A novel method for protein subcellular localization Combining residue-couple model and SVM

合集下载

Nature RNA_immunoprecipitation_protocol

Nature RNA_immunoprecipitation_protocol

© 2006 Nature Publishing Group /natureprotocols
RNA targets of multitargeted RNA-binding proteins (RBPs) can be studied by various methods including mobility shift assays, iterative in vitro selection techniques and computational approaches. These techniques, however, cannot be used to identify the cellular context within which mRNAs associate, nor can they be used to elucidate the dynamic composition of RNAs in ribonucleoprotein (RNP) complexes in response to physiological stimuli. But by combining biochemical and genomics procedures to isolate and identify RNAs associated with RNA-binding proteins, information regarding RNA–protein and RNA–RNA interactions can be examined more directly within a cellular context. Several protocols including the yeast three-hybrid system and immunoprecipitations that use physical or chemical cross-linking have been developed to address this issue. Cross-linking procedures in general, however, are limited by inefficiency and sequence biases. The approach outlined here, termed RNP immunoprecipitation−microarray (RIP-Chip), allows the identification of discrete subsets of RNAs associated with multi-targeted RNA-binding proteins and provides information regarding changes in the intracellular composition of mRNPs in response to physical, chemical or developmental inducements of living systems. Thus, RIP-Chip can be used to identify subsets of RNAs that have related functions and are potentially co-regulated, as well as proteins that are associated with them in RNP complexes. Using RIP-Chip, the identification and/or quantification of RNAs in RNP complexes can be accomplished within a few hours or days depending on the RNA detection methisolation and identification of mRNAs, microRNAs and protein components of ribonucleoprotein complexes from cell extracts

拟南芥alpha-Dioxygenase 2原核表达,纯化及亚细胞定位预测

拟南芥alpha-Dioxygenase 2原核表达,纯化及亚细胞定位预测

,2002 a,b)。van der Biezen .(1996)首次利用转
座子标签法从在番茄中分离到一个新基因,该基因
突变导致生长缓慢、植株矮小柔弱、结实率低等,因
而命名为
。Sanz (1998)通过差异显示
技术从烟草中分离到参与细菌诱导的超敏反应过程
的新基因,并命名为 (pathogen­induced oxyge­
spectively. A fusion protein about 98 kD was detected by SDS­PAGE and Western blot in the induced recombinant BL21 (DE3)­RIPL
codon+ strain, and it accounts for 38% of the total bacterial proteins. The target protein was purified by the GST chromatography col­
第5 期
张美祥等: 拟南芥 Alpha­dioxygenase 2 原核表达、纯化及亚细胞定位预测
817
们蛋白序列的相似性为 71.4%。拟南芥 DOX1 在蛋
白序列、表达模式与最初的从烟草中分离到的
相似,其表达受病原及抗逆过程诱导(Koeduka
,2005),拟南芥 DOX2 与最初从番茄中分离到
nase),到目前为止研究者已先后从番茄、烟草、拟南
芥、水稻、小麦等植 物中分离 到 18 个 同源基 因
(Hamberg
,2005), 并 将 其 统 一 归 类 为 al­
pha­dioxygenase。
拟南芥中脂肪酸 alpha­dioxygenase 存在两个拷

一种新的蛋白质亚细胞定位预测方法

一种新的蛋白质亚细胞定位预测方法

一种新的蛋白质亚细胞定位预测方法程昔恩;吴志诚【期刊名称】《计算机工程与应用》【年(卷),期】2012(048)006【摘要】It is one of basic problems of proteomics to identify the subcellular locations of a protein. It makes the problem more complicated that some proteins may simultaneously exist in two or more than two subcellular locations. Gene Ontology and pseudo amino acid composition are respectively employed to represent a protein as a real values vector. The idea of Ranking initiating from multi-label learning community is adopted to compute a score vector V, each component value of which indicates the probability that a protein of the corresponding subcellular location.The nearest neighbor algorithm is then employed to predict the number n of subcellular localization of human proteins. Finally, the n subcellular locations corresponding to the top n scores components in V are assign to the query protein.%蛋白质亚细胞定位是蛋白质组学基本问题之一.某些类型蛋白质可能存在于两个或两个以上的亚细胞位置,这类蛋白质的亚细胞定位问题更为复杂.分别利用Gene Ontology和伪氨基酸成分法,将一条蛋白质表示为一实值向量;采纳多标记学习中的Ranking思想,计算出一得分向量V,该向量的每一分量的值表示被预测蛋白质属于某个亚细胞位置的概率;利用最近邻算法预测蛋白质所属亚细胞位置的个数n,得分向量V中得分最高的n个分量对应的亚细胞位置即为预测的位置.【总页数】3页(P126-128)【作者】程昔恩;吴志诚【作者单位】景德镇陶瓷学院信息工程学院,江西景德镇333403;景德镇陶瓷学院信息工程学院,江西景德镇333403【正文语种】中文【中图分类】TP392【相关文献】1.一种基于最优局部信息融合的蛋白质亚细胞定位预测方法 [J], 张树波;赖剑煌;何建国2.一种新的蛋白质亚细胞定位预测训练集构造方法 [J], 曹隽喆;顾宏;贺建军3.一种新的蛋白质结构类预测方法 [J], 李楠;李春4.基于最优分割位点的蛋白质亚细胞位点预测方法 [J], 王伟;郑小琪;窦永超;刘太岗;赵娟;王军5.CL-RBF:一种基于改进ML-RBF的蛋白质亚细胞多点定位预测算法 [J], 薛卫; 洪晓宇; 胡雪娇; 陈行健; 张梁因版权原因,仅展示原文概要,查看原文内容请购买。

糖尿病SLC30A8蛋白及其相互作用蛋白初步

糖尿病SLC30A8蛋白及其相互作用蛋白初步
【Abstract】 Objective To analyze the SLC30A8 protein function and build the 11 protein interaction networks,provide theoretic foundation for the drug target discovery and novel drug design. Methods NCBI database and protein interaction databases were used to collect proteins interacting with the SLC30A8. ProtParam software,HPRD,GO database,and STRING databases were used to identify their functions,physical and chemical properties,subcellular location and build protein interaction networks. Results 11 kinds of protein function,physical and chemical properties,and subcellular localization were identified and proteins interaction network were built. SLC30A8-related protein interaction networks were analyzed. Conclusions SLC30A8 promotes zinc from the cytoplasm into the cell,maintains the balance of intracellular zinc ions. Proteins directly interacting with the SLC30A8 are mainly membrane transport proteins,transcription factors, enzymes,integral membrane protein,RNA-binding proteins and growth factors.

肽合成中多对二硫键的形成策略及分析方法_周艳荣

肽合成中多对二硫键的形成策略及分析方法_周艳荣
如果肽中含三对二硫键则第一步氧化形成三种异构体然后分别对三种异构体进行氧化通过测活确定正确二硫键配对的肽或在第一步氧化后测定二硫键的配对方式然后再氧化含特定二硫键桥的中间肽
838
文章编号 :299‘>9998F8998Q93>9838>98






!"##"$% &’ (&)#"*+’)!),-
-.]0 :.U>/] b9G 8>88>:.>/ !G0,+/>!-.Q *>#c Qd>//0:,BeD$ N0aG9/T %[[#T[6C[ B#D K>8.:9 *IT f>b>G>::> &*T N>]>,]. ;T !" #$$ N9?0: ! (>/] # ( Q9/9-9g./, bG98 %&’() )"*+#"() ?0/98BeD$ W.9Qd08.,-G+T %[[#T@%6 [[%[
在多肽合成中, 二硫键的正确形成往往是合成中的关键, 含一对二硫键( 分子内或分子间) 的多肽合成往往比较容易选 择反应的条件, 既可采用温和的氧化条件, 如空气、 A4%) 氧 汞盐、 铊盐 化等, 也可采用较为激烈的反应条件, 如 &8、 +8)8、 等, 得到的产物也容易分离纯化。然而, 由于含两对二硫键以 上的多肽氧化会形成多种异构体,一对二硫键形成条件已不 适用, 折叠的影响因素较多, 折叠条件的选择在合成大量富含 二硫键的多肽中尤为重要。 本文结合文献及工作实践, 对这类 肽合成中的氧化策略、 氧化条件( 氧化还原体系、 缓冲体系、 盐 浓度、 肽浓度、 反应温度) 进行讨论, 并就该类肽中二硫键的分 析方法作一介绍。 中含三对二硫键, 则第一步氧化形成三种异构体, 然后分别对 三种异构体进行氧化, 通过测活确定正确二硫键配对的肽, 或 在第一步氧化后, 测定二硫键的配对方式, 然后再氧化含特定 二硫键桥的中间肽。 关于保护基位置的选择, 根据作者经验, 以偏中间的巯基 从 对保护较佳, ’ 及 * 端巯基暴露有利于提高巯基的自由度, 全交叉二硫 而提高该步的氧化效率。在合成芋螺毒素 4!A ( 键) 时, 就采用了用 BKN 基保护 8 、 Z 位的半胱氨 酸 巯 基 \^]。 我 们 在 合 成 4!* 的 突 变 体 时 采 用 了 2 、 ^ 位 及 8、 Z 巯基保护, 结果表明以 8 、 Z 保护所得终产物效率高。 在 碘 氧 化 去 除 BKN 保 护 基 时 , 溶剂、 碘浓度、 反应时间、 肽浓度的选择很重要, 溶剂一般选择乙酸、 甲醇、 水或其混合 形成分子内二硫键时 体, 碘的浓度宜为巯基摩尔数的 3_^ 倍, 肽浓度不宜过高。 合成天然多肽一般来说选用一步氧化,因为氧化产物多 是具有热力学稳定的构象, 氧化产率也较高, 如一些含二至三 对的天然芋螺毒素主要采用一步法

GTPase

GTPase

M OLECULAR AND C ELLULAR B IOLOGY,Sept.2010,p.4324–4338Vol.30,No.17 0270-7306/10/$12.00doi:10.1128/MCB.01646-09Copyright©2010,American Society for Microbiology.All Rights Reserved.Regulation of the Rho Family Small GTPase Wrch-1/RhoU by C-Terminal Tyrosine Phosphorylation Requires Srcᰔ†Jamie K.Alan,1Anastacia C.Berzat,2,3‡Brian J.Dewar,4Lee M.Graves,1,4and Adrienne D.Cox1,2,3,5*Department of Pharmacology,1Curriculum in Genetics and Molecular Biology,2Department of Radiation Oncology,3 Curriculum in Toxicology,4and Lineberger Comprehensive Cancer Center,5University of North Carolina atChapel Hill,Chapel Hill,North Carolina27599Received22December2009/Returned for modification15February2010/Accepted4June2010Wrch-1is an atypical Rho family small GTPase with roles in migration,epithelial cell morphogenesis,osteoclastogenesis,and oncogenic transformation.Here,we observed rapid relocalization of Wrch-1from theplasma membrane upon serum stimulation.Studies revealed a requirement for serum-stimulated tyrosinephosphorylation of Wrch-1at residue Y254within its C-terminal membrane targeting domain,mediated by thenonreceptor tyrosine kinase Src.Genetic or pharmacological loss of Src kinase activity blocked both phos-phorylation and relocalization of Wrch-1.Functionally,Y254was required for proper Wrch-1modulation ofcystogenesis in three-dimensional culture,and the phospho-deficient mutant,Y254F,was enhanced in Wrch-1-mediated anchorage-independent growth.Mechanistically,C-terminal tyrosine phosphorylation and subse-quent relocalization of Wrch-1downregulated its ability to interact with and activate its effectors by decreasingactive Wrch-1-GTP,perhaps by altering proximity to a GEF or GAP.Phospho-deficient Wrch-1(Y254F)remained at the plasma membrane and GTP bound and continued to recruit and activate its effector PAK,evenupon serum stimulation.In contrast,a phospho-mimetic mutant,Y254E,was constitutively endosomallylocalized and GDP bound and failed to recruit PAK unless mutated to be constitutively active/GAP insensitive.C-terminal tyrosine phosphorylation thus represents a new paradigm in posttranslational control of smallGTPase localization,activation,and biological function.Rho family proteins are Ras-related small GTPases that regulate cytoskeletal organization and dynamics,cell adhesion, motility,trafficking,proliferation,and survival(20).They func-tion as tightly regulated molecular switches,cycling between an active GTP-bound state and an inactive GDP-bound state. Rho GTPases are also regulated by their subcellular localiza-tion,directed by sequences and posttranslational modifica-tions,such as an isoprenoid lipid attached permanently to their C-terminal membrane targeting regions(1),and a second sig-nal,such as a polybasic region or a palmitate fatty acid(34). Rho-guanine nucleotide dissociation inhibitors(RhoGDIs) bind prenyl groups and sequester Rho proteins from mem-branes(19,42).Interaction of the GTP-bound proteins with their downstream effectors at specific locations then elicits their biological functions.Wrch-1,also designated RhoU or Wrch1,is an atypical member of the Cdc42subgroup of Rho GTPases that induce the formation of actin microspikes andfilopodia.Although it shares57%sequence identity with Cdc42and61%sequence identity with its closest relative,Chp/Wrch-2,Wrch-1shares only partially overlapping localization and effector interactions with them and is regulated in a distinct manner.Like Cdc42, Wrch-1activates PAK1and JNK(13,44),induces formation of filopodia(34,35),and both morphological(8)and growth transformation in multiple cell types(5,8).Wrch-1also regu-lates focal adhesion turnover(13,31),negatively regulates tight junction kinetics(8),plays a required role in epithelial morpho-genesis(8),and modulates osteoclastogenesis(9,10,31). Initially discovered as a Wnt-responsive gene capable of phenocopying Wnt morphological transformation(43,44), Wrch-1is transcriptionally regulated by Wnt(36),RANKL (10),and STAT3(36),and it is upregulated in some cancers but downregulated in others(22).Thus,modulation of Wrch-1 activity at the level of expression is a common event.However, because it is a GTP-binding protein,a more dynamic regula-tion of Wrch-1activity is also required.Wrch-1is thought to be largely GTP bound due to a high intrinsic exchange rate(2,39),and no regulatory GEFs or GAPs have yet been identified.However,a putative dominant negative mutant of Wrch-1,T63N,does not behave like the wild type(34),so at least one GEF may be important to activate Wrch-1.Also,mutationally activated(Q107L,analo-gous to Q61L in Ras or Cdc42)Wrch-1is more active than wild-type Wrch-1(5,8,9,31,44),so one or more GAPs remain to be identified.Finally,Wrch-1contains a negative regulatory 46-amino-acid N-terminal extension(39),and interaction with Grb2or phospholipase C␥1(35,39)may help to relieve auto-inhibition(39).In addition to these modes of regulation,Wrch-1function requires posttranslational lipid modification of its C-terminal membrane targeting domain.Unusually,Wrch-1is not preny-*Corresponding author.Mailing address:Departments of Radia-tion Oncology and Pharmacology,Lineberger Comprehensive Cancer Center,450West Drive,University of North Carolina at Chapel Hill, Chapel Hill,NC27599-7295.Phone:(919)966-7712.Fax:(919)966-9673.E-mail:adrienne_cox@.†Supplemental material for this article may be found at http://mcb/.‡Present address:Memorial Sloan-Kettering Cancer Center,1275 York Ave.,Box572,New York,NY10065.ᰔPublished ahead of print on14June2010.4324lated but is modified by palmitoylation(5),a dynamically reg-ulated lipid modification(29)required for both its subcellular localization and biological activities(5,8).Lacking a prenyl group,Wrch-1does not bind RhoGDI(4).Both prenylation and the polybasic region of Cdc42are required for its proper localization and function(46),but the identities of additional signals governing Wrch-1are unknown.There is increasing evidence that C-terminal serine/threo-nine phosphorylation of small GTPases near the isoprenoid moiety is required for both their localization and specific func-tions.In response to protein kinase C(PKC)-mediated phos-phorylation of Ser181in its C-terminal membrane targeting domain,K-Ras4B translocates from the plasma membrane to the mitochondria,where it then promotes apoptosis instead of proliferation(6).RalA is a target of Aurora A kinase-mediated phosphorylation at Ser194(47)and PP2A A␤-mediated de-phosphorylation(7);phosphorylation of this site depletes RalA from the plasma membrane(27).Rap1is phosphory-lated on Ser180by protein kinase A(26,32),and RhoA local-ization and modulation of cell spreading and migration is reg-ulated by PKA-mediated phosphorylation on Ser188(17,25), which promotes its binding to RhoGDI(17).PKC␣(28,33) and ROCK(33)stimulate phosphorylation of Rnd3/RhoE, which results in translocation,and PKC-mediated phosphory-lation is required for Rnd3to modulate the Rho/ROCK path-way(28).TC10is phosphorylated by cyclin-dependent kinase5 on Thr197,which regulates its association with lipid rafts,a requirement for its ability to modulate insulin-stimulated GLUT4translocation(30).Thus,there is significant evidence for the functional importance of C-terminal serine/threonine modification of small GTPases.Here,we sought to determine which C-terminal elements in addition to palmitoylation contribute to the regulation of Wrch-1subcellular localization and biological activity.The minimal C-terminal membrane targeting sequence of Wrch-1 does not contain a suitable serine or threonine residue but contains a potentially phosphorylatable tyrosine residue.Fur-ther,subcellular localization is often dynamically regulated by external signals such as those supplied by serum factors that stimulate engagement of growth factor receptor tyrosine ki-nases and their associated nonreceptor tyrosine kinase part-ners(41).In this report,we describe our discovery that serum stimulates Src-mediated tyrosine phosphorylation of Wrch-1 and its translocation from the plasma membrane and that a specific tyrosine residue in the C-terminal membrane targeting domain of Wrch-1regulates its subcellular localization,GTP/ GDP-binding status,effector activation,and biological activi-ties.Thus,we have identified C-terminal tyrosine phosphory-lation as a novel mechanism for regulation of small GTPase activity.MATERIALS AND METHODSMolecular constructs.Mammalian expression constructs for greenfluorescent protein(GFP)-tagged and hemagglutinin(HA)epitope-tagged human Wrch-1 (wild-type[WT]and Q107L)have been described previously(5).Phospho-deficient Wrch-1(Y254F)and phospho-mimetic Wrch-1(Y254E)were generated by site-directed mutagenesis in both WT and Q107L backgrounds.The GFP-fusion protein of the C-terminal9amino acids(the“9-aa tail”)of Wrch-1 expressed from pEGFP has been described previously(5).An additional GFP fusion containing the C-terminal19amino acids(19-aa tail)was generated by site-directed mutagenesis in the same manner.All sequences were verified by the Genome Analysis Facility at UNC-CH.GFP-PAK-PBD and GST-PAK-PBD were kind gifts from Channing Der(UNC-CH)and Keith Burridge(UNC-CH), respectively.WT Src,constitutively active Src(Y528F),and kinase-deficient Src(K297R)were expressed from the pUSE vector,all from Upstate Biotech-nology.Bacterial expression constructs and purification of GST-Wrch-1protein have been described previously(38).Cell culture,transfections,and retroviral infection.H1299non-small cell lung cancer(NSCLC)cells were grown in Dulbecco’s modified Eagle medium(high glucose;DMEM-H;GIBCO/Invitrogen)supplemented with10%fetal bovine serum(FBS;Sigma)and1%penicillin-streptomycin(P/S)(“complete culture medium”)and maintained in5%CO2at37°C.H1299cells were transfected with expression constructs encoding Wrch-1,Src,or PAK-PBD proteins by using TransIT-LT1(Mirus)according to the manufacturer’s instructions.For localiza-tion assays,cells were transfected transiently and used24h after transfection. For selection of stably expressing cell lines,cells were grown in complete medium supplemented with the appropriate antibiotic for5to7days,after whichϾ50 colonies were pooled for use.MDCKII cells,generously provided by Robert Nicholas(UNC-CH),were grown as above and supplemented with1%nonessential amino acids(NEAA; Invitrogen)(“complete medium”).MDCKII cell lines stably expressing Wrch-1 were generated by retroviral infection.Retrovirus was collected following CaCl2-mediated transfection of pBabe-HAII-puro,pVPack-Gag/Pol,and pVPack-Ampho(Stratagene)expression vectors into293T cells.Cells were infected by exposure to retroviral supernant containing8␮g/ml of Polybrene(American Bioanalytical)and maintained in puromycin for10days,after which the colonies were pooled for use.SYF mouse embryofibroblast cells(MEFs)genetically lacking Src,Yes,and Fyn,or YF cells lacking Yes and Fyn but retaining Src(23),were grown in complete culture medium as described above.Cells were transiently transfected with constructs encoding HA-Wrch-1proteins by using TransIT-LT1(Mirus) according to the manufacturer’s instructions.Fluorescence,immunofluorescence,confocal microscopy,and localization as-says.Cells were transfected transiently with pEGFP-Wrch-1expression vectors or pEGFP-PAK-PBD as indicated above and grown in complete medium for 24h.The cells were then either grown further overnight in complete medium (“basal conditions”),or serum starved overnight in0%serum(“serum starved”), or serum starved overnight in0%serum and then stimulated for2,5,or15min with fresh serum-containing complete medium(“starved and stimulated”).For some experiments,cells were treated for1h with the Src family kinase inhibitor SU6656(Sigma)or dimethyl sulfoxide vehicle prior to serum stimulation.Fol-lowing incubation with Alexa647-transferrin(Molecular Probes),cells werefixed and then visualized for GFP-Wrch-1(green)or transferrin(red).HA-Wrch-1 was visualized by staining with a primary anti-HA antibody(Covance)and a secondary anti-mouse antibody conjugated to Alexa647(Invitrogen).Confocal microscopy was performed on an Olympus Fluoview500laser scanning confocal imaging system,configured with an IX81fluorescence microscopefitted with a PlanApo60ϫoil objective.Antibodies and immunoblot analysis.Western blot analyses were carried out as described previously(8).Briefly,cells were lysed in magnesium lysis buffer (MLB)containing1ϫprotease inhibitor cocktail(Roche)with or without100␮M pervanadate,lysates were cleared by centrifugation,and protein concentra-tions were determined using the DC Lowry protein assay(Bio-Rad).Twenty micrograms of protein for each sample,prepared in5ϫLaemmli sample buffer, was resolved by using12%SDS-PAGE.Proteins were transferred to polyvinyl-idene difluoride membranes(PVDF;Millipore),blocked overnight in3%fish gelatin,and then probed for Wrch-1by a1-h incubation with the following primary antibodies:anti-HA epitope(HA.11;Covance),antiphosphotyrosine (pY100from Cell Signaling Technologies[CST]or pY20from Santa Cruz Biotechnology),anti-GFP(JL8;Clontech),anti-PAK1/2/3(CST),anti-phospho-PAK1(Thr423)/-PAK2(Thr402)(CST),anti-Pyk2(50),or anti-phospho-Pyk2(Tyr402)(Biosource).Anti-␤-actin(Sigma)was used to demonstrate equiv-alent loading.Washed membranes were incubated in anti-mouse or anti-rabbit IgG–horseradish peroxidase(HRP;Amersham Biosciences)or anti-mouse kappa light chain–HRP(Zymed),washed again,and developed using Super-Signal West Dura extended duration substrate(Pierce). Immunoprecipitation.H1299cells expressing GFP-or HA-tagged Wrch-1 were lysed in MLB with protease inhibitor,with or without pervanadate,as described above,at24h after transfection.Lysates were precleared with protein A/G beads(Santa Cruz)and then incubated overnight with anti-GFP or anti-HA antibody.After18h,the protein-antibody complexes were recovered using protein A/G beads.Beads were collected and washed with MLB and resus-pended in Laemmli sample buffer,the precipitated proteins were resolved onV OL.30,2010Src-MEDIATED TYROSINE PHOSPHORYLATION OF Wrch-14325SDS-PAGE,and immunoblot analysis for phosphotyrosine was performed as described above.In vitro tyrosine kinase assay.Recombinant Wrch-1protein was used as a substrate for purified Src kinase in a standard in vitro kinase assay.Bacterially expressed GST-Wrch or GST alone was incubated for40min at30°C with or without0.8␮g of purified recombinant Src protein(Upstate Biotechnology)in Src kinase reaction buffer(100mM Tris[pH7.2],125mM MgCl2,25mM MnCl2, 2mM EGTA,100␮M Na3VO4,and2mM dithiothreitol)containing[␥-32P]ATP (10␮Ci per reaction mixture).Reactions were terminated by the addition of4ϫsample buffer and then heated at95°C for5min.Protein samples were separated by10%SDS-PAGE and visualized by Coomassie blue staining.Incorporation of radiolabel was determined by audioradiography.Anchorage-independent growth transformation assay.Single-cell suspensions of MDCK cells(3.5ϫ103cells per35-mm dish)were suspended in0.4%agar (BD Biosciences)in complete medium and layered on top of0.6%agar as described previously(8).After14days,colonies were stained with3-(4,5-di-methylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT;Sigma)after count-ing of small colonies(6to15cell diameters)and large colonies(Ͼ15cell diameters),and the average number of each type of colony on triplicate dishes was quantified.A one-way analysis of variance(ANOVA)and Tukey’s post hoc test were performed;P values ofϽ0.01were considered significant.Epithelial morphogenesis cyst formation assay.MDCKII cells stably express-ing HA-tagged Wrch-1proteins were allowed to form cysts in three-dimensional (3D)collagen matrices as described previously(8).Briefly,monodispersed MDCKII cells were allowed to grow and form multicellular cyst structures on collagen I gels for10days,when the cultures were treated with collagenase type VII(C-2399;Sigma).Cyst structures werefixed and permeabilized,then incu-bated withfluorescent Texas Red-phalloidin(Molecular Probes)and mounted for imaging on an Olympus Fluoview confocal microscope as indicated above.Multiple x-y and x-z scans were acquired for each3D collagen gel.We have shown previously that tightly regulated endogenous Wrch-1activity is critical for proper cystogenesis on3D collagen I matrices in these cells(8).In each of three replicate experiments,25multicellular cyst structures were evaluated for each Wrch-1-expressing cell line and binned into one of the following three groups: normal cysts(cysts containing a single lumen)or one of two groups of abnormal cysts(either cysts containing no lumen or cysts containing multiple lumens),as we have done previously(8).Student’s t test was performed to determine sig-nificance;P values ofϽ0.01were considered significant.Wrch-1activation assay.H1299cells were transiently transfected with either pCGN vector only or pCGN vectors encoding HA-Wrch-1,HA-Wrch-1(Y254F), HA-Wrch-1(Y254E),HA-Wrch-1(Q107L),HA-Wrch-1(Q107L/Y254F),or HA-Wrch-1(Q107L/Y254E),by using TransIT LT1as described above.Cells were serum starved overnight,or serum starved and then serum stimulated for2,5,or 15min as described above.Cells were then washed twice with ice-cold phos-phate-buffered saline(pH7.4)and lysed in MLB as described above.Equal volumes were removed from each lysate for total protein analysis.To each lysate, glutathione-agarose beads containing40␮g of GST-p21-activated kinase(PAK) GTPase-binding domain fusion protein(GST-PAK-PBD)were added and incu-bated at4°C for60min with rocking.Agarose-GST-PAK-PBD and associated Wrch-1was pelleted and washed three times with500␮l wash buffer(25mmol/ liter Tris[pH7.5],40mmol/liter sodium chloride,and30mmol/liter magnesium chloride).Final pellets were resuspended in1ϫprotein sample buffer and re-solved on SDS-PAGE.HA-Wrch-1was detected using anti-HA antibody(Co-vance).RESULTSWrch-1rapidly relocalizes from the plasma membrane in response to serum stimulation.We have shown previously that the atypical Rho family small GTPase Wrch-1localizes to both plasma membrane and internal compartments,including en-dosomal membranes(5).Because localization of other pro-teins,including some Rho GTPases,can be directed by exter-nal stimuli,such as serum-stimulated engagement of growth factor receptors,and is therefore dynamically regulated,we sought to determine whether Wrch-1localization was similarly regulated.We serum starved overnight H1299NSCLC cells transiently expressing enhanced GFP(EGFP)-tagged Wrch-1 (designated GFP-Wrch-1)and stimulated them with serum for 15min.Under basal or serum-starved conditions(Fig.1,left and middle columns),Wrch-1was localized both to the plasma membrane and to endosomal membranes(marked by trans-ferrin;middle row).In contrast,upon serum stimulation, Wrch-1underwent rapid loss from the plasma membrane(Fig. 1,right column)but continued to localize to endosomes.Time-lapse video taken over the same time course confirmed relo-calization of Wrch-1from the plasma membrane(see Movie S1in the supplemental material).These data indicate that Wrch-1subcellular localization is both dynamic and regulated by upstream growth signals.Relocalization is dependent on the presence of a tyrosine at position254in the Wrch-1C-terminal membrane targeting domain.We had previously determined that the carboxy-terminal9amino acids of Wrch-1are sufficient for its proper subcellular localization(5)and that mutation of the palmi-toylated cysteine residues therein results in cytosolic accu-mulation(5).Within this sequence of WWKKYCCFV,the single tyrosine at residue254(Y254)stood out.We hypothesized that Y254becomes tyrosine phosphorylated in response to serum stimulation and that this phosphoryation event modulates Wrch-1 subcellular localization and activity.We therefore generated a Y3F mutation at position254[des-ignated Wrch-1(Y254F)].The putatively phospho-deficient Wrch-1(Y254F)was resistant to serum-stimulated relocalization: the plasma membrane-localized pool of Wrch-1(Y254F)re-mained on the plasma membrane even after serum stimulation (Fig.2A;see also Movie S2in the supplemental material). Further,cells expressing this nonphosphorylatable mutant dis-played an exaggeratedly rounded phenotype under basal se-rum-containing conditions,similar to that seen in serum-starved cells expressing only WT Wrch-1(Fig.2A)and consistent with a serum-dependent event altering both Wrch-1 phosphorylation and cytoskeletal organization.These results were also consistent with our hypothesis that Wrch-1becomes tyrosine phosphorylated on Y254in response to serum stimulation and that this phosphorylation eventis FIG.1.Wrch-1rapidly relocalizes upon serum stimulation.H1299 NSCLC cells transiently transfected to express GFP-Wrch-1were grown in complete culture medium,then serum starved overnight,or first serum starved and then serum stimulated.Prior to serum stimu-lation,the treated cells were incubated for1h with AlexaFluor647-labeled transferrin to mark endosomal compartments.After15min of serum stimulation,cells werefixed and then subjected to confocal microscopy for visualization of GFP-Wrch-1(green)or transferrin (red).Bars,20␮m.4326ALAN ET AL.M OL.C ELL.B IOL.responsible for serum-stimulated relocalization of Wrch-1.To confirm that Wrch-1was tyrosine phosphorylated at Y254,we immunoprecipitated Wrch-1proteins from H1299cells with anti-HA antibody and probed for phosphotyrosine.Wrch-1but not Wrch-1(Y254F)was detected by anti-phosphotyrosine an-tibody following serum stimulation(Fig.2B),confirming that Wrch-1is tyrosine phosphorylated and suggesting that Y254is the major site of serum-stimulated phosphorylation. Interestingly,we also determined that basal subcellular lo-calization of Wrch-1requires less targeting information than does either tyrosine phosphorylation or serum-stimulated re-localization.The last9amino acids in the C terminus of Wrch-1are sufficient for proper basal localization of Wrch-1 (5).Surprisingly,this short sequence(the9-aa tail)(Fig.3A) was not sufficient to support either tyrosine phosphorylation (Fig.3B)or serum-stimulated relocalization(Fig.3C).We speculated that9amino acids was an insufficient length to allow binding of the kinase.We generated an additional GFP-fusion protein comprising19amino acids of the Wrch-1C terminus(the 19-aa tail),which still contained only a single tyrosine residue (Fig.3A),which was sufficient both for tyrosine phosphorylation (Fig.3B)and for relocalization(Fig.3C).Interestingly,expression of the19-aa tail resulted in altered cell size and shape,suggesting that it may sequester proteins important for regulating the actin cytoskeleton.Together,these data indicate that relocalization of Wrch-1requires additional sequences compared to basal local-ization,perhaps to allow efficient kinase binding to the region encompassing Y254.Src can phosphorylate Wrch-1,and Src tyrosine kinase ac-tivity is required for both tyrosine phosphorylation and se-rum-stimulated relocalization of Wrch-1.The nonreceptor ty-rosine kinase Src,which transmits signaling from several serum-responsive growth factor receptor tyrosine kinases,has many substrates,not all of which have been defined clearly.We speculated that Src could phosphorylate Wrch-1on Y254,and indeed we found that the Src family kinase inhibitorSU6656 FIG.2.Wrch-1is tyrosine phosphorylated on Y254in response to serum,and this phosphorylation is required for serum-stimulated relocal-ization.(A)Nonphosphorylatable Wrch-1(Y254F)is resistant to serum-stimulated relocalization.H1299cells expressing either GFP-Wrch-1or GFP-Wrch-1(Y254F)were grown,treated,and evaluated as described for Fig.1.Bars,20␮m.(B)Serum-stimulated tyrosine phosphorylation of Y254.H1299cell lysates from cells expressing empty vector(VO),HA-Wrch-1,or HA-Wrch-1(Y254F)were incubated with anti-HA antibody. Immunoprecipitated(IP)Wrch-1was detected by immunoblotting(IB)with anti-HA,and phosphotyrosine(p-Tyr)on Wrch-1was detected by immunoblotting with antiphosphotyrosine antibody.The bands above and below the Wrch-1band represent immunoglobulin heavy chain and light chains,respectively.Apparent molecular masses are shown(in kilodaltons).V OL.30,2010Src-MEDIATED TYROSINE PHOSPHORYLATION OF Wrch-14327effectively blocked tyrosine phosphorylation of Wrch-1(Fig.4A).At the concentration that we used (5␮M),SU6656is reported to inhibit the related tyrosine kinases Src,Yes,Fyn,and Lyn (3).Therefore,we analyzed Wrch-1phosphorylation in MEF cells from mice genetically deficient in Src,Yes,and Fyn (SYF;Src Ϫ/Ϫ)(Lyn is restricted to hematopoietic cells)or deficient in Yes and Fyn but not Src (YF;Src ϩ/ϩ).Wrch-1was tyrosine phosphorylated in YF MEFs expressing Src but not in SYF MEFs lacking Src,indicating that Src is required for tyrosine phosphorylation of Wrch-1(Fig.4B).These data in-dicate that Src functions upstream of Wrch-1to mediate its tyrosine phosphorylation.To address whether Src kinase en-zymatic activity is required,we cotransfected H1299cells with HA-Wrch-1or HA-Wrch-1(Y254F)along with either consti-tutively active or kinase-deficient Src.Wrch-1was tyrosine phosphorylated in the presence of kinase-active Src(Y528F)but not kinase-deficient Src(K297R),and the Y254residue of Wrch-1was required for this phosphorylation,as Wrch-1(Y254F)was not phosphorylated regardless of Src kinase activity (Fig.4C).These data support a requirement for Src kinase activity in order for Wrch-1to become tyrosine phos-phorylated on Y254.To address whether the phosphorylation is direct or indirect,we performed an in vitro kinase assay with recombinant purified Src and GST-Wrch-1.Wrch-1was di-rectly phosphorylated by Src in vitro (Fig.4D),consistent with the possibility that it may be phosphorylated directly in vivo .Having established that Wrch-1could be a substrate of Src kinase activity,and that the Y254residue of Wrch-1that is required for serum-stimulated relocalization is also the major site of Src-stimulated phosphorylation,we wished to test whether Src kinase activity is required for serum-stimulated relocalization of Wrch-1.We serum-starved cells expressing GFP-Wrch-1,treated them with SU6656(5␮M)for 1h,then serum-stimulated them as above.SU6656prevented Wrch-1relocalization in response to serum stimulation (Fig.4E).Col-lectively,these results indicate that Src tyrosine kinaseactivityFIG.3.The C-terminal 19amino acids of Wrch-1are sufficient for Wrch-1to become tyrosine phosphorylated and to be relocalized in response to serum.(A)Schematic of the 19-aa and 9-aa tails .Shown are the GFP-tagged sequences extended with 9or 19amino acids of the C terminus of Wrch-1.Y254is the only tyrosine residue present in each fusion protein.(B)Serum-stimulated tyrosine phosphorylation of the C-terminal 19but not 9amino acids of Wrch-1.H1299cell lysates expressing either empty vector (GFP),GFP-Wrch-1(FL),GFP fused to the 19-aa tail of Wrch-1,or GFP fused to the 9-aa tail ofWrch-1were incubated with anti-GFP antibody to immunoprecipitate Wrch-1.Immunoprecipitated (IP)Wrch-1was then detected by immunoblotting (IB)with anti-GFP antibody,and phosphotyrosine Wrch-1was detected by immunoblotting with anti-phosphotyrosine antibody.(C)The C-terminal 19amino acids of Wrch-1are sufficient for serum-stimulated relocalization .H1299cells as in panel B were grown,treated,and evaluated as described for Fig.1.Bars,20␮m.4328ALAN ET AL.M OL .C ELL .B IOL .FIG.4.Src activity is required in vivo for tyrosine phosphorylation of Wrch-1.(A)The Src family tyrosine kinase inhibitor SU6656prevents Wrch-1tyrosine phosphorylation in response to serum stimulation.H1299cells expressing HA-Wrch-1were serum starved overnight,treated with 5␮M SU6656for 1h,and serum stimulated for 5min.Lysates were subjected to immunoprecipitation (IP)with anti-HA to retrieve HA-Wrch-1,followed by immunoblotting (IB)for Wrch-1(anti-HA)or phosphotyrosine (anti-p-Tyr).Bands above and below the Wrch-1band represent the Ig heavy chain and light chain,respectively.(B)Endogenous Src is required for serum-stimulated Wrch-1tyrosine phosphorylation .SYF Ϫ/ϪMEFs (MEFs lacking Src,Yes,and Fyn)and YF Ϫ/ϪMEFs (MEFs retaining Src but lacking Yes and Fyn)expressing either HA-Wrch-1or nonphos-phorylatable HA-Wrch-1(Y254F)were starved overnight and then serum stimulated for 5min.The resulting cell lysates were probed for phosphotyrosine on Wrch-1as described for panel A.(C)Src kinase activity is required for tyrosine phosphorylation of Wrch-1.H1299cells were cotransfected with empty vector,HA-Wrch-1,or nonphosphorylatable HA-Wrch-1(Y254F)along with either empty vector,kinase-active Src (Src Y528F),or kinase-deficient Src (Src K297R).Phosphotyrosine on Wrch-1was detected as shown in panel A.(D)Src directly phosphorylates Wrch-1in vitro .Purified recombinant GST-Wrch-1protein was incubated with purified recombinant Src tyrosine kinase protein and [32P]ATP.Total protein was detected by Coomassie blue staining,and [32P]ATP incorporation was detected by autoradiography.(E)Inhibition of Src kinase with SU6656blocks serum-stimulated relocalization of Wrch-1.H1299cells expressing GFP-Wrch-1or nonphosphorylatable GFP-Wrch-1(Y254F)were grown in complete culture medium (basal)and then either serum starved overnight (serum starved)or first serum starved and then serum stimulated for 15min,with or without 1h of pretreatment with 5␮M SU6656.Cells were visualized as for Fig.1for Wrch-1(green)or transferrin (red).Bars,20␮m.V OL .30,2010Src-MEDIATED TYROSINE PHOSPHORYLATION OF Wrch-14329。

蛋白芯片技术(protein chip technology)

Protein chip technology Heng ZhuÃand Michael SnyderÃyMicroarray technology has become a crucial tool for large-scale and high-throughput biology.It allows fast,easy and parallel detection of thousands of addressable elements in a single experiment.In the past few years,protein microarray technology has shown its great potential in basic research,diagnostics and drug discovery.It has been applied to analyse antibody±antigen,protein±protein,protein±nucleic-acid, protein±lipid and protein±small-molecule interactions,as well as enzyme±substrate interactions.Recent progress in the®eld of protein chips includes surface chemistry,capture molecule attachment,protein labeling and detection methods,high-throughput protein/antibody production,and applications to analyse entire proteomes.AddressesÃDepartment of Molecular,Cellular,and Developmental Biology andy Department of Molecular Biophysics and Biochemistry,Yale University,New Haven,CT06520,USAe-mail:michael.snyder@Current Opinion in Chemical Biology2003,7:55±63This review comes from a themed issue onProteomics and genomicsEdited by Matthew Bogyo and James Hurley1367-5931/03/$±see front matterß2003Elsevier Science Ltd.All rights reserved.DOI10.1016/S1367-5931(02)00005-4AbbreviationsAFM atomic force microscopeGST glutathione-S-transferasePDMS polydimethylsiloxanePI phosphatidylinositidePVDF poly(vinylidene¯uoride)SAM self-assembled monolayerSELDI surface-enhanced laser desorption/ionizationSPR surface plasmon resonanceIntroductionThe past ten years have witnessed a fascinating growth in the®eld of large-scale and high-throughput biology, resulting in a new era of technology development and the collection and analysis of information.The challenges ahead are to elucidate the function of every encoded gene and protein in an organism and to understand the basic cellular events mediating complex processes and those causing diseases[1±4].Miniaturized and parallel assay systems,especially microarray-based analyses,are crucial to large-scale and high-throughput biological analysis,as they are a rapid and economic way to interpret gene function[3,5,6],as demonstrated by DNA microarray approaches[7,8].In a microarray format,capture mole-cules are immobilized in a very small area,and probed for various biochemical activities.High signal intensities and optimal signal-to-noise ratios can be achieved under ambient analyte conditions[3].The microarray format has become the leading technology that enables fast,easy and parallel detection of thousands of addressable ele-ments and side-by-side measurements.Despite the success of DNA microarrays in gene expres-sion pro®ling and mutation mapping,it is the activity of encoded proteins that directly manifest gene function. Thus,one would expect protein microarrays,in which proteins are prepared,arrayed and analysed at high spatial density,to be particularly powerful for analysing gene function,regulation and a variety of other applications. Proteins are more challenging to prepare for the micro-array format than DNA,and protein functionality is often dependent on the state of proteins,such as post-transla-tional modi®cations,partnership with other proteins, protein subcellular localization,and reversible covalent modi®cations(e.g.phosphorylation).Nonetheless,in recent years there have been considerable achievements in preparing microarrays containing over100proteins and even an entire proteome[1,2,9±11].Alternative array formats have also been developed including tissue arrays [12],living cell arrays[13 ,14 ],peptide arrays[1,15±17,18 ],antibody/antigen arrays[19 ,20],protein arrays [21,22,23 ±25 ],carbohydrate arrays[26 ,27 ],and small-molecule arrays[28 ].However,technological challenges in the®eld of protein microarrays still remain. In this review,we discuss recent progress in the®eld of protein chips,including surface chemistry,capture mole-cule attachment,protein labeling and detection methods, high-throughput protein/antibody production,and appli-cations to analyse protein families and entire proteomes. Manufacture of protein chipsIt is important that protein chips retain proteins in an active state at high densities,are compatible with most commercial arrayers and scanners,and can be printed in such a fashion that the proteins remain in a moisturized environment.Soft substrates such as polystryrene,poly-(vinylidene¯uoride)(PVDF),and nitrocellulose mem-branes,which have been used to attach proteins in traditional biochemical analyses(e.g.immunoblot and phage display),are often not compatible for protein microarrays[2,16,22].These surfaces often do not allow a suitable high protein density,the spotted material may spread on the surface,and/or they may not allow optimal signal to noise ratios[1,3,9,11].Thus,most projectshave55turned to using glass microscope slides or other materials that have been derivatized to attach proteins on their surface at high density.These slides have low ¯uores-cence background and are compatible with most assays.Different types of protein chipsA variety of types of chip have been designed,including 3D surface structures,nanowell and plain glass chips (Table 1).Polyacrylamide gel packet and agarose thin ®lm microarrays,patterned by using photolithography technology on a glass surface,have been created by Guschin et al.[29]and Afanassiev et al.[30],respectively (Figure 1).Because both acrylamide and agarose form highly porous and hydrophilic matrixes,capture mole-cules,such as DNA,proteins and antibodies,can readily diffuse into the porous structure and are immobilized by cross-linking to the reactive ligands modi®ed in the matrixes.Analytes are then added to these 3D arrays to carry out the biochemical assays [29].Because of the formation of 3D matrixes on the glass surface,the capa-city of protein immobilization is much higher than that on a 2D surface;the homogeneous water environment mini-mizes protein denaturing and thereby helps keep proteins in their active states.In addition to the sophisticated processes of creating such 3D matrixes,the major dis-advantage of the 3D arrays is that it is more dif®cult to change buffers and recover trapped molecules from the matrix microarrays [4].In contrast to 3D arrays,Zhu et al.[24 ]fabricated an open structure,namely nanowells,on a polydimethylsiloxane (PDMS)surface supported by the standard glass slides.The nanowells signi®cantly reduce evaporation and mini-mize cross-contamination and background.Because of the open nanowell structure,different components and buffers can be sequentially added,which is crucial for multiple-step biochemical assays.In addition,captured molecules can be easily recovered from the nanowells.When covered with gold in the nanowells,it is expected that high-throughput mass spectrometry and surface plasmon reso-nance (SPR)analyses can be performed.The biggest disadvantage of this technology it that specialized equip-ment is required to load the nanowells at high density.Many groups now directly array proteins and antibodies/antigens onto plain glass surface [19 ,20,23 ,25 ,31 ,32].To keep proteins in a wet environment during the print-ing process,high percent glycerol (30±40%)is used in sample buffer and the spotting is carried out in a humid-ity-controlled environment [23 ,25 ].Surface chemistryTo attach proteins to a solid substrate,the surface of the substrate has to be modi®ed to achieve the maximum binding capacity (Figure 2).A convenient method is to coattheglasssurfacewithathinnitrocellulosemembraneor poly-L -lysine such that proteins can be passively adsorbedTable 1Comparison of current antibody/protein microarrays.Surface Attachment AdvantageDisadvantageReferences PVDF Adsorption and absorptionNo protein modification requirement,high protein binding capacityNon-specific protein attachment in random orientation [2,16]NitrocelluloseAdsorption and absorptionNo protein modification requirement,high protein binding capacityNon-specific binding,high background [20,22]Low-density arraysPoly-lysine coated AdsorptionNo protein modification requirement Non-specific adsorption [19 ]Aldehyde-activatedCovalent cross-linkingHigh-density and strong protein attachmentRandom orientation of surface-attached proteins [23 ,25 ]High-resolution detection methods available Epoxy-activated Covalent cross-linking High-density and strong protein attachment Random orientation of surface-attached proteins[24 ]High-resolution detection methods available Avidin coated Affinity binding Strong,specific and high-density protein attachment,low-background Proteins have to be biotinylated [58]Ni-NTA coated Affinity binding Strong,specific and high-density protein attachment,low-background,uniform orientation of surface attached proteinsProteins have to be Hisx6tagged[25 ]Gold-coated siliconCovalent cross-linkingStrong and high-density protein attachment,low-background.Can be easily coupled with SPR and mass-spectrometry Random orientation of surface attached proteins,tough tofabricate,not commercially available [18 ,35]PDMS nanowell Covalent cross-linkingStrong and high-density proteinattachment,well suited for sophisticated biochemical analysesRandom orientation of surface attached proteins[24 ]3D gel pad and agarose thin film Diffusion High protein binding capacity,no protein modification requirementTough to fabricate,not commercially available [29,30]DNA/RNA coatedHybridizationStrong,specific and high-density protein attachment,low-background,uniform orientation of surface attached proteinsSophisticated in vitroproduction of labeled proteins[59]56Proteomics and genomicsto the modi®ed surface through non-speci®c interactions [20,22,33].The attached proteins lay on the surface in random orientation and can be washed off under stringent washing conditions.In addition,the noise level is usually higher because of the non-speci®c adsorption/absorption.To achieve more speci®c and stronger protein attach-ment,several groups have created reactive surfaces on glass that can covalently cross-link to proteins [23 ±25 ].In general,a bifunctional silane cross-linker is used to form a self-assembled monolayer (SAM),which has one functional group that reacts with the hydroxyl groups on glass surface,and another free one that can either directly react with primary amine groups of proteins (i.e.aldehyde or epoxy groups)or can be further chemically modi®ed to reach maximum speci®city [28 ,34].Gold-coated glass surface is another variation [18 ,35].To form a SAM on gold surface,bifunctional thio-alkylene is usually used,Figure 1YY YYY YYYAntigen chipProtein chipLigand chipApplications of protein microarrays.There are two general types of protein microarray:analytical and functional protein microarrays.Analytical microarrays involve a high-density array of affinity reagents (e.g.antibodies or antigens)that are used for detecting proteins in a complex mixture.Functional protein chips are constructed by immobilizing large numbers of purified proteins on a solid surface.Unlike the antibody±antigen chips,protein chips have enormous potential in assaying for a wide range of biochemical activities (e.g.protein±protein,protein±lipid,protein±nucleic-acid,and enzyme±substrate interactions),as well as drug and drug target identification.Small molecule and carbohydrate microarrays are other types of analytical microarrays that have been demonstrated to be capable of studying protein binding activities to ligands and carbohydrates.Protein chip technology Zhu and Snyder 57which has a SH-group that reacts with gold,and another free one that reacts with capture molecules.The advan-tage of using gold-coated surface is that SPR and mass spectrometry can potentially be integrated as detection methods to monitor the dynamics of the reactions,or to identify the captured molecules,respectively[18 ,35,36]. This approach provides the opportunity to study dynamics of biochemical reactions in a high-throughput fashion,and has great potential in drug and drug-target discovery and biomedical research[36].In the above covalent cross-linking approaches,because the reactive ligands also exist in the side chains of proteins,it is plausible that the attached proteins attach to the surface in a random fashion,which may alter the native conformation of proteins,reduce the activity of proteins,or make them inaccessible to probes(Figure2). Perhaps the best means of protein attachment is through highly speci®c af®nity interactions[3,24 ,25 ].Proteins fused with a high-af®nity tag at their amino or carboxy terminus are linked to the surface of the chip via this tag, and hence,all of the attached proteins should orient uniformly away from the surface(Figure2)[25 ].Using this method,immobilized proteins/antibodies are more likely to remain in their native conformation,while the analytes have easier access to the active sites of proteins. This approach was®rst successfully demonstrated in attaching5800fusion proteins containing a His tag onto a nickel-coated glass slide[25 ].It should also be pos-sible to use other af®nity methods such as glutathione/ glutathione-S-transferase(GST)and phosphonate/serine esterase cutinase ligand/protein tags[37].Protein delivery systemsAlthough a96-format dot blot instrument has been used to create low-density protein arrays on®lters[9,22,33], high-density protein microarrays(>30000spots per slide) can be achieved using robotic contact printing tools, such as those developed for creating DNA microarrays [23 ,25 ].The contact printing arrayers deliver sub-nanoliter sample volume directly to the surface using tiny pins with or without capillary slots.Because these contact printing robots cannot align their pins to the pre-fabricated structures and need to touch the surface,non-contact robotic printers,which use ink-jet technology, were used to deposit nanoliter to picoliter protein dro-plets to polyacrylamide gel packets[21]and nanowells [24 ].Although the current Packard ink-jet microarrayer can be slowed when spotting many different samples and the shearing force during drop formation may damage some samples[1],it is not restricted to the surface structure and is well suited for more complicated bio-chemical assays.Recently,electrospray deposition tech-nology was applied to deliver dry proteins to a dextran-grafted surface[38].This technology further reduced the spot size from$150m m to$30m m.Figure2VV V VVVVV VVVVVVVVVV VVV VVV VVV VVV VVVYVV VV VV VV VV VV V V VVYCovalent cross-linking Adsorption/absorptionDiffusionComparison of different protein attachment methods.Proteins can be attached to various kinds of surface via diffusion,adsorption/absorption, covalent cross-linking and affinity interaction.Except affinity attachment,proteins are usually laid on the surface in a random fashion,which may alter the native conformation of proteins,reduce the activity of proteins,or make them inaccessible to probes.However,when proteins are attached to the surface via their affinity tags,it is very likely that every protein molecule uniformly attaches to the surface and,therefore,proteins are more likely to remain in their native conformation,while the analytes have easier access to the active sites(indicated by the red dots)of proteins.58Proteomics and genomicsProbe detection methodsFluorescence detection methods are generally the pre-ferred detection method(Table2)because they are simple,safe,extremely sensitive and can have very high resolution[1,3].They are also compatible with standard microarray scanners.Typically,a chip is either directly probed with a¯uorescent molecule(e.g.protein or small molecule)or in two step by®rst using a tagged probe (e.g.biotin),which can then be detected in a second step using a¯uorescently labeled af®nity reagent(e.g.strep-tavidin).Another¯uorescent labeling method is rolling circle ampli®cation(RCA),which is extremely sensitive [39].However,other detection methods can also be used. For example,ELISA was®rst used to detect proteins for both®lter arrays[20,40]and glass arrays[29].Ge[22],and Zhu et al.[24 ]have used radioisotope labeling to study protein±protein,protein±DNA,protein±drug interactions on®lter arrays,and kinase±substrate interactions in nano-wells,respectively.Because labeling molecules can sometimes affect protein activity and are restricted to the available detection channels,non-labeling methods have advantages as a direct detection approach for antibody microarrays. SELDI(surface-enhanced laser desorption/ionization) mass spectrometry has been used to detect low-density arrays of captured proteins[41].Captured proteins on an array of metal surface(SELDI protein array)were vapour-ized using a laser beam,followed by the analysis of mass spectrometry data to reveal the identities of these pro-teins.The atomic force microscopy(AFM)method takes advantage of surface topological changes to identify the captured proteins on an antibody array[42].When the immobilized rabbit IgG on a gold surface bound to its complimentary antibodies,goat ant-rabbit IgG,AFM could detect the height increase,and therefore,revealed the binding activities.To study the kinetics of antigen±antibody interactions, however,real-time detection methods will be useful.SPR has matured as a versatile detection tool to study the kinetics of receptor±ligand interactions with a wide range of molecular weights,af®nities and binding rates[43±45]. Although the commercially available SPR chips are lim-ited to a few channels,Myszka and Rich[46]described a sensor surface with64individual immobilization sites in a single¯ow cell.Alternatively,Sapsford et al.[47]devel-oped an antibody array biosenor to study the kinetics of antigen binding using a planar waveguide as the detection method.More importantly,they demonstrated that sig-ni®cant signal intensity could be achieved from spots as small as200m m in diameter.It is therefore expected that the latter approach is well suited for high-throughput and parallel kinetics studies.Two functional classes of protein microarraysThere are two general types of protein microarrays. Firstly,analytical microarrays in which antibodies,anti-body mimics or other proteins are arrayed and used to measure the presence and concentrations of proteins in a complex mixtures.Secondly,functional protein microar-rays,in which sets of proteins or even an entire proteome are prepared and arrayed for a wide range of biochemical activities.Analytical microarraysAnalytical microarrays involve a high density array of af®nity reagents that are used for detecting proteins in a complex mixture.They have enormous potential for monitoring protein expression on a large-scale,a process that is sometimes termed protein pro®ling.Antibody microarraysThe most common form of analytical arrays are antibo-dies/antibody mimic arrays in which antibodies(or similar reagents)that bind speci®c antigens are arrayed on a glass slide at high density.A lysate is passed over the array and the bound antigen is detected after washing.Detection is usually carried out by using labeled lysates or using a second antibody that recognizes the antigen of interest. The biggest challenge with these methods is producing reagents that identify the protein of interest and with high enough speci®city in a high-throughput fashion. Antibodies are the traditional reagent of choice for detect-ing proteins in complex mixtures.However,polyclonal sera are often not speci®c and are expensive to produce,Table2Summary of current detection methods used in protein microarray experiments.Detection Probe labeling Data acquirement Real time Resolution References ELISA Enzyme-linked antibodies CCD imaging No Low[20,28 ,38] Isotropic labeling Radio isotope-labeled analyte X-ray film or phosphoimager No High[22,23 ,24 ] Sandwich immunoassay Fluorescently labeled antibodies Laser scanning No High[20]SPR Not necessary Refractive index change Yes Low[41±44]Non-contact AFM Not necessary Surface topological change No High[40]Planar waveguide Fluorescently labeled antibodies CCD imaging Yes High[45]SELDI Not necessary Mass spectrometry No Low[39]Electro-chemical Metal-coupled analyte Conductivity measurement Yes Medium[60]Protein chip technology Zhu and Snyder59and the conventional hybridoma method of producing highly speci®c monoclonal antibodies is also time-con-suming,laborious and costly.Recently,alternative meth-ods,such as phage antibody-display,ribosome display, SELEX(systematic evolution of ligands by exponential enrichment),mRNA display,and af®body display,have been developed to expedite the production of antibodies and/or antibody mimics[1±3,9].All of these approaches involve the construction of large repertoires of viable regions with potential binding activity,which are then selected by multiple rounds of af®nity puri®cations.The binding af®nity of the resulting candidate clones can be further improved using maturation strategies.However, the ideal selection system,which is not only fast,robust, sensitive,and of low cost,but automated and minimized, is yet to be fully developed[3,9].In spite of the challenge in obtaining speci®c antibodies, several studies using antibodies have recently appeared. In one of the largest studies to date,Sreekumar et al.[31 ] spotted146distinct antibodies on glass to monitor the alternations of protein quantity in LoVo colon carcinoma cells.Their results revealed radiation-induced up-regula-tion of many interesting proteins,including p53,DNA fragmentation factor40and45,tumour necrosis factor-related ligand,as well as down-regulated proteins.The most signi®cant problem with antibody arrays is speci®city.Proteins are often present in a very large dynamic range(106);thus,reagents that might have high af®nity for one protein,but are low af®nity for another will still exhibit detection of the lower af®nity protein if it is much more prevalent.Haab et al.[19 ]have investigated the ability of115well-characterized antibody±antigen pairs to react in high-density microarrays on modi®ed glass slides.30%of the pairs showed the expected linear relationships,indicating that a fraction of the antibodies were suitable for quantitative analysis.To avoid this problem,many groups have turned to using sandwich assays,in which the®rst antibody is spotted on the array and then the antigen is detected with a second antibody that recognizes a different part of the proteins.This approach dramatically increases the speci®city of the antigen detection,but required that a least two high-quality antibodies exist for each antigen to be detected. Other analytical microarraysIn addition to antibody microarrays,other analytical microarrays have been developed.These include micro-arrays for pro®ling antibodies in a patient's serum,essen-tially the reciprocal of that described above.Joos and colleagues[20]used18diagnostic markers for autoim-mune diseases to form an autogen microarray and screened for antigen±antibody interactions.Hiller et al.[48]arrayed94puri®ed allergen molecules,which included most common allergen sources,on glass slides to miniaturize the allergy test.These allergen molecules were not restricted to proteins,but also included peptides and small molecules.The allergen microarrays were speci®cally used to determine and monitor allergic patients'IgE reactivity pro®les to large numbers of dis-ease-causing allergens in single measurements.Only minute amounts of serum were required.Potential new leads to allergic diseases were revealed,and some of them have been con®rmed using the traditional skin tests.To characterize autoantibody responses,Robinson et al.[49] robotically arrayed hundreds of autoantigens,including proteins,peptides,and other biomolecules,in eight dis-tinct human autoimmune diseases onto glass slides to form the autoantigen microarrays.These arrays were incubated with patient serum samples to de®ne the pathogenesis of autoantibody responses in human auto-immune diseases.To explore the possibility of quantita-tive measurement of serum-speci®c IgE using protein chip format,Kim et al.[50]used puri®ed dermatopha-goides pteronyssinus(Dp)-speci®c IgE to detect allergens in serum challenged with Dp,egg white,milk,soybean and wheat.These authors were able to demonstrate that quantitative measurement of allergen in a protein mixture could be achieved.Functional protein chipsFunctional protein chips are constructed by immobilizing large numbers of puri®ed proteins on a solid surface. Unlike the antibody chips,which are mainly developed for diagnostics and pro®ling of protein and epitope expression,protein chips have enormous potential in basic research,as well as drug and drug target identi®ca-tion(Figure1).For example,both the Mrksich[18 ]and Schreiber[23 ]groups have demonstrated the potential of using protein microarrays to conduct enzymatic assays to identify downstream targets of kinases.However,the ®rst great obstacle to overcome is the puri®cation of large numbers of proteins in a high-throughput manner.High-throughput protein productionTo analyse the biochemical activities of as many proteins as possible,many research groups and companies have contributed tremendous effort in developing high-throughput protein puri®cation methods.The combina-tion of recombinant proteins and af®nity puri®cation has been used to purify proteins from various host cells, including lines from Escherichia coli,yeast,insects and humans[9,25 ,40,51,52].Leuking et al.[11]cloned cDNAs from human fetal brain tissues as C-terminal Hisx6-tagged fusions.The Hisx6 tags were used®rst as an indicator of in-frame fusion proteins and then served as an af®nity tag for high-throughput protein puri®cation from E.coli.In a later report,LaBaer and colleagues[51]created a system (FLEXP)that performs from cDNA cloning to protein production from E.coli in a fully automated fashion.In a test case,$80%of336random cDNA clones could60Proteomics and genomicssuccessfully purify fusion proteins in full length.Because the puri®cation process was automated,at least1000 proteins could be puri®ed in one day.However,because eukaryotic proteins expressed in prokaryotic systems are not post-translationally modi®ed,our group has devel-oped a high-throughout protein puri®cation method from the budding yeast[25 ].The yeast genes were cloned as N-terminal GST:Hisx6fusions,and puri®ed using the GST af®nity tags.In two weeks,>6500yeast proteins could be puri®ed individually from3ml culture.For the same reason,Albala et al.[52]chose72unique human cDNA clones to create an array of recombinant baculo-viruses,from which42%of the clones produced soluble fusion proteins in a96-well format.Alternatively,proteins can be produced using cell-free expression systems.For example,Keefe and Szostak[53] established a mRNA display system,in which each pro-tein was in vitro translated and covalently linked through its carboxy terminus to the3H end of its coding mRNA. More interestingly,He and Taussig[54]created a protein in situ array(PISA),which combines the protein produc-tion and immobilization in one step.Although the experi-ment was performed in microtiter dishes,it is plausible that the system can be easily automated and applied to a microarray format.Applications of functional protein chipsFunctional protein chips like traditional assays performed in microtiter plates[55]are suitable for a wide variety of biochemical analyses.Unlike microtiter plates,however, they are much more amenable to high-throughput studies and use small amounts of reagents.In early proof-of-concept studies,MacBeath and Schreiber[23 ]fabri-cated protein microarrays with three puri®ed proteins at high density,and performed protein±protein,protein±ligand,and kinase±substrate interactions using three test systems.Likewise,Mirzabekov and co-workers[29] demonstrated that proteins immobilized in the gel pads could still show their enzymatic activities.Studies analysing large sets of proteins have recently been ing a PDMS nanowell chip mounted on glass slides,Zhu et al.[24 ]analysed the activity of119 yeast kinases for17different substrates.The substrates were®rst covalently immobilized to individual nanowells, and individual protein kinases with radio-labeled ATP were incubated with the substrates.After washing away the kinases and unincorporated ATP,the nanowell chips were analysed for phosphorylated substrates using a phosphoimager.Not only known kinase±substrate inter-actions were identi®ed,but also many novel activities were revealed.This included the unexpected discovery that one-fourth of yeast protein kinases are capable of phosphorylating their substrates on tyrosine,even though the kinases are members of the Ser±Thr family of protein kinases.Because the ultimate goal of proteomics is to study biochemical activities of every protein encoded by an organism,Zhu et al.[25 ]prepared the®rst proteome chip.They cloned$94%(>5800of6200)of the yeast open reading frames in a yeast expression vector that expresses the proteins as N-terminal GST-Hisx6double tagged fusions and developed a high-throughput yeast protein puri®cation method to individually purify pro-teins.80%of yeast proteins are full length and of suf®-cient quantities that they are detectable by most assays. The proteins were puri®ed using the GST tags and were then attached to Ni-NTA-coated glass slides using the HisX6tags.In our initial study,the chips were probed with Cy3-labeled calmodulin and various phosphatidyli-nositides(PIs).Calmodulin is a highly conserved calcium-binding protein that regulates many signaling pathways and has many known binding partners.In addition to identifying known interactions,33novel binding proteins were detected.Sequence comparison revealed a novel binding motif that was related,but distinct from,the previous known calmodulin-binding motif.To demon-strate that proteome chips could be used to globally probe for novel activities,the chips were incubated with®ve different PIs,which are important secondary messengers that regulate diverse cellular processes[56].150novel lipid-binding proteins were identi®ed,49of which exhib-ited preferential binding to PIs.These results convin-cingly showed that proteins immobilized on a surface were able to bind to low molecular weight compounds. This suggests that an entire proteome can be immobi-lized on a glass surface to directly screen for interactions with proteins and small molecules.Peptide arraysIt is of great interest and importance to identify epitopes in proteins that de®ne the core activity.To study the substrates of the nonreceptor tyrosine kinase c-Src, Houseman et al.[18 ]immobilized9-mer peptide sub-strates on a gold-coated glass surface to form a high-density peptide microarray,and characterized the phos-phorylation of the peptide using SPR,¯uorescence and phosphoimaging.They could also quantitatively evaluate the effect of three known inhibitors of the kinase. Although their work was still primitive,the authors demonstrated the potentials of coupling peptide chips with various detection methods to quantitatively study dynamics of enzyme±substrate interactions,and applica-tions in drug discovery.Our group has also designed20 17-mer peptide substrates and covalently immobilized them to epoxy-activated glass surface(unpublished data). 120yeast kinases were screened for their preferred sub-strates.Because peptides are much shorter and more stable than proteins,high-density peptide microarrays can be fabri-cated by direct synthesis of peptides on a surface using photolithography or light-directed synthesis[15,57].Protein chip technology Zhu and Snyder61。

亚细胞定位的maker

亚细胞定位的maker【原创版】目录一、什么是亚细胞定位二、亚细胞定位的作用和意义三、亚细胞定位的实验方法四、亚细胞定位的应用实例五、亚细胞定位的研究发展趋势正文一、什么是亚细胞定位亚细胞定位(Subcellular Localization)是一种实验技术,用于确定蛋白质或表达产物在细胞内的具体位置。

细胞可以分成多个细胞器或者细胞区域,如细胞膜、细胞质、细胞核、线粒体、高尔基体、叶绿体、内质网等,这些细胞器被称为亚细胞。

亚细胞定位的目的是了解蛋白质在细胞内的分布,从而揭示其功能和作用机制。

二、亚细胞定位的作用和意义亚细胞定位对于研究蛋白质功能具有重要作用。

通过亚细胞定位,可以确定蛋白质在细胞内的具体位置,从而揭示其功能和作用机制。

此外,亚细胞定位还可以为药物筛选、疾病诊断和治疗提供有力的依据。

准确地了解某种蛋白功能,需要对其蛋白结构进行分析,同时清楚其亚细胞定位。

三、亚细胞定位的实验方法亚细胞定位实验通常使用荧光显微镜进行观察。

实验过程中,需要构建融合蛋白载体,将有荧光标记的蛋白质与载体融合,然后通过表达载体,使荧光标记的蛋白质在细胞内表达。

在荧光显微镜下,如果看到细胞内某一部位存在荧光信号,则说明与荧光标记的蛋白质融合的蛋白也存在于该部位,这样就达到了确定蛋白质亚细胞定位的目的。

四、亚细胞定位的应用实例亚细胞定位在生物学研究中具有广泛的应用。

例如,通过亚细胞定位可以研究线粒体在细胞内的位置和功能,进一步揭示线粒体在能量代谢和细胞凋亡中的作用。

此外,亚细胞定位还可以用于研究细胞膜转运蛋白、离子通道和受体蛋白等。

五、亚细胞定位的研究发展趋势随着科学技术的发展,亚细胞定位技术不断完善和提高。

未来,亚细胞定位将在生物学研究中发挥越来越重要的作用,为揭示蛋白质功能和疾病机制提供有力支持。

植物细胞中叶绿体蛋白质的亚细胞定位方法

Botanical Research 植物学研究, 2020, 9(3), 268-273Published Online May 2020 in Hans. /journal/brhttps:///10.12677/br.2020.93032Progress on Methods of SubcellularLocalization of Chloroplast Protein inPlant CellsYan Wang, Jie Na*School of Life Sciences, Liaoning Normal University, Dalian LiaoningReceived: Apr. 20th, 2020; accepted: May 19th, 2020; published: May 26th, 2020AbstractChloroplast plays an important role in photosynthesis. Chloroplast proteins distribute in different structures in chloroplast according to their various functions. Proteins could be effective and take part in cell activities only after they are transported to special loci. Therefore, it is necessary to know the subcellular localization of chloroplast protein to understand the relationship between chlorop-last structure and function. Recently, in the research of plant protein localization, some experimen-tal methods assisted by bioinformatic prediction were used to increase the accuracy of protein loca-lization analysis. Basic theory and main steps of these two methods were summarized in this paper in order to provide an effective method for chloroplast protein subcellular localization research.KeywordsChloroplast Protein, Subcellular Localization Methods植物细胞中叶绿体蛋白质的亚细胞定位方法王颜,那杰*辽宁师范大学生命科学学院,辽宁大连收稿日期:2020年4月20日;录用日期:2020年5月19日;发布日期:2020年5月26日摘要叶绿体是植物光合作用的重要场所。

  1. 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
  2. 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
  3. 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
sunzhr@
Abstract
Subcellular localization is a key functional characteristic of proteins. An automatic, reliable and efficient prediction system for protein subcellular localization is needed for large-scale genome analysis. In this paper, we introduce a novel subcellular prediction method combining boosting algorithm with probabilistic neural network algorithm. This new approach provided superior prediction performance compared with existing methods. The total prediction accuracy on Reinhardt and Hubbard’ s dataset reached up to 92.8% for prokaryotic protein sequences and 81.4% for eukaryotic protein sequences under 5-fold cross validation. On our new dataset, the total accuracy achieved 83.2%. This novel method provides superior prediction performance compared with existing algorithms based on amino acid composition and can be a complementing method to other existing methods based on sorting signals. Keywords: Subcellular localization; Boosting;
High throughout genome sequencing projects are producing an enormous amount of raw nucleic acid sequences and protein sequences. The next step is to analysis these sequences begging for finding new gene functions and key regulatory pathways. As a hot topic in genome science, genome function annotation including the assignment of a function for a potential gene in the raw sequence is a vitally important work in genome research. Subcellular location is a key function characteristic of potential gene expressing the protein because the protein functions in the specific location in the intact cells to maintain the cell survival. As a result, the knowledge of protein subcellular location could provide useful information for the gene function prediction. However, subcelluar localization analysis based on experiment is time consuming and could not be — — — — — — — — — — — — — — —
guojian99@
Yuanlie Lin
Faculty of Mathematical Sciences Tsinghua University Beijing, 100084, China
ylin@
Zhirong Sun
Faculty of Institute of Bioinformatics Tsinghua Beijing, 100084, China
Probabilistic neural network; Amino acid composition;
1
Introduction
performed for genome scale proteins. With the rapidly increasing number of sequences in database, it is highly necessary to develop an accurate, reliable and efficient system for protein subcellular localization automatically. Several efforts have been made in the prediction of protein subcellular localization. Up to now mainly two categories of prediction methods have been proposed. One was mainly based on the existence of sorting signals in N-terminal sequences (Nakai, 2000) including signal peptides, mitochondrial targeting peptides and chloroplast transit peptides (Nielsen et al, 1997, 1999). For the improvement of this method, Emanuelsson et al (Emanuelsson et al, 2000) proposed an integrated prediction system with artificial neural network based on individual sorting signal predictions. This system could be use to find cleavage sites in sorting signals and simulate the real sorting process to a certain extent. Nevertheless, the prediction accuracy of those methods based on sorting signals was highly correlated with the quality of protein N-terminal sequence assignment. Unfortunately, it is usually unreliable to annotate the N-terminal using known gene identification methods (Frishman et al, 1999). As a result, the prediction accuracy and reliability decreased when signals were missing or only partially included. The other category of methods was mainly based on the amino acid composition of protein sequences in different subcellular localizations. This approach was first suggested by Nakashima and Nishikwa (1994). They found that the intracellular and the extra cellular proteins could be discriminated with high accuracy only by amino acid composition. From then on, different statistical methods and machine learning methods have been used based on amino acid composition of protein sequences to improve prediction accuracy. Cedano et al (1997) adopted a statistical method with Mahalanobis distance for prediction. Reinhardt and Hubbard (1998) predicted subcellular locations with neural networks and reached the accuracy 66% for eukaryotic sequences and 81% for prokaryotic sequences. Chou et al (1999) proposed the covariant discriminant algorithm on the same prokaryotic dataset as Reinhardt et al. and achieved a total accuracy of 87%. Hua and Sun (2001) constructed a prediction system using support vector machine (SVM)— — a new machine learning method based on the statistical learning theory— — on the same prokaryotic and eukaryotic datasets. The prediction accuracy of Hua et al has reached up to 91.4% for prokaryotic proteins and 79.4% for eukaryotic proteins.
相关文档
最新文档