首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Kir5.1 is an inwardly rectifying K+ channel subunit whose functional role has not been fully elucidated. Expression and distribution of Kir5.1 in retina were examined with a specific polyclonal antibody. Kir5.1 immunoreactivity was detected in glial Müller cells and in some retinal neurons. In the Kir5.1-positive neurons the expression of glutamic acid decarboxylase (GAD65) was detected, suggesting that they may be GABAergic-amacrine cells. In Müller cells, spots of Kir5.1 immunoreactivity distributed diffusely at the cell body and in the distal portions, where Kir4.1 immunoreactivity largely overlapped. In addition, Kir4.1 immunoreactivity without Kir5.1 was strongly concentrated at the endfoot of Müller cells facing the vitreous surface or in the processes surrounding vessels. The immunoprecipitant obtained from retina with anti-Kir4.1 antibody contained Kir5.1. These results suggest that heterotetrameric Kir4.1/Kir5.1 channels may exist in the cell body and distal portion of Müller cells, whereas homomeric Kir4.1 channels are clustered in the endfeet and surrounding vessels. It is possible that homomeric Kir4.1 and heteromeric Kir4.1/Kir5.1 channels play different functional roles in the K+-buffering action of Müller cells. inwardly rectifying potassium channel; heteromerization; glial Müller cells; amacrine cells; potassium siphoning  相似文献   

2.
Extracellular potassium concentration, [K+]o, and intracellular calcium, [Ca2+]i, rise during neuron excitation, seizures and spreading depression. Astrocytes probably restrain the rise of K+ in a way that is only partly understood. To examine the effect of glial K+ uptake, we used a model neuron equipped with Na+, K+, Ca2+ and Cl conductances, ion pumps and ion exchangers, surrounded by interstitial space and glia. The glial membrane was either “passive”, incorporating only leak channels and an ion exchange pump, or it had rectifying K+ channels. We computed ion fluxes, concentration changes and osmotic volume changes. Increase of [K+]o stimulated the glial uptake by the glial 3Na/2K ion pump. The [K+]o flux through glial leak and rectifier channels was outward as long as the driving potential was outwardly directed, but it turned inward when rising [K+]o/[K+]i ratio reversed the driving potential. Adjustments of glial membrane parameters influenced the neuronal firing patterns, the length of paroxysmal afterdischarge and the ignition point of spreading depression. We conclude that voltage gated K+ currents can boost the effectiveness of the glial “potassium buffer” and that this buffer function is important even at moderate or low levels of excitation, but especially so in pathological states.  相似文献   

3.
The maintenance of cellular ion homeostasis is crucial for optimal neural function and thus it is of great importance to understand its regulation. Glial cells are extensively coupled by gap junctions forming a network that is suggested to serve as a spatial buffer for potassium (K+) ions. We have investigated the role of glial spatial buffering in the regulation of extracellular K+ concentration ([K+]o) within the locust metathoracic ganglion by pharmacologically inhibiting gap junctions. Using K+-sensitive microelectrodes, we measured [K+]o near the ventilatory neuropile while simultaneously recording the ventilatory rhythm as a model of neural circuit function. We found that blockade of gap junctions with either carbenoxolone (CBX), 18β-glycyrrhetinic acid (18β-GA) or meclofenamic acid (MFA) reliably induced repetitive [K+]o surges and caused a progressive impairment in the ability to maintain baseline [K+]o levels throughout the treatment period. We also show that a low dose of CBX that did not induce surging activity increased the vulnerability of locust neural tissue to spreading depression (SD) induced by Na+/K+-ATPase inhibition with ouabain. CBX pre-treatment increased the number of SD events induced by ouabain and hindered the recovery of [K+]o back to baseline levels between events. Our results suggest that glial spatial buffering through gap junctions plays an essential role in the regulation of [K+]o under normal conditions and also contributes to a component of [K+]o clearance following physiologically elevated levels of [K+]o.  相似文献   

4.
Normal vision depends on the correct function of retinal neurons and glia and it is impaired in the course of diabetic retinopathy. Müller cells, the main glial cells of the retina, suffer morphological and functional alterations during diabetes participating in the pathological retinal dysfunction. Recently, we showed that Müller cells express the pleiotropic protein potassium channel interacting protein 3 (KChIP3), an integral component of the voltage-gated K+ channels KV4. Here, we sought to analyze the role of KChIP3 in the molecular mechanisms underlying hyperglycemia-induced phenotypic changes in the glial elements of the retina. The expression and function of KChIp3 was analyzed in vitro in rat Müller primary cultures grown under control (5.6 mM) or high glucose (25 mM) (diabetic-like) conditions. We show the up-regulation of KChIP3 expression in Müller cell cultures under high glucose conditions and demonstrate a previously unknown interaction between the KV4 channel and KChIP3 in Müller cells. We show evidence for the expression of a 4-AP-sensitive transient outward voltage-gated K+ current and an alteration in the inactivation of the macroscopic outward K+ currents expressed in high glucose-cultured Müller cells. Our data support the notion that induction of KChIP3 and functional changes of KV4 channels in Müller cells could exert a physiological role in the onset of diabetic retinopathy.  相似文献   

5.
The inwardly rectifying potassium channel (Kir), Kir4.1 mediates spatial K+-buffering in the CNS. In this process the channel is potentially exposed to a large range of extracellular K+ concentrations ([K+]o). We found that Kir4.1 is regulated by K+o. Increased [K+]o leads to a slow (mins) increase in the whole-cell currents of Xenopus oocytes expressing Kir4.1. Conversely, removing K+ from the bath solution results in a slow decrease of the currents. This regulation is not coupled to the pHi-sensitive gate of the channel, nor does it require the presence of K67, a residue necessary for K+o-dependent regulation of Kir1.1. The voltage-dependent blockers Cs+ and Ba2+ substitute for K+ and prevent deactivation of the channel in the absence of K+o. Cs+ blocks and regulates the channel with similar affinity, consistent with the regulatory sites being in the selectivity-filter of the channel. Although both Rb+ and NH4+ permeate Kir4.1, only Rb+ is able to regulate the channel. We conclude that Kir4.1 is regulated by ions interacting with specific sites in the selectivity filter. Using a kinetic model of the permeation process we show the plausibility of the channel’s sensing the extracellular ionic environment through changes in the selectivity occupancy pattern, and that it is feasible for an ion with the selectivity properties of NH4+ to permeate the channel without inducing these changes.  相似文献   

6.
In a career that has spanned 45 years and shows no signs of slowing down, Dr Bruce Ransom has devoted considerable time and energy to studying regulation of interstitial K+. When Bruce commenced his studies in 1969 virtually nothing was known of the functions of glial cells, but Bruce’s research contributed to the physiological assignation of function to mammalian astrocytes, namely interstitial K+ buffering. The experiments that I describe in this review concern the response of the membrane potential (Em) of in vivo cat cortical astrocytes to changes in [K+]o, an experimental manoeuvre that was achieved in two different ways. The first involved recording the Em of an astrocyte while the initial aCSF was switched to one with different K+, whereas in the second series of experiments the cortex was stimulated and the response of the astrocyte Em to the K+ released from neighbouring neurons was recorded. The astrocytes responded in a qualitatively predictable manner, but quantitatively the changes were not as predicted by the Nernst equation. Elevations in interstitial K+ are not sustained and K+ returns to baseline rapidly due to the buffering capacity of astrocytes, a phenomenon studied by Bruce, and his son Chris, published 27 years after Bruce’s initial publications. Thus, a lifetime spent investigating K+ buffering has seen enormous advances in glial research, from the time cells were identified as ‘presumed’ glial cells or ‘silent cells’, to the present day, where glial cells are recognised as contributing to every important physiological brain function.  相似文献   

7.
Neuronal activity results in release of K+ into the extracellular space of the central nervous system. If the excess K+ is allowed to accumulate, neuronal firing will be compromised by the ensuing neuronal membrane depolarization. The surrounding glial cells are involved in clearing K+ from the extracellular space by molecular mechanism(s), the identity of which have been a matter of controversy for over half a century. Kir4.1-mediated spatial buffering of K+ has been promoted as a major contributor to K+ removal although its quantitative and temporal contribution has remained undefined. We discuss the biophysical and experimental challenges regarding determination of the contribution of Kir4.1 to extracellular K+ management during neuronal activity. It is concluded that 1) the geometry of the experimental preparation is crucial for detection of Kir4.1-mediated spatial buffering and 2) Kir4.1 enacts spatial buffering of K+ during but not after neuronal activity.  相似文献   

8.
Neuronal activity results in release of K+ into the extracellular space of the central nervous system. If the excess K+ is allowed to accumulate, neuronal firing will be compromised by the ensuing neuronal membrane depolarization. The surrounding glial cells are involved in clearing K+ from the extracellular space by molecular mechanism(s), the identity of which have been a matter of controversy for over half a century. Kir4.1-mediated spatial buffering of K+ has been promoted as a major contributor to K+ removal although its quantitative and temporal contribution has remained undefined. We discuss the biophysical and experimental challenges regarding determination of the contribution of Kir4.1 to extracellular K+ management during neuronal activity. It is concluded that 1) the geometry of the experimental preparation is crucial for detection of Kir4.1-mediated spatial buffering and 2) Kir4.1 enacts spatial buffering of K+ during but not after neuronal activity.  相似文献   

9.
The effects of changes in extracellular K+ concentration ([K+]o) on the resting membrane potential, the input resistance and 86Rb efflux (as a marker of K+ efflux) were examined with use of the cultured mouse neuroblastoma cells (N-18 clone). The results obtained are as follows. (1) The membrane potential was depolarized, with an increase in [K+]o at concentrations above 10–20 mM at a rate of 55–58 mV per 10-fold change in [K+]o, but practically unchanged with varying [K+]o below this concentration. (2) Above the critical [K+]o of 10–20 mM, the input membrane resistance decreased sharply by a factor of 14?15 with an increase in [K+]o. A similar decrease in the resistance occurred even under the conditions that the membrane potential was held at control level (about ?55 mV) by a steady-state current passage. (3) Elimination of Na+ and Cl? from the external solution brought about practically no change in the membrane potential. (4) A fractional escape rate of 86Rb from N-18 cells remained constant at relatively low level (0.125%/min on average) in the low [K+]o range, but increased sharply with increasing [K+]o above 15 mM (e.g., approx. 3.4- and 4.5-fold at 30 and 100 mM [K+]o, respectively). (5) The high K+-induced 86Rb efflux was not practically inhibited by 1 mM tetraethylammonium or 0.1 mM 4-aminopyridine, indicating that the K+ channels activated by an elevation of [K+]o are not the delayed (voltage-dependent) K+ channels. The present results favoured the conclusion that N-18 cells carry K+ channels which open at high [K+]o but are closed at low [K+]o including the physiological range for the mouse neuroblastoma cells (around 5.4 mM). This conclusion leads to the notion that in the mouse neuroblastoma N-18 cells the K+ permeability does not mainly contribute to determining the resting membrane potential under physiological conditions.  相似文献   

10.
Single inward rectifier K+ channels were studied in Xenopus laevis embryonic myocytes. We have characterized in detail the channel which is most frequently observed (Kir) although we routinely observe three other smaller current levels with the properties of inward rectifier K+ channels (Kir(0.3), Kir(0.5) and Kir(0.7)). For Kir, slope conductances of inward currents were 10.3, 20.3, and 27.9 pS, in 60, 120 and 200 mM [K+] o respectively. Extracellular Ba2+ blocked the normally high channel activity in a concentration-dependent manner (K A = 7.8 μm, −90 mV). In whole-cell recordings of inward rectifier K+ current, marked voltage dependence of Ba2+ block over the physiological range of potentials was observed. We also examined current rectification. Following step depolarizations to voltages positive to E K , outward currents through Kir channels were not observed even when the cytoplasmic face of excised patches were exposed to Mg2+-free solution at pH 9.1. This was probably also true for Kir(0.3), Kir(0.5) and Kir(0.7) channels. We then examined the possibility of modulation of Kir channel activity and found neither ATP nor GTP-γS had any effect on Kir channel activity when added to the solution perfusing the cytoplasmic face of a patch. Kinetic analysis revealed Kir channels with a single open state (mean dwell time 72 msec) and two closed states (time constants 1.4, 79 msec). These results suggest that the native Kir channels of Xenopus myocytes have similar properties to the cloned strong inward rectifier K+ channels, in terms of conductance, kinetics and barium block but does show some differences in the effects of modulators of channel activity. Furthermore, skeletal muscle may contain either different inward rectifier channels or a single-channel type which can exist in stable subconductance states. Received: 16 September 1996/Revised: 14 March 1997  相似文献   

11.
Experimental evidences point out the participation of nonsynaptic mechanisms (e.g., fluctuations in extracellular ions) in epileptiform bursting and spreading depression (SD). During these abnormal oscillatory patterns, it is observed an increase of extracellular potassium concentration [K+]o and a decrease of extracellular calcium concentration [Ca2+]o which raises the neuronal excitability. However, whether the high [K+]o triggers and propagates these abnormal neuronal activities or plays a secondary role into this process is unclear. To better understand the influence of extracellular potassium dynamics in these oscillatory patterns, the experimental conditions of high [K+]o and zero [Ca2+]o were replicated in an extended Golomb model where we added important regulatory mechanisms of ion concentration as Na+-K+ pump, ion diffusion and glial buffering. Within these conditions, simulations of the cell model exhibit seizure-like discharges (ictal bursting). The SD was elicited by the interruption of the Na+−K+ pump activity, mimicking the effect of cellular hypoxia (an experimental protocol to elicit SD, the hypoxia-induced SD). We used the bifurcation theory and the fast-slow method to analyze the interference of K+ dynamics in the cellular excitability. This analysis indicates that the system loses its stability at a high [K+]o, transiting to an elevated state of neuronal excitability. Effects of high [K+]o are observed in different stages of ictal bursting and SD. In the initial stage, the increase of [K+]o creates favorable conditions to trigger both oscillatory patterns. During the neuronal activity, a continuous growth of [K+]o by outward K+ flow depresses K+ currents in a positive feedback way. At the last stage, due to the depression of K+ currents, the Na+-K+ pump is the main mechanism in the end of neuronal activity. Thus, this work suggests that [K+]o dynamics may play a fundamental role in these abnormal oscillatory patterns.  相似文献   

12.
To explore non-synaptic mechanisms in paroxysmal discharges, we used a computer model of a simplified hippocampal pyramidal cell, surrounded by interstitial space and a “glial-endothelial” buffer system. Ion channels for Na+, K+, Ca2+ and Cl , ion antiport 3Na/Ca, and “active” ion pumps were represented in the neuron membrane. The glia had “leak” conductances and an ion pump. Fluxes, concentration changes and cell swelling were computed. The neuron was stimulated by injecting current. Afterdischarge (AD) followed stimulation if depolarization due to rising interstitial K+ concentration ([K+]o) activated persistent Na+ current (I Na,P). AD was either simple or self-regenerating; either regular (tonic) or burst-type (clonic); and always self-limiting. Self-regenerating AD required sufficient I Na,P to ensure re-excitation. Burst firing depended on activation of dendritic Ca2+ currents and Ca-dependent K+ current. Varying glial buffer function influenced [K+]o accumulation and afterdischarge duration. Variations in Na+ and K+ currents influenced the threshold and the duration of AD. The data show that high [K+]o and intrinsic membrane currents can produce the feedback of self-regenerating afterdischarges without synaptic input. The simulated discharge resembles neuron behavior during paroxysmal firing in living brain tissue. Action Editor: David Terman  相似文献   

13.
This work demonstrates that extracellular Na+ modulates the cloned inwardly rectifying K+ channels Kir4.1 and Kir4.1-Kir5.1. Whole-cell patch clamp studies on astrocytes have previously indicated that inward potassium currents are regulated by external Na+. We expressed Kir4.1 and Kir4.1-Kir5.1 in Xenopus oocytes to disclose if Kir4.1 and/or Kir4.1-Kir5.1 at the molecular level are responsible for the observed effect of [Na+]o and to investigate the regulatory mechanism of external cations further. Our results showed that Na+ has a biphasic modulatory effect on both Kir4.1 and Kir4.1-Kir5.1 currents. Depending on the Na+-concentration and applied voltage, the inward Kir4.1/Kir4.1-Kir5.1 currents are either enhanced or reduced by extracellular Na+. The Na+ activation was voltage-independent, whereas the Na+-induced reduction of the Kir4.1 and Kir4.1-Kir5.1 currents was both concentration-, time- and voltage-dependent. Our data indicate that the biphasic effect of extracellular Na+on the Kir4.1 and Kir4.1-Kir5.1 channels is caused by two separate mechanisms.  相似文献   

14.
Glial cells in the diseased nervous system undergo a process known as reactive gliosis. Gliosis of retinal Müller glial cells is characterized by an upregulation of glial fibrillary acidic protein and frequently by a reduction of inward K+ current amplitudes. Purinergic signaling is assumed to be involved in gliotic processes. As previously shown, lack of the nucleotide receptor P2Y1 leads to an altered regulation of K+ currents in Müller cells of the ischemic retina. Here, we asked first whether this effect is mediated by the IP3 receptor subtype 2 (IP3R2) known as the major downstream signaling target of P2Y1 in Müller cells. The second question was whether lack of IP3R2 affects neuronal survival in the control and ischemic retina. Ischemia was induced in wild type and IP3R2-deficient (IP 3 R2 ?/?) mice by transient elevation of the intraocular pressure. Immunostaining and TUNEL labelling were used to quantify neuronal cell loss. The downregulation of inward K+ currents in Müller cells from ischemic IP 3 R2 ?/? retinae was less strong than in wild type animals. The reduction of the number of cells in the ganglion cell layer and of calretinin- and calbindin-positive cells 7 days after ischemia was similar in wild type and IP 3 R2 ?/? mice. However, IP3R2 deficiency led to an increased number of TUNEL-positive cells in the outer nuclear layer at 1 day and to an enhanced postischemic loss of photoreceptors 7 days after ischemia. This implies that IP3R2 is involved in some but not all aspects of signaling in Müller cells after an ischemic insult.  相似文献   

15.
It has been known for more than three decades that outward Kir currents (IK1) increase with increasing extracellular K+ concentration ([K+]o). Although this increase in IK1 can have significant impacts under pathophysiological cardiac conditions, where [K+]o can be as high as 18 mm and thus predispose the heart to re-entrant ventricular arrhythmias, the underlying mechanism has remained unclear. Here, we show that the steep [K+]o dependence of Kir2.1-mediated outward IK1 was due to [K+]o-dependent inhibition of outward IK1 by extracellular Na+ and Ca2+. This could be accounted for by Na+/Ca2+ inhibition of IK1 through screening of local negative surface charges. Consistent with this, extracellular Na+ and Ca2+ reduced the outward single-channel current and did not increase open-state noise or decrease the mean open time. In addition, neutralizing negative surface charges with a carboxylate esterifying agent inhibited outward IK1 in a similar [K+]o-dependent manner as Na+/Ca2+. Site-directed mutagenesis studies identified Asp114 and Glu153 as the source of surface charges. Reducing K+ activation and surface electrostatic effects in an R148Y mutant mimicked the action of extracellular Na+ and Ca2+, suggesting that in addition to exerting a surface electrostatic effect, Na+ and Ca2+ might inhibit outward IK1 by inhibiting K+ activation. This study identified interactions of K+ with Na+ and Ca2+ that are important for the [K+]o dependence of Kir2.1-mediated outward IK1.  相似文献   

16.
In whole-cell recording, the conductance of the plasma membrane of protoplasts isolated from mesophyll cells of leaves of oat (Avena sativa) was greater for inward than outward current. The inward current in both the whole-cell mode and with isolated patches was dependent on [K+]o. When the membrane voltage was more positive than −50 millivolts, the membrane conductance in the whole-cell mode was low, and K+ channels in cell-attached or outside-out patches had a low probability of being open. At a membrane voltage more negative than −50 millivolts, the membrane conductance increased by sevenfold in the whole-cell mode, and the probability of the channels being open increased. The inward current was highly selective for K+ compared with Cs+, Na+, choline or Cl. Low concentrations of [Cs+]o or [Na+]o blocked the inward current in a strongly voltage-dependent fashion. Comparison of single-channel with the macroscopic current yields an estimate of about 200 inwardly rectifying K+ channels per cell at a density of 0.035 per square micrometer. At physiological membrane voltages and [K+]o about 10 millimolar, the influx through these channels is sufficient to increase the internal [K+] by 2 millimolar per minute. These K+ channels are activated by membrane voltages in the normal physiological range and could contribute to K+ uptake whenever the membrane is more negative than the K+ equilibrium potential.  相似文献   

17.
Patch-clamp experiments were performed on satellite glial cells wrapped around sympathetic neurons in the rabbit coeliac ganglion. With the cleaning method used, the glial cells could be kept in place and were directly accessible to the patch-clamp pipettes. Whole-cell recordings showed that glial cells had almost ohmic properties. Their resting potential (–79.1±1.2 mV) was found to be very nearly the same as the K+ reversal potential and 20 mV more negative than that of the neurons they encapsulated. Unitary currents from ionic channels present in the glial membrane were recorded in the cell-attached configuration with pipettes filled with various amounts of K+, Na+ and gluconate. Only K+-selective channels with slight inwardly rectifying properties (in the presence of 150 mM [K+]0) were detected. These channels were active (P 0=0.7–0.8) at the cell resting potential. The channel conductance, but not its opening probability, was dependent on the [K+] in the pipette. Cl-selective channels (outwardly rectifying and large conductance channels) were detected in excised patches.The properties of the K+ channels (increased inward current with [K+] and detectable outward current at low [K+]) are well suited for siphoning the K+ released by active neurons.  相似文献   

18.
Extracellular acidification and reduction of extracellular K+ are known to decrease the currents of some voltage-gated potassium channels. Although the macroscopic conductance of WT hKv1.5 channels is not very sensitive to [K+]o at pH 7.4, it is very sensitive to [K+]o at pH 6.4, and in the mutant, H463G, the removal of K+ o virtually eliminates the current at pH 7.4. We investigated the mechanism of current regulation by K+ o in the Kv1.5 H463G mutant channel at pH 7.4 and the wild-type channel at pH 6.4 by taking advantage of Na+ permeation through inactivated channels. Although the H463G currents were abolished in zero [K+]o, robust Na+ tail currents through inactivated channels were observed. The appearnnce of H463G Na+ currents with a slow rising phase on repolarization after a very brief depolarization (2 ms) suggests that channels could activate directly from closed-inactivated states. In wild-type channels, when intracellular K+ was replaced by NMG+ and the inward Na+ current was recorded, addition of 1 mM K+ prevented inactivation, but changing pH from 7.4 to 6.4 reversed this action. The data support the idea that C-type inactivation mediated at R487 in Kv1.5 channels is influenced by H463 in the outer pore. We conclude that both acidification and reduction of [K+]o inhibit Kv1.5 channels through a common mechananism (i.e., by increasing channel inactivation, which occurs in the resting state or develops very rapidly after activation).  相似文献   

19.
Glialcells express inwardly rectifying K+ (Kir) channels, whichplay a critical role in the buffering of extracellular K+.Kir4.1 is the only Kir channel so far shown to be expressed in brainglial cells. We examined the distribution of Kir4.1 in rat brain with aspecific antibody. The Kir4.1 immunostaining distributed broadly butnot diffusely in the brain. It was strong in some regions such as theglomerular layer of the olfactory bulb, the Bergmann glia in thecerebellum, the ependyma, and pia mater, while little activity wasdetected in white matter of the corpus callosum or cerebellar peduncle.In the olfactory bulb, Kir4.1 immunoreactivity was detected in ascattered manner in about one-half of the glial fibrillary acidicprotein-positive astrocytes. Immunoelectron microscopic examinationrevealed that Kir4.1 channels were enriched on the processes ofastrocytes wrapping synapses and blood vessels. These data suggest thatKir4.1 is expressed in a limited population of brain astrocytes and may play a specific role in the glial K+-buffering action.

  相似文献   

20.
Inwardly rectifying potassium (Kir) channels in Müller glia play a critical role in the spatial buffering of potassium ions that accumulate during retinal activity. To this end, Kir channels show a polarized subcellular distribution with the predominant channel subunit in Müller glia, Kir4.1, clustered in the endfeet of these cells at the inner limiting membrane. However, the molecular mechanisms underlying their distribution have yet to be identified. Here, we show that laminin, agrin and alpha-dystroglycan (DG) codistribute with Kir4.1 at the inner limiting membrane in the retina and that laminin-1 induces the clustering of alpha-DG, syntrophin and Kir4.1 in Müller cell cultures. In addition, we found that alpha-DG clusters were enriched for agrin and sought to investigate the role of agrin in their formation using recombinant C-agrins. Both C-agrin 4,8 and C-agrin 0,0 failed to induce alpha-DG clustering and neither of them potentiated the alpha-DG clustering induced by laminin-1. Finally, our data reveal that deletion of the PDZ-ligand domain of Kir4.1 prevents their laminin-induced clustering. These findings indicate that both laminin-1 and alpha-DG are involved in the distribution of Kir4.1 to specific Müller cell membrane domains and that this process occurs via a PDZ-domain-mediated interaction. Thus, in the basal lamina laminin is an essential regulator involved in clearing excess potassium released during neuronal activity, thereby contributing to the maintenance of normal synaptic transmission in the retina.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号