请求东芝2505请求维修c4b0TB2931HQ的参数资料,谢谢。

您是不是在找:
买家还在看:
当前位置:
关注行业资讯
【侠光电子】 TB2931HQ 原厂原装进口拆机元件 质量保
detail3e达人选购¥5800.00¥0.65¥1.60¥0.65¥0.95
detail3e周边优质供应商广东省深圳市广东省东莞市广东省广州市广东省汕头市
同参数产品
同参数产品
同参数产品
同参数产品
& & & & & && & & & 【温馨提示】【银行汇款订单】:客户朋友与我们商议好交易订单后,可以跟我们咨询银行帐号,汇款后需将您的【具体地址】,以、QQ或者手机知,以便我们安排将货物发送给您。广东省内可以使用【物流公司代收货款】,没有【网银或者】的客户朋友也同样可以在本店购买电子元器件。一、因型号众多,未能一一上传。请有需要的客户朋友通过【】、【QQ:】、【手机:】咨询我们,我们也一定会在第一时间回复您。很期待您的光临合作!二、因个别元器件的价格会随着市场行情变动,为避免未能及时更新,给客户朋友带来不必要的麻烦,在拍买物品前,请和我们核实价格和数量。三、电子产品由于厂家的【生产批次】不同,对元器件的【MARK】可能有些不一致,如果您收到的元器件与我们上的元器件照片有所不符【如:型号标记、生产日期标记】,但是元器件的【封装、功能】等方面并未改变,这些都是属于正常的现象。当然,如果您还有任何疑问,也可以咨询我们,我们也将会为您解答,直到您满意为止。四、电子元器件是属于精密型较高的电子产品。当您的电器电路出现故障时,请先检查线路及故障原因。本店售出的产品,如证实存在质量问题,请务必在收到货物的7天内通知我们,非人为因素【如上机、客户操作失误或其他人为因素】可凭故障检测结果,向我们无条件退货、退款。若签收后的七天内,未向我们提出异议的,我们也将视为认可,认可之后的订单,我们可就不接受退换货请求,请各位客户朋友留意了哦!非货物本身质量问题,恕不退换。五、由于快递公司较多,请您向我们指定想要发送的快递公司,以免货物无法到达。本店站在客户的角度,主要发【申通快递】为主,可为广大客户朋友节省高额的运费负担,不过不足之处就是快递的速度较慢,还望各位客户朋友体谅。另有【顺丰快递】【圆通快递】【龙邦快递】等等可供选择,如果是急用元件的客户朋友,本店推荐以【顺丰快递】为主,以免耽误您的工程。谢谢合作!& & & & & & & & & & &
慧聪网厂家深圳市南山区侠光电子商行为您提供【侠光电子】 TB2931HQ 原厂原装进口拆机元件 质量保的详细产品价格、产品图片等产品介绍信息,您可以直接联系厂家获取【侠光电子】 TB2931HQ 原厂原装进口拆机元件 质量保的具体资料,联系时请说明是在慧聪网看到的。
detail3e相关商品推荐¥850.00¥2800.00¥5500.00¥2800.00¥2800.00¥2800.00¥250.00¥780.00热门商品推荐 ¥850.00 ¥2800.00 ¥5500.00 ¥2800.00 ¥2800.00 ¥2800.00 ¥250.00 ¥780.00
detail3e店内热门商品面议面议面议面议
detail3e销相关资源销热门产品搜索销相关热门专题
提示:您在慧聪网上采购商品属于商业贸易行为。以上所展示的信息由卖家自行提供,内容的真实性、准确性和合法性由发布卖家负责,请意识到互联网交易中的风险是客观存在的。推荐使用,保障您的交易安全!
所在地:广东省&&
联系人:蔡伟坚 & 先生
075 ******
请供应商联系我
手机号不能为空
姓名不能为空
请供应商联系我
您对该公司的咨询信息已成功提交请注意接听供应商电话。
detail3e关于拆机
detail3e同类其他品牌
detail3e您是不是在找
您采购的产品:
请输入采购产品
您的手机号码:
请输入手机号码
*采购产品:
请输入采购产品
*采购数量/单位:
请输入采购数量
请选择单位
*采购截止日期:
请输入正确的手机号码
请输入验证码
*短信验证码:
<input id="valid_Code1" maxlength="6" placeholder="请输入验证码" name="VALIDCODE" class="codeInput" onkeyup="this.value=this.value.replace(/\D/g,'')" onkeypress="if(event.keyCode
57) event.returnValue =" type="text">
免费获取验证码
为了安全,请输入验证码,我们将优先处理您的需求!
请输入验证码
发送成功!
慧聪已收到您的需求,我们会尽快通知卖家联系您,同时会派出采购专员1对1为您提供服务,请您耐心等待!
075 ******
联系人:蔡伟坚&总经理室 总经理
公司名称:深圳市南山区侠光电子商行
备注:点击关注按钮后才可自动收到卖家电话
请输入正确的手机号码
请输入验证码
*短信验证码:
免费获取验证码
为了安全,请输入验证码,我们将优先处理您的需求!
请输入验证码
按字母分类 :PMCID: PMC4179688Potential of caveolae in the therapy of cardiovascular and neurological diseases,1 ,2 ,2,&#x02020; and
1,*&#x02020;2Department of Neuroscience, Karolinska Institutet, Stockholm, SwedenEdited by: Annemiek Van Spriel, Radboud Institute for Molecular Life Sciences, NetherlandsReviewed by: Alex M. Dopico, The University of Tennessee Health Science Center, USA; Cristina Moreno Vadillo, Maastricht University, Netherlands*Correspondence: Rafael Franco, Departament de Bioqu&#x000mica i Biologia Molecular, Facultat de Biologia, Diagonal 645, 08028 Barcelona, Spain e-mail: ; Email: This article was submitted to Membrane Physiology and Membrane Biophysics, a section of the journal Frontiers in Physiology.&#x02020;These authors have contributed equally to this work.
This article has been
other articles in PMC.Caveolae are membrane micro-domains enriched in cholesterol, sphingolipids and caveolins, which are transmembrane proteins with a hairpin-like structure. Caveolae participate in receptor-mediated trafficking of cell surface receptors and receptor-mediated signaling. Furthermore, caveolae participate in clathrin-independent endocytosis of membrane receptors. On the one hand, caveolins are involved in vascular and cardiac dysfunction. Also, neurological abnormalities in caveolin-1 knockout mice and a link between caveolin-1 gene haplotypes and neurodegenerative diseases have been reported. The aim of this article is to present the rationale for considering caveolae as potential targets in cardiovascular and neurological diseases.Keywords: arrhythmia, dementia, gene therapy, GPCR, ischemia reperfusion, neurodegeneration, Parkinson's disease, stem cellCaveolins are a family of proteins with a hairpin-like structure. This structure is necessary to form unique membrane micro-domains known as caveolae. To our knowledge Yamada () first identified caveolae in gallbladder epithelium. The author observed: &#x0201c;The free cell surface between microvilli shows larger cave-like depressions, likewise representing caveolae intracellulares, containing a dense material.&#x0201d; Twenty years later, Dulhunty and Franzini-Armstrong () provided a detailed account of the appearance of caveolae using freeze-fracture replicas of the cell surface membrane of frog skeletal muscles. They defined the structure as: &#x0201c;elliptical invaginations of the plasmalemma which open to the outside by a narrow &#x02018;neck&#x02019; of approximately 20 nm.&#x0201d; Rothberg et al. () identified and named as caveolin the key component in such membrane micro-domains. Caveolins are a family of proteins with similar structure and, to date, three members have been identified. The first member, VIP21-caveolin or caveolin-1, is widely expressed in tissues, especially in adipocytes, fibroblasts, epithelial cells and vascul caveolin-2 interacts with and is co-expressed with caveolin-1; and, M-caveolin or caveolin-3, is mainly expressed in striated (skeletal and smooth) muscle myocytes (Scherer et al., ; Tang et al., ; Way and Parton, ; see Gratton et al.,
and Gazzerro et al.,
for review). Caveolae are enriched in cholesterol and sphingolipids (Severs, ; Rothberg et al., ; Parton, ) and the high content in cholesterol allows manipulating the system using sterol-binding agents (e.g., methyl-&#x003b2;-cyclodextrin or filipin). These compounds have been indeed invaluable to study the physiological role of caveolae. Vertical domains that usually span both cell membrane leaflets and are enriched in cholesterol and sphingolipids are known as lipids rafts. Caveolae are considered lipid rafts in as much as they contain caveolins.The two most obvious roles of caveolae are to recruit membrane proteins at specific membrane domains and to participate in protein internalization (recently reviewed in Shvets et al., ). Also they can regulate numerous enzyme activities, including that of adenylate cyclase (AC), eNitric oxide synthase (eNOS), and several kinases and serine/threonine phosphatases (Toya et al., ; Carman et al., ; Razani and Lisanti, ; Hnasko and Lisanti, ). Hence, caveolins/caveolae are not just organizers or scaffolds that localize signaling proteins but regulators of important cell events. This perspective article focuses on the therapeutic potential of targeting caveolins/caveolae in cardiovascular and neurological diseases.Altered endothelium appears as a common factor in a variety of serious diseases (Berman et al., ; Harrison, ; Lerman and Burnett, ; Quyyumi, ; Vincent et al., ; Salmon and Satchell, ). Caveolins and caveolae are very relevant to maintain the endothelial cell membrane integrity in both structure and function (Williams and Lisanti, ; Yuan and Rigor, ). Atherosclerosis, a frequent disease in Western societies, is due to deposition of cholesterol-rich lipoproteins in the endothelium of blood vessels. Transcytosis, which is a vesicle-mediated mechanism of transcellular transport of molecules, is very important to remove lipid deposits and to avoid endothelial activation and vessel occlusion. Recent evidence shows that caveolin-1 and caveolae are involved in metabolic switching, endothelial transcytosis and regulate vascular inflammation (Pavlides et al., ; Shiroto et al., ).Often, heart abnormalities course with abnormal calcium handling, for instance, in atrial fibrillation (Hove-Madsen et al., ) and hypertrophy (Gwathmey and Morgan, ; Cuneo and Grassi de Gende, ). Phospholipase C and hetero-trimeric Gq proteins regulate intracellular calcium concentrations. In an elegant study Guo et al. () identified Gq proteins in caveolae and reported that adult caveolin-3-containing ventricular cardiomyocytes show oscillating Ca2+ waves that are extinguished by blocking the interaction between caveolin-3 and the &#x003b1; subunit of Gq proteins. Therefore, caveolin-3 is directly involved in regulating contractility and may be a target for heart hypertrophy.Mutations in the caveolin-1 gene and decreased expression of caveolin-1 have been identified in patients with pulmonary arterial hypertension (Desai, ), a disease with high morbidity. Caveolin-1 null mice display a marked reduction in life span due to a combination of cardiac hypertrophy, pulmonary fibrosis and pulmonary hypertension (Park et al., ). Interestingly, double KO mice for caveolin-1 and the myocyte-selective subtype, caveolin-3, are viable but display severe cardiomyopathy (Park et al., ). Although Feiner et al. () could not demonstrate significantly different levels of caveolin-3 in failing hearts, a significant correlation existed in human failing hearts between levels of caveolin-3 and Ca2+-ATPase, a marker of the heart-failure phenotype. To our knowledge, Fujimoto in () was the first to identify an ion pump (Ca2+) in caveolae. Since then several studies have confirmed a link between caveolins/caveolae and ion carriers. In cardiomyocytes the Na+/Ca2+ exchanger is very important for heart functionality. The exchanger may interact with caveolin-1 but may also form macromolecular complexes with caveolin-3 and annexin A5 (Bossuyt et al., ,). In left ventricular myocardial samples from human failing hearts the annexin A5-interaction site in the exchanger is not accessible and the interaction between the carrier and caveolin-3 is reduced. The data suggests that caveolin-3-containing structures are relevant for Ca2+ handling in cardiac cells (Camors et al., ). The involvement of caveolae in different cardiovascular diseases makes caveolin-based therapeutic approaches an attractive possibility to combat myocardial ischemia, heart failure and pulmonary hypertension (see Fridolfsson and Patel, ).Cumulative evidence in the last decade has shown that key proteins in cardiomyocyte function interact with caveolins. Thus, caveolins not only participate in membrane positioning but also in ion channel regulation. Many ion transporters interact with caveolins and/or are located in caveolae (see Balijepalli and Kamp, , for review). The human inward rectifying voltage-gated HERG K+ channel (Trudeau et al., ) interacts with caveolin-1 and reduces its activity when caveolin is up-regulated (Lin et al., ). Cell surface expression and degradation of HERG is also controlled by caveolin-3 via a complex of these two proteins with the Nedd4-2 ubiquitin ligase (Guo et al., ) and via dynamin-mediated endocytosis (Massaeli et al., ). Caveolin-3 interacts with another K+ inward rectifying channel (Kir2.1) whose current densities are affected by mutations in caveolin-3 (Vaidyanathan et al., ). Mutations in the cardiac hNa(v)1.5 channel lead to cardiac pheno interestingly, caveolin-3 mutations identified in patients with inherited long-QT syndrome result in enhanced currents via this specific sodium channel (Vatta et al., , see Wilde and Brugada, , for review). Mutations in the caveolin-3 gene are also related to sudden infant death syndrome, with higher risk in carriers with further mutations in the hNa(v)1.5 gene (Arnestad et al., ; Cronk et al., ). Finally, caveolae may regulate ion handling by recruiting ion transporters and regulatory molecules (Yarbrough et al., ; Shibata et al., ; Palygin et al., ). A computer-based investigation supports the possibility that accumulation of ions in caveolae may lead to delayed-repolarization-induced arrhythmias (Besse et al., ). It is also of interest that caveolin-3 may be linked to and alters the function of hyperpolarization-activated cyclic nucleotide-gated channel 4, which regulates cardiac pacemaker activity (Ye et al., ). Balijepalli and Kamp () provide a detailed account of the role of caveolae in arrhythmogenesis.As neurological diseases are very diverse we present below a few examples of results that support a link between caveolins/caveolae and the two most prominent neurodegenerative disorders in developed countries: Parkinson's and Alzheimer's diseases.&#x003b1;-synuclein is a protein that often accumulates in the brain of Parkinson's disease patients (Polymeropoulos et al., ; Arawaka et al., ; Takeda et al., ). &#x003b1;-synuclein seems to cause neurodegeneration by interacting with signaling proteins and/or altering receptor-mediated signaling pathways. Examples of proteins targeted by &#x003b1;-synuclein are protein kinase C, extracellular signal-regulated kinase (ERK) and phospholipase D (Ostrerova et al., ; Iwata et al., ; Ahn et al., ). Biochemical studies using &#x003b1;-synuclein-overexpressing human neuroblastoma cells show a correlation between altered ERK signaling and deregulation of caveolin-1 expression (Hashimoto et al., ). In parkinsonian patients but not in controls, six homozygous haplotypes of the caveolin-1 gene have been identified Darvish et al. (). Furthermore, the leucine-rich repeat kinase 2 (LRRK2) is located in the neck of caveolae in a human cell model (Alegre-Abarrategui et al., ). Mutations in the gene of this kinase are linked to Parkinson's disease (Di Fonzo et al., ; Gilks et al., ). Overall, the results establish a link between caveolin-1 and molecular hallmarks of Parkinson's disease.Alzheimer's disease has two pathological hallmarks: intracellular neurofibrillary tangles, made up of aberrantly phosphorylated tau protein and plaques, made up of amyloidogenic (aberrant) processing of the amyloid precursor protein (APP), that leads to A&#x000 peptide. The enzyme that cleaves APP to give A&#x000 (BACE-1) physically associates to lipid-raft proteins (Hattori et al., ). Caveolin-3 is up-regulated in glial cells surrounding plaques and enhances the amyloidogenic route of APP processing (Nishiyama et al., ). Furthermore, caveolins physically associate to presenilins (Nishiyama et al., ) that are also relevant molecules in the pathophysiology of Alzheimer's disease. Therefore, caveolae were suspected to be involved in processing of APP, a transmembrane protein requiring integrity of cell surface mechanisms for a correct physiological function. The first indication for this view came in 1998 from the identification of the A&#x000 peptide in detergent-insoluble compartments (Lee et al., ). More recently caveolin-1 knockout mice are considered a non-mutational model for Alzheimer's disease based on the accelerated aging and neurodegeneration phenotype (Head et al., ). The mice displayed astrogliosis and increased A&#x003b2; and hyperphosphorylated tau and, noteworthy, expression of caveolin-1 in neurons from the animals led to a significant decrease in A&#x003b2; expression (Head et al., ).Caveolins/caveolae are now considered as therapeutic targets for a variety of diseases. Inhibitors are more frequent than activators as drugs targeting enzymes. Drugs targeting receptors are more often antagonists (blockers) than agonists (activators). Matters in the case of caveolins/caveolae are not as straightforward as caveolins have neither orthosteric binding sites for function regulation nor regulatory sites. To our knowledge, no small molecule targeting caveolins has been developed. Difficulties in targeting this type of membrane proteins likely underlie this lack of drug development and limit the use of caveolins as drug targets. One way to circumvent this issue is the use of anti-caveolin antibodies (Oh and Schnitzer, ; Gumbleton et al., ). Another limitation is the absence of suitable readouts for caveolin action. In sharp contrast there are highly suitable and high throughput possibilities for both enzymes and receptors. A significant alternative intervention to either increase or decrease caveolin expression would be gene or cell therapy, direct targeting of caveolin using antisense and siRNA approaches, modulation of cellular cholesterol levels or caveolar lipid content and the use of inhibitory peptides derived from caveolin scaffolding domains.Regulation of cholesterol levels or caveolar lipid content may thus be attempted for therapeutic purposes. The use of statins such as 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors decrease caveolin-1 levels (Kusama et al., ), which can involve blockade of the production of mevalonate, an intermediate in isoprenoid and cholesterol synthesis (Kirschmeier et al., ). Simvastatin may also alter lipid raft composition (Zhuang et al., ).Despite the fact that gene therapy has not delivered the promised results, new viral vectors may be suitable to target caveolae with efficacy in the affected tissue. This would prevent side effects due to indiscriminate targeting of multiple tissues. Viruses can use caveolae-dependent or &#x02013;independent mechanisms to enter the cell. One may take advantage of this fact to design therapeutic strategies using viral vectors that target caveolae (see next section). Analogously, polymeric structures may target caveolae and interfere with caveolae-mediated physiological or pathological actions. As an example, polysorbitol-based transporter delivery of small interfering RNA use caveolae for cell entry (Islam et al., ). Dendrimers successful in delivering genes via caveolae (Huang et al., ) merit attention for either enhancing or depressing caveolae-mediated events. Similarly, complexes formed by protamine, dextran, and solid lipid nanoparticles may target caveolae after intravenous administration to mice (Delgado et al., ).Cell therapy is providing new hopes for a variety of diseases. Cell therapy using autologous stem cells engineered to express specific caveolins may be considered a further possibility to colonize tissues with caveolins/caveolae-related pathologies. To colonize lungs, Ghaedi et al. () have been successful in developing caveolin-1 expressing alveolar epithelial cells from human induced pluripotent stem (iPS) cells. Similarly, other iPS-derived cells may have the potential to colonize endothelium in different tissues or, alternatively, iPS cells may differentiate into neurons (Chung et al., ) or, eventually into cardiomyocytes. To increase safety of cell therapy, generation of tumor-free iPS cell has been recently possible (Phillips, ; Zhang et al., ).Caveolins/caveolae may be indirectly targeted by blocking membrane fusion events related to endocytosis. Dynasore, an inhibitor of the GTPase activity of dynamin may prevent the entry into cells of papilloma viruses that require caveolae- and clathrin-mediated endocytosis (Abban et al., ). Paramyxoviruses (S&#x000e1;nchez-Felipe et al., ) and neurotropic viruses such as Japanese encephalitis virus (Zhu et al., ) also use a caveolae and dynamin-dependent mechanism to enter into cells. Therefore, these viruses constitute a basis to design caveolae-targeting vectors.Skeletal muscle expressing the Pro104Leu mutant of caveolin-3 leads to atrophy, and mice with this mutation serve as a model of limb-girdle muscular dystrophy 1C (Minetti et al., ; Galbiati et al., ; Hagiwara et al., ). The transforming growth factor type I &#x003b2; receptor kinase inhibitor, Ki26894, is able to restore both in vitro muscle cell deficiencies and muscle atrophy and weakness displayed by mutant mice (Ohsawa et al., ). A similar intervention could be envisaged for restoring cardiomyocyte function in patients with cardiac atrophy.A further possibility is to modulate proteins located in caveolae to activate caveolae-mediated restorative events. Members of the G-protein-coupled receptor (GPCR) superfamily are a relevant example. GPCRs are targets of approximately 40% of compounds used in human therapy. Agonist and/or antagonist modulation of their activity can lead to increases or decreases in the expression levels of caveolins. In addition, it seems possible that GPCRs can form heteroreceptor complexes with caveolins. Interactions may modify the pharmacological properties in turn making possible the design of compounds selective for GPCR protomers present in such complexes. Examples of the direct link between caveolae and GPCRs are given in the next section.Many proteins and receptors (e.g., GPCR) contain putative caveolin binding domains (Couet et al., ). For instance, caveolin-1 interacting proteins contain the canonical caveolin-1 binding domain, &#x003d5;X&#x003d5;XXXX&#x003d5; or &#x003d5;XXXX&#x003d5;XX&#x003d5; (where &#x003d5; = Trp, Phe or Tyr). In some pathologies, reducing the ability of caveolins to couple to the signaling machinery at the inner plasma membrane may result in an efficacious intervention. For instance, the introduction of synthetic caveolin-scaffolding-domain peptides into cells may inhibit caveolin-protein interactions. In support of this, internalization of the caveolin scaffolding domain may be achieved by fusion of the domain with a 16-amino acid peptide of the Drosophila antennapedia homeodomain. By this approach platelet activating-factor-induced NO production and microvasculature permeability was reduced in tumor bearing animals (Zhu et al., ). It should be noted that neovascularization is required in initial steps of metastatic colonization of tissues and that caveolin-1 regulates metastasis in bladder cancer (Thomas et al., ).Use of indirect caveolin-modulating strategies may also be effective against cardiovascular and neurological diseases. As detailed in the previous and the following sections, the bi-directional relationship that caveolins have with a number of interacting proteins and receptors could be exploited to re-expressing or targeting caveolins for up- and down-regulation. Examples would be targeting some GPCRs. The use of GPCR-selective agonist or antagonist, many of which are in current clinical use, may also affect directly (via receptor-caveolin interactions) or indirectly (via second messenger and signal cascade activation, e.g., MAPK) caveolin expression levels. Via activation of some GPCRs we could control or re-program caveolin expression levels to explore therapeutic outcomes in heart and brain.Cumulative evidence points to caveolae and caveolins as important regulators of GPCR traffic and function thus raising therapeutic potential in targeting caveolae or GPCRs in caveolae (see Figure ). Caveolins form homo-oligomers (Monier et al., ; Sargiacomo et al., ) and interact with G proteins (Li et al., ). GPCRs are not homogeneously distributed on the cell surface and a significant amount of receptors are in caveolae (Gin&#x000e9;s et al., ). GPCRs may even interact with caveolins (Burgue&#x000f1;o et al., , ). Even important elements in GPCR-mediated signaling such as G protein-coupled receptor kinases (GRKs) have binding motives for caveolins and the interaction regulates GRK function (Carman et al., ). Depending on cell type, and probably on caveolin subtype, GPCR agonists may enrich receptors in caveolae or do the opposite. In fact, agonist-induced activation of adenosine receptors may recruit them into caveolae for caveolae-mediated internalization (Gin&#x000e9;s et al., ; Escriche et al., ). In contrast, in cardiomyocytes, these adenosine receptors are enriched in caveolae until activation leads to translocation out of caveolae (Lasley et al., ; Lasley and Smart, ). This differential behavior may be taken into account when targeting caveolae via GPCRs.Scheme of caveolins/caveolae participation in the cell biology of GPCRs. Caveolins/caveolae regulate agonist binding and signaling and GPCR traffic. Some interventions with therapeutic potential are: (A) targeting caveolin-binding domains in the GPCR. ...Specific G-protein-related signaling components are enriched in lipid rafts/caveolae meaning that these structures affect G-protein-coupling efficacy and signaling selectivity (see Chini and Parenti,
and Insel et al., , for review). An exhaustive review of the reports linking caveolins/caveolae to the biology of GPCRs is out of the scope of the present perspective article. Some few examples will, however, be provided to give a hint of the relevant connections between the receptors and caveolae. Localization of the &#x003b1;1A adrenergic GPCR in lipid rafts restricts their conformation and basal activity while allowing a substantial coupling to the G protein and a robust signaling upon agonist activation (Lei et al., ). Caveolin-2 participates in receptor signaling even in a simple system constituted by human embryonic kidney (HEK-293) cells expressing a GPCR (D1) for the neurotransmitter dopamine (Yu et al., ). Caveolin is also involved in agonist-induced recruitment and internalization of a GPCR for the regulatory molecule adenosine (subtype 1, A1 receptor) (Gin&#x000e9;s et al., ). In fact, Escriche et al. () provided morphological evidence of caveolae-mediated internalization, endosomal sorting and A1 receptor recycling. Very relevant for caveolae-based drug discovery, Klaasse et al. () reported small-molecule allosteric modulators of adenosine A1 GPCRs that affect internalization of the receptor. Adenosine deaminase, also interacts with A1 GPCRs, enhances signaling and appears to be an allosteric modulator of caveolae-mediated receptor internalization (Gin&#x000e9;s et al., ). Caveolae disruption by cholesterol depletion alters the regulation by adenosine subtype 2 (A2A) of anion secretion in epithelial cells (Lam et al., ). Adenosine A2A GPCRs are up-regulated in atrial fibrillation and their blockade results in restoring the abnormal calcium handling in cardiomyocytes from patients (Hove-Madsen et al., ; Llach et al., ). Adenosine A2A GPCRs may also form homodimers or heteromers with other receptors. The complexes are unique entities with specific signaling properties (Hillion et al., ; Canals et al., ). Therefore, differential expression of A2A GPCR monomer/homomers/heteromers in membrane micro-domains is an interesting possibility for altered functional properties that should be further explored. Endothelin subtype B receptor in endothelial cells is mainly present in caveolae and its activation by endothelin leads to rapid caveolae-dependent internalization. It is likely that activation of such receptors present in caveolae leads to rapid caveolae-mediated trafficking (Oh et al., ).The selective advantage of reperfusion after ischemic injury to minimize the consequences of a second ischemic episode has been known for long (see Yellon et al.,
for review). From the finding that the increase in extracellular adenosine was helpful in anoxic heart conditions (Jacob and Berne, ; Ely and Berne, ) several studies have shown that adenosine (Kitakaze et al., ; Pelleg, ) and other compounds acting on GPCRs (Avkiran and Haworth, ; Minguet et al., ; Dragasis et al., ) regulate heart-ischemia-reperfusion and preconditioning. As reviewed by Stary et al. () caveolins are necessary for ischemic preconditioning. Targeting for instance opioid receptors may help in enhancing the reperfusion benefits. Opioid-induced preconditioning alters the architecture of the myocyte and increases the number of caveolae (Tsutsumi et al., ; Stary et al., ). The use of knockout animals for caveolin-3 has shown that this protein is essential for in vivo opioid-induced preconditioning (Tsutsumi et al., ).Future work will help on understanding how caveolin-protein and caveolin-GPCR interactions may help in combating cardiovascular and neurological diseases.Conflict of interest statementThe authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.This work has been supported by the Swedish Medical Research Council (62X-), Karolinska Institutets Forskningsstiftelser 2013 and AFA-2014 to KF; by grants from the Swedish Royal Academy of Sciences (Stiftelsen B. von Beskows Fond and Stiftelsen Hierta-Retzius stipendiefond) and Karolinska Institutets Forskningsstiftelser 2013 to Dasiel O. Borroto-Escuela. Dasiel O. Borroto-Escuela belongs to &#x0201c;Academia de Bi&#x000f3;logos Cubanos.&#x0201d;AbbreviationsGPCRG-protein-coupled receptorGRKG protein-coupled receptor kinases.Abban C. Y., Bradbury N. A., Meneses P. I. (2008). HPV16 and BPV1 infection can be blocked by the dynamin inhibitor dynasore. Am. J. Ther. 15, 304&#x
10.1097/MJT.0b013e []
[] []Ahn B. H., Rhim H., Kim S. Y., Sung Y. M., Lee M. Y., Choi J. Y., et al.
(2002). alpha-Synuclein interacts with phospholipase D isozymes and inhibits pervanadate-induced phospholipase D activation in human embryonic kidney-293 cells. J. Biol. Chem. 277, 12334&#x
10.1074/jbc.M
[] []Alegre-Abarrategui J., Christian H., Lufino M. M., Mutihac R., Venda L. L., Ansorge O., et al.
(2009). LRRK2 regulates autophagic activity and localizes to specific membrane microdomains in a novel human genomic reporter cellular model. Hum. Mol. Genet. 18, 4022&#x
10.1093/hmg/ddp346 []
[] []Arawaka S., Saito Y., Murayama S., Mori H. (1998). Lewy body in neurodegeneration with brain iron accumulation type 1 is immunoreactive for alpha-synuclein. Neurology
51, 887&#x
10.1212/WNL.51.3.887
[] []Arnestad M., Crotti L., Rognum T. O., Insolia R., Pedrazzini M., Ferrandi C., et al.
(2007). Prevalence of long-QT syndrome gene variants in sudden infant death syndrome. Circulation
115, 361&#x
10.1161/CIRCULATIONAHA.106.658021
[] []Avkiran M., Haworth R. S. (2003). Regulatory effects of G protein-coupled receptors on cardiac sarcolemmal Na+/H+ exchanger activity: signalling and significance. Cardiovasc. Res. 57, 942&#x
[] []Balijepalli R. C., Kamp T. J. (2008). Caveolae, ion channels and cardiac arrhythmias. Prog. Biophys. Mol. Biol. 98, 149&#x
10.1016/j.pbiomolbio. []
[] []Berman J. S., Beer D. J., Theodore A. C., Kornfeld H., Bernardo J., Center D. M. (1990). Lymphocyte recruitment to the lung. Am. Rev. Respir. Dis. 142, 238&#x
10.1164/ajrccm/142.1.238
[] []Besse I. M., Mitchell C. C., Hund T. J., Shibata E. F. (2011). A computational investigation of cardiac caveolae as a source of persistent sodium current. Front. Physiol. 2:87
10.3389/fphys. []
[] []Bossuyt J., Taylor B. E., James-Kracke M., Hale C. C. (2002a). The cardiac sodium-calcium exchanger associates with caveolin-3. Ann. N.Y. Acad. Sci. 976, 197&#x
10.1111/j.02.tb04741.x
[] []Bossuyt J., Taylor B. E., James-Kracke M., Hale C. C. (2002b). Evidence for cardiac sodium-calcium exchanger association with caveolin-3. FEBS Lett. 511, 113&#x
10.1111/j.02.tb04741.x
[] []Burgue&#x000f1;o J., Canela E. I., Mallol J., Lluis C., Franco R., Ciruela F. (2004). Mutual regulation between metabotropic glutamate type 1alpha receptor and caveolin-proteins: from traffick to constitutive activity. Exp. Cell Res. 300, 23&#x02013;34
10.1016/j.yexcr.
[] []Burgue&#x000f1;o J., Enrich C., Canela E. I., Mallol J., Lluis C., Franco R., et al.
(2003). Metabotropic glutamate type 1alpha receptor localizes in low-density caveolin-rich plasma membrane fractions. J. Neurochem. 86, 785&#x
[]Camors E., Charue D., Trouv&#x000e9; P., Monceau V., Loyer X., Russo-Marie F., et al.
(2006). Association of annexin A5 with Na+/Ca2+ exchanger and caveolin-3 in non-failing and failing human heart. J. Mol. Cell. Cardiol. 40, 47&#x02013;55
10.1016/j.yjmcc.
[] []Canals M., Burgue&#x000f1;o J., Marcellino D., Cabello N., Canela E. I., Mallol J., et al.
(2004). Homodimerization of adenosine A2A receptors: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J. Neurochem. 88, 726&#x
10.1046/j.03.02200.x
[] []Carman C. V., Lisanti M. P., Benovic J. L. (1999). Regulation of G protein-coupled receptor kinases by caveolin. J. Biol. Chem. 274, 8858&#x
10.1074/jbc.274.13.8858
[] []Chini B., Parenti M. (2004). G-protein coupled receptors in lipid rafts and caveolae: how, when and why do they go there?
J. Mol. Endocrinol. 32, 325&#x
10.1677/jme.0.0320325
[] []Chung C. Y., Khurana V., Auluck P. K., Tardiff D. F., Mazzulli J. R., Soldner F., et al.
(2013). Identification and rescue of &#x003b1;-synuclein toxicity in Parkinson patient-derived neurons. Science
342, 983&#x
10.1126/science.1245296 []
[] []Couet J., Li S., Okamoto T., Ikezu T., Lisanti M. P. (1997). Identification of peptide and protein ligands for the caveolin-scaffolding domain. Implications for the interaction of caveolin-with caveolae-associated proteins. J. Biol. Chem. 272, 6525&#x
10.1074/jbc.272.10.6525
[] []Cronk L. B., Ye B., Kaku T., Tester D. J., Vatta M., Makielski J. C., et al.
(2007). Novel mechanism for sudden infant death syndrome: persistent late sodium current secondary to mutations in caveolin-3. Heart Rhythm. 4, 161&#x
10.1016/j.hrthm. []
[] []Cuneo M. E., Grassi de Gende A. O. (1988). Cardiac sarcoplasmic reticulum characteristics in hypertrophic hearts from spontaneously hypertensive rats. Basic Res. Cardiol. 83, 286&#x
10.1007/BF
[] []Darvish H., Heidari A., Hosseinkhani S., Movafagh A., Khaligh A., Jamshidi J., et al.
(2013). Biased homozygous haplotypes across the human caveolin-1 upstream purine complex in Parkinson's disease. J. Mol. Neurosci. 51, 389&#x
10.-013-0021-9
[] []Delgado D., Gasc&#x000f3;n A. R., Del Pozo-Rodr&#x000guez A., Echevarr&#x000a E., Ruiz de Garibay A. P., Rodr&#x000guez J. M., et al.
(2012). Dextran-protamine-solid lipid nanoparticles as a non-viral vector for gene therapy: in vitro characterization and in vivo transfection after intravenous administration to mice. Int. J. Pharm. 425, 35&#x02013;43
10.1016/j.ijpharm.
[] []Desai A. A. (2012). Novel inheritable caveolin-1 mutations in pulmonary arterial hypertension. Circ. Cardiovasc. Genet. 5, 706&#x
10.1161/CIRCGENETICS.112.965335
[] []Di Fonzo A., Roh&#x000e9; C. F., Ferreira J., Chien H. F., Vacca L., Stocchi F., et al.
(2005). Italian parkinson genetics network. A frequent LRRK2 gene mutation associated with autosomal dominant Parkinson's disease. Lancet
365, 412&#x
[] []Dragasis S., Bassiakou E., Iacovidou N., Papadimitriou L., Andreas Steen P., Gulati A., et al.
(2013). The role of opioid receptor agonists in ischemic preconditioning. Eur. J. Pharmacol. 720, 401&#x
10.1016/j.ejphar.
[] []Dulhunty A. F., Franzini-Armstrong C. (1975). The relative contributions of the folds and caveolae to the surface membrane of frog skeletal muscle fibres at different sarcomere lengths. J. Physiol. 250, 513&#x
[]Ely S. W., Berne R. M. (1992). Protective effects of adenosine in myocardial ischemia. Circulation
85, 893&#x
10.1161/01.CIR.85.3.893
[] []Escriche M., Burgue&#x000f1;o J., Ciruela F., Canela E. I., Mallol J., Enrich C., et al.
(2003). Ligand-induced caveolae-mediated internalization of A1 adenosine receptors: morphological evidence of endosomal sorting and receptor recycling. Exp. Cell Res. 285, 72&#x02013;90
[] []Feiner E. C., Chung P., Jasmin J. F., Zhang J., Whitaker-Menezes D., Myers V., et al.
(2011). Left ventricular dysfunction in murine models of heart failure and in failing human heart is associated with a selective decrease in the expression of caveolin-3. J. Card. Fail. 17, 253&#x
10.1016/j.cardfail. []
[] []Fridolfsson H. N., Patel H. H. (2013). Caveolin-and caveolae in age associated cardiovascular disease. J. Geriatr. Cardiol. 10, 66-74
10.3969/j.issn.13.01.011 []
[] []Fujimoto T. (1993). Calcium pump of the plasma membrane is localized in caveolae. J. Cell Biol. 120, 1147&#x
10.1083/jcb.120.5.1147 []
[] []Galbiati F., Volonte D., Minetti C., Bregman D. B., Lisanti M. P. (2000). Limb-girdle muscular dystrophy (LGMD-1C) mutants of caveolin-3 undergo ubiquitination and proteasomal degradation. Treatment with proteasomal inhibitors blocks the dominant negative effect of LGMD-1C mutanta and rescues wild-type caveolin-3. J. Biol Chem. 275, 37702&#x
10.1074/jbc.M
[] []Gazzerro E., Sotgia F., Bruno C., Lisanti M. P., Minetti C. (2010). Caveolinopathies: from the biology of caveolin-3 to human diseases. Eur. J. Hum. Genet. 18, 137&#x
10.1038/ejhg. []
[] []Ghaedi M., Calle E. A., Mendez J. J., Gard A. L., Balestrini J., Booth A., et al.
(2013). Human iPS cell-derived alveolar epithelium repopulates lung extracellular matrix. J. Clin. Invest. 123, 4950&#x
10.1172/JCI68793 []
[] []Gilks W. P., Abou-Sleiman P. M., Gandhi S., Jain S., Singleton A., Lees A. J., et al.
(2005). A common LRRK2 mutation in idiopathic Parkinson's disease. Lancet
365, 415&#x
[] []Gin&#x000e9;s S., Ciruela F., Burgue&#x000f1;o J., Casado V., Canela E. I., Mallol J., et al.
(2001). Involvement of caveolin-in ligand-induced recruitment and internalization of A1 adenosine receptor and adenosine deaminase in an epithelial cell line. Mol. Pharmacol. 59, 1314&#x
10.1124/mol.59.5.1314
[] []Gratton J. P., Bernatchez P., Sessa W. C. (2004). Caveolae and caveolins in the cardiovascular system. Circ. Res. 94, 1408&#x
10.1161/01.RES..56294.17
[] []Gumbleton M., Hollins A. J., Omidi Y., Campbell L., Taylor G. (2003). Targeting caveolae for vesicular drug transport. J. Control. Release
87, 139&#x
[] []Guo J., Wang T., Li X., Shallow H., Yang T., Li W., et al.
(2012). Cell surface expression of human ether-a-go-go-related gene (hERG) channels is regulated by caveolin-3 protein via the ubiquitin ligase Nedd4-2. J. Biol. Chem. 287, 33132&#x
10.1074/jbc.M112.389643 []
[] []Guo Y., Golebiewska U., Scarlata S. (2011). Modulation of Ca2+ activity in cardiomyocytes through caveolae-G&#x003b1;q interactions. Biophys. J. 100, 1599&#x
10.1016/j.bpj. []
[] []Gwathmey J. K., Morgan J. P. (1985). Altered calcium handling in experimental pressure-overload hypertrophy in the ferret. Circ. Res. 57, 836&#x
10.1161/01.RES.57.6.836
[] []Hagiwara Y., Sasaoka T., Araishi K., Imamura M., Yorifuji H., Nonaka I., et al.
(2000). Caveolin-3 deficiency causes muscle degeneration in mice. Hum. Mol. Genet. 9, 3047&#x
10.1093/hmg/9.20.3047
[] []Harrison D. G. (1991). Endothelial modulation of vascular tone: relevance to coronary angioplasty and restenosis. J. Am. Coll. Cardiol. 17(6 Suppl. B), 71B&#x02013;76B
10.97(91)90941-2
[] []Hashimoto M., Takenouchi T., Rockenstein E., Masliah E. (2003). Alpha-synuclein up-regulates expression of caveolin-1 and down-regulates extracellular signal-regulated kinase activity in B103 neuroblastoma cells: role in the pathogenesis of Parkinson's disease. J. Neurochem. 85, 1468&#x
10.1046/j.03.01791.x
[] []Hattori C., Asai M., Onishi H., Sasagawa N., Hashimoto Y., Saido T. C., et al.
(2006). BACE1 interacts with lipid raft proteins. J. Neurosci. Res. 84, 912&#x
10.1002/jnr.20981
[] []Head B. P., Peart J. N., Panneerselvam M., Yokoyama T., Pearn M. L., Niesman I. R., et al.
(2010). Loss of caveolin-1 accelerates neurodegeneration and aging. PLoS ONE
10.1371/journal.pone.0015697 []
[] []Hillion J., Canals M., Torvinen M., Casado V., Scott R., Terasmaa A., et al.
(2002). Coaggregation, cointernalization, and codesensitization of adenosine A2A receptors and dopamine D2 receptors. J. Biol. Chem. 277, 18091&#x
10.1074/jbc.M
[] []Hnasko R., Lisanti M. P. (2003). The biology of caveolae: lessons from caveolin knockout mice and implications for human disease. Mol. Interv.
10.1124/mi.3.8.445
[] []Hove-Madsen L., Llach A., Bayes-Genis A., Roura S., Rodriguez Font E., Aris A., et al.
(2004). Atrial fibrillation is associated with increased spontaneous calcium release from the sarcoplasmic reticulum in human atrial myocytes. Circulation
110, 1358&#x
10.1161/01.CIR..59876.87
[] []Hove-Madsen L., Prat-Vidal C., Llach A., Ciruela F., Casad&#x000f3; V., Lluis C., et al.
(2006). Adenosine A2A receptors are expressed in human atrial myocytes and modulate spontaneous sarcoplasmic reticulum calcium release. Cardiovasc. Res. 72, 292&#x
10.1016/j.cardiores.
[] []Huang H., Cao D., Qin L., Tian S., Liang Y., Pan S., et al.
(2014). Dilution-stable PAMAM G1-grafted polyrotaxane supermolecules deliver gene into cells through a caveolae-dependent pathway. Mol. Pharm. 11, 2323&#x
10.1021/mp5002608
[] []Insel P. A., Head B. P., Patel H. H., Roth D. M., Bundey R. A., Swaney J. S. (2005). Compartmentation of G-protein-coupled receptors and their signalling components in lipid rafts and caveolae. Biochem. Soc. Trans. 33, 1131&#x
10.1042/BST
[] []Islam M. A., Shin J. Y., Firdous J., Park T. E., Choi Y. J., Cho M. H., et al.
(2012). The role of osmotic polysorbitol-based transporter in RNAi silencing via caveolae-mediated endocytosis and COX-2 expression. Biomaterials
33, 8868&#x
10.1016/j.biomaterials.
[] []Iwata A., Maruyama M., Kanazawa I., Nukina N. (2001). Alphasynuclein affects the MAPK pathway and accelerates cell death. J. Biol. Chem. 276, 45320&#x
10.1074/jbc.M
[] []Jacob M. I., Berne R. M. (1961). Metabolism of adenosine by the isolated anoxic cat heart. Proc. Soc. Exp. Biol. Med. 107, 738&#x
10.7-107-26739
[] []Kirschmeier P. T., Whyte D., Wilson O., Bishop W. R., Pai J. K. (2001). In vivo prenylation analysis of Ras and Rho proteins. Meth. Enzymol. 332, 115&#x
[] []Kitakaze M., Hori M., Kamada T. (1993). Role of adenosine and its interaction with alpha adrenoceptor activity in ischaemic and reperfusion injury of the myocardium. Cardiovasc. Res. 27, 18&#x02013;27
10.1093/cvr/27.1.18
[] []Klaasse E. C., van den Hout G., Roerink S. F., de Grip W. J., IJzerman A. P., Beukers M. W. (2005). Allosteric modulators affect the internalization of human adenosine A1 receptors. Eur. J. Pharmacol. 522, 1&#x02013;8
10.1016/j.ejphar.
[] []Kusama T., Mukai M., Iwasaki T., Tatsuta M., Matsumoto Y., Akedo H., et al.
(2001). Inhibition of epidermal growth factor-induced RhoA translocation and invasion of human pancreatic cancer cells by 3-hydroxy-3-methylglutaryl-coenzyme a reductase inhibitors. Cancer Res. 61, 4885&#x
[]Lam R. S., Nahirney D., Duszyk M. (2009). Cholesterol-dependent regulation of adenosine A2A receptor-mediated anion secretion in colon epithelial cells. Exp. Cell Res. 315, 3028&#x
10.1016/j.yexcr.
