMEN

KCNQ (Kv7, "M-type") K+ channels and TRPC (transient receptor potential, "canonical") cation channels are coupled to neuronal discharge properties and are regulated via Gq/11-protein-mediated signals. Stimulation of Gq/11-coupled receptors both consumes phosphatidylinositol 4,5-bisphosphate (PIP2) via phosphalipase Cß hydrolysis and stimulates PIP2 synthesis via rises in Ca2+i and other signals. Using brain-slice electrophysiology and Ca2+ imaging from male and female mice, we characterized threshold K+ currents in dentate gyrus granule cells (DGGCs) and CA1 pyramidal cells, the effects of Gq/11-coupled muscarinic M1 acetylcholine (M1R) stimulation on M current and on neuronal discharge properties, and elucidated the intracellular signaling mechanisms involved. We observed disparate signaling cascades between DGGCs and CA1 neurons. DGGCs displayed M1R enhancement of M-current, rather than suppression, due to stimulation of PIP2 synthesis, which was paralleled by increased PIP2-gated G-protein coupled inwardly rectifying K+ currents as well. Deficiency of KCNQ2-containing M-channels ablated the M1R-induced enhancement of M-current in DGGCs. Simultaneously, M1R stimulation in DGGCs induced robust increases in [Ca2+]i, mostly due to TRPC currents, consistent with, and contributing to, neuronal depolarization and hyperexcitability. CA1 neurons did not display such multimodal signaling, but rather M current was suppressed by M1R stimulation in these cells, similar to the previously described actions of M1R stimulation on M-current in peripheral ganglia that mostly involves PIP2 depletion. Therefore, these results point to a pleiotropic network of cholinergic signals that direct cell-type-specific, precise control of hippocampal function with strong implications for hyperexcitability and epilepsy.SIGNIFICANCE STATEMENT At the neuronal membrane, protein signaling cascades consisting of ion channels and metabotropic receptors govern the electrical properties and neurotransmission of neuronal networks. Muscarinic acetylcholine receptors are G-protein-coupled metabotropic receptors that control the excitability of neurons through regulating ion channels, intracellular Ca2+ signals, and other second-messenger cascades. We have illuminated previously unknown actions of muscarinic stimulation on the excitability of hippocampal principal neurons that include M channels, TRPC (transient receptor potential, "canonical") cation channels, and powerful regulation of lipid metabolism. Our results show that these signaling pathways, and mechanisms of excitability, are starkly distinct between peripheral ganglia and brain, and even between different principal neurons in the hippocampus.


Asunto(s)
Potenciales de Acción , Región CA1 Hipocampal/metabolismo , Giro Dentado/metabolismo , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/metabolismo , Receptores Muscarínicos/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Giro Dentado/citología , Giro Dentado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Células Piramidales/metabolismo , Células Piramidales/fisiología
3.
J Biol Chem ; 294(15): 6094-6112, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30808708

RESUMEN

Calmodulin (CaM) conveys intracellular Ca2+ signals to KCNQ (Kv7, "M-type") K+ channels and many other ion channels. Whether this "calmodulation" involves a dramatic structural rearrangement or only slight perturbations of the CaM/KCNQ complex is as yet unclear. A consensus structural model of conformational shifts occurring between low nanomolar and physiologically high intracellular [Ca2+] is still under debate. Here, we used various techniques of biophysical chemical analyses to investigate the interactions between CaM and synthetic peptides corresponding to the A and B domains of the KCNQ4 subtype. We found that in the absence of CaM, the peptides are disordered, whereas Ca2+/CaM imposed helical structure on both KCNQ A and B domains. Isothermal titration calorimetry revealed that Ca2+/CaM has higher affinity for the B domain than for the A domain of KCNQ2-4 and much higher affinity for the B domain when prebound with the A domain. X-ray crystallography confirmed that these discrete peptides spontaneously form a complex with Ca2+/CaM, similar to previous reports of CaM binding KCNQ-AB domains that are linked together. Microscale thermophoresis and heteronuclear single-quantum coherence NMR spectroscopy indicated the C-lobe of Ca2+-free CaM to interact with the KCNQ4 B domain (Kd ∼10-20 µm), with increasing Ca2+ molar ratios shifting the CaM-B domain interactions via only the CaM C-lobe to also include the N-lobe. Our findings suggest that in response to increased Ca2+, CaM undergoes lobe switching that imposes a dramatic mutually induced conformational fit to both the proximal C terminus of KCNQ4 channels and CaM, likely underlying Ca2+-dependent regulation of KCNQ gating.


