首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 218 毫秒
1.
Fertilin是一种异二聚体精子表面蛋白.业已证明,它在精卵反应中起着重要作用.成熟的Fertilinβ亚基的氨基端93肽含有与整联蛋白结合的整联蛋白配体区.该整联蛋白配体区与蛇毒整联蛋白配体区高度同源,它们能与细胞表面的整联蛋白相结合.在此我们报道猴成熟Fertilinβ氨基端93肽基因的克隆和在大肠杆菌中的表达.用RT-PCR方法获得mmFβNTP93基因,克隆入表达载体pT7-7/hCGβ的EcoRI和BamHI位点,该基因与hCGβ基因一起表达融合蛋白mFβNTP93-hCGβ,Western blotting结果显示,融合蛋白的表观分子量为33kDa,能与抗hCG抗体专一性结合.由于mFβNTP93基因与hCGβ基因相连,因此我们推测,mFβNTP93基因已获得表达.  相似文献   

2.
目的:利用酵母双杂交技术筛选与雌激素受体(ER)αAF1转录激活结构域相互作用的蛋白,为乳腺癌发生、发展机制的研究奠定基础。方法:将编码ERαAF1的cDNA片段克隆到诱饵蛋白载体pGBKT7中,以构建的pGBKT7-ERα-AF1为表达靶蛋白的质粒,筛选人乳腺文库。将筛选到的含Ⅰ型胶原基因的质粒与表达ERα和ERβ不同结构域的质粒共转化酵母细胞,验证Ⅰ型胶原与ERαAF1作用的特异性。结果:经酶切鉴定,证实重组质粒pGBKT7-ERα-AF1含有目的基因片段;Western印迹证实ERαAF1在酵母中获得表达;酵母双杂交筛选得到与ERαAF1相互作用的Ⅰ型胶原蛋白。酵母细胞共转化实验证实,Ⅰ型胶原蛋白与ERα和ERβ的AF1结合,但与ERβ的DBD、AF2不结合。结论:Ⅰ型胶原与ERα和ERβ的AF1及ERβ的DBD存在相互作用。  相似文献   

3.
整联蛋白是广泛存在于真核细胞表面的完整的膜受体家族,包括由至少18种不同的α亚基及8种β亚基形成的20多种αβ异二聚体。整联蛋白配体主要有胶原蛋白、纤维结合蛋白、层粘连蛋白、玻连蛋白、血小板凝血酶敏感蛋白、胞间黏附分子、细胞反受体、补体蛋白,以及多种细菌和病毒蛋白,在介导血管内皮细胞和肿瘤细胞的黏附、淋巴细胞运输、肿瘤生长及感染等都有重要的作用。  相似文献   

4.
精子头后部(或赤道区)表面fertilin糖蛋白由相关的两个跨膜亚基α和β构成异二体形式。这两个亚基前体均含有金属蛋白酶区(met-alloprotease domain)和整联蛋白配体区(disin-tegrin domain),属于ADAMs gene家族。α和β前体分别在睾丸和附睾中从上述两区域连接处水解后,得到成熟型亚基。受精时,穿过透明带的顶体反应后精子借助β亚基的disintegrin肽段与卵母细胞表面的整联蛋白结合,同时fertilin结构发生变化,暴露出α亚基上潜在的融合肽段(90—111aa),并介导精子与卵母细胞发生质膜融合,最终完成受精过程。  相似文献   

5.
为了探讨机械拉伸应变对人肺上皮细胞表面整联蛋白α3 、α5和β1分布的调控作用,建立了体外周期性拉伸应变装置,并应用胞外基质蛋白——纤连蛋白(Fn)、胶原蛋白Ⅳ(Col Ⅳ)裱衬基底膜,激光共聚焦显微镜分析了在应变为15%,频率为40次/min的拉伸刺激下,正常人肺上皮细胞H727表面α3、α5和β1整联蛋白的再分布.结果表明:在人肺上皮细胞H727中,α3、α5和β1整联蛋白是对拉伸应变敏感的膜受体,周期性拉伸刺激可诱导其激活,使之发生分布的重组,并向基底层转移,形成局部粘附连接,增强了整联蛋白受体与其特异性配体的结合能力.结果提示:一个有效的局部粘附连接的形成是受体聚集和配体占据共同启动的一个协调反应,该反应可能参与了细胞响应机械应力的起始过程,可能进一步通过力-化学信号的耦合或张力整合的形式,最终对细胞的生物学行为产生影响.  相似文献   

6.
口蹄疫是由口蹄疫病毒(FMDV)引起的一种高度接触性传染病,主要侵害偶蹄动物。乳鼠常作为一种重要的实验动物模型用于FMDV的研究;整联蛋白αvβ6是FMDV的重要受体之一。为深入研究整联蛋白αvβ6在FMDV感染乳鼠中所发挥的作用,克隆了乳鼠整联蛋白αvβ6的两个亚基,并将其导入中国仓鼠卵巢细胞(Chinese hamster ovary,CHO-677)基因组中,构建了稳定表达乳鼠整联蛋白αv和β6亚基的细胞系CHO-677-mαvβ6,并分别选用两种不同血清型的野生型FMDV毒株Asia1/HN/CHA/06和O/BY/CHA/2010感染细胞系来分析细胞系对FMDV的易感性。首先通过PCR和间接免疫荧光试验证明了细胞系中整联蛋白αvβ6在基因水平成功导入,在蛋白水平成功表达。然后,通过实时荧光定量RT-PCR检测病毒RNA拷贝数,并结合TCID50试验测定了代表毒株在两个细胞上的生长曲线。结果表明,与亲本细胞CHO-677相比,细胞系CHO-677-mαvβ6对FMDV更易感,从αvβ6的功能性上进一步验证了细胞系被成功构建。  相似文献   

7.
HtrA2/Omi是一种线粒体丝氨酸蛋白酶, 在哺乳动物细胞中具有双重功能, 即诱导细胞凋亡和参与维持线粒体活性的动态平衡. PDZ结构域是最重要的蛋白质相互作用结构域之一, 参与多种生物学过程, 如细胞信号转导、蛋白质降解、细胞骨架组织等. 最近研究发现, HtrA2/Omi蛋白的PDZ结构域与配体的相互作用, 可以调节HtrA2/Omi蛋白自身的水解酶活性.以HtrA2/Omi PDZ结构域为研究对象, 用酵母双杂交系统验证性筛选PDZ结构域结合配体文库, 快速研究该结构域的结合特性, 并在人类全蛋白质组范围内预测并发现该结构域新的相互作用蛋白, 最后分析这些新的相互作用所能够形成的最小相互作用网络来评估其可信度. 研究结果揭示了HtrA2/Omi PDZ结构域新的结合特性, 即: 不仅能够结合已报道的II类PDZ配体而且还可以结合I类和III类PDZ配体, 并且配体-3位氨基酸具有一定范围内的可变性. 而且, 发现了7个新的HtrA2/Omi PDZ结构域相互作用蛋白, 为进一步阐明HtrA2/Omi蛋白的生物学功能提供了重要线索. 同时证明了验证性筛选目的结构域结合配体文库, 这一结构域结合特性研究新策略的实用性和高效性.  相似文献   

8.
VLDL-受体的配体结合结构域结构分析   总被引:1,自引:1,他引:0  
极低密度脂蛋白受体(VLDL-R)的配体结合域具有8个富含半胱氨酸的配体结合重复序列(ligand-binding repeats,LBR),被认为是与配体结合的部位。该受体与含7个类似重复序列的低密度脂蛋白受体(LDL-R)的配体结合特性明显不同。为了明确VLDL-R中8个LBR在配体结合中的作用并探讨结合位点的结构,本研究采用计算机辅助蛋白质结构预测方法,在二级结构分析的基础上,通过同源建模方法预测受体N-端328个氨基酸的配体结合域空间结构,结果显示该区域呈现弧形口袋样结构,其中前3个LBR结构紧凑,呈棒状,负电荷相对集中,推测这一特征结构是配体结合的重要结构基础,结构分析同时表明在LBR5与LBR6之间连接区的弹性结构可以赋予结合位点一定的伸缩性,利于其与不同配体的结合。本研究预测结果首次提出了VLDL-R配体结构域结构及结合位点的结构特征,并与已有的实验结果一致。  相似文献   

9.
蔡婷  查锡良 《生命的化学》1999,19(6):257-260
整联蛋白是由α、β两种跨膜亚基组成的异源二聚体,是介导胞外基质(ECM)粘附的主要细胞表面受体家族。整联蛋白介导的粘附作用参与调节多种细胞功能。近十年间对整联蛋白的研究已从发现其家族的新成员转至描述各种整联蛋白的具体功能,主要热点在于:1.整联蛋白是细胞迁移的关键效应分子;2.整联蛋白与生长因子受体合作促进细胞的增殖,其原因至少部分是由于锚定依赖在细胞周期中由G1到S期所起的作用;3.粘附是细胞退出细胞周期开始分化的必要条件;4.当粘附的组织细胞脱离ECM时,则丧失了存活信号而发生凋亡[1]。近…  相似文献   

10.
整合素的构象变化与亲和力调控   总被引:2,自引:0,他引:2  
整合素(integrin)是由α、β两个亚单位通过非共价键连接而组成的异源二聚体。每种α、β亚单位都是含有多种结构域的大分子量Ⅰ型穿膜糖蛋白。它在细胞与细胞间、细胞与基质间相互作用的过程中发挥着十分关键的作用。整合素多种结构域的空间排列决定了其构象特征,而整合素的不同构象状态与其亲和力呈高度相关。对αVβ133整合素晶体结构的解析使我们对整合素的结构与功能有了更进一步的理解。  相似文献   

11.
Integrin-ligand interactions are regulated in a complex manner by divalent cations, and previous studies have identified ligand-competent, stimulatory, and inhibitory cation-binding sites. In collagen-binding integrins, such as alpha2beta1, ligand recognition takes place exclusively at the alpha subunit I domain. However, activation of the alphaI domain depends on its interaction with a structurally similar domain in the beta subunit known as the I-like or betaI domain. The top face of the betaI domain contains three cation-binding sites: the metal-ion dependent adhesion site (MIDAS), the ADMIDAS (adjacent to MIDAS), and LIMBS (ligand-associated metal-binding site). The role of these sites in controlling ligand binding to the alphaI domain has yet to be elucidated. Mutation of the MIDAS or LIMBS completely blocked collagen binding to alpha2beta1; in contrast mutation of the ADMIDAS reduced ligand recognition but this effect could be overcome by the activating monoclonal antibody TS2/16. Hence, the MIDAS and LIMBS appear to be essential for the interaction between alphaI and betaI, whereas occupancy of the ADMIDAS has an allosteric effect on the conformation of betaI. An activating mutation in the alpha2 I domain partially restored ligand binding to the MIDAS and LIMBS mutants. Analysis of the effects of Ca(2+), Mg(2+), and Mn(2+) on ligand binding to these mutants showed that the MIDAS is a ligand-competent site through which Mn(2+) stimulates ligand binding, whereas the LIMBS is a stimulatory Ca(2+)-binding site, occupancy of which increases the affinity of Mg(2+) for the MIDAS.  相似文献   

12.
The Asp of the RGD motif of the ligand coordinates with the β I domain metal ion dependent adhesion site (MIDAS) divalent cation, emphasizing the importance of the MIDAS in ligand binding. There appears to be two distinct groups of integrins that differ in their ligand binding affinity and adhesion ability. These differences may be due to a specific residue associated with the MIDAS, particularly the β3 residue Ala252 and corresponding Ala in the β1 integrin compared to the analogous Asp residue in the β2 and β7 integrins. Interestingly, mutations in the adjacent to MIDAS (ADMIDAS) of integrins α4β7 and αLβ2 increased the binding and adhesion abilities compared to the wild-type, while the same mutations in the α2β1, α5β1, αVβ3, and αIIbβ3 integrins demonstrated decreased ligand binding and adhesion. We introduced a mutation in the αIIbβ3 to convert this MIDAS associated Ala252 to Asp. By combination of this mutant with mutations of one or two ADMIDAS residues, we studied the effects of this residue on ligand binding and adhesion. Then, we performed molecular dynamics simulations on the wild-type and mutant αIIbβ3 integrin β I domains, and investigated the dynamics of metal ion binding sites in different integrin-RGD complexes. We found that the tendency of calculated binding free energies was in excellent agreement with the experimental results, suggesting that the variation in this MIDAS associated residue accounts for the differences in ligand binding and adhesion among different integrins, and it accounts for the conflicting results of ADMIDAS mutations within different integrins. This study sheds more light on the role of the MIDAS associated residue pertaining to ligand binding and adhesion and suggests that this residue may play a pivotal role in integrin-mediated cell rolling and firm adhesion.  相似文献   

13.
Raborn J  Wang W  Luo BH 《Biochemistry》2011,50(12):2084-2091
The ability of αIIbβ3 to bind ligands and undergo outside-in signaling is regulated by three divalent cation binding sites in the β I domain. Specifically, the metal ion-dependent adhesion site (MIDAS) and the synergistic metal binding site (SyMBS) are thought to be required for ligand binding due to their synergy between Ca(2+) and Mg(2+). The adjacent to MIDAS (ADMIDAS) is an important ligand binding regulatory site that also acts as a critical link between the β I and hybrid domains for signaling. Mutations in this site have provided conflicting results for ligand binding and adhesion in different integrins. We have mutated the β3 SyMBS and ADMIDAS. The SyMBS mutant abolished ligand binding and outside-in signaling, but when an activating glycosylation mutation in the αIIb Calf 2 domain was introduced, the ligand binding affinity and signaling were restored. Thus, the SyMBS is important but not absolutely required for integrin bidirectional signaling. The ADMIDAS mutants showed reduced ligand binding affinity and abolished outside-in signaling, and the activating glycosylation mutation could fully restore integrin signaling of the ADMIDAS mutant. We propose that the ADMIDAS ion stabilizes the low-affinity state when the integrin headpiece is in the closed conformation, whereas it stabilizes the high-affinity state when the headpiece is in the open conformation with the swung-out hybrid domain.  相似文献   

14.
Three divalent cation binding sites in the integrin β I domain have been shown to regulate ligand binding and adhesion. However, the degree of ligand binding and adhesion varies among integrins. The αLβ2 and α4β7 integrins show an increase in ligand binding affinity and adhesion when one of their ADMIDAS (adjacent to MIDAS, or the metal ion-dependent adhesion site) residues is mutated. By contrast, the α2β1, α5β1, and αIIbβ3 integrins show a decrease in binding affinity and adhesion when their ADMIDAS is mutated. Our study here indicated that integrin αVβ3 had lower affinity when the ADMIDAS was mutated. By comparing the primary sequences of these integrin subunits, we propose that one residue associated with the MIDAS (β3 Ala(252)) may account for these differences. In the β1 integrin subunit, the corresponding residue is also Ala, whereas in both β2 and β7 integrin subunits, it is Asp. We mutated the β3 residue Ala(252) to Asp and combined this mutant with mutations of one or two ADMIDAS residues. The mutant A252D showed reduced ligand binding affinity and adhesion. The ligand binding affinity and adhesion were increased when this A252D mutant was paired with mutations of one ADMIDAS residue. But when paired with mutations of two ADMIDAS residues the mutant nearly abolished ligand-binding ability, which was restored by the activating glycosylation mutation. Our study suggests that the variation of this residue contributes to the different ligand binding affinities and adhesion abilities among different integrin families.  相似文献   

15.
Integrins are a family of α/β heterodimeric adhesion metalloprotein receptors and their functions are highly dependent on and regulated by different divalent cations. Recently advanced studies have revolutionized our perception of integrin metal ion-binding sites and their specific functions. Ligand binding to integrins is bridged by a divalent cation bound at the MIDAS motif on top of either α I domain in I domain-containing integrins or β I domain in α I domain-less integrins. The MIDAS motif in β I domain is flanked by ADMIDAS and SyMBS, the other two crucial metal ion binding sites playing pivotal roles in the regulation of integrin affinity and bidirectional signaling across the plasma membrane. The β-propeller domain of α subunit contains three or four β-hairpin loop-like Ca2+-binding motifs that have essential roles in integrin biogenesis. The function of another Ca2+-binding motif located at the genu of α subunit remains elusive. Here, we provide an overview of the integrin metal ion-binding sites and discuss their roles in the regulation of integrin functions.  相似文献   

16.
Integrins are a family of α/β heterodimeric adhesion metalloprotein receptors and their functions are highly dependent on and regulated by different divalent cations. Recently advanced studies have revolutionized our perception of integrin metal ion-binding sites and their specific functions. Ligand binding to integrins is bridged by a divalent cation bound at the MIDAS motif on top of either α I domain in I domain-containing integrins or β I domain in α I domain-less integrins. The MIDAS motif in β I domain is flanked by ADMIDAS and SyMBS, the other two crucial metal ion binding sites playing pivotal roles in the regulation of integrin affinity and bidirectional signaling across the plasma membrane. The β-propeller domain of α subunit contains three or four β-hairpin loop-like Ca2+-binding motifs that have essential roles in integrin biogenesis. The function of another Ca2+-binding motif located at the genu of α subunit remains elusive. Here, we provide an overview of the integrin metal ion-binding sites and discuss their roles in the regulation of integrin functions.  相似文献   

17.
We have applied hydrogen-deuterium exchange mass spectrometry, in conjunction with differential scanning calorimetry and protein stability analysis, to examine solution dynamics of the integrin α1 I domain induced by the binding of divalent cations, full-length type IV collagen, or a function-blocking monoclonal antibody. These studies revealed features of integrin activation and α1I-ligand complexes that were not detected by static crystallographic data. Mg2+ and Mn2+ stabilized α1I but differed in their effects on exchange rates in the αC helix. Ca2+ impacted α1I conformational dynamics without altering its gross thermal stability. Interaction with collagen affected the exchange rates in just one of three metal ion-dependent adhesion site (MIDAS) loops, suggesting that MIDAS loop 2 plays a primary role in mediating ligand binding. Collagen also induced changes consistent with increased unfolding in both the αC and allosteric C-terminal helices of α1I. The antibody AQC2, which binds to α1I in a ligand-mimetic manner, also reduced exchange in MIDAS loop 2 and increased exchange in αC, but it did not impact the C-terminal region. This is the first study to directly demonstrate the conformational changes induced upon binding of an integrin I domain to a full-length collagen ligand, and it demonstrates the utility of the deuterium exchange mass spectrometry method to study the solution dynamics of integrin/ligand and integrin/metal ion interactions. Based on the ligand and metal ion binding data, we propose a model for collagen-binding integrin activation that explains the differing abilities of Mg2+, Mn2+, and Ca2+ to activate I domain-containing integrins.  相似文献   

18.
Conformational changes in integrins are important for efficient ligand binding during activation. We proposed that the I domain of the integrin lymphocyte function-associated antigen 1 (LFA-1) could exist in both open and closed conformations and generated constitutively activated LFA-1 by locking the I domain in the open conformation. Here we provide structural and biochemical evidence to validate conformational change in the I domain of LFA-1 upon activation. Two monoclonal antibodies to alpha(L), HI111 and CBR LFA-1/1, bind wild-type LFA-1 well, but their binding is significantly reduced when LFA-1 is locked in the open conformation. Furthermore, this reduction in monoclonal antibody binding also occurs when LFA-1 is activated by divalent cations. HI111 maps to the top region of the I domain that is close to the putative ligand-binding site surrounding the MIDAS (metal ion-dependent adhesion site). The epitope of CBR LFA-1/1 is at the C-terminal segment of the I domain that links to the beta-propeller, and undergoes a large movement between the open and closed conformations. Our data demonstrate that these two regions undergo significant conformational changes during LFA-1 activation and that the I domain of activated LFA-1 adopts a similar tertiary structure as the predicted locked open form.  相似文献   

19.
Mao D  Lü S  Li N  Zhang Y  Long M 《PloS one》2011,6(8):e24188
β2 integrin of lymphocyte function-associated antigen-1 (LFA-1) or macrophage-1 antigen (Mac-1) binds to their common ligand of intercellular adhesion molecule-1 (ICAM-1) and mediates leukocyte-endothelial cell (EC) adhesions in inflammation cascade. Although the two integrins are known to have distinct functions, the corresponding micro-structural bases remain unclear. Here (steered-)molecular dynamics simulations were employed to elucidate the conformational stability of α subunit I domains of LFA-1 and Mac-1 in different affinity states and relevant I domain-ICAM-1 interaction features. Compared with low affinity (LA) Mac-1, the LA LFA-1 I domain was unstable in the presence or absence of ICAM-1 ligand, stemming from diverse orientations of its α7-helix with different motifs of zipper-like hydrophobic junction between α1- and α7-helices. Meanwhile, spontaneous transition of LFA-1 I domain from LA state to intermediate affinity (IA) state was first visualized. All the LA, IA, and high affinity (HA) states of LFA-1 I domain and HA Mac-1 I domain were able to bind to ICAM-1 ligand effectively, while LA Mac-1 I domain was unfavorable for binding ligand presumably due to the specific orientation of S144 side-chain that capped the MIDAS ion. These results furthered our understanding in correlating the structural bases with their functions of LFA-1 and Mac-1 integrins from the viewpoint of I domain conformational stability and of the characteristics of I domain-ICAM-1 interactions.  相似文献   

20.
Murcia M  Jirouskova M  Li J  Coller BS  Filizola M 《Proteins》2008,71(4):1779-1791
A combination of experimental and computational approaches was used to provide a structural context for the role of the beta3 integrin subunit ligand-associated metal binding site (LIMBS) in the binding of physiological ligands to beta3 integrins. Specifically, we have carried out (1) adhesion assays on cells expressing normal alphaIIbeta3, normal alphaVbeta3, or the corresponding beta3 D217A LIMBS mutants; and (2) equilibrium and nonequilibrium (steered) molecular dynamics (MD) simulations of eptifibatide in complex with either a fully hydrated normal alphaIIbeta3 integrin fragment (alphaIIb beta-propeller and the beta3 betaA (I-like), hybrid, and PSI domains) or the equivalent beta3 D217A mutant. Normal alphaIIbeta3 expressing cells adhered to immobilized fibrinogen and echistatin, whereas cells expressing the alphaIIbeta3 D217A LIMBS mutant failed to adhere to either ligand. Similarly, the equivalent alphaVbeta3 mutant was unable to support adhesion to vitronectin or fibrinogen. The alphaIIbeta3 D217A mutation increased the binding of mAb AP5, which recognizes a ligand-induced binding site (LIBS) in the beta3 PSI domain, indicating that this mutation induced allosteric changes in the protein. Steered MD simulating the unbinding of eptifibatide from either normal alphaIIbeta3 or the equivalent beta3 D217A mutant suggested that the reduction in ligand binding caused by the LIMBS mutant required the loss of both the LIMBS and the metal ion-dependent adhesion site (MIDAS) metal ions. Our computational results indicate that the LIMBS plays a crucial role in ligand binding to alphaIIbeta3 by virtue of its effects on the coordination of the MIDAS.  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