首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 156 毫秒
1.
目的研究G蛋白偶联受体(G protein-coupled receptors)激酶5(GPCR kinase-5,GRK5)基因缺陷和老化交互作用对阿尔茨海默病(Alzheimer's disease,AD)早期的病理改变-海马内肿胀轴突丛(swollen axonal clusters,SACs)出现和积累的影响。方法选取5、6、7、9、12~13、18—19月龄雌性GRK5基因敲除小鼠(GRK5 Knockout,GRK5KO)作为观察对象,另选取年龄匹配的雌性野生型(wild type,WT)小鼠为对照,每个年龄段GRK5KO和WT小鼠各4只。用抗人神经原纤维缠结(neurofibrillary tangles,NFTs)特异性抗体的免疫荧光染色方法观察海马内SACs的变化。结果所有小鼠随着年龄增长,海马内SACs逐渐增加;GRKSKO小鼠组海马内NFT^+ SACs数量较WT型小鼠组显著增加(P〈0.01);双因素方差分析显示遗传性GRK5基因缺陷和老化双因素对海马内NFT^+SACs的影响有显著协同效应(P〈0.01)。结论在促进早期AD病理发生的过程中,GRK5缺陷和老化双因素共同加剧了雌性GRKSKO小鼠海马内SACs的形成与积累。  相似文献   

2.
目的研究体内G蛋白偶联受体激酶5(GRK5)缺陷是否会加剧转瑞典突变淀粉样肽前体蛋白基因(TgAPPsw,Tg2576)小鼠海马内的病理改变。方法将具有C57/BL6遗传背景的GRK5缺陷/敲除(GRK5KO)杂合子与具有相同遗传背景的Tg2576小鼠杂交,以产生野生型(WT)、GRK5KO杂合子型、转淀粉样肽前体蛋白(APP)基因型以及转基因&敲除(Double)型4种基因型小鼠。用免疫荧光(IF)染色方法来观察这些动物海马内肿胀轴突丛(SACs)和A8沉积量变化。结果IF染色结果定量分析显示,Tg2576小鼠被灭活一个拷贝的GRK5基因后导致海马内SACs和A8沉积量均显著增加。结论体内GRK5缺陷加剧了AD动物海马内的病理改变。  相似文献   

3.
目的 阿尔茨海默病(AD)是最常见的老年痴呆症。年龄和性别是AD发病过程中最重要的两个因素。随着年龄的增长,临床前和临床症状相继出现,而焦虑是AD发展过程中典型的早期症状之一。此外,AD在女性中的发病率高于男性。然而,尽管在AD患者发病中观察到了这些差异,但仍然缺少对动物模型的评价。方法 本文选择APP London突变(Val717Ile) AD转基因小鼠为研究对象。该突变是APP中最早描述的突变之一,与AD的早期发病有关。为了探究APP V717I小鼠在AD进展过程中是否存在性别差异,本文利用旷场试验、跳台试验、Morris水迷宫等动物行为试验对非认知症状和认知症状进行了评价。结果 本研究发现,雌性转基因小鼠在6月龄时表现出明显的焦虑,而雄性转基因小鼠仅在10月龄时表现出焦虑。在6月龄时,雄性和雌性转基因小鼠都没有表现出认知缺陷;但是,在10月龄时,两种性别都表现出明显的认知障碍。结论 这些结果表明,APP V717I Tg小鼠在记忆障碍发生前表现出焦虑样活动;在AD发展过程中,APP V717I转基因小鼠在年龄和性别上有明显的差异。本研究为AD发病过程中的年龄和性别差异提供了有利...  相似文献   

4.
该文旨在研究雌激素缺乏不同时间段对APP/PS1双转基因小鼠学习记忆及海马区细胞增殖和成熟的影响及探究潜在的机制。将3月龄APP/PS1双转基因AD雌性小鼠行双侧卵巢切除(AD-OVX),以假手术AD小鼠(AD-Sham)及同月龄正常野生型小鼠(WT)作为对照,于术后1周(模拟绝经早期)和3月(模拟绝经中晚期), Morris水迷宫行为测试结果显示,在APP/PS1双转基因AD小鼠中, OVX后1周, AD-OVX组与AD-Sham组比较,其逃避潜伏期、搜索路径以及穿越平台的次数无明显差异(P0.05);而OVX后3月, AD-OVX组小鼠找到平台的时间和搜索路径显著延长(P0.05),穿越平台的次数也相应减少(P0.05);子宫重量结果、EDU细胞增殖状况、老年斑、脑内NeuN蛋白和芳香酶的变化水平分别显示,在APP/PS1双转基因AD小鼠中, OVX后1周, AD-OVX组与ADSham组比较,循环雌激素水平无明显变化;小鼠脑内未见老年斑;小鼠海马区新生阳性细胞数量和NeuN的表达反应性增多(P0.05);此时小鼠脑内芳香酶表达也呈反应性升高(P0.05)。而OVX后3月, AD-OVX组小鼠循环雌激素水平明显降低(P0.05);脑内老年斑显著增加(P0.05);小鼠海马区新生阳性细胞数量和NeuN的表达减少(P0.05);此时小鼠脑内芳香酶水平也显著降低(P0.05)。以上结果说明,雌激素缺乏早期可反应性地增加痴呆小鼠海马区细胞的增殖和成熟,对小鼠学习记忆无影响;但随着雌激素缺乏时间的延长,痴呆小鼠出现学习记忆的损害及海马区细胞增殖和成熟减少;该作用可能与脑内芳香酶水平的变化密切相关。  相似文献   

5.
目的探讨胆碱能神经系统是否是抗抑郁药氟西汀(Fluoxetine,FLXT)改善阿尔茨海默病(Alzheimer's disease,AD)患者大脑空间学习记忆能力的作用靶点。方法随机选取18月龄的雄性APP/PS1双转基因AD小鼠20只分为阳性对照组(APP/PSI组)和FLXT组,分别给予为期4周的腹腔注射生理盐水和FLXT,并随机选取18月龄同窝生野生型(wild type,WT)小鼠10只作为阴性对照组(WT组),该组不给于药物干预。运用Morris水迷宫实验对三组小鼠的空间学习记忆能力进行检测,采用免疫组织化学染色和紫外分光光度法对三组小鼠大脑内β-淀粉样蛋白(amyloidβ-protein,Aβ)沉积情况和乙醜胆碱(acetylcholine,Ach)的含量、乙酰胆碱脂酶(acetylcholinesterase,AchE)及胆碱乙酰化酶(choline acetyl transferase,ChAT)活性等进行检测。结果水迷宫实验显示,APP/PS1小鼠逃避潜伏期显著长于WT小鼠,FLXT处理可明显缩短APP/PS1小鼠逃避潜伏期;免疫组织化学染色显示,WT小鼠脑内未见明显Aβ沉积,而APP/PSI小鼠海马内可见大量Aβ沉积,FLXT处理可明显减少APP/PS1小鼠海马内Aβ沉积;对Ach含量、AchE和ChAT活性检测显示,APP/PS1小鼠大脑皮质及海马内的Ach含量、AchE活性及皮质内的CHAT活性均较WT小鼠降低,FLXT处理可明显抑制APP/PS1小鼠大脑皮质及海马内Ach含量、AchE活性的降低,但对ChAT活性没明显的作用。结论胆碱能系统可能是FLXT作用于AD大脑的作用靶点之一,即FLXT可能通过增加AD大脑胆碱能神经系统的神经功能活性,进而增加Ach的含量,从而改善AD大脑的空间学习记忆能力。  相似文献   

6.
小鼠肺腺癌模型的建立及肿瘤病理分析   总被引:2,自引:0,他引:2  
目的用乙基亚硝脲(ENU)在BABL/c小鼠建立肺腺癌模型并对ENU所诱发的肺腺癌进行病理观察。方法妊娠17d的SPF级母鼠腹腔接受ENU或缓冲液注射,在子代鼠的鼠龄满32周时收获其全肺标本,对肺组织进行常规石蜡半连续切片,HE染色,镜下观察肿瘤病理。结果 ENU经胎盘一次性诱发子代鼠多发性肺肿瘤形成,病理显示这些肿瘤为处于不同发展阶段的腺瘤和腺癌,腺癌的类型有细支气管肺泡癌样腺癌(雌性:5/6,雄性:4/6)和分化不等的腺癌(雌性:4/6,雄性:5/6),诱癌频率在雌、雄性小鼠均为5/6,癌变频率在雌性16/43,雄性12/31。结论成功建立了小鼠肺腺癌模型,肿瘤病理的多样性提示癌变机制在分子水平的复杂性。  相似文献   

7.
目的:观察性别对小鼠H22腹水型肝癌生长情况的影响,研究不同性别动物对肝癌耐受性的差异。方法:取30只8周龄昆明鼠,雌雄各半,随机分为四组,实验组每组10只,对照组每组5只,腹腔接种小鼠H22肝癌细胞,建立小鼠H22腹水型肝癌模型。每天测量小鼠体重并记录生存时间,直至实验组小鼠全部死亡,比较性别因素对小鼠H22腹水型肝癌的生存期是否存在差异。结果:小鼠接种瘤细胞后,逐渐产生腹水,体重增加。雌性小鼠的体重增加比雄性小鼠显著,P=0.049。雄性小鼠生存后期体重出现下降,呈明显恶液质状态。雌性小鼠的体重、腹水增加虽然较雄性动物明显,但生存期却并不少于雄性鼠,反而比雄性小鼠略长,P=0.1567。结论:性别对小鼠H22腹水型肝癌的生长有一定的差异,雌性小鼠的耐受性优于雄性小鼠。  相似文献   

8.
目的分析和研究阿尔兹海默病(AD)转基因模型小鼠(APP/PS1)活体脑部影像学特征。方法本研究利用7.0 T高场强磁共振成像(MRI)技术,对1、3、5、7、9和11月龄AD转基因小鼠模型及对照组活体脑组织的微观结构变化进行对比研究。定量分析了脑组织顶叶皮层及海马区的横向弛豫时间(T2)、表观扩散系数(ADC)和各向异性分数(FA)随AD小鼠年龄变化情况。结果从9月龄开始AD转基因小鼠顶叶皮层及海马区可见散点状低信号区,并且随年龄增长逐渐增多;AD转基因小鼠顶叶皮层和海马区的T2磁豫时间在1~9月龄过程中有减小趋势,但与对照组无显著性差异;顶叶皮层及海马区ADC值的计算结果表明,7~11月龄AD转基因小鼠的ADC值明显下降(P≤0.05);同样APP/PS1小鼠的FA值从5月龄就开始降低(P≤0.05),并且这种差异一直持续到11月龄。结论高场强MRI能够显示AD病变出现早期小鼠顶叶皮层及海马区FA值的明显改变,揭示FA值对早期痴呆症临床诊断具有一定的参考价值。  相似文献   

9.
β-淀粉样蛋白(Aβ)在脑内的沉积被认为是阿尔茨海默病(AD)发病的始动因素之一.β-淀粉样蛋白前体蛋白裂解酶1(BACE1)是Aβ产生过程中重要的蛋白酶.BACE1在细胞内的表达与成熟受多种因素调节.BACE1与缺血缺氧、炎症应激等多种AD早期的分子病理变化相关,BACE1、Aβ及其相关细胞因子可能成为体液生物学标记物,为临床早期诊断AD提供新的手段.  相似文献   

10.
目的探讨胎次、性别是否对成年小鼠海马齿状回神经发生及学习记忆产生影响。方法运用Morris水迷宫系统检测第1~3胎成年小鼠的学习记忆能力,腹腔注射BrdU,标记神经干细胞,检测不同胎次、性别小鼠海马齿状回中的神经发生的差异。结果 (1)在同性别、不同胎次成年小鼠间,第2胎的学习记忆能力(LMA)均显著地高于第1、3胎的,其影响规律为LMA2〉LMA1〉LMA3,且P〈0.05;在同胎次、不同性别成年小鼠间,雌性小鼠的LMA均高于雄性小鼠的,但其差异无显著性(P〉0.05)。(2)在同性别、不同胎次成年小鼠间,第2胎海马DG新生神经细胞的数量(N)均高于第1、3胎的,其影响规律分别为NF2〉NF3〉NF1和NM2〉NM1〉NM3,但其差异无显著性(P〉0.05);在同胎次、不同性别成年小鼠间,雌性小鼠的N均高于雄性小鼠的,但其差异无显著性(P〉0.05)。结论胎次、性别对实验动物神经发生及学习记忆等方面产生的影响是肯定的。因此,在使用实验动物时,应予以充分考虑,尽量使用胎次、性别相同的。  相似文献   

11.
G protein-coupled receptor kinase 5 (GRK5) deficiency has been linked recently to early Alzheimer disease (AD), but the mechanism by which GRK5 deficiency may contribute to AD pathogenesis remains elusive. Here we report that overexpression of dominant negative mutant of GRK5 (dnGRK5) in a cholinergic neuronal cell line led to decreased acetylcholine (ACh) release. This reduction was fully corrected by pertussis toxin, atropine (a nonselective muscarinic antagonist), or methoctramine (a selective M2/M4 muscarinic receptor antagonist). Consistent with results in cultured cells, high potassium-evoked ACh release in hippocampal slices from young GRK5 knock-out mice was significantly reduced compared with wild type littermates, and this reduced ACh release was also fully corrected by methoctramine. In addition, following treatment with the nonselective muscarinic agonist oxotremorine-M, M2, and M4 receptors underwent significantly reduced internalization in GRK5KO slices compared with wild type slices, as assessed by plasma membrane retention of receptor immunoreactivity, whereas M1 receptor internalization was not affected by loss of GRK5 expression. Moreover, Western blotting revealed no synaptic or cholinergic degenerative changes in young GRK5 knock-out mice. Altogether, these results suggest that GRK5 deficiency leads to a reduced hippocampal ACh release and cholinergic hypofunction by selective impairment of desensitization of presynaptic M2/M4 autoreceptors. Because this nonstructural cholinergic hypofunction precedes the hippocampal cholinergic hypofunction associated with structural cholinergic degeneration and cognitive decline in aged GRK5 knock-out mice, this nonstructural alteration may be an early event contributing to cholinergic degeneration in AD.G protein-coupled receptor kinase-5 (GRK5)2 is one of the seven GRK family members whose primary function is to desensitize G protein-coupled receptors (GPCRs) (1, 2). We recently reported that increased soluble β-amyloid decreases membrane (functional) levels of GRK5 in vitro, and this membrane GRK5 deficiency occurs in vivo as well in an Alzheimer disease (AD) transgenic model (3) and in postmortem human AD brain samples (4). Moreover, the aged GRK5 knock-out (GRK5KO) mouse, which models this GRK5 deficiency in the absence of exogenous mutant human β-amyloid precursor protein (β-APP) or any other known AD-related genes (i.e. presenilins or tau), develops axonal defects and mild cholinergic degeneration with associated amnestic mild cognitive impairment (5). When Swedish mutant βAPP is overexpressed in the GRK5KO mice by cross-breeding with Swedish APP transgenic mice, the aged double mutant mice display significantly exaggerated brain inflammation (6). These accumulating data strongly suggest that GRK5 deficiency significantly contributes to AD pathogenesis, although the precise molecular mechanisms remain to be delineated.Mounting evidence indicates that the substrate spectrum of broadly expressed GRKs (i.e. GRK2/3/5/6) can significantly overlap for some receptors, suggesting that a lack of one of these members may have only a limited impact on GPCR regulation (2). On the other hand, compensation for loss of a particular GRK member by others in vivo can be incomplete or selective for other receptor types. For example, GRK2KO and GRK6KO mice have been shown to display selective impairments of adrenergic and dopaminergic receptor desensitization, respectively (7, 8). Findings from different GRK isoform-targeted animals strongly support the conclusion that although redundancy exists between GRK isoforms, each isoform has its own selective substrates; should one GRK be deficient or inactivated, desensitization of its selective substrates will be impaired (1). For GRK5 in particular, previous studies have demonstrated that GRK5KO mice display selectively impaired desensitization of muscarinic acetylcholine receptors (mAChRs) (9, 10).To date, five mAChR subtypes have been identified, with M1, M3, and M5 receptors being Gq/11-coupled, and M2 and M4 receptors being Gi/o-coupled (11). In hippocampal memory circuits, M2 receptor (M2R) is primarily a presynaptic autoreceptor that inhibits ACh release (12, 13), whereas M1R is postsynaptic and is believed to be critical in memory processes involving an interaction between the cerebral cortex and hippocampus (11). In AD, there is a selective loss of cholinergic neurons that leads to a cholinergic hypofunction, primarily a hypoactivity of postsynaptic nicotinic and M1 muscarinic receptors (14). GRK5KO mice, when challenged with nonselective muscarinic agonists, display augmented hypothermia, hypoactivity, tremor, and salivation, as well as antinociceptive changes (9). These behavioral changes are typical M2 and/or M4 receptor-mediated functions, according to the findings from muscarinic receptor subtype knock-out mice (11, 15). Therefore, GRK5 deficiency in vivo may selectively impair M2/M4R desensitization. If so, the resulting presynaptic M2/M4R hyperactivity would overly inhibit ACh release from cholinergic neurons and eventually compromise the learning and memory function. This study was undertaken to investigate the impact of GRK5 deficiency on ACh release and desensitization of mAChR subtypes using GRK5-deficient models both in vitro and in hippocampal slices from the GRK5KO mice.  相似文献   

12.
Alzheimer disease (AD) and stroke are two leading causes of age-associated dementia. Increasing evidence points to vascular damage as an early contributor to the development of AD and AD-like pathology. In this review, we discuss the role of G protein-coupled receptor kinase 2 (GRK2) as it relates to individuals affected by AD and how the cardiovasculature plays a role in AD pathogenesis. The possible involvement of GRKs in AD pathogenesis is an interesting notion, which may help bridge the gap in our understanding of the heart–brain connection in relation to neurovisceral damage and vascular complications in AD, since kinases of this family are known to regulate numerous receptor functions both in the brain, myocardium, and elsewhere. The aim of this review is to discuss our findings of overexpression of GRK2 in the context of the early pathogenesis of AD, because increased levels of GRK2 immunoreactivity were found in vulnerable neurons of AD patients as well as in a two-vessel occlusion (2-VO) mammalian model of ischaemia. Also, we consider the consequences for this overexpression as a loss of G-protein coupled receptor (GPCR) regulation, as well as suggest a potential role for GPCRs and GRKs in a unifying theory of AD pathogenesis, particularly in the context of cerebrovascular disease. We synthesize this newer information and attempt to put it into context with GRKs as regulators of diverse physiological cellular functions that could be appropriate targets for future pharmacological intervention.  相似文献   

13.
G protein-coupled receptor kinase 5 (GRK5) is a serine/threonine kinase whose dysfunction results in cognitive impairment and Alzheimer-like pathology, including tau hyperphosphorylation. However, the mechanisms whereby GRK5 influences tau phosphorylation remain incompletely understood. In the current study, we showed that GRK5 influenced the phosphorylation of tau via glycogen synthase kinase 3β (GSK3β). The activity of both tau and GSK3β in the hippocampus was increased in aged GRK5-knockout mice, which is consistent with what occurs in APP/PS1 transgenic mice. Furthermore, GRK5 regulated the activity of GSK3β and phosphorylated tau in vitro. Regardless of changes of GRK5 protein levels, tau hyperphosphorylation remained reduced after GSK3β activity was inhibited, suggesting that GRK5 may specifically influence tau hyperphosphorylation by modulating GSK3β activity. Taken together, our findings suggest that GRK5 deficiency contributes to the pathogenesis of Alzheimer's disease by influencing the hyperphosphorylation of tau through the activation of GSK3β.  相似文献   

14.
15.
G-protein coupled receptor kinase-5 (GRK5) is a serine/threonine kinase discovered for its role in the regulation of G-protein coupled receptor signaling. Recent studies have shown that GRK5 is also an important regulator of signaling pathways stimulated by non-GPCRs. This study was undertaken to determine the physiological role of GRK5 in Toll-like receptor-4-induced inflammatory signaling pathways in vivo and in vitro. Using mice genetically deficient in GRK5 (GRK5(-/-) ) we demonstrate here that GRK5 is an important positive regulator of lipopolysaccharide (LPS, a TLR4 agonist)-induced inflammatory cytokine and chemokine production in vivo. Consistent with this role, LPS-induced neutrophil infiltration in the lungs (assessed by myeloperoxidase activity) was markedly attenuated in the GRK5(-/-) mice compared to the GRK5(+/+) mice. Similar to the in vivo studies, primary macrophages from GRK5(-/-) mice showed attenuated cytokine production in response to LPS. Our results also identify TLR4-induced NFκB pathway in macrophages to be selectively regulated by GRK5. LPS-induced IκBα phosphorylation, NFκB p65 nuclear translocation, and NFκB binding were markedly attenuated in GRK5(-/-) macrophages. Together, our findings demonstrate that GRK5 is a positive regulator of TLR4-induced IκBα-NFκB pathway as well as a key modulator of LPS-induced inflammatory response.  相似文献   

16.
Membrane G protein-coupled receptor kinase 5 (GRK5) deficiency is linked to Alzheimer disease, yet its precise roles in the disease pathogenesis remain to be delineated. We have previously demonstrated that GRK5 deficiency selectively impairs desensitization of presynaptic M2 autoreceptors, which causes presynaptic M2 hyperactivity and inhibits acetylcholine release. Here we report that inactivation of one copy of Grk5 gene in transgenic mice overexpressing β-amyloid precursor protein (APP) carrying Swedish mutations (Tg2576 or APPsw) resulted in significantly increased β-amyloid (Aβ) accumulation, including increased Aβ(+) plaque burdens and soluble Aβ in brain lysates and interstitial fluid (ISF). In addition, secreted β-APP fragment (sAPPβ) also increased, whereas full-length APP level did not change, suggesting an alteration in favor of β-amyloidogenic APP processing in these animals. Reversely, perfusion of methoctramine, a selective M2 antagonist, fully corrected the difference between the control and GRK5-deficient APPsw mice for ISF Aβ. In contrast, a cholinesterase inhibitor, eserine, although significantly decreasing the ISF Aβ in both control and GRK5-deficient APPsw mice, failed to correct the difference between them. However, combining eserine with methoctramine additively reduced the ISF Aβ further in both animals. Altogether, these findings indicate that GRK5 deficiency accelerates β-amyloidogenic APP processing and Aβ accumulation in APPsw mice via impaired cholinergic activity and that presynaptic M2 hyperactivity is the specific target for eliminating the pathologic impact of GRK5 deficiency. Moreover, a combination of an M2 antagonist and a cholinesterase inhibitor may reach the maximal disease-modifying effect for both amyloid pathology and cholinergic dysfunction.  相似文献   

17.
G protein-coupled receptor kinases (GRKs) mediate desensitization of agonist-occupied G protein-coupled receptors (GPCRs). Here we report that GRK5 contains a DNA-binding nuclear localization sequence (NLS) and that its nuclear localization is regulated by GPCR activation, results that suggest potential nuclear functions for GRK5. As assessed by fluorescence confocal microscopy, transfected and endogenous GRK5 is present in the nuclei of HEp2 cells. Mutation of basic residues in the catalytic domain of GRK5 (between amino acids 388 and 395) results in the nuclear exclusion of the mutant enzyme (GRK5(Delta)(NLS)), demonstrating that GRK5 contains a functional NLS. The nuclear localization of GRK5 is subject to dynamic regulation. Calcium ionophore treatment or activation of Gq-coupled muscarinic-M3 receptors promotes the nuclear export of the kinase in a Ca(2+)/calmodulin (Ca(2+)/CaM)-dependent fashion. Ca(2+)/CaM binding to the N-terminal CaM binding site of GRK5 mediates this effect. Furthermore, GRK5, but not GRK5(Delta)(NLS) or GRK2, binds specifically and directly to DNA in vitro. Consistent with their presence in the nuclei of transfected cells, all the GRK4, but not GRK2, subfamily members contain putative NLSs. These results suggest that the GRK4 subfamily of GRKs may play a signaling role in the nucleus and that GRK4 and GRK2 subfamily members perform divergent cellular functions.  相似文献   

18.
G-protein coupled receptor kinase-5 (GRK5) is a recently described NFκB regulator in TLR4 signaling pathway. To determine whether the role of GRK5 is MyD88- or TRIF-dependent, we injected wild type and GRK5 knockout mice with Pam3CSK4 (MyD88-dependent TLR1/2 ligand) and Poly(I:C) (TRIF-dependent TLR3 ligand) and examined the in vivo systemic inflammatory response. Our results demonstrate that GRK5 regulates IL-12p40 and G-CSF via a mechanism that is common to both MyD88 and TRIF. However, GRK5 regulates IL-5 and MCP-1 in a MyD88-dependent but TNFα in a TRIF-dependent manner. Together, our results demonstrate multiple roles of GRK5 in TLR signaling.  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