Asunto(s)
Calcio/química , Calmodulina/química , Canales de Potasio KCNQ/química , Animales , Células CHO , Calcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Cricetulus , Cristalografía por Rayos X , Humanos , Activación del Canal Iónico , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Dominios Proteicos , Estructura Secundaria de Proteína
4.
Hippocampus ; 30(5): 435-455, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31621989

RESUMEN

M-type (KCNQ2/3) K+ channels play dominant roles in regulation of active and passive neuronal discharge properties such as resting membrane potential, spike-frequency adaptation, and hyper-excitatory states. However, plasticity of M-channel expression and function in nongenetic forms of epileptogenesis are still not well understood. Using transgenic mice with an EGFP reporter to detect expression maps of KCNQ2 mRNA, we assayed hyperexcitability-induced alterations in KCNQ2 transcription across subregions of the hippocampus. Pilocarpine and pentylenetetrazol chemoconvulsant models of seizure induction were used, and brain tissue examined 48 hr later. We observed increases in KCNQ2 mRNA in CA1 and CA3 pyramidal neurons after chemoconvulsant-induced hyperexcitability at 48 hr, but no significant change was observed in dentate gyrus (DG) granule cells. Using chromogenic in situ hybridization assays, changes to KCNQ3 transcription were not detected after hyper-excitation challenge, but the results for KCNQ2 paralleled those using the KCNQ2-mRNA reporter mice. In mice 7 days after pilocarpine challenge, levels of KCNQ2 mRNA were similar in all regions to those from control mice. In brain-slice electrophysiology recordings, CA1 pyramidal neurons demonstrated increased M-current amplitudes 48 hr after hyperexcitability; however, there were no significant changes to DG granule cell M-current amplitude. Traumatic brain injury induced significantly greater KCNQ2 expression in the hippocampal hemisphere that was ipsilateral to the trauma. In vivo, after a secondary challenge with subconvulsant dose of pentylenetetrazole, control mice were susceptible to tonic-clonic seizures, whereas mice administered the M-channel opener retigabine were protected from such seizures. This study demonstrates that increased excitatory activity promotes KCNQ2 upregulation in the hippocampus in a cell-type specific manner. Such novel ion channel expressional plasticity may serve as a compensatory mechanism after a hyperexcitable event, at least in the short term. The upregulation described could be potentially leveraged in anticonvulsant enhancement of KCNQ2 channels as therapeutic target for preventing onset of epileptogenic seizures.


Asunto(s)
Hipocampo/metabolismo , Canal de Potasio KCNQ2/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Femenino , Hipocampo/efectos de los fármacos , Canal de Potasio KCNQ2/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Pilocarpina/farmacología
5.
J Biol Chem ; 293(50): 19411-19428, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30348901

RESUMEN

Phosphatidylinositol 4,5-bisphosphate (PIP2) in the plasma membrane regulates the function of many ion channels, including M-type (potassium voltage-gated channel subfamily Q member (KCNQ), Kv7) K+ channels; however, the molecular mechanisms involved remain unclear. To this end, we here focused on the KCNQ3 subtype that has the highest apparent affinity for PIP2 and performed extensive mutagenesis in regions suggested to be involved in PIP2 interactions among the KCNQ family. Using perforated patch-clamp recordings of heterologously transfected tissue culture cells, total internal reflection fluorescence microscopy, and the zebrafish (Danio rerio) voltage-sensitive phosphatase to deplete PIP2 as a probe, we found that PIP2 regulates KCNQ3 channels through four different domains: 1) the A-B helix linker that we previously identified as important for both KCNQ2 and KCNQ3, 2) the junction between S6 and the A helix, 3) the S2-S3 linker, and 4) the S4-S5 linker. We also found that the apparent strength of PIP2 interactions within any of these domains was not coupled to the voltage dependence of channel activation. Extensive homology modeling and docking simulations with the WT or mutant KCNQ3 channels and PIP2 were consistent with the experimental data. Our results indicate that PIP2 modulates KCNQ3 channel function by interacting synergistically with a minimum of four cytoplasmic domains.


Asunto(s)
Citoplasma/metabolismo , Canal de Potasio KCNQ3/química , Canal de Potasio KCNQ3/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Cricetulus , Humanos , Canal de Potasio KCNQ3/genética , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos
7.
J Neurosci ; 35(5): 2101-11, 2015 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25653366

RESUMEN

Cerebral ischemic stroke is a worldwide cause of mortality/morbidity and thus an important focus of research to decrease the severity of brain injury. Therapeutic options for acute stroke are still limited. In neurons throughout the brain, "M-type" K(+) currents, underlain by KCNQ subunits 2-5, play dominant roles in control over excitability, and are thus implicated in myriad neurological and psychiatric disorders. Although KCNQ channel openers, such as retigabine, have emerged as anti-epilepsy drugs, their effects on ischemic injury remain unknown. Here, we investigated the protective effects of M-channel openers on stroke-induced brain injury in mouse photothrombotic and middle cerebral artery occlusion (MCAo) models. Both photothrombosis and MCAo led to rapid, predictable, and consistently sized necrotic brain lesions, inflammatory responses, and behavioral deficits. Administration of three distinct M-channel openers at 0-6 h after ischemic injury significantly decreased brain infarct size and inflammation, and prevented neurological dysfunction, although they were more effective when administered 0-3 h poststroke. Thus, we show beneficial effects against stroke-induced brain injury and neuronal death through pharmacological regulation of ion channels that control neuronal excitability.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Canales de Potasio KCNQ/antagonistas & inhibidores , Animales , Antracenos/administración & dosificación , Antracenos/farmacología , Antracenos/uso terapéutico , Carbamatos/administración & dosificación , Carbamatos/farmacología , Carbamatos/uso terapéutico , Quimioterapia Combinada , Canales de Potasio KCNQ/agonistas , Masculino , Ratones , Ratones Endogámicos C57BL , Fenilendiaminas/administración & dosificación , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Bloqueadores de los Canales de Potasio/administración & dosificación , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico
8.
J Physiol ; 594(1): 31-7, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25653013

RESUMEN

A-kinase anchoring proteins (AKAPs) have emerged as a converging point of diverse signals to achieve spatiotemporal resolution of directed cellular regulation. With the extensive studies of AKAP79/150 in regulation of ion channel activity, the major questions to be posed centre on the mechanism and functional role of synergistic regulation of ion channels by such signalling proteins. In this review, we summarize recent discoveries of AKAP79/150-mediated modulation of voltage-gated neuronal M-type (KCNQ, Kv7) K(+) channels and L-type CaV 1 Ca(2+) channels, on both short- and longer-term time scales, highlighting the dynamics of the macromolecular signalling complexes in brain and peripheral nerve We also discuss several models for the possible mechanisms of these multi-protein assemblies and how they serve the agenda of the neurons in which they occur.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Encéfalo/metabolismo , Nervios Periféricos/metabolismo , Transducción de Señal , Animales , Canales de Calcio Tipo L/metabolismo , Humanos , Canales de Potasio KCNQ/metabolismo , Nervios Periféricos/fisiología
9.
Neurotherapeutics ; 20(3): 853-869, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36976493

RESUMEN

We investigated whether pharmacological increase of "M-type" (KCNQ, Kv7) K + channel currents by the M-channel opener, retigabine (RTG), acutely after repetitive traumatic brain injuries (rTBIs), prevents or reduces their long-term detrimental effects. rTBIs were studied using a blast shock air wave mouse model. Animals were monitored by video and electroencephalogram (EEG) records for nine months after the last injury to assess the occurrence of post-traumatic seizures (PTS), post-traumatic epilepsy (PTE), sleep-wake cycle architecture alterations, and the power of the EEG signals. We evaluated the development of long-term changes in the brain associated with various neurodegenerative diseases in mice by examining transactive response DNA-binding protein 43 (TDP-43) expression and nerve fiber damage ~ 2 years after the rTBIs. We observed acute RTG treatment to reduce the duration of PTS and impair the development of PTE. Acute RTG treatment also prevented post-injury hypersomnia, nerve fiber damage, and cortical TDP-43 accumulation and translocation from the nucleus to the cytoplasm. Mice that developed PTE displayed impaired rapid eye movement (REM) sleep, and there were significant correlations between seizure duration and time spent in the different stages of the sleep-wake cycle. We observed acute RTG treatment to impair injury-induced reduction of age-related increase in gamma frequency power of the EGG, which has been suggested to be necessary for a healthy aged brain. The data show that RTG, administered acutely post-TBI, is a promising, novel therapeutic option to blunt/prevent several long-term effects of rTBIs. Furthermore, our results show a direct relationship between sleep architecture and PTE.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Epilepsia Postraumática , Ratones , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Carbamatos/farmacología , Carbamatos/uso terapéutico
10.
Biophys J ; 102(11): 2499-509, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22713565

