Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Structure ; 28(2): 169-184.e5, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31806353

RESUMEN

Polycystin-2 (PC2) is a transient receptor potential (TRP) channel present in ciliary membranes of the kidney. PC2 shares a transmembrane fold with other TRP channels, in addition to an extracellular domain found in TRPP and TRPML channels. Using molecular dynamics (MD) simulations and cryoelectron microscopy we identify and characterize PIP2 and cholesterol interactions with PC2. PC2 is revealed to have a PIP binding site close to the equivalent vanilloid/lipid binding site in the TRPV1 channel. A 3.0-Å structure reveals a binding site for cholesterol on PC2. Cholesterol interactions with the channel at this site are characterized by MD simulations. The two classes of lipid binding sites are compared with sites observed in other TRPs and in Kv channels. These findings suggest PC2, in common with other ion channels, may be modulated by both PIPs and cholesterol, and position PC2 within an emerging model of the roles of lipids in the regulation and organization of ciliary membranes.


Asunto(s)
Colesterol/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/metabolismo , Animales , Sitios de Unión , Microscopía por Crioelectrón , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Estructura Secundaria de Proteína , Células Sf9
2.
J Gen Physiol ; 149(8): 799-811, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28720589

RESUMEN

G protein-gated inwardly rectifying potassium (GIRK) channels control neuronal excitability in the brain and are implicated in several different neurological diseases. The anionic phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) is an essential cofactor for GIRK channel gating, but the precise mechanism by which PIP2 opens GIRK channels remains poorly understood. Previous structural studies have revealed several highly conserved, positively charged residues in the "tether helix" (C-linker) that interact with the negatively charged PIP2 However, these crystal structures of neuronal GIRK channels in complex with PIP2 provide only snapshots of PIP2's interaction with the channel and thus lack details about the gating transitions triggered by PIP2 binding. Here, our functional studies reveal that one of these conserved basic residues in GIRK2, Lys200 (6'K), supports a complex and dynamic interaction with PIP2 When Lys200 is mutated to an uncharged amino acid, it activates the channel by enhancing the interaction with PIP2 Atomistic molecular dynamic simulations of neuronal GIRK2 with the same 6' substitution reveal an open GIRK2 channel with PIP2 molecules adopting novel positions. This dynamic interaction with PIP2 may explain the intrinsic low open probability of GIRK channels and the mechanism underlying activation by G protein Gßγ subunits and ethanol.

3.
Channels (Austin) ; 11(5): 482-486, 2017 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-28723241

RESUMEN

The TREK-2 (KCNK10) K2P potassium channel can be regulated by variety of polymodal stimuli including pressure. In a recent study, we demonstrated that this mechanosensitive K+ channel responds to changes in membrane tension by undergoing a major structural change from its 'down' state to the more expanded 'up' state conformation. These changes are mostly restricted to the lower part of the protein within the bilayer, but are allosterically coupled to the primary gating mechanism located within the selectivity filter. However, any such structural changes within the filter also have the potential to alter ionic selectivity and there are reports that some K2Ps, including TREK channels, exhibit a dynamic ionic selectivity. In this addendum to our previous study we have therefore examined whether the selectivity of TREK-2 is altered by stretch activation. Our results reveal that the filter remains stable and highly selective for K+ over Na+ during stretch activation, and that permeability to a range of other cations (Rb+, Cs+ and NH4+) also does not change. The asymmetric structural changes that occur during stretch activation therefore allow the channel to respond to changes in membrane tension without a loss of K+ selectivity.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/metabolismo , Sitios de Unión , Línea Celular , Humanos , Activación del Canal Iónico , Iones , Simulación de Dinámica Molecular , Unión Proteica , Estrés Mecánico
4.
Structure ; 25(5): 708-718.e2, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28392258

RESUMEN

The mechanosensitive two-pore domain (K2P) K+ channels (TREK-1, TREK-2, and TRAAK) are important for mechanical and thermal nociception. However, the mechanisms underlying their gating by membrane stretch remain controversial. Here we use molecular dynamics simulations to examine their behavior in a lipid bilayer. We show that TREK-2 moves from the "down" to "up" conformation in direct response to membrane stretch, and examine the role of the transmembrane pressure profile in this process. Furthermore, we show how state-dependent interactions with lipids affect the movement of TREK-2, and how stretch influences both the inner pore and selectivity filter. Finally, we present functional studies that demonstrate why direct pore block by lipid tails does not represent the principal mechanism of mechanogating. Overall, this study provides a dynamic structural insight into K2P channel mechanosensitivity and illustrates how the structure of a eukaryotic mechanosensitive ion channel responds to changes in forces within the bilayer.


Asunto(s)
Activación del Canal Iónico , Canales de Potasio de Dominio Poro en Tándem/química , Humanos , Membrana Dobles de Lípidos/química , Mecanotransducción Celular , Canales de Potasio de Dominio Poro en Tándem/metabolismo
5.
Structure ; 24(12): 2207-2216, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27866853

RESUMEN

Ion channels play key roles in cell membranes, and recent advances are yielding an increasing number of structures. However, their functional relevance is often unclear and better tools are required for their functional annotation. In sub-nanometer pores such as ion channels, hydrophobic gating has been shown to promote dewetting to produce a functionally closed (i.e., non-conductive) state. Using the serotonin receptor (5-HT3R) structure as an example, we demonstrate the use of molecular dynamics to aid the functional annotation of channel structures via simulation of the behavior of water within the pore. Three increasingly complex simulation analyses are described: water equilibrium densities; single-ion free-energy profiles; and computational electrophysiology. All three approaches correctly predict the 5-HT3R crystal structure to represent a functionally closed (i.e., non-conductive) state. We also illustrate the application of water equilibrium density simulations to annotate different conformational states of a glycine receptor.


Asunto(s)
Canales Iónicos/química , Receptores de Serotonina 5-HT3/química , Agua/química , Membrana Celular/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Simulación de Dinámica Molecular , Conformación Proteica
6.
J Gen Physiol ; 147(6): 497-505, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27241700

RESUMEN

The TREK subfamily of two-pore domain (K2P) K(+) channels exhibit polymodal gating by a wide range of physical and chemical stimuli. Crystal structures now exist for these channels in two main states referred to as the "up" and "down" conformations. However, recent studies have resulted in contradictory and mutually exclusive conclusions about the functional (i.e., conductive) status of these two conformations. To address this problem, we have used the state-dependent TREK-2 inhibitor norfluoxetine that can only bind to the down state, thereby allowing us to distinguish between these two conformations when activated by different stimuli. Our results reconcile these previously contradictory gating models by demonstrating that activation by pressure, temperature, voltage, and pH produce more than one structurally distinct open state and reveal that channel activation does not simply involve switching between the up and down conformations. These results also highlight the diversity of structural mechanisms that K2P channels use to integrate polymodal gating signals.


Asunto(s)
Activación del Canal Iónico , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Animales , Humanos , Canales de Potasio de Dominio Poro en Tándem/química , Xenopus
7.
Biochem Soc Trans ; 43(2): 146-50, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25849908

RESUMEN

Gating in channels and nanopores plays a key role in regulating flow of ions across membranes. Molecular simulations provide a 'computational microscope' which enables us to examine the physical nature of gating mechanisms at the level of the single channel molecule. Water enclosed within the confines of a nanoscale pore may exhibit unexpected behaviour. In particular, if the molecular surfaces lining the pore are hydrophobic this promotes de-wetting of the pore. De-wetting is observed as stochastic liquid-vapour transitions within a pore, and may lead to functional closure of a pore to the flow of ions and/or water. Such behaviour was first observed in simulations of simple model nanopores and referred to as 'hydrophobic gating'. Simulations of both the nicotinic acetylcholine receptor and of TWIK-1 potassium channels (the latter alongside experimental studies) suggest hydrophobic gating may occur in some biological ion channels. Current studies are focused on designing hydrophobic gates into biomimetic nanopores.


Asunto(s)
Biomimética , Activación del Canal Iónico , Canales Iónicos/química , Nanoporos , Membrana Celular/química , Membrana Celular/metabolismo , Simulación por Computador , Interacciones Hidrofóbicas e Hidrofílicas , Canales Iónicos/metabolismo , Proteínas de Transporte de Membrana/química , Modelos Moleculares , Nanotecnología , Canales de Potasio de Dominio Poro en Tándem/química , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Agua/química
8.
Science ; 347(6227): 1256-9, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25766236

RESUMEN

TREK-2 (KCNK10/K2P10), a two-pore domain potassium (K2P) channel, is gated by multiple stimuli such as stretch, fatty acids, and pH and by several drugs. However, the mechanisms that control channel gating are unclear. Here we present crystal structures of the human TREK-2 channel (up to 3.4 angstrom resolution) in two conformations and in complex with norfluoxetine, the active metabolite of fluoxetine (Prozac) and a state-dependent blocker of TREK channels. Norfluoxetine binds within intramembrane fenestrations found in only one of these two conformations. Channel activation by arachidonic acid and mechanical stretch involves conversion between these states through movement of the pore-lining helices. These results provide an explanation for TREK channel mechanosensitivity, regulation by diverse stimuli, and possible off-target effects of the serotonin reuptake inhibitor Prozac.


Asunto(s)
Activación del Canal Iónico , Canales de Potasio de Dominio Poro en Tándem/química , Secuencia de Aminoácidos , Ácido Araquidónico/farmacología , Sitios de Unión , Cristalografía por Rayos X , Fluoxetina/análogos & derivados , Fluoxetina/química , Fluoxetina/metabolismo , Fluoxetina/farmacología , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Conformación Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
9.
Channels (Austin) ; 9(1): 44-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25487004

RESUMEN

Several recent ion channel structures have revealed large side portals, or 'fenestrations' at the interface between their transmembrane helices that potentially expose the ion conduction pathway to the lipid core of the bilayer. In a recent study we demonstrated that functional activity of the TWIK-1 K2P channel is influenced by the presence of hydrophobic residues deep within the inner pore. These residues are located near the fenestrations in the TWIK-1 structure and promote dewetting of the pore by forming a hydrophobic barrier to ion conduction. During our previous MD simulations, lipid tails were observed to enter these fenestrations. In this addendum to that study, we investigate lipid contribution to the dewetting process. Our results demonstrate that lipid tails from both the upper and lower leaflets can occupy the fenestrations and partially penetrate into the pore. The lipid tails do not sterically occlude the pore, but there is an inverse correlation between the presence of water within the hydrophobic barrier and the number of lipids tails within the lining of the pore. However, dewetting still occurs in the absence of lipids tails, and pore hydration appears to be determined primarily by those side-chains lining the narrowest part of the pore cavity.


Asunto(s)
Lípidos/química , Canales de Potasio de Dominio Poro en Tándem/química , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Conformación Proteica
10.
J Mol Biol ; 427(1): 121-30, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25106689

RESUMEN

Biological ion channels are nanoscale transmembrane pores. When water and ions are enclosed within the narrow confines of a sub-nanometer hydrophobic pore, they exhibit behavior not evident from macroscopic descriptions. At this nanoscopic level, the unfavorable interaction between the lining of a hydrophobic pore and water may lead to stochastic liquid-vapor transitions. These transient vapor states are "dewetted", i.e. effectively devoid of water molecules within all or part of the pore, thus leading to an energetic barrier to ion conduction. This process, termed "hydrophobic gating", was first observed in molecular dynamics simulations of model nanopores, where the principles underlying hydrophobic gating (i.e., changes in diameter, polarity, or transmembrane voltage) have now been extensively validated. Computational, structural, and functional studies now indicate that biological ion channels may also exploit hydrophobic gating to regulate ion flow within their pores. Here we review the evidence for this process and propose that this unusual behavior of water represents an increasingly important element in understanding the relationship between ion channel structure and function.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Animales , Humanos , Modelos Moleculares
11.
Nat Commun ; 5: 4377, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-25001086

RESUMEN

Recent X-ray crystal structures of the two-pore domain (K2P) family of potassium channels have revealed a unique structural architecture at the point where the cytoplasmic bundle-crossing gate is found in most other tetrameric K(+) channels. However, despite the apparently open nature of the inner pore in the TWIK-1 (K2P1/KCNK1) crystal structure, the reasons underlying its low levels of functional activity remain unclear. In this study, we use a combination of molecular dynamics simulations and functional validation to demonstrate that TWIK-1 possesses a hydrophobic barrier deep within the inner pore, and that stochastic dewetting of this hydrophobic constriction acts as a major barrier to ion conduction. These results not only provide an important insight into the mechanisms which control TWIK-1 channel activity, but also have important implications for our understanding of how ion permeation may be controlled in similar ion channels and pores.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/química , Animales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Conformación Proteica , Agua , Xenopus
12.
J Neurosci ; 32(6): 2086-99, 2012 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-22323721

RESUMEN

We studied N-(2-aminoethyl)-N-(4-(benzyloxy)-3-methoxybenzyl)thiophene-2-carboxamide hydrochloride (M8-B), a selective and potent antagonist of the transient receptor potential melastatin-8 (TRPM8) channel. In vitro, M8-B blocked cold-induced and TRPM8-agonist-induced activation of rat, human, and murine TRPM8 channels, including those on primary sensory neurons. In vivo, M8-B decreased deep body temperature (T(b)) in Trpm8(+/+) mice and rats, but not in Trpm8(-/-) mice, thus suggesting an on-target action. Intravenous administration of M8-B was more effective in decreasing T(b) in rats than intrathecal or intracerebroventricular administration, indicating a peripheral action. M8-B attenuated cold-induced c-Fos expression in the lateral parabrachial nucleus, thus indicating a site of action within the cutaneous cooling neural pathway to thermoeffectors, presumably on sensory neurons. A low intravenous dose of M8-B did not affect T(b) at either a constantly high or a constantly low ambient temperature (T(a)), but the same dose readily decreased T(b) if rats were kept at a high T(a) during the M8-B infusion and transferred to a low T(a) immediately thereafter. These data suggest that both a successful delivery of M8-B to the skin (high cutaneous perfusion) and the activation of cutaneous TRPM8 channels (by cold) are required for the hypothermic action of M8-B. At tail-skin temperatures <23°C, the magnitude of the M8-B-induced decrease in T(b) was inversely related to skin temperature, thus suggesting that M8-B blocks thermal (cold) activation of TRPM8. M8-B affected all thermoeffectors studied (thermopreferendum, tail-skin vasoconstriction, and brown fat thermogenesis), thus suggesting that TRPM8 is a universal cold receptor in the thermoregulation system.


Asunto(s)
Temperatura Corporal/fisiología , Frío , Ganglios Espinales/fisiología , Tiritona/fisiología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/deficiencia , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/fisiología , Conducta Animal/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Ganglios Espinales/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Preparaciones Farmacéuticas/administración & dosificación , Ratas , Ratas Wistar , Tiritona/efectos de los fármacos , Tiofenos/farmacología
13.
Proc Natl Acad Sci U S A ; 107(23): 10702-7, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20498043

RESUMEN

Emerging evidence suggests that the neurotransmitter acetylcholine (ACh) negatively regulates the development of the neuromuscular junction, but it is not clear if ACh exerts its effects exclusively through muscle ACh receptors (AChRs). Here, we used genetic methods to remove AChRs selectively from muscle. Similar to the effects of blocking ACh biosynthesis, eliminating postsynaptic AChRs increased motor axon branching and expanded innervation territory, suggesting that ACh negatively regulates synaptic growth through postsynaptic AChRs. However, in contrast to the effects of blocking ACh biosynthesis, eliminating postsynaptic AChRs in agrin-deficient mice failed to restore deficits in pre- and postsynaptic differentiation, suggesting that ACh negatively regulates synaptic differentiation through nonpostsynaptic receptors. Consistent with this idea, the ACh agonist carbachol inhibited presynaptic specialization of motorneurons in vitro. Together, these data suggest that ACh negatively regulates axon growth and presynaptic specialization at the neuromuscular junction through distinct cellular mechanisms.


Asunto(s)
Acetilcolina/metabolismo , Unión Neuromuscular/metabolismo , Acetilación , Acetilcolina/agonistas , Animales , Carbacol/farmacología , Diferenciación Celular , Agonistas Colinérgicos/farmacología , Ratones , Unión Neuromuscular/citología , Unión Neuromuscular/efectos de los fármacos , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo
14.
Nat Neurosci ; 12(8): 988-95, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19561601

RESUMEN

Ethanol modifies neural activity in the brain by modulating ion channels. Ethanol activates G protein-gated inwardly rectifying K(+) channels, but the molecular mechanism is not well understood. Here, we used a crystal structure of a mouse inward rectifier containing a bound alcohol and structure-based mutagenesis to probe a putative alcohol-binding pocket located in the cytoplasmic domains of GIRK channels. Substitutions with bulkier side-chains in the alcohol-binding pocket reduced or eliminated activation by alcohols. By contrast, alcohols inhibited constitutively open channels, such as IRK1 or GIRK2 engineered to strongly bind PIP(2). Mutations in the hydrophobic alcohol-binding pocket of these channels had no effect on alcohol-dependent inhibition, suggesting an alternate site is involved in inhibition. Comparison of high-resolution structures of inwardly rectifying K(+) channels suggests a model for activation of GIRK channels using this hydrophobic alcohol-binding pocket. These results provide a tool for developing therapeutic compounds that could mitigate the effects of alcohol.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/efectos de los fármacos , Animales , Sitios de Unión/fisiología , Bovinos , Línea Celular , Cristalografía por Rayos X , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/química , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Humanos , Ratones , Modelos Moleculares , Mutagénesis , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Ratas
15.
J Physiol ; 580(Pt 1): 51-65, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17185339

RESUMEN

Many neurotransmitters and hormones signal by stimulating G protein-coupled neurotransmitter receptors (GPCRs), which activate G proteins and their downstream effectors. Whether these signalling proteins diffuse freely within the plasma membrane is not well understood. Recent studies have suggested that direct protein-protein interactions exist between GPCRs, G proteins and G protein-gated inwardly rectifying potassium (GIRK or Kir3) channels. Here, we used fluorescence resonance energy transfer (FRET) combined with total internal reflection fluorescence microscopy to investigate whether proteins within this signalling pathway move within 100 A of each other in the plasma membrane of living cells. GABA(B) R1 and R2 receptors, Kir3 channels, Galphao subunits and regulators of G protein signalling (RGS4) proteins were each fused to cyan fluorescent protein (CFP) or yellow fluorescent protein (YFP) and first assessed for functional expression in HEK293 cells. The presence of the fluorophore did not significantly alter the signalling properties of these proteins. Possible FRET was then investigated for different protein pair combinations. As a positive control, FRET was measured between tagged GABA(B) R1 and R2 subunits ( approximately 12% FRET), which are known to form heterodimers. We measured significant FRET between tagged RGS4 and GABA(B) R1 or R2 subunits ( approximately 13% FRET), and between Galphao and GABA(B) R1 or R2 subunits ( approximately 10% FRET). Surprisingly, FRET also occurred between tagged Kir3.2a/Kir3.4 channels and GABA(B) R1 or R2 subunits ( approximately 10% FRET). FRET was not detected between Kir3.2a and RGS4 nor between Kir3.2a and Galphao. These data are discussed in terms of a model in which GABA(B) receptors, G proteins, RGS4 proteins and Kir3 channels are closely associated in a signalling complex.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Proteínas RGS/fisiología , Receptores de GABA-B/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/ultraestructura , AMP Cíclico/metabolismo , Electrofisiología , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Humanos , Interpretación de Imagen Asistida por Computador , Modelos Moleculares , Técnicas de Placa-Clamp , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Técnicas de Cultivo de Tejidos , Transfección
16.
Neuron ; 46(4): 569-79, 2005 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-15944126

RESUMEN

Synapse formation requires interactions between pre- and postsynaptic cells to establish the connection of a presynaptic nerve terminal with the neurotransmitter receptor-rich postsynaptic apparatus. At developing vertebrate neuromuscular junctions, acetylcholine receptor (AChR) clusters of nascent postsynaptic apparatus are not apposed by presynaptic nerve terminals. Two opposing activities subsequently promote the formation of synapses: positive signals stabilize the innervated AChR clusters, whereas negative signals disperse those that are not innervated. Although the nerve-derived protein agrin has been suggested to be a positive signal, the negative signals remain elusive. Here, we show that cyclin-dependent kinase 5 (Cdk5) is activated by ACh agonists and is required for the ACh agonist-induced dispersion of the AChR clusters that have not been stabilized by agrin. Genetic elimination of Cdk5 or blocking ACh production prevents the dispersion of AChR clusters in agrin mutants. Therefore, we propose that ACh negatively regulates neuromuscular synapse formation through a Cdk5-dependent mechanism.


Asunto(s)
Acetilcolina/fisiología , Quinasas Ciclina-Dependientes/metabolismo , Inhibición Neural/fisiología , Unión Neuromuscular/fisiología , Agregación de Receptores/fisiología , Receptores Colinérgicos/fisiología , Agrina/deficiencia , Agrina/farmacología , Animales , Western Blotting/métodos , Bungarotoxinas/farmacocinética , Carbacol/farmacología , Carbocianinas/farmacocinética , Línea Celular , Colina O-Acetiltransferasa/deficiencia , Agonistas Colinérgicos/farmacología , Quinasa 5 Dependiente de la Ciclina , Diafragma/citología , Interacciones Farmacológicas , Embrión de Mamíferos , Femenino , Proteínas de Homeodominio , Inmunohistoquímica/métodos , Inmunoprecipitación , Hibridación in Situ/métodos , Ratones , Ratones Noqueados , Muscarina/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Liso/efectos de los fármacos , Músculo Liso/embriología , Inhibición Neural/efectos de los fármacos , Embarazo , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Agregación de Receptores/efectos de los fármacos , Roscovitina , Sinaptofisina/metabolismo , Factores de Tiempo , Factores de Transcripción/deficiencia
17.
J Biol Chem ; 280(1): 669-76, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15492008

RESUMEN

The mitogen-activated protein kinases (MAPK) have been the subject of many studies to identify signaling pathways that promote cell survival or death. In cultured cardiac myocytes, p38 MAPK promotes cell survival or death depending on whether it is activated by mitogen-activated protein kinase kinase 6 (MKK6) or MKK3, respectively. The objectives of the current study were to examine the effects of MKK6-mediated p38 activation in the heart in vivo. Accordingly, we generated transgenic (TG) mice that overexpress wild type MKK6 in a cardiac-restricted manner. Although p38 was about 17-fold more active in TG than non-transgenic (NTG) mouse hearts, TG mouse hearts were morphologically and functionally similar to those of NTG littermates. However, upon transient ischemia followed by reperfusion, the MKK6 TG mouse hearts exhibited significantly better functional recovery and less injury than NTG mouse hearts. Because MKK6 increases levels of the protective small heat shock protein, alpha B-crystallin (alpha BC), in cultured cardiac myocytes, we examined alpha BC levels in the mouse hearts. The level of alpha BC was 2-fold higher in MKK6 TG than NTG mouse hearts. Moreover, ischemia followed by reperfusion induced a 6.4-fold increase in alpha BC levels in the mitochondrial fractions of TG mouse hearts but no increase in alpha BC levels in any of the other fractions analyzed. These alterations in alpha BC expression and localization suggest possible mechanisms of cardioprotection in MKK6 TG mouse hearts.


Asunto(s)
MAP Quinasa Quinasa 6/biosíntesis , Infarto del Miocardio/enzimología , Daño por Reperfusión Miocárdica/enzimología , Animales , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Humanos , Técnicas In Vitro , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , MAP Quinasa Quinasa 6/genética , Ratones , Ratones Transgénicos , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/fisiopatología , Especificidad de Órganos , Recuperación de la Función , Cadena B de alfa-Cristalina/metabolismo
18.
Traffic ; 5(10): 798-813, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15355515

RESUMEN

Myosin-VI has been implicated in endocytic trafficking at both the clathrin-coated and uncoated vesicle stages. The identification of alternative splice forms led to the suggestion that splicing defines the vesicle type to which myosin-VI is recruited. In contrast to this hypothesis, we find that in all cell types examined, myosin-VI is associated with uncoated endocytic vesicles, regardless of splice form. GIPC, a PDZ-domain containing adapter protein, co-assembles with myosin-VI on these vesicles. Myosin-VI is only recruited to clathrin-coated vesicles in cells that express high levels of Dab2, a clathrin-binding adapter protein. Overexpression of Dab2 is sufficient to reroute myosin-VI to clathrin-coated pits in cells where myosin-VI is normally associated with uncoated vesicles. In normal rat kidney (NRK) cells, which express high endogenous levels of Dab2, splicing of the globular tail domain further modulates targeting of ectopically expressed myosin-VI. Although myosin-VI can be recruited to clathrin-coated pits, we find no requirement for myosin-VI motor activity in endocytosis in NRK cells. Instead, our data suggest that myosin-VI recruitment to clathrin-coated pits may be an early step in the recruitment of GIPC to the vesicle surface.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Empalme Alternativo/fisiología , Cadenas Pesadas de Miosina/metabolismo , Vesículas Transportadoras/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Empalme Alternativo/genética , Animales , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular , Chlorocebus aethiops , Cartilla de ADN , ADN Complementario/genética , Proteínas Fluorescentes Verdes , Humanos , Immunoblotting , Inmunohistoquímica , Microscopía Fluorescente , Cadenas Pesadas de Miosina/genética , Neuropéptidos/metabolismo , Transporte de Proteínas/fisiología , Ratas , Porcinos
19.
Am J Physiol Heart Circ Physiol ; 284(2): H575-83, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12414446

RESUMEN

Familial hypertrophic cardiomyopathy (HCM) is an autosomal dominant disease characterized by varying degrees of ventricular hypertrophy and myofibrillar disarray. Mutations in cardiac contractile proteins cause HCM. However, there is an unexplained wide variability in the clinical phenotype, and it is likely that there are multiple contributing factors. Because mitochondrial dysfunction has been described in heart disease, we tested the hypothesis that mitochondrial dysfunction contributes to the varying HCM phenotypes. Mitochondrial function was assessed in two transgenic models of HCM: mice with a mutant myosin heavy chain gene (MyHC) or with a mutant cardiac troponin T (R92Q) gene. Despite mitochondrial ultrastructural abnormalities in both models, the rate of state 3 respiration was significantly decreased only in the mutant MyHC mice by approximately 23%. Notably, this decrease in state 3 respiration preceded hemodynamic dysfunction. The maximum activity of alpha-ketogutarate dehydrogenase as assayed in isolated disrupted mitochondria was decreased by 28% compared with isolated control mitochondria. In addition, complexes I and IV were decreased in mutant MyHC transgenic mice. Inhibition of beta-adrenergic receptor kinase, which is elevated in mutant MyHC mouse hearts, can prevent mitochondrial respiratory impairment in mutant MyHC mice. Thus our results suggest that mitochondria may contribute to the hemodynamic dysfunction seen in some forms of HCM and offer a plausible mechanism responsible for some of the heterogeneity of the disease phenotypes.


Asunto(s)
Cardiomiopatía Hipertrófica/fisiopatología , Mitocondrias Cardíacas/fisiología , Animales , Cardiomiopatía Hipertrófica/patología , Complejo I de Transporte de Electrón , Complejo IV de Transporte de Electrones/metabolismo , Hemodinámica , Hibridación Genética , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos/genética , Mitocondrias Cardíacas/ultraestructura , Cadenas Pesadas de Miosina/genética , NADH NADPH Oxidorreductasas/metabolismo , Consumo de Oxígeno , Troponina T/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA