首页 | 本学科首页   官方微博 | 高级检索  
文章检索
  按 检索   检索词:      
出版年份:   被引次数:   他引次数: 提示:输入*表示无穷大
  收费全文   8篇
  免费   1篇
基础医学   2篇
内科学   1篇
神经病学   2篇
眼科学   3篇
药学   1篇
  2015年   3篇
  2014年   1篇
  2009年   1篇
  2007年   2篇
  2006年   1篇
  2003年   1篇
排序方式: 共有9条查询结果,搜索用时 15 毫秒
1
1.
目的在背根神经节(dorsal root ganglion,DRG)中等大小感觉神经元中可以观察到钙激活氯离子流(I_(Cl(Ca)))。在坐骨神经损伤模型中,在大多数大中神经元上诱导出类似的氯离子流。本文旨在探讨引起这个离子流的分子基础。方法使用常规的定量RT-PCR方法检测在DRG中三个基因家族的表达,这三个基因家族都具有诱导I_(Cl(Ca))的特点。结果在成年小鼠的DRG中,分别显示了在正常状态和坐骨神经损伤3天后CLCA,Bestrophin和Tweety基因家族成员的转录产物。结论mBestl和Tweety2可能在损伤诱导的DRG神经元I_(Cl(Ca))中发挥作用。  相似文献   
2.
This is to review the current state of knowledge on the functional and clinical aspects of bestrophin 1, a prominent member of a family of proteins involved in the control and properties of the light peak of the EOG. Initially human bestrophin 1 gene (BEST1) mutations were identified to underlie Best vitelliform macular dystrophy (VMD), a dominantly inherited, juvenile-onset form of macular degeneration. In the recent past the phenotypical spectrum of retinal disorders associated with BEST1 mutations has been extended and the term bestrophinopathies was coined. The physiological role of bestrophin 1 is still not completely understood but has been linked to the generation of a transepithelial chloride current by controlling voltage-dependent calcium channels (VDCC). Dysfunction of bestrophin 1 may result in abnormal ion and fluid transport by the retinal pigment epithelium (RPE) disturbing and even disrupting direct interactions between the RPE and the photoreceptors.  相似文献   
3.
目的在背根神经节(dorsalrootganglion,DRG)中等大小感觉神经元中可以观察到钙激活氯离子流(ICl(Ca))。在坐骨神经损伤模型中,在大多数大中神经元上诱导出类似的氯离子流。本文旨在探讨引起这个离子流的分子基础。方法使用常规的定量RT-PCR方法检测在DRG中三个基因家族的表达,这三个基因家族都具有诱导,ICl(Ca)的特点。结果在成年小鼠的DRG中,分别显示了在正常状态和坐骨神经损伤3天后CLCA,Bestrophin和Tweety基因家族成员的转录产物。结论mBestl和Tweety2可能在损伤诱导的DRG神经元,ICl(Ca)中发挥作用。  相似文献   
4.
In response to cell swelling, volume-regulated anion channels (VRACs) participate in a process known as regulatory volume decrease (RVD). Only recently, first insight into the molecular identity of mammalian VRACs was obtained by the discovery of the leucine-rich repeats containing 8A (LRRC8A) gene. Here, we show that bestrophin 1 (BEST1) but not LRRC8A is crucial for volume regulation in human retinal pigment epithelium (RPE) cells. Whole-cell patch-clamp recordings in RPE derived from human-induced pluripotent stem cells (hiPSC) exhibit an outwardly rectifying chloride current with characteristic functional properties of VRACs. This current is severely reduced in hiPSC-RPE cells derived from macular dystrophy patients with pathologic BEST1 mutations. Disruption of the orthologous mouse gene (Best1−/−) does not result in obvious retinal pathology but leads to a severe subfertility phenotype in agreement with minor endogenous expression of Best1 in murine RPE but highly abundant expression in mouse testis. Sperm from Best1−/− mice showed reduced motility and abnormal sperm morphology, indicating an inability in RVD. Together, our data suggest that the molecular identity of VRACs is more complex—that is, instead of a single ubiquitous channel, VRACs could be formed by cell type- or tissue-specific subunit composition. Our findings provide the basis to further examine VRAC diversity in normal and diseased cell physiology, which is key to exploring novel therapeutic approaches in VRAC-associated pathologies.Tight regulation of cell volume is fundamental to proper cell function and survival. In general, rapid water influx across cell membranes leads to cell swelling, which in turn activates net efflux of K+ and Cl, thereby triggering the release of osmotically obligated water from the cell. Essential to this process is the activation of a current primarily carried by chloride ions (Iswell). This current is gated by volume-regulated anion channels (VRACs) returning the cell to a controlled state of homeostatic integrity, a complex mechanism commonly referred to as regulatory volume decrease (RVD) (1, 2). Although VRACs share common features in almost all cell types, it is unclear whether there is one ubiquitous channel or a diversity of chloride channels with slightly differing functional properties. In this context, three families of proteins—the Ca2+- and/or volume-sensitive anoctamins, bestrophins, and the recently discovered LRRC8s—are presently at the center of interest (37).Bestrophin 1 (BEST1), a member of the human bestrophin family of four paralogous genes, encodes an integral membrane protein strongly expressed in the human retinal pigment epithelium (RPE) (8). Mutations in BEST1 have been associated with various macular dystrophies most prominently represented by Best disease (BD), a central retinopathy with autosomal dominant inheritance but variable penetrance and expressivity (9, 10). Key features of BD pathology include a striking lipofuscin accumulation in the macular RPE (11) and an abnormal light peak (LP)/dark trough ratio in the electro-oculogram (EOG) reflective of an impaired RPE (12). The abnormalities in the LP were suggested to be compatible with a function of BEST1 as a Ca2+-activated Cl channel (CaCC) (13, 14).Addressing BEST1 function, several studies have suggested a role of the protein in distinct basic cellular processes such as Ca2+ homeostasis, neurotransmitter release, and cell volume regulation. These studies mostly relied on BEST1 overexpression in HEK293 cells or conducted in vitro experiments with isolated cells from existing Best1-deficient mouse lines. In summarizing these data, BEST1 was shown to be (i) a calcium sensor localized to the endoplasmic reticulum (ER) of mouse RPE (15), (ii) an intracellular Cl channel activating anoctamin 1 (ANO1) located at the plasma membrane of mouse trachea (5), (iii) a modulator of voltage-gated Ca2+ channels in murine RPE (16), and (iv) a channel for tonic GABA or slow glutamate release in mouse glia cells and astrocytes (17, 18). To date, the functional role of Best1 has not been determined in the mouse testis, the site of highest endogenous Best1 expression in the mouse (19). In addition, using patient-derived hiRPE cells, the role of BEST1 in mediating ER calcium release and/or uptake was shown (20). In contrast, two independent studies in S2R+ cells from Drosophila melanogaster strongly suggested the invertebrate Drosophila Best1 (dBest1) to act as a volume-regulated chloride channel but with biophysical characteristics clearly distinct from a vertebrate VRAC (3, 21). By small interfering RNA (siRNA)-mediated knockdown of BEST1 in HEK293 cells (6) and mouse Best1 (mBest1) gene disruption in murine peritoneal macrophages (22), two studies could not show a functional effect of BEST1 on Iswell, thus questioning this protein as a candidate for mammalian VRAC in these cell types. Instead, two studies identified the LRRC8A gene as an essential component of a VRAC in various cultured cell lines (6, 7). In these latter studies, the authors propose a scenario where LRRC8A and the isoforms LRRC8B to LRRC8E form variable cell type-specific hexamers, explaining the variability of VRAC properties in different cell types.Together, the rather disparate reports on BEST 1 function underscore the need to further clarify its role in mammalian VRACs. To this end, we focused on two tissues with strong endogenous BEST1 protein expression—namely, human RPE (8) and mouse sperm (19). Major insight into BEST1 function was gained from (i) RPE cell culture models established via hiPSC technology from a healthy donor and two macular dystrophy patients with established pathologic mutations in BEST1 and (ii) a mouse strain deficient for Best1, the murine ortholog of the human BEST1 gene. When exposed to hypo-osmotic challenge, both the mutant hiPSC-RPE cells and Best1-deficient mouse spermatozoa exhibited severe phenotypes, suggesting BEST1 as a crucial component of VRAC function in these cell types. In addition, membrane rupture experiments and voltage-clamp recordings in oocytes from Xenopus laevis, coexpressing aquaporin-1 (AQP1) and BEST1 from mouse and human, respectively, demonstrated identical functional properties of the mammalian BEST1 orthologs.  相似文献   
5.
Cultured retinal pigment epithelial (RPE) cells are commonly used as a model of the tissue to study their involvement in visual diseases. Unfortunately, cultured RPE often lose their differentiated phenotype reducing their usefulness as a model of the RPE in vivo. In this study, we used a Ca++-switch protocol to initiate the patterned expression of several phenotypic and functional markers of RPE differentiation. Cultured RPE cells from adult donors were maintained through at least six serial passages prior to assay to minimize their differentiated properties. The cells were then subjected to the Ca++-switch protocol and maintained at confluence for up to 4 months. Paired control and Ca++-switch cells were examined for phenotype, pigmentation, and the expression of tyrosinase, CRABP, myocilin, and bestrophin by western blot analysis. The Ca++-switch protocol led to a rapid restriction of N-cadherin to lateral cell borders, and to expression of tyrosinase by day 4. After 8 weeks, the experimental RPE monolayers began to accumulate visible pigment, and after 12 weeks CRABP expression was observed. Myocilin was observed at 4 months after the Ca++-switch but bestrophin was not detected at any time point. Our results suggest this protocol may drive epithelial morphogenesis in RPE cells. We note two specific differences in cells plated in low Ca++, reduced spreading on the substrate and coordinated development of cadherin adhesion when the Ca++-concentration is returned to normal. Thus, we suggest that this method produces phenotypic changes through multiple cell signalling pathways.  相似文献   
6.
Best macular dystrophy (BMD) is an autosomal dominant retinopathy caused by mutations in the VMD2 gene that encodes a chloride channel in the basolateral membrane of the retinal pigment epithelium (RPE). BMD patients were studied using optical coherence tomography (OCT) to understand the disease process in the macula leading to vision loss. BMD patients (ages 5-61), representing four families with known VMD2 mutations, were included. OCT scans were recorded in the central retina and longitudinal reflectivity profiles were analysed. The central retina in BMD showed different OCT abnormalities at or near the level of the highly reflective deep retinal band termed the outer retina-choroid complex (ORCC). Two types of ORCC change were noted to occur either separately or together: (1) splitting with or without intervening hyporeflective areas; and (2) elevation. Longitudinal study of a BMD patient indicated that such abnormalities were dynamic and changed in type and degree with time. The pathogenetic sequence in BMD may begin with defective fluid transport across the RPE secondary to the channelopathy in the basolateral membrane. In the macula, this leads to an abnormal interface with adjacent structures at both apical and basal surfaces of the RPE. The disease process results in detachments of the neurosensory retina, such as in central serous chorioretinopathy, and sub-RPE pathology resembling some stages of age-related macular degeneration, with eventual loss of photoreceptors, inner retina and central vision.  相似文献   
7.
Bestrophin是最新发现的一类Cl-通道,其同源序列广泛存在于动物、真菌、原核生物。Bestrophin受到Ca2+、细胞容积和自身结构的调节。在嗅觉传导、视网膜色素上皮细胞液体的转运以及细胞的增殖中发挥了重要作用。  相似文献   
8.
9.
1
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