RESUMEN

Two mechanisms have been postulated to underlie KCNQ3 homomeric current amplitudes, which are small compared with those of KCNQ4 homomers and KCNQ2/Q3 heteromers. The first involves differential channel expression governed by the D-helix within the C-terminus. The second suggests similar channel surface expression but an intrinsically unstable KCNQ3 pore. Here, we find H2O2-enhanced oligomerization of KCNQ4 subunits, as reported by nondenaturing polyacrylamide gel electrophoresis, at C643 at the end of the D-helix, where KCNQ3 possesses a histidine. However, H2O2-mediated enhancement of KCNQ4 currents was identical in the C643A mutant, and KCNQ3 H646C produced homomeric or heteromeric (with KCNQ2) currents similar to those of wild-type KCNQ3, ruling out this divergent residue as underlying the small KCNQ3 amplitudes. In KcsA, F103 in S6 is critical for pore-mediated destabilization of the conductive pathway. We found that mutations at the analogous F344 in KCNQ3 dramatically decreased the KCNQ3 currents. Total internal reflection fluorescence imaging revealed only minor differential surface expression among the wild-type and mutant channels. Homology modeling suggests that the effects of the F344 mutants arise from the disruption of the interaction between F344 and A315 in the pore helix. These data support a secondary role of the C-terminus, compared with pore helix-S6 interactions, in governing KCNQ3 current amplitudes.


Asunto(s)
Activación del Canal Iónico , Canal de Potasio KCNQ3/química , Canal de Potasio KCNQ3/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cisteína/metabolismo , Disulfuros/metabolismo , Conductividad Eléctrica , Peróxido de Hidrógeno/farmacología , Activación del Canal Iónico/efectos de los fármacos , Canal de Potasio KCNQ3/genética , Microscopía Fluorescente , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad
11.
Biophys J ; 102(11): 2489-98, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22713564

RESUMEN

KCNQ3 homomeric channels yield very small macroscopic currents compared with other KCNQ channels or KCNQ2/3 heteromers. Two disparate regions of the channels--the C-terminus and the pore region--have been implicated in governing KCNQ current amplitudes. We previously showed that the C-terminus plays a secondary role compared with the pore region. Here, we confirm the critical role of the pore region in determining KCNQ3 currents. We find that mutations at the 312 position in the pore helix of KCNQ3 (I312E, I312K, and I312R) dramatically decreased KCNQ3 homomeric currents as well as heteromeric KCNQ2/3 currents. Evidence that these mutants were expressed in the heteromers includes shifted TEA sensitivity compared with KCNQ2 homomers. To test for differential membrane protein expression, we performed total internal reflection fluorescence imaging, which revealed only small differences that do not underlie the differences in macroscopic currents. To determine whether this mechanism generalizes to other KCNQ channels, we tested the effects of analogous mutations at the conserved I273 position in KCNQ2, with similar results. Finally, we performed homology modeling of the pore region of wild-type and mutant KCNQ3 channels to investigate the putative structural mechanism mediating these results. The modeling suggests that the lack of current in I312E, I312K, and I312R KCNQ3 channels is due to pore helix-selectivity filter interactions that lock the selectivity filter in a nonconductive conformation.


Asunto(s)
Activación del Canal Iónico , Canal de Potasio KCNQ3/química , Canal de Potasio KCNQ3/metabolismo , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Conductividad Eléctrica , Interacciones Hidrofóbicas e Hidrofílicas , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Multimerización de Proteína , Estabilidad Proteica , Estructura Terciaria de Proteína , Homología Estructural de Proteína , Relación Estructura-Actividad
12.
J Neurosci ; 31(19): 7199-211, 2011 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-21562284

