Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Hum Mol Genet ; 30(23): 2300-2314, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34245260

RESUMEN

Here, we report on six unrelated individuals, all presenting with early-onset global developmental delay, associated with impaired motor, speech and cognitive development, partly with developmental epileptic encephalopathy and physical dysmorphisms. All individuals carry heterozygous missense variants of KCND2, which encodes the voltage-gated potassium (Kv) channel α-subunit Kv4.2. The amino acid substitutions associated with the variants, p.(Glu323Lys) (E323K), p.(Pro403Ala) (P403A), p.(Val404Leu) (V404L) and p.(Val404Met) (V404M), affect sites known to be critical for channel gating. To unravel their likely pathogenicity, recombinant mutant channels were studied in the absence and presence of auxiliary ß-subunits under two-electrode voltage clamp in Xenopus oocytes. All channel mutants exhibited slowed and incomplete macroscopic inactivation, and the P403A variant in addition slowed activation. Co-expression of KChIP2 or DPP6 augmented the functional expression of both wild-type and mutant channels; however, the auxiliary ß-subunit-mediated gating modifications differed from wild type and among mutants. To simulate the putative setting in the affected individuals, heteromeric Kv4.2 channels (wild type + mutant) were studied as ternary complexes (containing both KChIP2 and DPP6). In the heteromeric ternary configuration, the E323K variant exhibited only marginal functional alterations compared to homomeric wild-type ternary, compatible with mild loss-of-function. By contrast, the P403A, V404L and V404M variants displayed strong gating impairment in the heteromeric ternary configuration, compatible with loss-of-function or gain-of-function. Our results support the etiological involvement of Kv4.2 channel gating impairment in early-onset monogenic global developmental delay. In addition, they suggest that gain-of-function mechanisms associated with a substitution of V404 increase epileptic seizure susceptibility.


Asunto(s)
Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/metabolismo , Variación Genética , Activación del Canal Iónico , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo , Alelos , Sustitución de Aminoácidos , Biomarcadores , Discapacidades del Desarrollo/diagnóstico , Susceptibilidad a Enfermedades , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Fenotipo , Subunidades de Proteína , Canales de Potasio Shal/química
2.
Adv Physiol Educ ; 47(1): 97-116, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36476117

RESUMEN

Vascular diseases of the legs are highly prevalent and constitute an important part of medical curricula. The understanding of these diseases relies on strongly interwoven aspects of vascular physiology and vascular medicine. We aimed to connect these within a horizontally integrated laboratory class on vascular physiology of the leg that was designed in cooperation between the departments of physiology and vascular surgery. Conceptually, we applied examination techniques of vascular medicine to visualize physiological parameters that are altered by the most frequent diseases. This facilitates integrative discussions on malfunctions, trains diagnostic skills, and bridges to vascular medicine. In four experiments, we use oscillometry and impedance venous occlusion plethysmography to address key aspects of the arterial and venous system of the legs: 1) arterial pulse wave, 2) arterial systolic blood pressure, 3) venous capacitance and venous outflow, and 4) reactive hyperemia. After the experiments, physiological vascular function, the associated diseases, their impact on the recorded parameters, and diagnostic options are discussed. To allow reproduction, we describe the course structure and the experimental setup in detail. We present the experimental data of a cohort of medical students and document learning success and student satisfaction. All experiments were feasible and provided robust data on physiologically and clinically relevant vascular functions. The activity was perceived positively by the students and led to a substantial improvement of knowledge. With this work, we offer a template for reproduction or variation of a proven concept of horizontally integrated teaching of vascular physiology of the leg.NEW & NOTEWORTHY This article presents an integrative laboratory class on vascular physiology bridging to vascular medicine. The four experiments rely on oscillometry and venous occlusion plethysmography. We describe in detail this new class regarding structure, experimental setup, and experimental procedure, and we give insight into the applied materials. Moreover, we present the experimental data of 74 students and a quantitative evaluation of the students' learning success and acceptance.


Asunto(s)
Cardiología , Fisiología , Humanos , Pletismografía/métodos , Venas/fisiología , Presión Sanguínea
3.
J Biol Chem ; 295(34): 12099-12110, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32641494

RESUMEN

Voltage-gated potassium (Kv) channels of the Kv4 subfamily associate with Kv channel-interacting proteins (KChIPs), which leads to enhanced surface expression and shapes the inactivation gating of these channels. KChIP3 has been reported to also interact with the late endosomal/lysosomal membrane glycoprotein CLN3 (ceroid lipofuscinosis neuronal 3), which is modified because of gene mutation in juvenile neuronal ceroid lipofuscinosis (JNCL). The present study was undertaken to find out whether and how CLN3, by its interaction with KChIP3, may indirectly modulate Kv4.2 channel expression and function. To this end, we expressed KChIP3 and CLN3, either individually or simultaneously, together with Kv4.2 in HEK 293 cells. We performed co-immunoprecipitation experiments and found a lower amount of KChIP3 bound to Kv4.2 in the presence of CLN3. In whole-cell patch-clamp experiments, we examined the effects of CLN3 co-expression on the KChIP3-mediated modulation of Kv4.2 channels. Simultaneous co-expression of CLN3 and KChIP3 with Kv4.2 resulted in a suppression of the typical KChIP3-mediated modulation; i.e. we observed less increase in current density, less slowing of macroscopic current decay, less acceleration of recovery from inactivation, and a less positively shifted voltage dependence of steady-state inactivation. The suppression of the KChIP3-mediated modulation of Kv4.2 channels was weaker for the JNCL-related missense mutant CLN3R334C and for a JNCL-related C-terminal deletion mutant (CLN3ΔC). Our data support the notion that CLN3 is involved in Kv4.2/KChIP3 somatodendritic A-type channel formation, trafficking, and function, a feature that may be lost in JNCL.


Asunto(s)
Regulación de la Expresión Génica/genética , Proteínas de Interacción con los Canales Kv , Glicoproteínas de Membrana , Chaperonas Moleculares , Mutación Missense , Lipofuscinosis Ceroideas Neuronales , Proteínas Represoras , Canales de Potasio Shal , Sustitución de Aminoácidos , Células HEK293 , Humanos , Proteínas de Interacción con los Canales Kv/genética , Proteínas de Interacción con los Canales Kv/metabolismo , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Lipofuscinosis Ceroideas Neuronales/patología , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Canales de Potasio Shal/biosíntesis , Canales de Potasio Shal/genética
4.
Pflugers Arch ; 469(11): 1457-1470, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28735419

RESUMEN

The transient outward current (I to) in the human heart is mediated by Kv4.3 channels complexed with Kv channel interacting protein (KChIP) 2, a cytoplasmic Ca2+-binding EF-hand protein known to modulate Kv4.3 inactivation gating upon heterologous co-expression. We studied Kv4.3 channels co-expressed with wild-type (wt) or EF-hand-mutated (ΔEF) KChIP2 in human embryonic kidney (HEK) 293 cells. Co-expression took place in the absence or presence of BAPTA-AM, and macroscopic currents were recorded in the whole-cell patch-clamp configuration with different free Ca2+ concentrations in the patch-pipette. Our data indicate that Ca2+ is not necessary for Kv4.3/KChIP2 complex formation. The Kv4.3/KChIP2-mediated current decay was faster and the recovery of Kv4.3/KChIP2 channels from inactivation slower with 50 µM Ca2+ than with BAPTA (nominal Ca2+-free) in the patch-pipette. The apparent Ca2+-mediated slowing of recovery kinetics was still observed when EF-hand 4 of KChIP2 was mutated (ΔEF4) but not when EF-hand 2 (ΔEF2) was mutated, and turned into a Ca2+-mediated acceleration of recovery kinetics when EF-hand 3 (ΔEF3) was mutated. In the presence of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor KN-93 cytoplasmic Ca2+ (50 µM) induced an acceleration of Kv4.3/KChIP2 recovery kinetics, which was still observed when EF-hand 2 was mutated (ΔEF2) but not when EF-hand 3 (ΔEF3) or EF-hand 4 (ΔEF4) was mutated. Our results support the notion that binding of Ca2+ to KChIP2 EF-hands can acutely modulate Kv4.3/KChIP2 channel inactivation gating, but the Ca2+-dependent gating modulation depends on CaMKII action. Our findings speak for an acute modulation of I to kinetics and frequency-dependent I to availability in cardiomyocytes under conditions with elevated Ca2+ levels and CaMKII activity.


Asunto(s)
Calcio/metabolismo , Citoplasma/metabolismo , Proteínas de Interacción con los Canales Kv/metabolismo , Canales de Potasio Shal/metabolismo , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular , Citoplasma/efectos de los fármacos , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Cinética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp/métodos , Sulfonamidas/farmacología
5.
Biophys J ; 110(1): 157-75, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26745419

RESUMEN

We studied the kinetics and structural determinants of closed-state inactivation (CSI) in Kv4.2 channels, considering a multistep process and the possibility that both intra- and intersubunit dynamic binding (i.e., loss and restoration of physical contact) may occur between the S4-S5 linker, including the initial S5 segment (S4S5), and the S6 gate. We expressed Kv4.2 channels in Xenopus oocytes and measured the onset of low-voltage inactivation under two-electrode voltage clamp. Indicative of a transitory state, the onset kinetics were best described by a double-exponential function. To examine the involvement of individual S4S5 and S6 amino acid residues in dynamic binding, we studied S4S5 and S6 single alanine mutants and corresponding double mutants. Both transitory and steady-state inactivation were modified by these mutations, and we quantified the mutational effects based on apparent affinities for the respective inactivated states. Double-mutant cycle analyses revealed strong functional coupling of the S6 residues V404 and I412 to all tested S4S5 residues. To examine whether dynamic S4S5/S6 binding occurs within individual α-subunits or between neighboring α-subunits, we performed a double-mutant cycle analysis with Kv4.2 tandem-dimer constructs. The constructs carried either an S4S5/S6 double mutation in the first α-subunit and no mutation in the second (concatenated) α-subunit or an S4S5 point mutation in the first α-subunit and an S6 point mutation in the second α-subunit. Our results support the notion that CSI in Kv4.2 channels is a multistep process that involves dynamic binding both within individual α-subunits and between neighboring α-subunits.


Asunto(s)
Activación del Canal Iónico , Subunidades de Proteína/metabolismo , Canales de Potasio Shal/química , Canales de Potasio Shal/metabolismo , Animales , Humanos , Cinética , Modelos Moleculares , Mutación Puntual , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Canales de Potasio Shal/genética , Xenopus/genética
6.
FASEB J ; 28(5): 2177-90, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24451387

RESUMEN

HCN channels are important regulators of neuronal excitability. The proper function of these channels is governed by various mechanisms, including post-translational modifications of channel subunits. Here, we provide evidence that ubiquitination via a ubiquitin ligase, neuronal precursor cell expressed developmentally downregulated (Nedd)-4-2, is involved in the regulation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. We identified a PY motif (L/PPxY), the characteristic binding motif for Nedd4-2 in the C terminus of the HCN1 subunit, and showed that HCN1 and Nedd4-2 interacted both in vivo (rat hippocampus, neocortex, and cerebellum) and in vitro [human embryonic kidney 293 (HEK293) cells], resulting in increased HCN1 ubiquitination. Elimination of the PY motif reduced, but did not abolish, Nedd4-2 binding, which further involved a stretch of ∼100 aa downstream in the HCN1 C terminus. Coexpression of Nedd4-2 and HCN1 drastically reduced the HCN1-mediated h-current amplitude (85-92%) in Xenopus laevis oocytes and reduced surface expression (34%) of HCN1 channels in HEK293 cells, thereby opposing effects of tetratricopeptide repeat-containing Rab8b interacting protein (TRIP8b)-(1a-4), an auxiliary subunit that promotes HCN1 surface expression. Regulation may further include N-glycosylation of HCN1 channels, which is significantly enhanced by TRIP8b(1a-4), but may be reduced by Nedd4-2. Taken together, our data indicate that Nedd4-2 plays an important role in the regulation of HCN1 trafficking and may compete with TRIP8b(1a-4) in this process.


Asunto(s)
Membrana Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Regulación de la Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Secuencias de Aminoácidos , Animales , Encéfalo/metabolismo , Regulación hacia Abajo , Electrofisiología , Femenino , Glicosilación , Células HEK293 , Humanos , Ubiquitina-Proteína Ligasas Nedd4 , Oocitos/citología , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas de Xenopus , Xenopus laevis
7.
Adv Physiol Educ ; 38(3): 253-64, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25179616

RESUMEN

The generation and conduction of neuronal action potentials (APs) were the subjects of a cell physiology exercise for first-year medical students. In this activity, students demonstrated the all-or-none nature of AP generation, measured conduction velocity, and examined the dependence of the threshold stimulus amplitude on stimulus duration. For this purpose, they used the median giant nerve fiber (MGF) in the ventral nerve cord of the common earthworm (Lumbricus terrestris). Here, we introduce a specialized stimulation and recording chamber that the nonanesthetized earthworm enters completely unforced. The worm resides in a narrow round duct with silver electrodes on the bottom such that individual APs of the MGF can be elicited and recorded superficially. Our experimental setup combines several advantages: it allows noninvasive single fiber AP measurements taken from a nonanesthetized animal that is yet restrained. Students performed the experiments with a high success rate. According to the data acquired by the students, the mean conduction velocity of the MGF was 30.2 m/s. From the amplitude-duration relationship for threshold stimulation, rheobase and chronaxie were graphically determined by the students according to Lapicque's method. The mean rheobase was 1.01 V, and the mean chronaxie was 0.06 ms. The acquired data and analysis results are of high quality, as deduced from critical examination based on the law of Weiss. In addition, we provide video material, which was also used in the practical course.


Asunto(s)
Fibras Nerviosas/fisiología , Oligoquetos/fisiología , Animales
8.
J Physiol ; 589(Pt 3): 461-79, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21098008

RESUMEN

Inactivation of voltage-gated ion channels is an intrinsic auto-regulatory process necessary to govern the occurrence and shape of action potentials and establish firing patterns in excitable tissues. Inactivation may occur from the open state (open-state inactivation, OSI) at strongly depolarized membrane potentials, or from pre-open closed states (closed-state inactivation, CSI) at hyperpolarized and modestly depolarized membrane potentials. Voltage-gated Na(+), K(+), Ca(2+) and non-selective cationic channels utilize both OSI and CSI. Whereas there are detailed mechanistic descriptions of OSI, much less is known about the molecular basis of CSI. Here, we review evidence for CSI in voltage-gated cationic channels (VGCCs) and recent findings that shed light on the molecular mechanisms of CSI in voltage-gated K(+) (Kv) channels. Particularly, complementary observations suggest that the S4 voltage sensor, the S4S5 linker and the main S6 activation gate are instrumental in the installment of CSI in Kv4 channels. According to this hypothesis, the voltage sensor may adopt a distinct conformation to drive CSI and, depending on the stability of the interactions between the voltage sensor and the pore domain, a closed-inactivated state results from rearrangements in the selectivity filter or failure of the activation gate to open. Kv4 channel CSI may efficiently exploit the dynamics of the subthreshold membrane potential to regulate spiking properties in excitable tissues.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Potenciales de la Membrana/fisiología , Animales , Humanos , Canales de Potasio Shal/fisiología
9.
J Biol Chem ; 285(37): 28968-79, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20610389

RESUMEN

Cell adhesion molecules and neurotrophin receptors are crucial for the development and the function of the nervous system. Among downstream effectors of neurotrophin receptors and recognition molecules are ion channels. Here, we provide evidence that G protein-coupled inwardly rectifying K(+) channel Kir3.3 directly binds to the neural cell adhesion molecule (NCAM) and neurotrophin receptor TrkB. We identified the binding sites for NCAM and TrkB at the C-terminal intracellular domain of Kir3.3. The interaction between NCAM, TrkB, and Kir3.3 was supported by immunocytochemical co-localization of Kir3.3, NCAM, and/or TrkB at the surface of hippocampal neurons. Co-expression of TrkB and Kir3.1/3.3 in Xenopus oocytes increased the K(+) currents evoked by Kir3.1/3.3 channels. This current enhancement was reduced by the concomitant co-expression with NCAM. Both surface fluorescence measurements of microinjected oocytes and cell surface biotinylation of transfected CHO cells indicated that the cell membrane localization of Kir3.3 is regulated by TrkB and NCAM. Furthermore, the level of Kir3.3, but not of Kir3.2, at the plasma membranes was reduced in TrkB-deficient mice, supporting the notion that TrkB regulates the cell surface expression of Kir3.3. The premature expression of developmentally late appearing Kir3.1/3.3 in hippocampal neurons led to a reduction of NCAM-induced neurite outgrowth. Our observations indicate a decisive role for the neuronal K(+) channel in regulating NCAM-dependent neurite outgrowth and attribute a physiologically meaningful role to the functional interplay of Kir3.3, NCAM, and TrkB in ontogeny.


Asunto(s)
Membrana Celular/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Hipocampo/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuritas/metabolismo , Receptor trkB/metabolismo , Animales , Células CHO , Adhesión Celular/fisiología , Membrana Celular/genética , Cricetinae , Cricetulus , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Noqueados , Moléculas de Adhesión de Célula Nerviosa/genética , Oocitos , Unión Proteica/fisiología , Ratas , Receptor trkB/genética , Xenopus laevis
10.
J Mol Cell Cardiol ; 48(1): 140-51, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19744491

RESUMEN

The transient outward potassium current (I(to)) in cardiac myocytes is mainly mediated by members of the Kv4 subfamily of voltage-gated potassium channels. Several in vitro studies have shown that angiotensin II (Ang II), which plays an important role in the development of cardiac hypertrophy, rapidly downregulates Kv4.3 mRNA expression. However, it is not clear whether Ang II regulates I(to)in vivo and whether this regulation may depend on alterations in Kv4.3 gene expression. To address this question, we determined the effects of acute (24 h) and chronic (14 days) exogenous infusions of Ang II on I(to) and the expression of its channel subunits in the mouse left ventricle. Ang II rapidly increased blood pressure and reduced Kv4.2 but not Kv4.3 mRNA levels in the absence of cardiac hypertrophy. In response to chronically elevated Ang II levels cardiac hypertrophy developed, which was associated with a downregulation of Kv4.2 and Kv4.3 mRNA levels, and an upregulation of Kv1.4 mRNA levels. In contrast, neither KChIP2 mRNA levels nor amplitude or macroscopic inactivation kinetics of I(to) were affected by the acute or chronic Ang II treatments. Consistent with the unchanged I(to) amplitude, Kv4.2, Kv4.3, and KChIP protein expression levels were similar after chronic Ang II and sham treatment. Our findings demonstrate that elevations of Ang II concentrations that induce hypertension and cardiac hypertrophy do not alter the amplitude of I(to) in the mouse left ventricle. Furthermore, they suggest that functional expression of cardiac I(to) in mice is stabilized by KChIP2.


Asunto(s)
Angiotensina II/farmacología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Vasoconstrictores/farmacología , Animales , Western Blotting , Electrofisiología , Hemodinámica/efectos de los fármacos , Proteínas de Interacción con los Canales Kv/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Canales de Potasio/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Potasio Shal/metabolismo
11.
Biophys J ; 94(4): 1276-94, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17981906

RESUMEN

We examined the relationship between deactivation and inactivation in Kv4.2 channels. In particular, we were interested in the role of a Kv4.2 N-terminal domain and accessory subunits in controlling macroscopic gating kinetics and asked if the effects of N-terminal deletion and accessory subunit coexpression conform to a kinetic coupling of deactivation and inactivation. We expressed Kv4.2 wild-type channels and N-terminal deletion mutants in the absence and presence of Kv channel interacting proteins (KChIPs) and dipeptidyl aminopeptidase-like proteins (DPPs) in human embryonic kidney 293 cells. Kv4.2-mediated A-type currents at positive and deactivation tail currents at negative membrane potentials were recorded under whole-cell voltage-clamp and analyzed by multi-exponential fitting. The observed changes in Kv4.2 macroscopic inactivation kinetics caused by N-terminal deletion, accessory subunit coexpression, or a combination of the two maneuvers were compared with respective changes in deactivation kinetics. Extensive correlation analyses indicated that modulatory effects on deactivation closely parallel respective effects on inactivation, including both onset and recovery kinetics. Searching for the structural determinants, which control deactivation and inactivation, we found that in a Kv4.2 Delta 2-10 N-terminal deletion mutant both the initial rapid phase of macroscopic inactivation and tail current deactivation were slowed. On the other hand, the intermediate and slow phase of A-type current decay, recovery from inactivation, and tail current decay kinetics were accelerated in Kv4.2 Delta 2-10 by KChIP2 and DPPX. Thus, a Kv4.2 N-terminal domain, which may control both inactivation and deactivation, is not necessary for active modulation of current kinetics by accessory subunits. Our results further suggest distinct mechanisms for Kv4.2 gating modulation by KChIPs and DPPs.


Asunto(s)
Activación del Canal Iónico/fisiología , Riñón/fisiología , Modelos Biológicos , Modelos Químicos , Canales de Potasio Shal/química , Canales de Potasio Shal/fisiología , Línea Celular , Simulación por Computador , Humanos , Potenciales de la Membrana/fisiología , Conformación Proteica , Subunidades de Proteína , Canales de Potasio Shal/ultraestructura , Relación Estructura-Actividad
12.
Channels (Austin) ; 12(1): 187-200, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30070159

RESUMEN

Kv channel-interacting proteins (KChIPs) belong to the neuronal calcium sensor (NCS) family of Ca2+-binding EF-hand proteins. KChIPs constitute a group of specific auxiliary ß-subunits for Kv4 channels, the molecular substrate of transient potassium currents in both neuronal and non-neuronal tissues. Moreover, KChIPs can interact with presenilins to control ER calcium signaling and apoptosis, and with DNA to control gene transcription. Ca2+ binding via their EF-hands, with the consequence of conformationl changes, is well documented for KChIPs. Moreover, the Ca2+ dependence of the presenilin/KChIP complex may be related to Alzheimer's disease and the Ca2+ dependence of the DNA/KChIP complex to pain sensing. However, only in few cases could the Ca2+ binding to KChIPs be directly linked to the control of excitability in nerve and muscle cells known to express Kv4/KChIP channel complexes. This review summarizes current knowledge about the Ca2+ binding properties of KChIPs and the Ca2+ dependencies of macromolecular complexes containing KChIPs, including those with presenilins, DNA and especially Kv4 channels. The respective physiological or pathophysiolgical roles of Ca2+ binding to KChIPs are discussed.


Asunto(s)
Calcio/metabolismo , Proteínas de Interacción con los Canales Kv/metabolismo , Neuronas/metabolismo , Humanos
13.
PLoS One ; 13(1): e0191911, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29385176

RESUMEN

Kv4.2 channels mediate a subthreshold-activating somatodendritic A-type current (ISA) in hippocampal neurons. We examined the role of accessory Kv channel interacting protein (KChIP) binding in somatodendritic surface expression and activity-dependent decrease in the availability of Kv4.2 channels. For this purpose we transfected cultured hippocampal neurons with cDNA coding for Kv4.2 wild-type (wt) or KChIP binding-deficient Kv4.2 mutants. All channels were equipped with an externally accessible hemagglutinin (HA)-tag and an EGFP-tag, which was attached to the C-terminal end. Combined analyses of EGFP self-fluorescence, surface HA immunostaining and patch-clamp recordings demonstrated similar dendritic trafficking and functional surface expression for Kv4.2[wt]HA,EGFP and the KChIP binding-deficient Kv4.2[A14K]HA,EGFP. Coexpression of exogenous KChIP2 augmented the surface expression of Kv4.2[wt]HA,EGFP but not Kv4.2[A14K]HA,EGFP. Notably, activity-dependent decrease in availability was more pronounced in Kv4.2[wt]HA,EGFP + KChIP2 coexpressing than in Kv4.2[A14K]HA,EGFP + KChIP2 coexpressing neurons. Our results do not support the notion that accessory KChIP binding is a prerequisite for dendritic trafficking and functional surface expression of Kv4.2 channels, however, accessory KChIP binding may play a potential role in Kv4.2 modulation during intrinsic plasticity processes.


Asunto(s)
Hipocampo/metabolismo , Proteínas de Interacción con los Canales Kv/metabolismo , Canales de Potasio Shal/metabolismo , Potenciales de Acción , Animales , Células Cultivadas , Dendritas/metabolismo , Epítopos/genética , Epítopos/metabolismo , Hipocampo/citología , Humanos , Inmunohistoquímica , Proteínas de Interacción con los Canales Kv/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neuronas/metabolismo , Técnicas de Placa-Clamp , Unión Proteica , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canales de Potasio Shal/química , Canales de Potasio Shal/genética , Transfección
14.
PLoS One ; 12(1): e0171213, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28141821

RESUMEN

The transient outward current (Ito) in cardiomyocytes is largely mediated by Kv4 channels associated with Kv Channel Interacting Protein 2 (KChIP2). A knockout model has documented the critical role of KChIP2 in Ito expression. The present study was conducted to characterize in both sexes the dependence of Ito properties, including current magnitude, inactivation kinetics, recovery from inactivation and voltage dependence of inactivation, on the number of functional KChIP2 alleles. For this purpose we performed whole-cell patch-clamp experiments on isolated left ventricular cardiomyocytes from male and female mice which had different KChIP2 genotypes; i.e., wild-type (KChIP2+/+), heterozygous knockout (KChIP2+/-) or complete knockout of KChIP2 (KChIP2-/-). We found in both sexes a KChIP2 gene dosage effect (i.e., a proportionality between number of alleles and phenotype) on Ito magnitude, however, concerning other Ito properties, KChIP2+/- resembled KChIP2+/+. Only in the total absence of KChIP2 (KChIP2-/-) we observed a slowing of Ito kinetics, a slowing of recovery from inactivation and a negative shift of a portion of the voltage dependence of inactivation. In a minor fraction of KChIP2-/- myocytes Ito was completely lost. The distinct KChIP2 genotype dependences of Ito magnitude and inactivation kinetics, respectively, seen in cardiomyocytes were reproduced with two-electrode voltage-clamp experiments on Xenopus oocytes expressing Kv4.2 and different amounts of KChIP2. Our results corroborate the critical role of KChIP2 in controlling Ito properties. They demonstrate that the Kv4.2/KChIP2 interaction in cardiomyocytes is highly dynamic, with a clear KChIP2 gene dosage effect on Kv4 channel surface expression but not on inactivation gating.


Asunto(s)
Separación Celular , Activación del Canal Iónico/genética , Proteínas de Interacción con los Canales Kv/genética , Miocitos Cardíacos/metabolismo , Animales , Femenino , Genotipo , Cinética , Proteínas de Interacción con los Canales Kv/deficiencia , Proteínas de Interacción con los Canales Kv/metabolismo , Masculino , Ratones Endogámicos C57BL , Canales de Potasio Shal/metabolismo , Xenopus
15.
Oncotarget ; 7(43): 69703-69717, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27626485

RESUMEN

Development of drug resistance is an inevitable phenomenon in castration-resistant prostate cancer (CRPC) cells requiring novel therapeutic approaches. In this study, efficacy and toxicity of Rhizochalinin (Rhiz) - a novel sphingolipid-like marine compound - was evaluated in prostate cancer models, resistant to currently approved standard therapies. In vitro activity and mechanism of action of Rhiz were examined in the human prostate cancer cell lines PC-3, DU145, LNCaP, 22Rv1, and VCaP. Rhiz significantly reduced cell viability at low micromolar concentrations showing most pronounced effects in enzalutamide and abiraterone resistant AR-V7 positive cells. Caspase-dependent apoptosis, inhibition of pro-survival autophagy, downregulation of AR-V7, PSA and IGF-1 expression as well as inhibition of voltage-gated potassium channels were identified as mechanisms of action. Remarkably, Rhiz re-sensitized AR-V7 positive cells to enzalutamide and increased efficacy of taxanes.In vivo activity and toxicity were evaluated in PC-3 and 22Rv1 NOD SCID mouse xenograft models using i.p. administration. Rhiz significantly reduced growth of PC-3 and 22Rv1 tumor xenografts by 27.0% (p = 0.0156) and 46.8% (p = 0.047) compared with controls with an increased fraction of tumor cells showing apoptosis secondary to Rhiz exposure. In line with the in vitro data, Rhiz was most active in AR-V7 positive xenografts in vivo. In animals, no severe side effects were observed.In conclusion, Rhiz is a promising novel marine-derived compound characterized by a unique combination of anticancer properties. Its further clinical development is of high impact for patients suffering from drug resistant prostate cancer especially those harboring AR-V7 mediated resistance to enzalutamide and abiraterone.


Asunto(s)
Alcoholes Grasos/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Caspasas/fisiología , Línea Celular Tumoral , Docetaxel , Alcoholes Grasos/efectos adversos , Alcoholes Grasos/farmacología , Humanos , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Ratones , Bloqueadores de los Canales de Potasio/farmacología , Antígeno Prostático Específico/análisis , Taxoides/farmacología
16.
J Mol Med (Berl) ; 82(3): 189-96, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14714110

RESUMEN

We have found a novel nonsense mutation in the C-terminus of HERG in a four-generation Chinese family with long QT syndrome and investigated the molecular mechanism of this mutation in vitro. Six family members, including the proband, were clinically affected. Syncope and ventricular tachycardia of torsades de pointes were triggered by startling or emotional stress, and beta-adrenergic blockade treatment was ineffective. Haplotype analysis showed that only LQT2 markers cosegregated with the disease, and sequence analysis revealed a substitution of T with C at nucleotide position 2770 of the HERG gene (U04270), which creates a stop codon at amino acid position 863 (R863X) of the HERG protein, leading to a deletion of 296 amino acids. Whole cell patch clamp studies showed that the R863X HERG could not induce time-dependent current. Coexpression of R863X with wild-type HERG showed reduced current densities and accelerated voltage-dependent inactivation of HERG channels. Subcellular localization of R863X-EGFP revealed that the mutant did not traffic to the cell surface. These data suggest that R863X failed to form functional HERG channels, contributing to a prolongation of the QT interval and long QT syndrome with a dominant phenotype. These findings provide new insights into the structure-function relationships of the HERG C-terminus.


Asunto(s)
Proteínas de Transporte de Catión/genética , Electrofisiología/métodos , Síndrome de QT Prolongado/genética , Mutación , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Animales , Secuencia de Bases , Células CHO , Proteínas de Transporte de Catión/química , Membrana Celular/metabolismo , Clonación Molecular , Cricetinae , Análisis Mutacional de ADN , Canales de Potasio Éter-A-Go-Go , Femenino , Eliminación de Gen , Haplotipos , Humanos , Masculino , Microscopía Confocal , Modelos Genéticos , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Linaje , Fenotipo , Canales de Potasio/química , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Factores de Tiempo
17.
Cardiovasc Res ; 59(3): 612-9, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14499862

RESUMEN

OBJECTIVES: To identify the underlying genetic basis of a Chinese pedigree with Long QT syndrome, the causally related genes were screened in a family and the functional consequence of the identified gene mutation was evaluated in vitro. METHODS: Mutations in the five defined Long QT syndrome related genes were screened with polymerase chain reaction and single-strand conformation polymorphism methods and direct sequencing. The electrophysiological properties of the identified mutation were characterized in the Xenopus oocyte heterologous expression system. RESULTS: A novel missense mutation, G to A at position 154 in the KCNE1 gene was identified in a Chinese Long QT syndrome family, which leads to an amino acid substitution of arginine (R) for glycine (G) at position 52 (G52R-KCNE1). Of 26 family members (one DNA was not available), seven were mutation carriers and two of them with normal electrocardiogram. Compared with wild-type KCNE1/KCNQ1 channels, coexpression of G52R-KCNE1 with KCNQ1 in Xenopus oocytes did not amplify the KCNQ1 current amplitudes and slightly changed the activation kinetics of the KCNQ1 channels. Coexpression of KCNQ1 together with wild type KCNE1 and G52R-KCNE1 reduced the wild-type I(ks) current amplitude by 50%, whereas other biophysical properties of the I(ks) were not altered. CONCLUSIONS: Our findings indicate that glycine52 in the transmembrane domain is critical for KCNE1 function. The mutant G52R-KCNE1 has a dominant negative effect on I(ks) current, which reduces the I(ks) current amplitude and leads to a prolongation of the cardiac action potential. This could underlie the molecular mechanism of ventricular arrhythmias and sudden death in those patients.


Asunto(s)
Síndrome de QT Prolongado/genética , Mutación , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Adolescente , Adulto , Animales , Niño , China , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oocitos/metabolismo , Linaje , Polimorfismo Conformacional Retorcido-Simple , Canales de Potasio/metabolismo , Análisis de Secuencia de ADN , Transfección , Xenopus laevis
18.
Toxicon ; 43(8): 923-32, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15208026

RESUMEN

In order to find new peptide inhibitors for voltage-dependent potassium (Kv) channels, we examined the effects of venom from Theraphosa leblondi on Kv channel-mediated currents with the whole-cell patch-clamp technique. Both A-type currents in cultured hippocampal neurons and A-type currents recorded from HEK 293 cells transiently expressing recombinant Kv4.2 channels were selectively inhibited by T. leblondi venom. No venom activity was observed on recombinant Kv1.3, Kv1.4, Kv2.1 or Kv3.4 channels. We purified and sequenced three novel homologous peptides from this venom, which are related to previously identified Kv4 channel-specific peptide inhibitors and were named T. leblondi toxin (TLTx) 1, 2 and 3. The mode of action of TLTx1 on recombinant Kv4.2 channels was studied in more detail. TLTx1 inhibited Kv4.2-mediated currents with an IC50 of approximately 200 nM, and macroscopic current inactivation was slowed in the presence of TLTx1. Notably, TLTx1 also caused a shallower voltage dependence of Kv4.2 peak conductance and a shift of the activation midpoint to more positive potentials (DeltaV1/2 = +35 mV). TLTx1 caused a noticable slowing of Kv4.2 activation kinetics, and Kv4.2 deactivation kinetics were accelerated by TLTx1 as infered from Rb+ tail current measurements. Chimeric Kv2.1(4.2L3-4) channels, in which the linker region between S3 and S4 of the TLTx1-insensitive Kv2.1 channel was replaced by the corresponding Kv4.2 domain, were sensitive to TLTx1. Apparently, TLTx1 can act as a gating modifier of Kv4.2 channels.


Asunto(s)
Péptidos/farmacología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/efectos de los fármacos , Venenos de Araña/farmacología , Arañas/química , Secuencia de Aminoácidos , Animales , Transporte Biológico Activo/efectos de los fármacos , Células Cultivadas , Fraccionamiento Químico , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Electrofisiología , Guyana Francesa , Humanos , Cinética , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , Neuronas/metabolismo , Técnicas de Placa-Clamp , Péptidos/genética , Péptidos/metabolismo , Canales de Potasio/metabolismo , Alineación de Secuencia , Análisis de Secuencia de Proteína , Canales de Potasio Shal , Venenos de Araña/metabolismo
19.
Neuropharmacology ; 63(8): 1389-403, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22964468

RESUMEN

We examined the effects of the sulfonylurea compound NS5806 on neuronal A-type channel function. Using whole-cell patch-clamp we studied the effects of NS5806 on the somatodendritic A-type current (I(SA)) in cultured hippocampal neurons and the currents mediated by Kv4.2 channels coexpressed with different auxiliary ß-subunits, including both Kv channel interacting proteins (KChIPs) and dipeptidyl aminopeptidase-related proteins (DPPs), in HEK 293 cells. The amplitude of the I(SA) component in hippocampal neurons was reduced in the presence of 20 µM NS5806. I(SA) decay kinetics were slowed and the recovery kinetics accelerated, but the voltage dependence of steady-state inactivation was shifted to more negative potentials by NS5806. The peak amplitudes of currents mediated by ternary Kv4.2 channel complexes, associated with DPP6-S (short splice-variant) and either KChIP2, KChIP3 or KChIP4, were potentiated and their macroscopic inactivation slowed by NS5806, whereas the currents mediated by binary Kv4.2 channels, associated only with DPP6-S, were suppressed, and the NS5806-mediated slowing of macroscopic inactivation was less pronounced. Neither potentiation nor suppression and no effect on current decay kinetics in the presence of NS5806 were observed for Kv4.2 channels associated with KChIP3 and the N-type inactivation-conferring DPP6a splice-variant. For all recombinant channel complexes, NS5806 slowed the recovery from inactivation and shifted the voltage dependence of steady-state inactivation to more negative potentials. Our results demonstrate the activity of NS5806 on native I(SA) and possible molecular correlates in the form of recombinant Kv4.2 channels complexed with different KChIPs and DPPs, and they shed some light on the mechanism of NS5806 action.


Asunto(s)
Hipocampo/metabolismo , Compuestos de Fenilurea/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Shal/efectos de los fármacos , Tetrazoles/farmacología , Animales , Células Cultivadas , Interpretación Estadística de Datos , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/efectos de los fármacos , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/fisiología , Células HEK293 , Hipocampo/efectos de los fármacos , Humanos , Técnicas In Vitro , Cinética , Proteínas de Interacción con los Canales Kv/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Wistar
20.
Front Pharmacol ; 3: 100, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22654758

RESUMEN

In voltage-gated potassium (Kv) channels membrane depolarization causes movement of a voltage sensor domain. This conformational change of the protein is transmitted to the pore domain and eventually leads to pore opening. However, the voltage sensor domain may interact with two distinct gates in the pore domain: the activation gate (A-gate), involving the cytoplasmic S6 bundle crossing, and the pore gate (P-gate), located externally in the selectivity filter. How the voltage sensor moves and how tightly it interacts with these two gates on its way to adopt a relaxed conformation when the membrane is depolarized may critically determine the mode of Kv channel inactivation. In certain Kv channels, voltage sensor movement leads to a tight interaction with the P-gate, which may cause conformational changes that render the selectivity filter non-conductive ("P/C-type inactivation"). Other Kv channels may preferably undergo inactivation from pre-open closed-states during voltage sensor movement, because the voltage sensor temporarily uncouples from the A-gate. For this behavior, known as "preferential" closed-state inactivation, we introduce the term "A/C-type inactivation". Mechanistically, P/C- and A/C-type inactivation represent two forms of "voltage sensor inactivation."

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA