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   151篇
  免费   16篇
  国内免费   3篇
基础医学   27篇
口腔科学   7篇
临床医学   7篇
内科学   34篇
皮肤病学   1篇
神经病学   6篇
外科学   16篇
综合类   21篇
预防医学   11篇
眼科学   10篇
药学   15篇
中国医学   2篇
肿瘤学   13篇
  2024年   1篇
  2023年   3篇
  2022年   16篇
  2021年   10篇
  2020年   9篇
  2019年   7篇
  2018年   3篇
  2017年   6篇
  2016年   3篇
  2015年   5篇
  2014年   4篇
  2013年   5篇
  2012年   4篇
  2011年   8篇
  2010年   2篇
  2009年   4篇
  2008年   7篇
  2007年   5篇
  2006年   6篇
  2005年   8篇
  2004年   8篇
  2003年   1篇
  2002年   6篇
  2001年   6篇
  2000年   4篇
  1999年   2篇
  1998年   2篇
  1997年   5篇
  1996年   6篇
  1995年   3篇
  1994年   1篇
  1993年   3篇
  1992年   1篇
  1991年   2篇
  1990年   1篇
  1987年   1篇
  1986年   1篇
  1980年   1篇
排序方式: 共有170条查询结果,搜索用时 31 毫秒
1.
面对《医疗事故处理条例》的实施医院应采取的对策   总被引:3,自引:0,他引:3  
为实施《医疗事故处理条例》,必须加强医院自身建设,规范管理。并从强化教育、严格准入制度、健全规章制度,规范病案与物证管理,尊重患者知情同意权,及时化解纠纷,分摊风险责任,维护合法权益等八个方面进行研讨提出对策。  相似文献   
2.
用放射免疫方法测定了不同胎龄胎儿胃肠道P物质和胃动素的含量。结果发现P物质在第10周胎儿胃肠道即可测及。以26~30周为最高。胃动素同样于第10周胎儿胃肠道测及,而且也以26~30周含量最高。这说明含有P物质和胃动素的内分泌细胞和神经元及神经纤维以26~30周发育最快。  相似文献   
3.
4.
目的:初步探究Toll样受体4(Toll like receptor 4,TLR4)在慢性牙周炎诱发大鼠肝脏炎症中的作用。方法:将24只雄性Wistar大鼠随机分成两组,对照组和牙周炎组每组各12只,2%戊巴比妥钠麻醉下检测牙龈出血指数、牙周探诊深度及松动度;将20μL牙龈卟啉单胞菌脂多糖(Porphyromonas gingivalis lipopolysaccharide,Pg-LPS)(1 g/L)注射到大鼠双侧上颌第一磨牙龈沟及第一二磨牙龈间隙内,隔1 d注射1次,每周3次,连续6周。对照组不做处理。最后一次注射Pg-LPS 12 h后,2%戊巴比妥钠麻醉下检测牙周探诊深度、牙龈出血指数及松动度等3项牙周指标后将大鼠处死,取其上颌骨及肝脏,采用组织学方法、实时荧光定量聚合酶链反应(quantitative real-time polymerase chain reaction,qRT-PCR)、Western blot分别检测肝组织炎症反应情况及TLR4、肿瘤坏死因子-α(tumor necrosis factor,TNF-α)和白细胞介素-6(interleukin-6,IL-6)的基因和蛋白表达水平。结果:牙周炎组大鼠牙周探诊深度、牙龈出血指数及松动度均重于对照组;苏木精-伊红(hematoxylin-eosin,HE)染色结果显示慢性牙周炎大鼠肝组织内肝索结构紊乱,汇管区可见大量淋巴细胞浸润,大量肝细胞呈空泡样改变;qRT-PCR结果显示慢性牙周炎组大鼠肝组织TLR4、TNF-α和IL-6的mRNA表达水平较对照组升高;Western blot结果显示TLR4蛋白表达与基因表达一致。结论:TLR4在慢性牙周炎诱发大鼠肝脏炎症性改变的过程中发挥着重要的作用。  相似文献   
5.
肝癌和癌旁组织中核仁组成区嗜银蛋白表达的研究   总被引:2,自引:0,他引:2  
核仁组成区(NORs)是细胞核中的DNA环,与核仁的形成及变化密切相关,硝酸银染色能特异地显示细胞核内含有rRNA基因的NORs。我们应用该技术观察45例肝癌及癌旁组织中AgNORs表达特征,发现AgNORs之基本形态为团块、颗粒和混合三型;正常、癌旁和肝癌组织的AgNORs数分别为1.88±0.22、3.58±0.78和7.92±3.01,依次递增明显,差别显著,其形态和分布亦异化悬殊;AgNORs计数和形态与HCC分化程度呈正相关,与HBV感染(80%的癌旁组织为HBV肝炎后肝硬化)之间的关系未能确定。Ag-NORs检测实用性强,值得进一步研究和应用。  相似文献   
6.
目的 研究类弹性蛋白多肽(ELP)作为一种黏膜下注射制剂在内镜黏膜下剥离术(ESD)中的应用,比较不同质量浓度的ELP在ESD中的应用,并对其有效性和安全性进行评估.方法 40只新西兰大白兔随机分为2组(n=20).第1组观察不同注射液分离组织的有效性.依据不同质量浓度的ELP(0.05、0.025、0.012 5、0.005 g/ml)及对照组(甘油果糖)随机分为5个亚组(n=4),切取胃组织,在胃窦或胃体处分别注射不同质量浓度的ELP和甘油果糖2ml,观察并记录药物注射后0、5、10、30 min局部黏膜隆起高度及黏膜表面的改变.第2组测量不同注射液的注射压力.亚组分组同第1组,使用注射器/压力计组件分别测量2ml不同质量浓度ELP和甘油果糖的注射压力.结果 注射药物后30 min内,0.05、0.025 g/ml ELP维持黏膜隆起高度均大于甘油果糖,差异具有统计学意义(P<0.05);0.012 5 g/ml ELP维持黏膜隆起高度与甘油果糖相比,差异无统计学意义(P>0.05);0.005 g/ml ELP维持黏膜隆起高度小于甘油果糖,且差异具有统计学意义(P<0.05).同时,注射压力与浓度相关,2 ml ELP(0.05、0.025、0.012 5、0.005 yml)和甘油果糖注射后的平均注射压力分别为(332±36)、(223±24)、(174±22)、(142±19)、(269±17)kPa,0.025 g/ml ELP注射压力小于甘油果糖,差异有统计学意义(P<0.05).结论 ELP是一种安全有效的新型黏膜下注射制剂,推荐ESD术中注射ELP的最适质量浓度为0.025 g/ml.  相似文献   
7.
卢雯湉  任青玲 《中医学报》2020,35(5):1029-1033
肾为天癸之源、气血之根,女子以血为主,经、孕、产、乳皆以血为用。月经来潮及其周期性演变需在心肾交济、肝脾协调的整体配合下周而复始地完成。心肾燮理阴阳,肝脾协调气血,气血阴阳调摄有度,月经周期圆运动才能节律适常。故本病的治疗以心肾为核心,辨证论治兼调四脏,使得阴平阳秘,达到调经促孕的目的。任青玲教授在补养心肾调周法的基础上提出滋阴补阳序贯法,结合卵巢局部发生胰岛素抵抗的表现是卵巢局部“痰浊”的特点,对于多囊卵巢综合征伴胰岛素抵抗的治疗强调经前期以补阳为主,常用右归饮;经后期以滋阴为要,常用滋肾生肝饮;经间期温化痰湿以治标,常用苍附导痰汤。治疗上以补肾为主,兼以化痰、疏肝、活血等方法,配合周期不同阶段用药,使治疗更具有针对性。  相似文献   
8.
Li  Wentian  Lai  Kaitao  Chopra  Neha  Zheng  Zhaomin  Das  Abhirup  Diwan  Ashish D. 《European spine journal》2022,31(4):917-925
Purpose

Low back pain (LBP), a widely prevalent and costly disease around the world, is mainly caused by intervertebral disc (IVD) degeneration (IDD). Although numerous factors may trigger this degenerative process, microbiome dysbiosis has recently been implicated as one of the likely causes. However, the exact relationship between the microbiome and IDD is not well understood. This review summarizes the potential mechanisms and discusses microbiome dysbiosis’s possible influence on IDD and LBP.

Methods

Prospective literature review.

Results

Alterations in microbiome composition and host responses to the microbiota causing pathological bone development and involution, led to the concept of gut-bone marrow axis and gut-bone axis. Moreover, the concept of the gut-disc axis was also proposed to explain the microbiome’s role in IDD and LBP. According to the existing evidence, the microbiome could be an important factor for inducing and aggravating IDD through changing or regulating the outside and inside microenvironment of the IVD. Three potential mechanisms by which the gut microbiota can induce IVD and cause LBP are: (1) translocation of the bacteria across the gut epithelial barrier and into the IVD, (2) regulation of the mucosal and systemic immune system, and (3) regulation of nutrient absorption and metabolites formation at the gut epithelium and its diffusion into the IVD. Furthermore, to investigate whether IVD is initiated by pathogenic bacteria and establish the correlation between the presence of certain microbial groups with the disease in question, microbiome diversity analysis based on16S rRNA data can be used to characterise stool/blood microbiota from IVD patients.

Conclusion

Future studies on microbiome, fungi and viruses in IDD is necessary to revolutionize our thinking about their possible role in the development of IVD diseases. Furthermore, we believe that inflammation inhibition and interruption of amplification of cascade reaction in IVD by targeting the gut and IVD microbiome is worthwhile for the treatment of IDD and LBP.

Level of Evidence I

Diagnostic: individual cross-sectional studies with the consistently applied reference standard and blinding.

  相似文献   
9.
The neonatal Fc receptor (FcRn) is a major regulator of IgG and albumin homeostasis systemically and in the kidneys. We investigated the role of FcRn in the development of immune complex–mediated glomerular disease in mice. C57Bl/6 mice immunized with the noncollagenous domain of the α3 chain of type IV collagen (α3NC1) developed albuminuria associated with granular capillary loop deposition of exogenous antigen, mouse IgG, C3 and C5b-9, and podocyte injury. High-resolution imaging showed abundant IgG deposition in the expanded glomerular basement membrane, especially in regions corresponding to subepithelial electron dense deposits. FcRn-null and -humanized mice immunized with α3NC1 developed no albuminuria and had lower levels of serum IgG anti-α3NC1 antibodies and reduced glomerular deposition of IgG, antigen, and complement. Our results show that FcRn promotes the formation of subepithelial immune complexes and subsequent glomerular pathology leading to proteinuria, potentially by maintaining higher serum levels of pathogenic IgG antibodies. Therefore, reducing pathogenic IgG levels by pharmacologic inhibition of FcRn may provide a novel approach for the treatment of immune complex–mediated glomerular diseases. As proof of concept, we showed that a peptide inhibiting the interaction between human FcRn and human IgG accelerated the degradation of human IgG anti-α3NC1 autoantibodies injected into FCRN-humanized mice as effectively as genetic ablation of FcRn, thus preventing the glomerular deposition of immune complexes containing human IgG.The MHC class I–like neonatal Fc receptor (FcRn), a heterodimer comprising a heavy chain and β2-microglobulin light chain, is the major regulator of IgG and albumin homeostasis.1 Perinatally, FcRn mediates the transfer of IgG from mother to offspring, across the placenta in primates and trans-intestinally in suckling rodents. Throughout life, FcRn protects IgG and albumin from catabolism, explaining the unusually long t1/2 and high serum levels of these proteins. IgG and albumin taken up by cells by pinocytosis bind strongly to FcRn at pH 6.0–6.5 in endosomes. FcRn-bound ligands are then recycled to the plasma membrane, where they dissociate at pH 7.4, whereas IgG and albumin not bound to FcRn are targeted to lysosomes for degradation. FcRn is thought to promote some autoimmune diseases because it protects pathogenic IgG from degradation. For instance, Fcrn−/− mice are resistant to passive transfer of arthritis by K/BxN sera and autoimmune skin pathology induced by antibodies targeting autoantigens at the dermal–epidermal junction, although this protection can be overcome by excess autoantibodies.24In kidneys, FcRn is expressed in podocytes and proximal tubular epithelial cells.5 Overall, renal FcRn reclaims albumin but facilitates elimination of IgG.6 Tubular FcRn mediates IgG transcytosis.7 Podocytes use FcRn to clear IgG from the glomerular basement membrane (GBM).8 IgG accumulates in the glomeruli of aged Fcrn−/− mice due to impaired clearance of IgG from the GBM, and saturating this clearance mechanism by excess ligand potentiates the pathogenicity of nephrotoxic sera in wild-type mice. Podocyte FcRn has been postulated to be involved in the clearance of immune complexes (ICs) present in pathologic conditions such as membranous nephropathy.5 Expression of FcRn in human podocytes is increased in various immune-mediated glomerular diseases.9 Given its role in IgG and albumin handling in the kidneys and systemically, FcRn can be expected to influence the development of immune-mediated kidney diseases at multiple levels. This conjecture awaits experimental verification.To determine the role of FcRn in IgG-mediated glomerular disease, we asked how FcRn deficiency alters the course of disease in mice immunized with the NC1 domain of α3 type IV collagen (α3NC1). We chose this antigen because of its reported ability to induce disease in C57Bl/6 (B6) mice,10 corroborated in pilot studies (Supplemental Figure 1). Fcrn−/− mice are hypoalbuminemic due to impaired albumin recycling,11 and also exhibit reduced urinary albumin excretion.12 As a control for this potential confounder, we used FCRN-humanized mice, which have normal serum albumin because human FcRn recycles mouse albumin but not mouse IgG.13All mice immunized with α3NC1 developed circulating mouse IgG anti-α3NC1 antibodies, which reached the maximum titer about 6 weeks later and gradually declined thereafter. At all times, the levels of mouse IgG anti-α3NC1 antibodies in sera from Fcrn−/− mice and FCRN-humanized mice were approximately 50%–70% lower than those in wild-type mouse sera (Figure 1A). The results were similar for mouse IgG1, IgG2b, and IgG2c anti-α3NC1 antibodies (Supplemental Figure 2). Wild-type B6 mice immunized with α3NC1 started developing progressive albuminuria 8–10 weeks later (Figure 1B). By week 14, the urinary albumin creatinine ratio increased approximately 100-fold, and hypoalbuminemia developed (Figure 1C). Urinary albumin excretion in Fcrn−/− mice and FCRN-humanized mice immunized with α3NC1 was not significantly higher than in adjuvant-immunized control mice. No mice developed renal failure (Supplemental Figure 3).Open in a separate windowFigure 1.FcRn ablation reduces serum levels of mouse IgG anti-α3NC1 antibodies and prevents the development of albuminuria in α3NC1-immunized mice. (A) The left panel shows circulating mIgG anti-α3NC1 antibodies from C57Bl6 wild-type mice (○), Fcrn−/− mice (□), FCRN-humanized (hFCRN) mice (◇), and the control CFA group (△), which are assayed by indirect ELISA in plates coated with α3NC1 (100 ng/well). Mouse sera are diluted 1:5000. The right panel shows the significance of circulating mIgG anti-α3NC1 antibody differences among groups at week 12, as assessed by one-way ANOVA followed by Bonferroni post tests for pairwise comparisons. (B) The left panel shows that the urinary albumin creatinine ratio (mean±SEM) time course is monitored in C57Bl6 wild-type mice (○), Fcrn−/− mice (□), and hFCRN mice (◇) immunized with α3NC1 (n=5–8 mice in each group, from two separate experiments). Mice in the control group (△) are immunized with adjuvant alone (n=9). The right panel shows the urinary albumin creatinine ratio (mean±SEM) at 14 weeks, when mice are euthanized. The significance of differences among groups is assessed by one-way ANOVA followed by Bonferroni post tests for pairwise comparisons. (C) The left panel shows SDS-PAGE analysis of serum (0.5 µl/lane) and urine samples (2 µl/lane) from CFA-immunized control mice (a) and α3NC1-immunized wild-type mice (b), Fcrn−/− mice (c), and hFCRN mice (d) collected at week 14. The right panel presents a densitometric analysis of the relative levels of albumin in mouse serum samples showing that α3NC1-immunized wild-type mice developed hypoalbuminemia. *P<0.05 by two-tailed t test versus CFA-immunized wild-type mice; **P<0.01; ***P<0.001. ns, not significant; WT, wild type.At 14 weeks after α3NC1 immunization, kidneys examined by light microscopy showed mild glomerular pathology, with few crescents and relatively little inflammation (Figure 2A), similar to α3NC1-immunized DBA/1 mice with comparable albuminuria.14,15 Electron microscopy showed extensive subepithelial IC deposits surrounded by an expanded GBM and effacement of podocyte foot processes in α3NC1-immunized B6 mice, whereas Fcrn−/− mice had fewer subepithelial deposits (Figure 2B, Supplemental Figure 4). Immunofluorescence staining showed granular capillary loop deposition of mouse IgG, exogenous antigen, C3, and C5b-9, more intense in wild-type mice than in Fcrn−/− mice and FCRN-humanized mice (Figure 2, Ca–Cp, Supplemental Figure 5). A loss of nephrin staining, indicative of podocyte injury, occurred in α3NC1-immunized B6 mice but not in Fcrn−/− mice or FCRN-humanized mice (Figure 2, Cq–Ct).Open in a separate windowFigure 2.FcRn deficiency reduces formation of pathogenic subepithelial ICs. (A) Light microscopic evaluation of kidneys from adjuvant-immunized control mice (a) and α3NC1-immunized wild-type mice (b) and Fcrn−/− mice (c) revealed few pathogenic changes and the absence of glomerular inflammation (periodic acid–Schiff staining). (B) Transmission electron microscopy shows normal GBM (arrow) and podocyte foot processes in control mice (a), extensive subepithelial electron dense deposits (arrowhead), thickened GBM, and podocyte foot process effacement in α3NC1-immunized wild-type mice (b), and fewer IC deposits in the Fcrn−/− mice (c). (C) Immunofluorescence analysis of kidneys from adjuvant-immunized control mice (a, e, i, m, and q) and α3NC1-immunized wild-type mice (b, f, j, n, and r), FcRn−/− mice (c, g, k, o, and s), and hFCRN mice (d, h, l, p, and t) evaluate the deposition of mouse IgG (a–d), exogenous α3NC1 antigen stained by mAb RH34 (e–h), mouse C3c (i–l), C5b-9 (m–p), and nephrin staining (q–t) at 14 weeks. Wild-type mice exhibit linear-granular GBM deposition of mouse IgG and granular GBM deposition of exogenous antigen, C3, and C5b-9, which are attenuated in Fcrn−/− mice and hFCRN mice and essentially absent in control mice. Compared with control mice, α3NC1-immunized wild-type mice but not Fcrn−/− or hFCRN mice exhibit a loss of nephrin staining, indicative of podocyte injury. WT, wild type; EM, electron microscopy, PAS, periodic acid–Schiff. Original magnification, ×400 in A; ×2850 in B; ×200 in C.Because B6 mice immunized with bovine GBM NC1 hexamers have normal kidney function and histology despite linear GBM deposition of IgG autoantibodies binding to mouse α345(IV) collagen (Supplemental Figure 1), the question arises as to what causes proteinuria in α3NC1-immunized mice. Because the clinical presentation, morphology, and effector mechanisms depend on where ICs are localized in the capillary wall, we compared IgG distribution in α3NC1-immunized mice and mice injected with anti-α3NC1 antibodies modeling anti-GBM autoantibodies. The distribution and relative abundance of mouse IgG, as imaged by immunoperoxidase immunoelectron microscopy and stochastic optical reconstruction microscopy (STORM), a method for super-resolution fluorescence microscopy, were concordant. In α3NC1-immunized mice, IgG deposition was abundant in the areas of expanded GBM and especially in regions corresponding to the subepithelial dense deposits seen by routine electron microscopy. By contrast, in mice injected with α3NC1-specific anti-GBM mAb, the IgG was confined to an ultrastructurally normal GBM that lacked subepithelial deposits (Figure 3).Open in a separate windowFigure 3.Localization of IgG by high-resolution imaging. The localization of mouse IgG in glomerular capillary walls of wild-type mice immunized with α3NC1 (A, C–E), or intravenously injected with anti-mouse α3NC1 IgG mAb 8D1 (B, F–H) is determined by immunoperoxidase electron microscopy (A and B) and STORM imaging (C–H). In A, the GBM is irregularly thickened, and abundant electron dense peroxidase reaction product is present in discontinuous, subepithelial patterns beneath broadly effaced podocyte foot processes (arrows). In B, the peroxidase reaction product is diffusely present throughout the GBM (arrowhead), but less abundant compared with A. Electron dense deposits are absent, and podocyte foot process architecture appears normal. (C–E) By STORM imaging, anti-agrin (blue) identifies both normal and thickened areas of the GBM, both of which contain dense accumulations of mouse IgG throughout (red). The electron microscopy correlation in E shows GBM staining with respect to the podocytes and endothelial cells. (F–H) IgG mAb 8D1 (red) is present in the GBM, which shows no evidence of thickening. CL, capillary lumen; EM, electron microscopy En, endothelium;Po, podocyte.Subepithelial ICs, a hallmark of human membranous nephropathy (MN), form when IgG antibodies bind to podocyte antigens, such as phospholipase A2 receptor (PLA2R) and neutral endopeptidase (NEP), or to planted antigens, such as cationic BSA.1618 Subsequent expansion of the GBM, complement activation, and podocyte injury by C5b-9 cause proteinuria. Although it is unexpected, formation of subepithelial ICs in α3NC1-immunized mice may be explained by exogenous α3NC1 deposited in glomeruli acting as a planted antigen.19 Alternatively, anti-α3NC1 antibodies in complex with α3NC1 antigen may act as surrogate antipodocyte antibodies, because α3NC1-containing ICs bind to podocytes.20 After four immunizations with α3NC1 monomers, B6 mice and DBA/1 mice eventually develop crescentic GN by 26 and 10 weeks, respectively.10,14 The combination of subepithelial ICs and crescentic anti-GBM antibody GN was most recently described in a series of eight patients with circulating anti-α3NC1 autoantibodies but undetectable anti-PLA2R autoantibodies.21In contrast to wild-type B6 mice, congenic Fcrn−/− mice and FCRN-humanized mice did not develop albuminuria after α3NC1 immunization. Their resistance to proteinuria was associated with lower serum titers of anti-α3NC1 IgG antibodies and reduced glomerular deposition of IgG, antigen, C3, and C5b-9. Because C5b-9 is an essential mediator of podocyte damage and proteinuria by subepithelial ICs,22,23 reduced complement activation potentially explains the attenuated glomerular pathology in FcRn-deficient mice. The resistance of FCRN-humanized mice indicates that FcRn promotes IC-mediated glomerular disease due to its interaction with IgG rather than albumin. We propose that FcRn promotes the development of subepithelial ICs and subsequent glomerular injury primarily by maintaining higher serum levels of pathogenic IgG (Supplemental Figure 6). However, we cannot formally exclude a possible pathogenic role of podocyte FcRn, whose stimulation by ICs may induce maladaptive signaling.9 Future studies in mice with podocyte-specific ablation of FcRn would address this possibility.Our findings identify FcRn as a potential target for therapeutic intervention in IC-mediated glomerular diseases, typically treated with nonspecific immunosuppressants that are toxic and sometimes ineffective. More specific therapies include ablation of B cells by rituximab. In patients with idiopathic MN who respond to rituximab therapy, serum levels of anti-PLA2R IgG autoantibodies decline over a period of many months, and their disappearance is followed by resolution of proteinuria.24 The slow decline in proteinuria is problematic for patients already suffering from complications of nephrotic syndrome, who would benefit from ancillary therapies that reduce pathogenic IgG antibodies more rapidly. This may be achieved by inhibiting FcRn.One implementation of this concept is therapy with high-dose intravenous Ig (HD-IVIG). HD-IVIG accelerates the degradation of IgG by saturating FcRn,25 one of the mechanisms that explain the beneficial effects of HD-IVIG therapy in some autoimmune diseases.3 In pregnant women with circulating anti-NEP alloantibodies mediating antenatal MN, treatment with HD-IVIG reduces the titers of IgG alloantibodies by approximately 30% within 2–3 weeks.26 However, HD-IVIG is inefficient, because large amounts of IgG (1–2 g/kg) cause relatively modest reductions in pathogenic IgG titers. Specific FcRn inhibitors recapitulate this activity of HD-IVIG more effectively at lower doses. By reducing pathogenic IgG levels, function-blocking anti-FcRn mAbs ameliorate experimental myasthenia gravis in rats,27 and engineered IgG “Abdegs” that bind with high affinity to FcRn ameliorate arthritis transferred by K/BxN serum.28To assess the translational potential of our findings, we asked whether pharmacologic blockade of human FcRn can reproduce the effects of genetic FcRn deficiency. To this end, FCRN-humanized and Fcrn−/− mice were passively immunized with human IgG containing anti-α3NC1 (Goodpasture) autoantibodies. To inhibit human FcRn, we used a lysine analog of SYN1436 (Figure 4A),29 a peptide that binds with subnanomolar affinity to human FcRn, thus preventing IgG binding.30 In vivo, SYN1436 reduces IgG levels in cynomolgus monkeys by 80%.30 Serum anti-α3NC1 autoantibodies in FCRN-humanized mice treated with anti-FcRn peptide, but not with control peptide, sharply decreased to the same levels as in Fcrn−/− mice (Figure 4B), and were no longer detected after 4 days. In mice, human IgG elicits murine anti-human IgG antibodies, forming ICs that can deposit in glomeruli, as shown in active serum sickness models. Glomerular deposition of ICs containing human IgG was abolished in mice treated with anti-FcRn peptide, but not with control peptide (Figure 4C). Linear GBM deposition of human anti-GBM IgG was not observed, because the epitopes recognized by Goodpasture autoantibodies are completely inaccessible in the mouse GBM.31 These results provide proof of concept that therapies targeting human FcRn effectively lower serum levels of pathogenic human IgG autoantibodies, which could be beneficial in patients with IgG-mediated kidney diseases. Because FcRn also mediates the trans-placental transfer of IgG from mother to the fetus, FcRn inhibition may be particularly attractive for preventing antenatal MN caused by maternal anti-NEP alloantibodies.Open in a separate windowFigure 4.Pharmacologic blockade of human FcRn accelerates the catabolism of human IgG autoantibodies in FCRN-humanized mice. (A) Structure of a peptide that binds with high affinity to human FcRn, competitively inhibiting its interaction with human IgG (top). The control peptide (bottom) containing D-amino acids does not bind to human FcRn. Pen, Sar, and NMeLeu denote penicillamine, sarcosine, and N-methyl-leucine, respectively. (B) Serum level of human IgG anti-α3NC1 antibodies in FCRN-humanized mice treated with anti-FcRn peptide (▪) or control peptide (●) and in Fcrn−/− (▲) mice sera (n=3 in each group) is analyzed by indirect ELISA in plates coated with α3NC1 (100 ng/well). Mouse sera are diluted 1:500. (C) Kidney deposition of human IgG (a and b) and mouse IgG (c and d) in FCRN-humanized mice treated with control peptide (a and c) or anti-FcRn peptide (b and d) is evaluated by direct immunofluorescence staining. Treatment with anti-FcRn peptide prevents the glomerular deposition of ICs containing human IgG.  相似文献   
10.
hMLH1基因高甲基化在胃癌发生、发展中的作用   总被引:1,自引:0,他引:1  
基因启动子区CpG岛甲基化使许多基因失活,从而导致恶性肿瘤的发生和发展。目的:检测hMLHl基因启动子区CpG岛甲基化水平,探讨其胃癌发生、发展中的作用。方法:以甲基化特异性聚合酶链反应(MSP)检测41例胃癌、40例癌前病变和38例对照组织中hMLHl基因启动子区CpG岛甲基化状态,并分析其与胃癌患者临床病理特征的关系。结果:胃癌组织中hMLHl基因启动子区CpG岛甲基化阳性率为34.1%,显著高于癌前病变组的5.0%和对照组的0%(P〈0.05)。hMLHl基因甲基化阳性率与胃癌患者的年龄和肿瘤浸润深度有关(阳性率分别为46.4%对7.7%和55.0%对14.3%,P〈0.05),与性别、肿瘤分化程度和淋巴结转移无关(阳性率分别为34.8%对33.3%、28.0%对43.8%和38.1%对30.0%)。结论:胃癌组织中存在hMLHl基因启动子区CpG岛高甲基化,可能与胃癌的发生、发展有关.且可能在老年胃癌患者的肿瘤发生过程中起重要作用。  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