[] []Lasley R. D., Narayan P., Uittenbogaard A., Smart E. J. (2000). Activated cardiac adenosine A(1) receptors translocate out of caveolae. J. Biol. Chem. 275, 4417&#x
10.1074/jbc.275.6.4417
[] []Lasley R. D., Smart E. J. (2001). Cardiac myocyte adenosine receptors and caveolae. Trends Cardiovasc. Med. 11, 259&#x
[] []Lee S. J., Liyanage U., Bickel P. E., Xia W., Lansbury P. T., Jr., Kosik K. S. (1998). A detergent-insoluble membrane compartment contains A beta in vivo. Nat. Med. 4, 730&#x
10.1038/nm
[] []Lei B., Morris D. P., Smith M. P., Schwinn D. A. (2009). Lipid rafts constrain basal alpha(1A)-adrenergic receptor signaling by maintaining receptor in an inactive conformation. Cell. Signal. 21, 1532&#x
10.1016/j.cellsig.
[] []Lerman A., Burnett J. C., Jr. (1992). Intact and altered endothelium in regulation of vasomotion. Circulation
86(Suppl. 6), III12&#x02013;III19
[]Li S., Okamoto T., Chun M., Sargiacomo M., Casanova J. E., Hansen S. H., et al.
(1995). Evidence for a regulated interaction between heterotrimeric G proteins and caveolin. J. Biol. Chem. 270, 15693&#x
10.1074/jbc.270.26.15693
[] []Lin J., Lin S., Choy P. C., Shen X., Deng C., Kuang S., et al.
(2008). The regulation of the cardiac potassium channel (HERG) by caveolin-1. Biochem. Cell Biol. 86, 405&#x
[] []Llach A., Molina C. E., Prat-Vidal C., Fernandes J., Casad&#x000f3; V., Ciruela F., et al.
(2011). Abnormal calcium handling in atrial fibrillation is linked to up-regulation of adenosine A2A receptors. Eur. Heart J. 32, 721&#x
10.1093/eurheartj/ehq464
[] []Massaeli H., Sun T., Li X., Shallow H., Wu J., Xu J., et al.
(2010). Involvement of caveolin-in low K+-induced endocytic degradation of cell-surface human ether-a-go-go-related gene (hERG) channels. J. Biol. Chem. 285, 27259&#x
10.1074/jbc.M110.124909 []
[] []Minetti C., Sotgia F., Bruno C., Scartezzini P., Broda P., Bado M., et al.
(1998). Mutations in the caveolin-3 gene cause autosomal dominant limb-girdle muscular dystrophy. Nat. Genet. 18, 365&#x
10.1038/ng
[] []Minguet G., Brichant J. F., Joris J. (2012). Opioids and protection against ischemia-reperfusion injury: from experimental data to potential clinical applications. Acta Anaesthesiol. Belg. 63, 23&#x02013;34
[]Monier S., Parton R. G., Vogel F., Behlke J., Henske A., Kurzchalia T. (1995). VIP21-caveolin, a membrane protein constituent of the caveolar coat, oligomerizes in vivo and in vitro. Mol. Biol. Cell
10.1091/mbc.6.7.911 []
[] []Nishiyama K., Trapp B. D., Ikezu T., Ransohoff R. M., Tomita T., Iwatsubo T., et al.
(1999). Caveolin-3 upregulation activates beta-secretase-mediated cleavage of the amyloid precursor protein in Alzheimer's disease. J. Neurosci. 19, 6538&#x
[]Oh P., Horner T., Witkiewicz H., Schnitzer J. E. (2012). Endothelin induces rapid, dynamin-mediated budding of endothelial caveolae rich in ET-B. J. Biol. Chem. 287, 17353&#x
10.1074/jbc.M111.338897 []
[] []Oh P., Schnitzer J. E. (1999). Immunoisolation of caveolae with high affinity antibody binding to the oligomeric caveolin cage. J. Biol. Chem. 274, 23144&#x
10.1074/jbc.274.33.23144
[] []Ohsawa Y., Okada T., Nishimatsu S., Ishizaki M., Suga T., Fujino M., et al.
(2012). An inhibitor of transforming growth factor beta type I receptor ameliorates muscle atrophy in a mouse model of caveolin-3-deficient muscular dystrophy. Lab. Invest. 92, 1100&#x
10.1038/labinvest.2012.78
[] []Ostrerova N., Petrucelli L., Farrer M., Mehta M., Choi P., Hardy J., et al.
(1999). Alpha-synuclein shares physical and functional homology with 14-3-3 proteins. J. Neurosci. 19, 5782&#x
[]Palygin O. A., Pettus J. M., Shibata E. F. (2008). Regulation of caveolar cardiac sodium current by a single Gsalpha histidine residue. Am. J. Physiol. Heart Circ. Physiol. 294, H1693&#x0
10.1152/ajpheart.
[] []Park D. S., Cohen A. W., Frank P. G., Razani B., Lee H., Williams T. M., et al.
(2003). Caveolin-1 null (-/-) mice show dramatic reductions in life span. Biochemistry
42, 15124&#x
10.1021/bi0356348
[] []Park D. S., Woodman S. E., Schubert W., Cohen A. W., Frank P. G., Chandra M., et al.
(2002). Caveolin-1/3 double-knockout mice are viable, but lack both muscle and non-muscle caveolae, and develop a severe cardiomyopathic phenotype. Am. J. Pathol. 160, 2207&#x
10.-68-6 []
[] []Parton R. G. (1994). Ultrastructural localiza GM1 is concentrated in caveolae. J. Histochem. Cytochem. 42, 155&#x
10..8288861
[] []Pavlides S., Gutierrez-Pajares J. L., Iturrieta J., Lisanti M. P., Frank P. G. (2014). Endothelial caveolin-1 plays a major role in the development of atherosclerosis. Cell Tissue Res. 356, 147&#x
10.-013-1767-7
[] []Pelleg A. (1993). Mechanisms of action and therapeutic potential of adenosine and its analogues in the treatment of cardiac arrhythmias. Coron. Artery Dis. 4, 109&#x
[] []Phillips M. I. (2014). Tumor-free iPS stem cells for heart cells. Cell Cycle
10.4161/cc.28894 []
[] []Polymeropoulos M. H., Lavedan C., Leroy E., Ide S. E., Dehejia A., Dutra A., et al.
(1997). Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science
276, 2045&#x
10.1126/science.276.
[] []Quyyumi A. A. (1998). Endothelial function in health and disease: new insights into the genesis of cardiovascular disease. Am. J. Med. 105, 32S&#x02013;39S
[] []Razani B., Lisanti M. P. (2001). Two distinct caveolin-1 domains mediate the functional interaction of caveolin-1 with protein kinase A. Am. J. Physiol. Cell Physiol.
281, C1241&#x0
[]Rothberg K. G., Heuser J. E., Donzell W. C., Ying Y. S., Glenney J. R., Anderson R. G. (1992). Caveolin, a protein component of caveolae membrane coats. Cell
68, 673&#x
10.74(92)90143-Z
[] []Rothberg K. G., Ying Y. S., Kamen B. A., Anderson R. G. (1990). Cholesterol controls the clustering of the glycophospholipid-anchored membrane receptor for 5-methyltetrahydrofolate. J. Cell Biol. 111, 2931&#x
[]Salmon A. H., Satchell S. C. (2012). Endothelial glycocalyx dysfunction in disease: albuminuria and increased microvascular permeability. J. Pathol. 226, 562&#x
10.1002/path.3964
[] []S&#x000e1;nchez-Felipe L., Villar E., Mu&#x000f1;oz-Barroso I. (2014). Entry of newcastle disease virus into the host cell: role of acidic pH and endocytosis. Biochim. Biophys. Acta
1838, 300&#x
10.1016/j.bbamem.
[] []Sargiacomo M., Scherer P. E., Tang Z.-L., Kubler E., Song K. S., Sanders M. C., Lisanti M. P. (1995). Proc. Natl. Acad. Sci. U.S.A. 92, 9407&#x
10.1073/pnas.92.20.9407 []
[] []Scherer P. E., Okamoto T., Chun M., Nishimoto I., Lodish H. F., Lisanti M. P. (1996). Identification, sequence, and expression of caveolin-2 defines a caveolin-gene family. Proc. Natl. Acad. Sci. U.S.A. 93, 131&#x
10.1073/pnas.93.1.131 []
[] []Severs N. J. (1981). Plasma membrane cholesterol in myocardial muscle and capillary endothelial cells. Distribution of filipin-induced deformations in freeze-fracture. Eur. J. Cell Biol. 25, 289&#x
[]Shibata E. F., Brown T. L., Washburn Z. W., Bai J., Revak T. J., Butters C. A. (2006). Autonomic regulation of voltage-gated cardiac ion channels. J. Cardiovasc. Electrophysiol. 17, S34&#x02013;S42
10.1111/j.06.00387.x
[] []Shiroto T., Romero N., Sugiyama T., Sartoretto J. L., Kalwa H., Yan Z., et al.
(2014). Caveolin-1 is a critical determinant of autophagy, metabolic switching, and oxidative stress in vascular endothelium. PLoS ONE
10.1371/journal.pone.0087871 []
[] []Shvets E., Ludwig A., Nichols B. J. (2014). News from the caves: update on the structure and function of caveolae. Curr. Opin. Cell Biol. 29C, 99&#x
10.1016/j.ceb.
[] []Stary C. M., Tsutsumi Y. M., Patel P. M., Head B. P., Patel H. H., Roth D. M. (2012). Caveolins: targeting pro-survival signaling in the heart and brain. Front. Physiol. 3:393
10.3389/fphys. []
[] []Takeda A., Mallory M., Sundsmo M., Honer W., Hansen L., Masliah E. (1998). Abnormal accumulation of NACP/alpha-synuclein in neurodegenerative disorders. Am. J. Pathol. 152, 367&#x
[]Tang Z., Scherer P. E., Okamoto T., Song K., Chu C., Kohtz D. S., et al.
(1996). Molecular cloning of caveolin-3, a novel member of the caveolin-gene family expressed predominantly in muscle. J. Biol. Chem. 271, 2255&#x
10.1074/jbc.271.4.2255
[] []Thomas S., Overdevest J. B., Nitz M. D., Williams P. D., Owens C. R., Sanchez-Carbayo M., et al.
(2011). Src and caveolin-1 reciprocally regulate metastasis via a common downstream signaling pathway in bladder cancer. Cancer Res. 71, 832&#x
10.72.CAN-10-0730
[] []Toya Y., Schwencke C., Couet J., Lisanti M. P., Ishikawa Y. (1998). Inhibition of adenylyl cyclase by caveolin peptides. Endocrinol. 139, 2025&#x
10.1210/en.139.4.2025
[] []Trudeau M. C., Warmke J. W., Ganetzky B., Robertson G. A. (1995). HERG, a human inward rectifier in the voltage-gated potassium channel family. Science
269, 92&#x02013;95 Erratum in: Science 87.
[]Tsutsumi Y. M., Kawaraguchi Y., Niesman I. R., Patel H. H., Roth D. M. (2010). Opioid-induced preconditioning is dependent on caveolin-3 expression. Anesth. Analg. 111, 1117&#x
10.1213/ANE.0b013ea []
[] []Vaidyanathan R., Vega A. L., Song C., Zhou Q., Tan B. H., Berger S., et al.
(2013). The interaction of caveolin-3 protein with the potassium inward rectifier channel Kir2.1: physiology and pathology related to long QT syndrome 9 (LQT9). J. Biol. Chem. 288, 17472&#x
10.1074/jbc.M112.435370 []
[] []Vatta M., Ackerman M. J., Ye B., Makielski J. C., Ughanze E. E., Taylor E. W., et al.
(2006). Mutant caveolin-3 induces persistent late sodium current and is associated with long-QT syndrome. Circulation
114, 2104&#x
10.1161/CIRCULATIONAHA.106.635268
[] []Vincent A. M., Callaghan B. C., Smith A. L., Feldman E. L. (2011). Diabetic neuropathy: cellular mechanisms as therapeutic targets. Nat. Rev. Neurol. 7, 573&#x
10.1038/nrneurol.
[] []Way M., Parton R. G. (1996). M-caveolin, a muscle-specific caveolin-related protein. FEBS Lett. 378, 108&#x
10.93(96)82884-5
[] []Wilde A. A., Brugada R. (2011). Phenotypical manifestations of mutations in the genes encoding subunits of the cardiac sodium channel. Circ. Res. 108, 884&#x
10.1161/CIRCRESAHA.110.238469
[] []Williams T. M., Lisanti M. P. (2004). The Caveolin genes: from cell biology to medicine. Ann. Med. 36, 584&#x
[] []Yamada E. (1955). The fine structure of the gall bladder epithelium of the mouse. J. Biophys. Biochem. Cytol. 1, 445&#x
10.1083/jcb.1.5.445 []
[] []Yarbrough T. L., Lu T., Lee H. C., Shibata E. F. (2002). Localization of cardiac sodium channels in caveolin-rich membrane domains: regulation of sodium current amplitude. Circ. Res. 90, 443&#x
10.1161/hh
[] []Ye B., Balijepalli R. C., Foell J. D., Kroboth S., Ye Q., Luo Y. H., et al.
(2008). Caveolin-3 associates with and affects the function of hyperpolarization-activated cyclic nucleotide-gated channel 4. Biochemistry
47, 12312&#x
10.1021/bi8009295 []
[] []Yellon D. M., Baxter G. F., Garcia-Dorado D., Heusch G., Sumeray M. S. (1998). Ischaemic preconditioning: present position and future directions. Cardiovasc. Res. 37, 21&#x02013;33
[] []Yu P., Yang Z., Jones J. E., Wang Z., Owens S. A., Mueller S. C., et al.
(2004). D1 dopamine receptor signaling involves caveolin-2 in HEK-293 cells. Kidney Int. 66, 2167&#x
10.1111/j.04.66007.x
[] []Yuan S. Y., Rigor R. R., editors.
(eds.). (2010). Chapter 4: the endothelial barrier, in Regulation of Endothelial Barrier Function (San Rafael, CA: Morgan &#x00026; Claypool Life S ).Zhang L., Pan Y., Qin G., Chen L., Chatterjee T. K., Weintraub N. L., et al.
(2014). Inhibition of stearoyl-coA desaturase selectively eliminates tumorigenic Nanog-positive cells: improving the safety of iPS cell transplantation to myocardium. Cell Cycle
13, 762&#x
10.4161/cc.27677 []
[] []Zhu L., Schwegler-Berry D., Castranova V., He P. (2004). Internalization of caveolin-1 scaffolding domain facilitated by Antennapedia homeodomain attenuates PAF-induced increase in microvessel permeability. Am. J. Physiol. Heart Circ. Physiol. 286, H195&#x0
10.1152/ajpheart.
[] []Zhu Y. Z., Xu Q. Q., Wu D. G., Ren H., Zhao P., Lao W. G., et al.
(2012). Japanese encephalitis virus enters rat neuroblastoma cells via a pH-dependent, dynamin and caveolae-mediated endocytosis pathway. J. Virol. 86, 13407&#x
10.1128/JVI.00903-12 []
[] []Zhuang L., Kim J., Adam R. M., Solomon K. R., Freeman M. R. (2005). Cholesterol targeting alters lipid raft composition and cell survival in prostate cancer cells and xenografts. J. Clin. Invest. 115, 959&#x
10.1172/JCI []
[] []Articles from Frontiers in Physiology are provided here courtesy of Frontiers Media SA
)- subscription required
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
Show all items
CitePeer Related Articles

我要回帖

更多关于 东芝2505请求维修c4b0 的文章

 

随机推荐