RESUMEN

Voltage-gated M-type (KCNQ) K+ channels play critical roles in regulation of neuronal excitability. Previous work showed A-kinase-anchoring protein (AKAP)79/150-mediated protein kinase C (PKC) phosphorylation of M channels to be involved in M current (I(M)) suppression by muscarinic M1, but not bradykinin B2, receptors. In this study, we first explored whether purinergic and angiotensin suppression of I(M) in superior cervical ganglion (SCG) sympathetic neurons involves AKAP79/150. Transfection into rat SCG neurons of ΔA-AKAP79, which lacks the A domain necessary for PKC binding, or the absence of AKAP150 in AKAP150(-/-) mice, did not affect I(M) suppression by purinergic agonist or by bradykinin, but reduced I(M) suppression by muscarinic agonist and angiotensin II. Transfection of AKAP79, but not ΔA-AKAP79 or AKAP15, rescued suppression of I(M) by muscarinic receptors in AKAP150(-/-) neurons. We also tested association of AKAP79 with M(1), B(2), P2Y(6), and AT(1) receptors, and KCNQ2 and KCNQ3 channels, via Förster resonance energy transfer (FRET) on Chinese hamster ovary cells under total internal refection fluorescence microscopy, which revealed substantial FRET between AKAP79 and M1 or AT1 receptors, and with the channels, but only weak FRET with P2Y(6) or B2 receptors. The involvement of AKAP79/150 in G(q/11)-coupled muscarinic regulation of N- and L-type Ca2+) channels and by cAMP/protein kinase A was also studied. We found AKAP79/150 to not play a role in the former, but to be necessary for forskolin-induced upregulation of L-current. Thus, AKAP79/150 action correlates with the PIP(2) (phosphatidylinositol 4,5-bisphosphate)-depletion mode of I(M) suppression, but does not generalize to G(q/11)-mediated inhibition of N- or L-type Ca2+ channels.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Unión al GTP/metabolismo , Activación del Canal Iónico/fisiología , Neuronas/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas/citología , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/metabolismo
13.
J Biol Chem ; 286(1): 830-41, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21051544

RESUMEN

Phosphatidylinositol 4,5-bisphosphate (PIP(2)) regulates Ca(2+) (I(Ca)) and M-type K(+) currents in superior cervical ganglion sympathetic neurons. In those cells, M(1) muscarinic and AT(1) angiotensin types do not elicit Ca(2+)(i) signals and suppress both currents via depletion of PIP(2), whereas the B(2) bradykinin and P2Y purinergic types elicit robust IP(3)-mediated [Ca(2+)](i) rises and neither deplete PIP(2) nor inhibit I(Ca). We have suggested that this specificity arises from differential Ca(2+)(i) signals underlying receptor-specific stimulation of PIP(2) synthesis by phosphatidylinositol (PI) 4-kinase. Here, we investigate which PI 4-kinase isoform underlies this signal, whether stimulation of PI 4-phosphate 5-kinase is also required, and the origin of receptor-specific Ca(2+)(i) signals. Recordings of I(Ca) were used as a PIP(2) "biosensor." In control, stimulation of M(1), but not B(2) or P2Y, receptors robustly suppressed I(Ca). However, when PI 4-kinase IIIß, diacylglycerol kinase, Rho, or Rho-kinase was blocked, agonists of all three receptors robustly suppressed I(Ca). Overexpression of exogenous M(1) receptors yielded large [Ca(2+)](i) rises by muscarinic agonist, and transfection of wild-type IRBIT decreased Ca(2+)(i) signals, whereas dominant negative IRBIT-S68A had little effect on B(2) or P2Y responses but greatly increased muscarinic responses. We conclude that overlaid on microdomain organization is IRBIT, setting a "threshold" for [IP(3)], assisting in fidelity of receptor specificity.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Neuronas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Bovinos , Diacilglicerol Quinasa/metabolismo , Femenino , Masculino , Antígenos de Histocompatibilidad Menor , Agonistas Muscarínicos/farmacología , Neuronas/efectos de los fármacos , Ácidos Fosfatidicos/metabolismo , Fosfatidilinositol 4,5-Difosfato/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ratas , Especificidad por Sustrato , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
14.
Nat Rev Neurosci ; 8(12): 921-34, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17971783

RESUMEN

Over the past decade, there has been an explosion in the number of membrane transport proteins that have been shown to be sensitive to the abundance of phosphoinositides in the plasma membrane. These proteins include voltage-gated potassium and calcium channels, ion channels that mediate sensory and nociceptive responses, epithelial transport proteins and ionic exchangers. Each of the regulatory lipids is also under multifaceted regulatory control. Phosphoinositide modulation of membrane proteins in neurons often has a dramatic effect on neuronal excitability and synaptic transmitter release. The repertoire of lipid signalling mechanisms that regulate membrane proteins is intriguingly complex and provides a rich array of topics for neuroscience research.


Asunto(s)
Membrana Celular/química , Membrana Celular/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Fosfatidilinositoles/química , Fosfatidilinositoles/fisiología , Secuencia de Aminoácidos , Animales , Humanos , Transporte Iónico/fisiología , Datos de Secuencia Molecular
15.
J Neurosci ; 30(6): 2311-23, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-20147557

RESUMEN

M-type channels are localized to neuronal, cardiovascular, and epithelial tissues, where they play critical roles in control of excitability and K(+) transport, and are regulated by numerous receptors via G(q/11)-mediated signals. One pathway shown for KCNQ2 and muscarinic receptors uses PKC, recruited to the channels by A-kinase anchoring protein (AKAP)79/150. As M-type channels can be variously composed of KCNQ1-5 subunits, and M current is known to be regulated by Ca(2+)/calmodulin (CaM) and PIP(2), we probed the generality of AKAP79/150 actions among KCNQ1-5 channels, and the influence of Ca(2+)/CaM and PIP(2) on AKAP79/150 actions. We first examined which KCNQ subunits are targeted by AKAP79 in Chinese hamster ovary (CHO) cells heterologously expressing KCNQ1-5 subunits and AKAP79, using fluorescence resonance energy transfer (FRET) under total internal reflection fluorescence (TIRF) microscopy, and patch-clamp analysis. Donor-dequenching FRET between CFP-tagged KCNQ1-5 and YFP-tagged AKAP79 revealed association of KCNQ2-5, but not KCNQ1, with AKAP79. In parallel with these results, CHO cells stably expressing M(1) receptors studied under perforated patch-clamp showed cotransfection of AKAP79 to "sensitize" KCNQ2/3 heteromers and KCNQ2-5, but not KCNQ1, homomers to muscarinic inhibition, manifested by shifts in the dose-response relations to lower concentrations. The effect on KCNQ4 was abolished by the T553A mutation of the putative PKC phosphorylation site. We then probed the role of CaM and PIP(2) in these AKAP79 actions. TIRF/FRET experiments revealed cotransfection of wild-type, but not dominant-negative (DN), CaM that cannot bind Ca(2+), to disrupt the interaction of YFP-tagged AKAP79(1-153) with CFP-tagged KCNQ2-5. Tonic depletion of PIP(2) by cotransfection of a PIP(2) phosphatase had no effect, and sudden depletion of PIP(2) did not delocalize GFP-tagged AKAP79 from the membrane. Finally, patch-clamp experiments showed cotransfection of wild-type, but not DN, CaM to prevent the AKAP79-mediated sensitization of KCNQ2/3 heteromers to muscarinic inhibition. Thus, AKAP79 acts on KCNQ2-5, but not KCNQ1-containing channels, with effects disrupted by calcified CaM, but not by PIP(2) depletion.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Calcio/fisiología , Calmodulina/fisiología , Canales de Potasio KCNQ/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Animales , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Transferencia Resonante de Energía de Fluorescencia , Humanos , Canales de Potasio KCNQ/genética , Microscopía Fluorescente , Técnicas de Placa-Clamp , Fosfatidilinositol 4,5-Difosfato/fisiología , Multimerización de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transfección
16.
J Physiol ; 589(Pt 10): 2559-68, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21486761

RESUMEN

M-type (KCNQ) K⁺ channels are known to regulate excitability and firing properties of sympathetic neurons (SNs), but their role in regulating neurotransmitter release is unclear, requiring further study. We sought to use a physiological preparation in which SNs innervate primary cardiomyocytes to evaluate the direct role of M-channels in the release of noradrenaline (NA) from SNs. Co-cultures of rat SNs and mouse cardiomyocytes were prepared, and the contraction rate (CR) of the cardiomyocyte syncytium monitored by video microscopy. We excited the SNs with nicotine, acting on nicotinic acetylcholine receptors, and monitored the increase in CR in the presence or absence of the specific M-channel opener retigabine, or agonists of bradykinin B2 or purinergic P2Y receptors on the SNs. The maximal adrenergic effect on the CR was determined by application of isoproterenol (isoprenaline). To isolate the actions of B2 or P2Y receptor stimulation to the neurons, we prepared cardiomyocytes from B2 receptor or P2Y2 receptor knock-out mice, respectively. We found that co-application of retigabine strongly decreased the nicotine-induced increase in CR. Conversely, co-application of bradykinin or the P2Y-receptor agonist UTP augmented the nicotine-induced increase in CR to about half of the level produced by isoproterenol. All effects on the CR were wholly blocked by propranolol. Our data support the role of M-type K⁺ channels in the control of NA release by SNs at functional adrenergic synapses on cardiomyocytes.We conclude that physiological receptor agonists control the heart rate via the regulation of M-current in SNs.


Asunto(s)
Frecuencia Cardíaca/fisiología , Corazón/fisiología , Canales de Potasio KCNQ/fisiología , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Sistema Nervioso Simpático/fisiología , Animales , Carbamatos/farmacología , Cardiotónicos/farmacología , Células Cultivadas , Técnicas de Cocultivo , Estimulantes Ganglionares/farmacología , Corazón/efectos de los fármacos , Corazón/inervación , Frecuencia Cardíaca/efectos de los fármacos , Isoproterenol/farmacología , Canales de Potasio KCNQ/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Nicotina/farmacología , Fenilendiaminas/farmacología , Agonistas del Receptor Purinérgico P2Y/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Bradiquinina B2/agonistas , Sistema Nervioso Simpático/efectos de los fármacos
17.
Neuroimage ; 58(3): 761-9, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21787867

RESUMEN

Neuronal M-channels are low threshold, slowly activating and non-inactivating, voltage dependent K(+) channels that play a crucial role in controlling neuronal excitability. The native M-channel is composed of heteromeric or homomeric assemblies of subunits belonging to the Kv7/KCNQ family, with KCNQ2/3 heteromers being the most abundant form. KCNQ2 and KCNQ3 subunits have been found to be expressed in various neurons in the central and peripheral nervous system of rodents and humans. Previous evidence shows preferential localization of both subunits to axon initial segments, somata and nodes of Ranvier. In this work, we show the distribution and co-localization of KCNQ2 and KCNQ3 subunits throughout the hippocampal formation, via immunostaining experiments on unfixed rat brain slices and confocal microscopy. We find intense localization and colocalization to the axonal initial segment in several regions of the hippocampus, as well as staining for non-neuronal cells in the area of the lateral ventricle. We did not observe colocalization of KCNQ2 or KCNQ3 with the presynaptic protein, synaptophysin.


Asunto(s)
Química Encefálica , Hipocampo/química , Canal de Potasio KCNQ2/análisis , Canal de Potasio KCNQ3/análisis , Animales , Hipocampo/metabolismo , Inmunohistoquímica , Canal de Potasio KCNQ2/biosíntesis , Canal de Potasio KCNQ3/biosíntesis , Microscopía Confocal , Subunidades de Proteína/análisis , Subunidades de Proteína/biosíntesis , Ratas , Ratas Sprague-Dawley
18.
Mol Pain ; 7: 34, 2011 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-21569553

RESUMEN

BACKGROUND: The transient receptor potential vanilloid type1 (TRPV1) is expressed in nociceptive sensory neurons and is sensitive to phosphorylation. A-Kinase Anchoring Protein 79/150 (AKAP150) mediates phosphorylation of TRPV1 by Protein Kinases A and C, modulating channel activity. However, few studies have focused on the regulatory mechanisms that control AKAP150 association with TRPV1. In the present study, we identify a role for calcium/calmodulin in controlling AKAP150 association with, and sensitization of, TRPV1. RESULTS: In trigeminal neurons, intracellular accumulation of calcium reduced AKAP150 association with TRPV1 in a manner sensitive to calmodulin antagonism. This was also observed in transfected Chinese hamster ovary (CHO) cells, providing a model for conducting molecular analysis of the association. In CHO cells, the deletion of the C-terminal calmodulin-binding site of TRPV1 resulted in greater association with AKAP150, and increased channel activity. Furthermore, the co-expression of wild-type calmodulin in CHOs significantly reduced TRPV1 association with AKAP150, as evidenced by total internal reflective fluorescence-fluorescence resonance energy transfer (TIRF-FRET) analysis and electrophysiology. Finally, dominant-negative calmodulin co-expression increased TRPV1 association with AKAP150 and increased basal and PKA-sensitized channel activity. CONCLUSIONS: the results from these studies indicate that calcium/calmodulin interferes with the association of AKAP150 with TRPV1, potentially extending resensitization of the channel.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Sitios de Unión , Células CHO , Calmodulina/antagonistas & inhibidores , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Transferencia Resonante de Energía de Fluorescencia , Espacio Intracelular/metabolismo , Masculino , Unión Proteica , Ratas , Ratas Sprague-Dawley , Eliminación de Secuencia
19.
Front Neurosci ; 15: 681144, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489621

RESUMEN

We investigated the contribution of excitatory transient receptor potential canonical (TRPC) cation channels to posttraumatic hyperexcitability in the brain 7 days following controlled cortical impact model of traumatic brain injury (TBI) to the parietal cortex in male adult mice. We investigated if TRPC1/TRPC4/TRPC5 channel expression is upregulated in excitatory neurons after TBI in contribution to epileptogenic hyperexcitability in key hippocampal and cortical circuits that have substantial cholinergic innervation. This was tested by measuring TRPC1/TRPC4/TRPC5 protein and messenger RNA (mRNA) expression, assays of cholinergic function, neuronal Ca2+ imaging in brain slices, and seizure susceptibility after TBI. We found region-specific increases in expression of TRPC1, TRPC4, and TRPC5 subunits in the hippocampus and cortex following TBI. The dentate gyrus, CA3 region, and cortex all exhibited robust upregulation of TRPC4 mRNA and protein. TBI increased cFos activity in dentate gyrus granule cells (DGGCs) and layer 5 pyramidal neurons both at the time of TBI and 7 days post-TBI. DGGCs displayed greater magnitude and duration of acetylcholine-induced rises in intracellular Ca2+ in brain slices from mice subjected to TBI. The TBI mice also exhibited greater seizure susceptibility in response to pentylenetetrazol-induced kindling. Blockade of TRPC4/TRPC5 channels with M084 reduced neuronal hyperexcitation and impeded epileptogenic progression of kindling. We observed that the time-dependent upregulation of TRPC4/TRPC5-containing channels alters cholinergic responses and activity of principal neurons acting to increase proexcitatory sensitivity. The underlying mechanism includes acutely decreased acetylcholinesterase function, resulting in greater G q / 11-coupled muscarinic receptor activation of TRPC channels. Overall, our evidence suggests that TBI-induced plasticity of TRPC channels strongly contributes to overt hyperexcitability and primes the hippocampus and cortex for seizures.

20.
Neurotherapeutics ; 18(4): 2707-2721, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34608616

RESUMEN

Traumatic brain injury (TBI) remains one of the greatest public health concerns with increasing morbidity and mortality rates worldwide. Our group reported that stimulation of astrocyte mitochondrial metabolism by P2Y1 receptor agonists significantly reduced cerebral edema and reactive gliosis in a TBI model. Subsequent data on the pharmacokinetics (PK) and rapid metabolism of these compounds suggested that neuroprotection was likely mediated by a metabolite, AST-004, which binding data indicated was an adenosine A3 receptor (A3R) agonist. The neuroprotective efficacy of AST-004 was tested in a control closed cortical injury (CCCI) model of TBI in mice. Twenty-four (24) hours post-injury, mice subjected to CCCI and treated with AST-004 (0.22 mg/kg, injected 30 min post-trauma) exhibited significantly less secondary brain injury. These effects were quantified with less cell death (PSVue794 fluorescence) and loss of blood brain barrier breakdown (Evans blue extravasation assay), compared to vehicle-treated TBI mice. TBI-treated mice also exhibited significantly reduced neuroinflammatory markers, glial-fibrillary acidic protein (GFAP, astrogliosis) and ionized Ca2+-binding adaptor molecule 1 (Iba1, microgliosis), both at the mRNA (qRT-PCR) and protein (Western blot and immunofluorescence) levels, respectively. Four (4) weeks post-injury, both male and female TBI mice presented a significant reduction in freezing behavior during contextual fear conditioning (after foot shock). AST-004 treatment prevented this TBI-induced impairment in male mice, but did not significantly affect impairment in female mice. Impairment of spatial memory, assessed 24 and 48 h after the initial fear conditioning, was also reduced in AST-004-treated TBI-male mice. Female TBI mice did not exhibit memory impairment 24 and 48 h after contextual fear conditioning and similarly, AST-004-treated female TBI mice were comparable to sham mice. Finally, AST-004 treatments were found to increase in vivo ATP production in astrocytes (GFAP-targeted luciferase activity), consistent with the proposed mechanism of action. These data reveal AST-004 as a novel A3R agonist that increases astrocyte energy production and enhances their neuroprotective efficacy after brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Fármacos Neuroprotectores , Adenosina/metabolismo , Adenosina/farmacología , Animales , Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Gliosis/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroprotección , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA