Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(12): 2021-2031, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37903858

RESUMEN

S100A8/S100A9 is a proinflammatory mediator released by myeloid cells during many acute and chronic inflammatory disorders. However, the precise mechanism of its release from the cytosolic compartment of neutrophils is unclear. Here, we show that E-selectin-induced rapid S100A8/S100A9 release during inflammation occurs in an NLRP3 inflammasome-dependent fashion. Mechanistically, E-selectin engagement triggers Bruton's tyrosine kinase-dependent tyrosine phosphorylation of NLRP3. Concomitant potassium efflux via the voltage-gated potassium channel KV1.3 mediates ASC oligomerization. This is followed by caspase 1 cleavage and downstream activation of pore-forming gasdermin D, enabling cytosolic release of S100A8/S100A9. Strikingly, E-selectin-mediated gasdermin D pore formation does not result in cell death but is a transient process involving activation of the ESCRT III membrane repair machinery. These data clarify molecular mechanisms of controlled S100A8/S100A9 release from neutrophils and identify the NLRP3/gasdermin D axis as a rapid and reversible activation system in neutrophils during inflammation.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Gasderminas , Neutrófilos/metabolismo , Selectina E/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Inflamación/metabolismo
2.
Circ Res ; 134(10): 1348-1378, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38723033

RESUMEN

Loss or dysregulation of the normally precise control of heart rate via the autonomic nervous system plays a critical role during the development and progression of cardiovascular disease-including ischemic heart disease, heart failure, and arrhythmias. While the clinical significance of regulating changes in heart rate, known as the chronotropic effect, is undeniable, the mechanisms controlling these changes remain not fully understood. Heart rate acceleration and deceleration are mediated by increasing or decreasing the spontaneous firing rate of pacemaker cells in the sinoatrial node. During the transition from rest to activity, sympathetic neurons stimulate these cells by activating ß-adrenergic receptors and increasing intracellular cyclic adenosine monophosphate. The same signal transduction pathway is targeted by positive chronotropic drugs such as norepinephrine and dobutamine, which are used in the treatment of cardiogenic shock and severe heart failure. The cyclic adenosine monophosphate-sensitive hyperpolarization-activated current (If) in pacemaker cells is passed by hyperpolarization-activated cyclic nucleotide-gated cation channels and is critical for generating the autonomous heartbeat. In addition, this current has been suggested to play a central role in the chronotropic effect. Recent studies demonstrate that cyclic adenosine monophosphate-dependent regulation of HCN4 (hyperpolarization-activated cyclic nucleotide-gated cation channel isoform 4) acts to stabilize the heart rate, particularly during rapid rate transitions induced by the autonomic nervous system. The mechanism is based on creating a balance between firing and recently discovered nonfiring pacemaker cells in the sinoatrial node. In this way, hyperpolarization-activated cyclic nucleotide-gated cation channels may protect the heart from sinoatrial node dysfunction, secondary arrhythmia of the atria, and potentially fatal tachyarrhythmia of the ventricles. Here, we review the latest findings on sinoatrial node automaticity and discuss the physiological and pathophysiological role of HCN pacemaker channels in the chronotropic response and beyond.


Asunto(s)
Frecuencia Cardíaca , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Nodo Sinoatrial , Humanos , Animales , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/fisiopatología , Nodo Sinoatrial/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Relojes Biológicos
3.
Handb Exp Pharmacol ; 278: 277-304, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36894791

RESUMEN

Endo-lysosomes are membrane-bound acidic organelles that are involved in endocytosis, recycling, and degradation of extracellular and intracellular material. The membranes of endo-lysosomes express several Ca2+-permeable cation ion channels, including two-pore channels (TPC1-3) and transient receptor potential mucolipin channels (TRPML1-3). In this chapter, we will describe four different state-of-the-art Ca2+ imaging approaches, which are well-suited to investigate the function of endo-lysosomal cation channels. These techniques include (1) global cytosolic Ca2+ measurements, (2) peri-endo-lysosomal Ca2+ imaging using genetically encoded Ca2+ sensors that are directed to the cytosolic endo-lysosomal membrane surface, (3) Ca2+ imaging of endo-lysosomal cation channels, which are engineered in order to redirect them to the plasma membrane in combination with approaches 1 and 2, and (4) Ca2+ imaging by directing Ca2+ indicators to the endo-lysosomal lumen. Moreover, we will review useful small molecules, which can be used as valuable tools for endo-lysosomal Ca2+ imaging. Rather than providing complete protocols, we will discuss specific methodological issues related to endo-lysosomal Ca2+ imaging.


Asunto(s)
Calcio , Canales de Potencial de Receptor Transitorio , Humanos , Calcio/metabolismo , Lisosomas/metabolismo , Señalización del Calcio , Cationes/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(30): 18068-18078, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32661165

RESUMEN

Mast cells and basophils are main drivers of allergic reactions and anaphylaxis, for which prevalence is rapidly increasing. Activation of these cells leads to a tightly controlled release of inflammatory mediators stored in secretory granules. The release of these granules is dependent on intracellular calcium (Ca2+) signals. Ca2+ release from endolysosomal compartments is mediated via intracellular cation channels, such as two-pore channel (TPC) proteins. Here, we uncover a mechanism for how TPC1 regulates Ca2+ homeostasis and exocytosis in mast cells in vivo and ex vivo. Notably, in vivo TPC1 deficiency in mice leads to enhanced passive systemic anaphylaxis, reflected by increased drop in body temperature, most likely due to accelerated histamine-induced vasodilation. Ex vivo, mast cell-mediated histamine release and degranulation was augmented upon TPC1 inhibition, although mast cell numbers and size were diminished. Our results indicate an essential role of TPC1 in endolysosomal Ca2+ uptake and filling of endoplasmic reticulum Ca2+ stores, thereby regulating exocytosis in mast cells. Thus, pharmacological modulation of TPC1 might blaze a trail to develop new drugs against mast cell-related diseases, including allergic hypersensitivity.


Asunto(s)
Anafilaxia/etiología , Anafilaxia/metabolismo , Canales de Calcio/deficiencia , Susceptibilidad a Enfermedades , Mastocitos/inmunología , Mastocitos/metabolismo , Biomarcadores , Señalización del Calcio , Degranulación de la Célula , Citocinas/metabolismo , Predisposición Genética a la Enfermedad , Histamina/metabolismo , Inmunoglobulina E/inmunología , Mediadores de Inflamación/metabolismo
5.
Pflugers Arch ; 474(7): 649-663, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35556164

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are the molecular correlate of the If current and are critically involved in controlling neuronal excitability and the autonomous rhythm of the heart. The HCN4 isoform is the main HCN channel subtype expressed in the sinoatrial node (SAN), a tissue composed of specialized pacemaker cells responsible for generating the intrinsic heartbeat. More than 40 years ago, the If current was first discovered in rabbit SAN tissue. Along with this discovery, a theory was proposed that cyclic adenosine monophosphate-dependent modulation of If mediates heart rate regulation by the autonomic nervous system-a process called chronotropic effect. However, up to the present day, this classical theory could not be reliably validated. Recently, new concepts emerged confirming that HCN4 channels indeed play an important role in heart rate regulation. However, the cellular mechanism by which HCN4 controls heart rate turned out to be completely different than originally postulated. Here, we review the latest findings regarding the physiological role of HCN4 in the SAN. We describe a newly discovered mechanism underlying heart rate regulation by HCN4 at the tissue and single cell levels, and we discuss these observations in the context of results from previously studied HCN4 mouse models.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Nodo Sinoatrial , Animales , AMP Cíclico , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Frecuencia Cardíaca , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Ratones , Conejos
6.
Glia ; 69(4): 872-889, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33156956

RESUMEN

Astrocytes are an important component of the multipartite synapse and crucial for proper neuronal network function. Although small GTPases of the Rho family are powerful regulators of cellular morphology, the signaling modules of Rho-mediated pathways in astrocytes remain enigmatic. Here we demonstrated that the serotonin receptor 4 (5-HT4 R) is expressed in hippocampal astrocytes, both in vitro and in vivo. Through fluorescence microscopy, we established that 5-HT4 R activation triggered RhoA activity via Gα13 -mediated signaling, which boosted filamentous actin assembly, leading to morphological changes in hippocampal astrocytes. We investigated the effects of these 5-HT4 R-mediated changes in mixed cultures and in acute slices, in which 5-HT4 R was expressed exclusively in astrocytes. In both systems, 5-HT4 R-RhoA signaling changed glutamatergic synaptic transmission: It increased the frequency of miniature excitatory postsynaptic currents (mEPSCs) in mixed cultures and reduced the paired-pulse-ratio (PPR) of field excitatory postsynaptic potentials (fEPSPs) in acute slices. Overall, our present findings demonstrate that astrocytic 5-HT4 R-Gα13 -RhoA signaling is a previously unrecognized molecular pathway involved in the functional regulation of excitatory synaptic circuits.


Asunto(s)
Astrocitos , Serotonina , Potenciales Postsinápticos Excitadores , Hipocampo , Receptores de Serotonina/genética , Transmisión Sináptica
7.
Proc Natl Acad Sci U S A ; 114(20): 5259-5264, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28468800

RESUMEN

Retinitis pigmentosa (RP) is an inherited neurodegenerative disease, in which the death of mutant rod photoreceptors leads secondarily to the non-cell autonomous death of cone photoreceptors. Gene therapy is a promising treatment strategy. Unfortunately, current methods of gene delivery treat only a fraction of diseased cells, yielding retinas that are a mosaic of treated and untreated rods, as well as cones. In this study, we created two RP mouse models to test whether dying, untreated rods negatively impact treated, rescued rods. In one model, treated and untreated rods were segregated. In the second model, treated and untreated rods were diffusely intermixed, and their ratio was controlled to achieve low-, medium-, or high-efficiency rescue. Analysis of these mosaic retinas demonstrated that rescued rods (and cones) survive, even when they are greatly outnumbered by dying photoreceptors. On the other hand, the rescued photoreceptors did exhibit long-term defects in their outer segments (OSs), which were less severe when more photoreceptors were treated. In summary, our study suggests that even low-efficiency gene therapy may achieve stable survival of rescued photoreceptors in RP patients, albeit with OS dysgenesis.


Asunto(s)
Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Animales , Muerte Celular , Modelos Animales de Enfermedad , Terapia Genética/métodos , Ratones , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Rodopsina/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(41): E8595-E8602, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28923947

RESUMEN

Two-pore channels (TPCs) are endolysosomal cation channels. Two members exist in humans, TPC1 and TPC2. Functional roles associated with the ubiquitously expressed TPCs include VEGF-induced neoangiogenesis, LDL-cholesterol trafficking and degradation, physical endurance under fasting conditions, autophagy regulation, the acrosome reaction in sperm, cancer cell migration, and intracellular trafficking of pathogens such as Ebola virus or bacterial toxins (e.g., cholera toxin). In a genome-wide association study for variants associated with human pigmentation characteristics two coding variants of TPC2, rs35264875 (encoding M484L) and rs3829241 (encoding G734E), have been found to be associated with a shift from brown to blond hair color. In two recent follow-up studies a role for TPC2 in pigmentation has been further confirmed. However, these human polymorphic variants have not been functionally characterized until now. The development of endolysosomal patch-clamp techniques has made it possible to investigate directly ion channel activities and characteristics in isolated endolysosomal organelles. We applied this technique here to scrutinize channel characteristics of the polymorphic TPC2 variants in direct comparison with WT. We found that both polymorphisms lead to a gain of channel function by independent mechanisms. We next conducted a clinical study with more than 100 blond- and brown/black-haired individuals. We performed a genotype/phenotype analysis and subsequently isolated fibroblasts from WT and polymorphic variant carriers for endolysosomal patch-clamp experimentation to confirm key in vitro findings.


Asunto(s)
Canales de Calcio/genética , Cabello/química , Pigmentación/genética , Polimorfismo Genético , Canales de Calcio/fisiología , Estudio de Asociación del Genoma Completo , Células HEK293 , Cabello/metabolismo , Humanos , Técnicas de Placa-Clamp , Fenotipo
9.
EMBO J ; 34(13): 1743-58, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25872774

RESUMEN

The second messenger NAADP triggers Ca(2+) release from endo-lysosomes. Although two-pore channels (TPCs) have been proposed to be regulated by NAADP, recent studies have challenged this. By generating the first mouse line with demonstrable absence of both Tpcn1 and Tpcn2 expression (Tpcn1/2(-/-)), we show that the loss of endogenous TPCs abolished NAADP-dependent Ca(2+) responses as assessed by single-cell Ca(2+) imaging or patch-clamp of single endo-lysosomes. In contrast, currents stimulated by PI(3,5)P2 were only partially dependent on TPCs. In Tpcn1/2(-/-) cells, NAADP sensitivity was restored by re-expressing wild-type TPCs, but not by mutant versions with impaired Ca(2+)-permeability, nor by TRPML1. Another mouse line formerly reported as TPC-null likely expresses truncated TPCs, but we now show that these truncated proteins still support NAADP-induced Ca(2+) release. High-affinity [(32)P]NAADP binding still occurs in Tpcn1/2(-/-) tissue, suggesting that NAADP regulation is conferred by an accessory protein. Altogether, our data establish TPCs as Ca(2+)-permeable channels indispensable for NAADP signalling.


Asunto(s)
Canales de Calcio/genética , Calcio/metabolismo , NADP/análogos & derivados , Animales , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Células Cultivadas , Potenciales Evocados/efectos de los fármacos , Expresión Génica/fisiología , Concentración de Iones de Hidrógeno , Lisosomas/efectos de los fármacos , Lisosomas/fisiología , Ratones , Ratones Noqueados , NADP/metabolismo , NADP/farmacología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Hum Mol Genet ; 25(12): 2367-2377, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27033727

RESUMEN

Peripherin-2 is a glycomembrane protein exclusively expressed in the light-sensing compartments of rod and cone photoreceptors designated as outer segments (OS). Mutations in peripherin-2 are associated with degenerative retinal diseases either affecting rod or cone photoreceptors. While peripherin-2 has been extensively studied in rods, there is only little information on its supramolecular organization and function in cones. Recently, we have demonstrated that peripherin-2 interacts with the light detector rhodopsin in OS of rods. It remains unclear, however, if peripherin-2 also binds to cone opsins. Here, using a combination of co-immunoprecipitation analyses, transmission electron microscopy (TEM)-based immunolabeling experiments, and quantitative fluorescence resonance energy transfer (FRET) measurements in cone OS of wild type mice, we demonstrate that peripherin-2 binds to both, S-opsin and M-opsin. However, FRET-based quantification of the respective interactions indicated significantly less stringent binding of peripherin-2 to S-opsin compared to its interaction with M-opsin. Subsequent TEM-studies also showed less co-localization of peripherin-2 and S-opsin in cone OS compared to peripherin-2 and M-opsin. Furthermore, quantitative FRET analysis in acutely isolated cone OS revealed that the cone degeneration-causing V268I mutation in peripherin-2 selectively reduced binding to M-opsin without affecting the peripherin-2 interaction to S-opsin or rhodopsin. The differential binding of peripherin-2 to cone opsins and the mutant-specific interference with the peripherin-2/M-opsin binding points to a novel role of peripherin-2 in cones and might contribute to understanding the differential penetrance of certain peripherin-2 mutations in rods and cones. Finally, our results provide a proof-of-principle for quantitative FRET measurements of protein-protein interactions in cone OS.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Opsinas de los Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/genética , Animales , Antígenos de Neoplasias/genética , Opsinas de los Conos/genética , Transferencia Resonante de Energía de Fluorescencia , Humanos , Ratones , Microscopía Electrónica de Transmisión , Mutación , Unión Proteica , Retina/metabolismo , Retina/patología , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/patología , Rodopsina/genética , Rodopsina/metabolismo
11.
J Cell Sci ; 128(21): 4014-23, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26359296

RESUMEN

We show that the combination of an intracellular bi-partite calmodulin (CaM)-binding site and a distant assembly region affect how an ion channel is regulated by a membrane lipid. Our data reveal that regulation by phosphatidylinositol(4,5)bisphosphate (PIP2) and stabilization of assembled Kv7.2 subunits by intracellular coiled-coil regions far from the membrane are coupled molecular processes. Live-cell fluorescence energy transfer measurements and direct binding studies indicate that remote coiled-coil formation creates conditions for different CaM interaction modes, each conferring different PIP2 dependency to Kv7.2 channels. Disruption of coiled-coil formation by epilepsy-causing mutation decreases apparent CaM-binding affinity and interrupts CaM influence on PIP2 sensitivity.


Asunto(s)
Calmodulina/metabolismo , Canal de Potasio KCNQ2/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Calmodulina/genética , Línea Celular , Humanos , Canal de Potasio KCNQ2/genética , Mutación/genética , Unión Proteica
12.
Hum Mol Genet ; 23(22): 5989-97, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24963162

RESUMEN

Outer segments (OSs) of rod photoreceptors are cellular compartments specialized in the conversion of light into electrical signals. This process relies on the light-triggered change in the intracellular levels of cyclic guanosine monophosphate, which in turn controls the activity of cyclic nucleotide-gated (CNG) channels in the rod OS plasma membrane. The rod CNG channel is a macromolecular complex that in its core harbors the ion-conducting CNGA1 and CNGB1a subunits. To identify additional proteins of the complex that interact with the CNGB1a core subunit, we applied affinity purification of mouse retinal proteins followed by mass spectrometry. In combination with in vitro and in vivo co-immunoprecipitation and fluorescence resonance energy transfer (FRET), we found that the tetraspanin peripherin-2 links CNGB1a to the light-detector rhodopsin. Using immunoelectron microscopy, we found that this peripherin-2/rhodopsin/CNG channel complex localizes to the contact region between the disk rims and the plasma membrane. FRET measurements revealed that the fourth transmembrane domain (TM4) of peripherin-2 is required for the interaction with rhodopsin. Quantitatively, the binding affinity of the peripherin-2/rhodopsin interaction was in a similar range as that observed for rhodopsin dimers. Finally, we demonstrate that the p.G266D retinitis pigmentosa mutation found within TM4 selectively abolishes the binding of peripherin-2 to rhodopsin. This finding suggests that the specific disruption of the rhodopsin/peripherin-2 interaction in the p.G266D mutant might contribute to the pathophysiology in affected persons.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Periferinas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/metabolismo , Rodopsina/metabolismo , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Humanos , Ratones , Proteínas del Tejido Nervioso/genética , Periferinas/genética , Unión Proteica , Estructura Terciaria de Proteína , Retina/metabolismo , Retinitis Pigmentosa/genética , Rodopsina/genética
13.
Hum Mol Genet ; 23(6): 1538-50, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24163243

RESUMEN

Mutations in CACNA1F encoding the α1-subunit of the retinal Cav1.4 L-type calcium channel have been linked to Cav1.4 channelopathies including incomplete congenital stationary night blindness type 2A (CSNB2), Åland Island eye disease (AIED) and cone-rod dystrophy type 3 (CORDX3). Since CACNA1F is located on the X chromosome, Cav1.4 channelopathies are typically affecting male patients via X-chromosomal recessive inheritance. Occasionally, clinical symptoms have been observed in female carriers, too. It is currently unknown how these mutations lead to symptoms in carriers and how the retinal network in these females is affected. To investigate these clinically important issues, we compared retinal phenotypes in Cav1.4-deficient and Cav1.4 heterozygous mice and in human female carrier patients. Heterozygous Cacna1f carrier mice have a retinal mosaic consistent with differential X-chromosomal inactivation, characterized by adjacent vertical columns of affected and non-affected wild-type-like retinal network. Vertical columns in heterozygous mice are well comparable to either the wild-type retinal network of normal mice or to the retina of homozygous mice. Affected retinal columns display pronounced rod and cone photoreceptor synaptopathy and cone degeneration. These changes lead to vastly impaired vision-guided navigation under dark and normal light conditions and reduced retinal electroretinography (ERG) responses in Cacna1f carrier mice. Similar abnormal ERG responses were found in five human CACNA1F carriers, four of which had novel mutations. In conclusion, our data on Cav1.4 deficient mice and human female carriers of mutations in CACNA1F are consistent with a phenotype of mosaic CSNB2.


Asunto(s)
Canales de Calcio/genética , Enfermedades Hereditarias del Ojo/patología , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Miopía/patología , Ceguera Nocturna/patología , Retina/patología , Células Fotorreceptoras Retinianas Conos/patología , Animales , Canales de Calcio/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Modelos Animales de Enfermedad , Electrorretinografía , Enfermedades Hereditarias del Ojo/genética , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Heterocigoto , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación Missense , Miopía/genética , Ceguera Nocturna/genética , Fenotipo , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Cromosoma X , Inactivación del Cromosoma X
14.
Adv Exp Med Biol ; 854: 619-25, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427467

RESUMEN

Electroretinograms (ERGs) are commonly recorded at the cornea for an assessment of the functional status of the retina in mouse models. Full-field ERGs can be elicited by single-flash as well as flicker light stimulation although in most laboratories flicker ERGs are recorded much less frequently than singleflash ERGs. Whereas conventional single-flash ERGs contain information about layers, i.e., outer and inner retina, flicker ERGs permit functional assessment of the vertical pathways of the retina, i.e., rod system, cone ON-pathway, and cone OFF-pathway, when the responses are evoked at a relatively high luminance (0.5 log cd s/m(2)) with varying frequency (from 0.5 to 30 Hz) without any adapting background illumination. Therefore, both types of ERGs complement an in-depth functional characterization of the mouse retina, allowing for a discrimination of an underlying functional pathology. Here, we introduce the systematic interpretation of the single-flash and flicker ERGs by demonstrating several different patterns of functional phenotype in genetic mouse models, in which photoreceptors and/or bipolar cells are primarily or secondarily affected.


Asunto(s)
Adaptación a la Oscuridad/fisiología , Modelos Animales de Enfermedad , Electrorretinografía/métodos , Retina/fisiología , Animales , Adaptación a la Oscuridad/genética , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Humanos , Luz , Iluminación , Ratones Noqueados , Estimulación Luminosa , Retina/metabolismo , Transducina/genética , Transducina/metabolismo , Visión Ocular/genética , Visión Ocular/fisiología
15.
J Biol Chem ; 288(11): 7580-7589, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23382386

RESUMEN

Most ion channels consist of the principal ion-permeating core subunit(s) and accessory proteins that are assembled with the channel core. The biological functions of the latter proteins are diverse and include the regulation of the biophysical properties of the ion channel, its connection to signaling pathways and the control of its cell surface expression. There is recent evidence that native hyperpolarization-activated cyclic nucleotide-gated channel complexes (HCN1-4) also contain accessory subunits, among which TRIP8b (tetratricopeptide repeat-containing Rab8b-interacting protein) has been most extensively studied. Here, we identify KCTD3, a so far uncharacterized member of the potassium channel tetramerization-domain containing (KCTD) protein family as an HCN3-interacting protein. KCTD3 is widely expressed in brain and some non-neuronal tissues and colocalizes with HCN3 in specific regions of the brain including hypothalamus. Within the HCN channel family, KCTD3 specifically binds to HCN3 and leads to a profound up-regulation of cell surface expression and current density of this channel. HCN3 can also functionally interact with TRIP8b; however, we found no evidence for channel complexes containing both TRIP8b and KCTD3. The C terminus of HCN3 is crucially required for functional interaction with KCTD3. Replacement of the cytosolic C terminus of HCN2 by the corresponding domain of HCN3 renders HCN2 sensitive to regulation by KCTD3. The C-terminal-half of KCTD3 is sufficient for binding to HCN3. However, the complete protein including the N-terminal tetramerization domain is needed for HCN3 current up-regulation. Together, our experiments indicate that KCTD3 is an accessory subunit of native HCN3 complexes.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Regulación de la Expresión Génica , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Regulación hacia Arriba , Animales , Biofisica/métodos , Encéfalo/embriología , Encéfalo/metabolismo , Membrana Celular/metabolismo , Citosol/metabolismo , Electrofisiología/métodos , Vectores Genéticos , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Hipotálamo/metabolismo , Ratones , Microscopía Fluorescente/métodos , Técnicas de Placa-Clamp , Filogenia , Canales de Potasio/química , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Técnicas del Sistema de Dos Híbridos
16.
Circulation ; 128(24): 2585-94, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24218458

RESUMEN

BACKGROUND: Sinus node dysfunction (SND) is a major clinically relevant disease that is associated with sudden cardiac death and requires surgical implantation of electric pacemaker devices. Frequently, SND occurs in heart failure and hypertension, conditions that lead to electric instability of the heart. Although the pathologies of acquired SND have been studied extensively, little is known about the molecular and cellular mechanisms that cause congenital SND. METHODS AND RESULTS: Here, we show that the HCN1 protein is highly expressed in the sinoatrial node and is colocalized with HCN4, the main sinoatrial pacemaker channel isoform. To characterize the cardiac phenotype of HCN1-deficient mice, a detailed functional characterization of pacemaker mechanisms in single isolated sinoatrial node cells, explanted beating sinoatrial node preparation, telemetric in vivo electrocardiography, echocardiography, and in vivo electrophysiology was performed. On the basis of these experiments we demonstrate that mice lacking the pacemaker channel HCN1 display congenital SND characterized by bradycardia, sinus dysrhythmia, prolonged sinoatrial node recovery time, increased sinoatrial conduction time, and recurrent sinus pauses. As a consequence of SND, HCN1-deficient mice display a severely reduced cardiac output. CONCLUSIONS: We propose that HCN1 stabilizes the leading pacemaker region within the sinoatrial node and hence is crucial for stable heart rate and regular beat-to-beat variation. Furthermore, we suggest that HCN1-deficient mice may be a valuable genetic disease model for human SND.


Asunto(s)
Modelos Animales de Enfermedad , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/deficiencia , Canales de Potasio/deficiencia , Síndrome del Seno Enfermo/fisiopatología , Animales , Gasto Cardíaco/fisiología , Femenino , Frecuencia Cardíaca/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canales de Potasio/genética , Canales de Potasio/metabolismo , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/fisiopatología
18.
Eur Biophys J ; 43(2-3): 97-104, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24526306

RESUMEN

Patch clamp electrophysiology is the main technique to study mechanosensitive ion channels (MSCs), however, conventional patch clamping is laborious and success and output depends on the skills of the operator. Even though automated patch systems solve these problems for other ion channels, they could not be applied to MSCs. Here, we report on activation and single channel analysis of a bacterial mechanosensitive ion channel using an automated patch clamp system. With the automated system, we could patch not only giant unilamellar liposomes but also giant Escherichia coli (E. coli) spheroplasts. The tension sensitivity and channel kinetics data obtained in the automated system were in good agreement with that obtained from the conventional patch clamp. The findings will pave the way to high throughput fundamental and drug screening studies on mechanosensitive ion channels.


Asunto(s)
Automatización de Laboratorios/métodos , Proteínas de Escherichia coli/metabolismo , Canales Iónicos/metabolismo , Técnicas de Placa-Clamp/métodos , Escherichia coli/metabolismo , Esferoplastos/metabolismo
19.
Handb Exp Pharmacol ; 222: 659-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24756725

RESUMEN

TRPML3 belongs to the MCOLN (TRPML) subfamily of transient receptor potential (TRP) channels comprising three genes in mammals. Since the discovery of the pain sensing, capsaicin- and heat-activated vanilloid receptor (TRPV1), TRP channels have been found to be involved in regulating almost all kinds of our sensory modalities. Thus, TRP channel members are sensitive to heat or cold; they are involved in pain or osmosensation, vision, hearing, or taste sensation. Loss or mutation of TRPML1 can cause retina degeneration and eventually blindness in mice and men (mucolipidosis type IV). Gain-of-function mutations in TRPML3 cause deafness and circling behavior in mice. A special feature of TRPML channels is their intracellular expression. They mostly reside in membranes of organelles of the endolysosomal system such as early and late endosomes, recycling endosomes, lysosomes, or lysosome-related organelles. Although the physiological roles of TRPML channels within the endolysosomal system are far from being fully understood, it is speculated that they are involved in the regulation of endolysosomal pH, fusion/fission processes, trafficking, autophagy, and/or (hormone) secretion and exocytosis.


Asunto(s)
Canales de Potencial de Receptor Transitorio/metabolismo , Secuencia de Aminoácidos , Animales , Permeabilidad de la Membrana Celular , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Activación del Canal Iónico , Potenciales de la Membrana , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fenotipo , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/deficiencia , Canales de Potencial de Receptor Transitorio/genética
20.
J Clin Invest ; 134(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38557489

RESUMEN

Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.


Asunto(s)
Canales de Calcio , Calcio , Ratones , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Calcio/metabolismo , Páncreas/metabolismo , Exocitosis/fisiología , Vesículas Secretoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA