Improved protein fold assignment using support

合集下载

Zinc finger锌指

Zinc finger锌指

Zinc fingerZinc fingers are small protein structural motifs that can coordinate one or more zinc ions to help stabilize their folds. They can be classified into several different structural families (zinc finger proteins) and typically function as interaction modules that bind DNA, RNA, proteins, or small molecules. The name "zinc finger" was originally coined to describe the finger-like appearance of a diagram showing the hypothesized structure of the repeated unit in Xenopus laevis transcription factor IIIA.[1]Zinc fingers coordinate zinc ions with a combination of cysteine and histidine residues. They can be classified by the type and order of these zinc coordinating residues (e.g., Cys2His2, Cys4, and Cys6). A more systematic method classifies them into different "fold groups" based on the overall shape of the protein backbone in the folded domain. The most common "fold groups" of zinc fingers are the Cys2His2-like (the "classic zinc finger"), treble clef, and zinc ribbonEngineered zinc finger arraysGenerating arrays of engineered Cys2His2 zinc fingers is the most developed method for creating proteins capable of targeting desired genomic DNA sequences. The majority of engineered zinc finger arrays are based on the zinc finger domain of the murine transcription factor Zif268, although some groups have used zinc finger arrays based on the human transcription factor SP1. Zif268 has three individual zinc finger motifs that collectively bind a 9 bp sequence with high affinity.[4] The structure of this protein bound to DNA was solved in 1991[5] and stimulated a great deal of research into engineered zinc finger arrays. In 1994 and 1995, a number of groups used phage display to alter the specificity of a single zinc finger of Zif268.[6][7][8][9] Carlos F. Barbas et al. also reported the development of zinc finger technology in the patent literature and have been granted a number of patents that have been important for the commercial development of zinc finger technology.[10][11] Typical engineered zinc finger arrays have between 3 and 6 individual zinc finger motifs and bind target sites ranging from 9 basepairs to 18 basepairs in length. Arrays with 6 zinc finger motifs are particularly attractive because they bind a target site that is long enough to have a good chance of being unique in a mammalian genome.[12] There are two main methods currently used to generate engineered zinc finger arrays, modular assembly and a bacterial selection system, and there is some debate about which method is best suited for most applications.[13][14][edit] Modular assemblyThe most straightforward method to generate new zinc finger arrays is to combine smaller zinc finger "modules" of known specificity. The structure of the zinc finger protein Zif268 bound to DNA described by Pavletich and Pabo in their 1991 publication has been key to much of this work and describes the concept of obtaining fingers for each of the 64 possible base pair triplets and then mixing and matching these fingers to design proteins with any desired sequence specificity.[5] The most common modular assembly process involves combining separate zinc fingers that can each recognize a 3 basepair DNA sequence to generate 3-finger, 4-, 5-, or 6-finger arrays that recognize target sites ranging from 9 basepairs to 18 basepairs in length. Another method uses 2-finger modules to generate zinc finger arrays with up to six individual zinc fingers.[15] The Barbas Laboratory of The ScrippsResearch Institute used phage display to develop and characterize zinc finger domains that recognize most DNA triplet sequences[16][17][18] while another group isolated and characterized individual fingers from the human genome.[19] A potential drawback with modular assembly in general is that specificities of individual zinc finger can overlap and can depend on the context of the surrounding zinc fingers and DNA. A recent study demonstrated that a high proportion of 3-finger zinc finger arrays generated by modular assembly fail togenerate zinc finger nucleases with both 3-finger arrays and 4-finger arrays and observed a much higher success rate with 4-finger arrays.[21] A variant of modular assembly that takes the context of neighboring fingers into account has also been reported and this method tends to yield proteins with improved performance relative to standard modular assembly.[22][edit] Selection methodsNumerous selection methods have been used to generate zinc finger arrays capable of targeting desired sequences. Initial selection efforts utilized phage display to select proteins that bound a given DNA target from a large pool of partially randomized zinc finger arrays. This technique is difficult to use on more than a single zinc finger at a time, so a multi-step processes that generated a completely optimized 3-finger array by adding and optimizing a single zinc finger at a time was developed.[23] More recent efforts have utilized yeast one-hybrid systems, bacterial one-hybrid and two-hybrid systems, and mammalian cells. A promising new method to select novel 3-finger zinc finger arrays utilizes a bacterial two-hybrid system and has been dubbed "OPEN" by its creators.[24] This system combines pre-selected pools of individual zinc fingers that were each selected to bind a given triplet and then utilizes a second round of selection to obtain 3-finger arrays capable of binding a desired 9-bp sequence. This system was developed by the Zinc Finger Consortium as an alternative to commercial sources of engineered zinc finger arrays. It is somewhat difficult to directly compare the binding properties of proteins generated with this method to proteins generated by modular assembly as the specificity profiles of proteins generated by the OPEN method have never been reported.[edit] ApplicationsEngineered zinc finger arrays can then be used in numerous applications such as artificial transcription factors, zinc finger methylases, zinc finger recombinases, and Zinc finger nucleases.[25] While initial studies with another DNA-binding domain from bacterial TAL effectors show promise,[26][27][28][29] it remains to be seen whether these domains are suitable for some or all of the applications where engineered zinc fingers are currently used. Artificial transcription factors with engineered zinc finger arrays have been used in numerous scientific studies, and an artificial transcription factor that activates expression of VEGF is currently being evaluated in humans as a potential treatment for several clinical indications. Zinc finger nucleases have become useful reagents for manipulating genomes of many higher organisms including Drosophila melanogaster, Caenorhabditis elegans, tobacco, corn,[15]zebrafish,[30] various types of mammalian cells,[31] and rats.[32] An ongoing clinical trial is evaluating Zinc finger nucleases that disrupt the CCR5 gene in CD4+ human T-cells as a potential treatment for HIV/AIDS.[33]Phage displayPhage display is a method for the study of protein–protein, protein–peptide, and protein–DNA interactions that uses bacteriophages to connect proteins with the genetic information that encodes them.[1] Phage Display was originally invented by George P. Smith in 1985 and he demonstrated the display of peptides on filamentous phage by fusing the peptide of interest on to gene3 of filamentous phage.[1] This technology was further developed and improved by groups at the MRC Laboratory of Molecular Biology with Winter and McCafferty and The Scripps Research Institute with Lerner and Barbas for display of proteins like antibodies for therapeutic protein engineering. The connection between genotype and phenotype enables large libraries of proteins to be screened and amplified in a process called in vitro selection, which is analogous to natural selection. The most common bacteriophages used in phage display are M13 and fd filamentous phage,[2][3] though T4,[4]T7, and λ phage have also been used.[edit] PrincipleLike the two-hybrid system, phage display is used for the high-throughput screening of protein interactions. In the case of M13 filamentous phage display, the DNA encoding the protein or peptide of interest is ligated into the pIII or pVIII gene, encoding either the minor or major coat protein, respectively. Multiple cloning sites are sometimes used to ensure thatthe fragments are inserted in all three possible frames so that the cDNA fragment is translated in the proper frame. The phage gene and insert DNA hybrid is then transformed into Escherichia coli (E. coli) bacterial cells such as TG1, SS320, ER2738, or XL1-Blue E. coli. If a "phagemid" vector is used (a simplified display construct vector) phage particles will not be released from the E. coli cells until they are infected with helper phage, which enables packaging of the phage DNA and assembly of the mature virions with the relevant protein fragment as part of their outer coat on either the minor (pIII) or major (pVIII) coat protein.By immobilizing a relevant DNA or protein target(s) to the surface of a well, a phage that displays a protein that binds to one of those targets on its surface will remain while others are removed by washing. Those that remain can be eluted, used to produce more phage (by bacterial infection with helper phage) and so produce a phage mixture that is enriched with relevant (i.e. binding) phage. The repeated cycling of these steps is referred to as 'panning', in reference to the enrichment of a sample of gold by removing undesirable materials.Phage eluted in the final step can be used to infect a suitable bacterial host, from which the phagemids can be collected and the relevant DNA sequence excised and sequenced to identify the relevant, interacting proteins or protein fragments.The use of a helper phage can be eliminated by using 'bacterial packaging cell line' technology.[5][edit] General protocol1.Target proteins or DNA sequences are immobilised to the wells of a microtiter plate.2.Many genetic sequences are expressed in a bacteriophage library in the form of fusions withthe bacteriophage coat protein, so that they are displayed on the surface of the viral particle.The protein displayed corresponds to the genetic sequence within the phage.3.This phage-display library is added to the dish and after allowing the phage time to bind, thedish is washed.4.Phage-displaying proteins that interact with the target molecules remain attached to thedish, while all others are washed away.5.Attached phage may be eluted and used to create more phage by infection of suitablebacterial hosts. The new phage constitutes an enriched mixture, containing considerably less irrelevant phage (i.e. non-binding) than were present in the initial mixture.6.The DNA within the interacting phage contains the sequences of interacting proteins, andfollowing further bacterial-based amplification, can be sequenced to identify the relevant,interacting proteins or protein fragments.[edit] ApplicationsThe applications of this technology include determination of interaction partners of a protein (which would be used as the immobilised phage "bait" with a DNA library consisting of all coding sequences of a cell, tissue or organism) so that new functions or mechanisms of function of that protein may be inferred.[6] The technique is also used to determine tumour antigens (for use in diagnosis and therapeutic targeting)[7] and in searching for protein–DNA interactions[8] using specially-constructed DNA libraries with randomised segments.Phage display is also a widely used method for in vitro protein evolution (also called protein engineering). As such, phage display is a useful tool in drug discovery. It is used for finding new ligands (enzyme inhibitors, receptor agonists and antagonists) to target proteins.[9][10][11] Invention of antibody phage display by laboratories at the MRC Laboratory of Molecular Biology led by Greg Winter and John McCafferty and at The Scripps Research Institute led by Richard Lerner and Carlos F. Barbas revolutionised antibody drug discovery.[12][13] In 1991, The Scripps group reported the first display and selection of human antibodies on phage.[14] This initial study described the rapid isolation of human antibody Fab fragements that bound tetanus toxin and the method was then extended to rapidly clone human anti-HIV-1 antibodies for vaccine design and therapy.[15][16][17][18][19] Following the pioneering disclosures of these laboratories phage display of antibody libraries became a powerful method for both studying the immune response as well as a method to rapidly select and evolve human antibodies for therapy. Antibody phage display was later used by Carlos F. Barbas at The Scripps Research Institute to create the first synthetic human antibody libraries, thereby allowing human antibodies to be created in vitro from synthetic diversity elements.[20][21][22][23] Antibody libraries displaying millions of different antibodies on phage are frequently used in the pharmaceutical industry for isolation of highly specific therapeutic antibody leads, for development into primarily anti-cancer or anti-inflammatory antibody drugs. One of the most successful was HUMIRA (adalimumab), discovered by Cambridge Antibody Technology as D2E7 and developed and marketed by Abbott Laboratories. HUMIRA, an antibody to TNF alpha, was the world's first fully human antibody,[24] which achieved annual sales exceeding $1bn.[25]Competing methods for in vitro protein evolution are yeast display, bacterial display, ribosome display, and mRNA display.。

蛋白质的结构、分类及预测

蛋白质的结构、分类及预测
4
结构域是在超二级结构的基础上形成的, 通常由50-300个氨基酸残基组成,其特点 是在三维空间可以明显区分和相对独立, 并且具有一定的生物功能。 模体或基序(motif)是结构域的亚单位, 通常由2~3二级结构单位组成。 较大的蛋白质分子一般含有两个以上的结 构域,其间以柔性的铰链(hinge)相连,
25
蛋白质结构的可视化软件
Weblab Viewlite 5.0 (DS ViewerPro 5.0 ) RASMOL 2.7.2.1 Swiss-PdbViewer 3.7 CHIME 2.6 INSIGHTII Cn3D (NCBI格式)
26
Worms (Schematic)
27
通过Entrez检索
通过PDB id进 行检索
35
VAST and VAST Search
URL:
/Structure/VAST/vast.shtml
用于确定相似的蛋白三维结构
– 已知结构:pre-calculated – 新解析结构:VAST搜索
24
PDB格式文件的主要内容
结构名称、编号、简单说明、递交日期 化合物名称、来源、测定方法、分辨率 结构递交者姓名、单位、联系地址 相关文献作者、题目、刊物、日期 结构测定和修正注释 一级结构、二级结构、二硫键、复合物信息 构晶胞参数、旋转矩阵 原子坐标 二硫键配对标记 文件结束标记
对PDB数据解释和分类的数据库
MMDB (Molecular Modeling DataBase )
URL: /Structure/MMDB/m mdb.shtml 数据:PDB数据库中的实验来源数据 数据格式:ASN.1 可视工具:Cn3D (See In 3D) VAST (Vector Alignment Search Tool) 矢量同源 比较搜索工具 34

Autodesk Fabrication CAMduct 2013 Service Pack 4 E

Autodesk Fabrication CAMduct 2013 Service Pack 4 E

Autodesk®Fabrication CAMduct™ 2013Service Pack 4 Enhancement ListImprovements made in Service Pack 4 build 3.01.193:∙All SP1, SP2 and SP3 enhancements incorporated (see below)∙Improved file compatibility for Farley.exe.∙Enhanced pierced holes visibility in takeoff developments.∙Improved coil line processor option text installation.∙Enhanced NC Part Request by Type columns.∙Improved stability of pattern CID 850.∙Enhancements made to various post processors.∙Improved scripting functionality to allow users to return values from catalogued item options.∙Stability improvement made to ancillary database components to allow the assignment of lone fabrication tables.Improvements made in Service Pack 3 build 3.01.154:∙All SP1 and SP2 enhancements incorporated (see below)∙Enhanced Scripting to support “Group” prefix when changing Item Specification or Material∙Improved import functionality when specifications contain alternate connectors and seams.∙Enhanced status time/dates to report out in local time rather than UTC format.∙More consistently apply collar seams from the specification to various round pattern developments.∙Enhanced ancillary database display to support sorting.∙Update position of Splitter holes in developments of Radius Elbow and Breeches piece.∙Enhanced hole development positions and notching when using connectors with –ve Turnover.∙Enhanced Mach3 post processor for plate detection support in Z axis for varying heights for pierce and cut.∙Enhanced Mitsubishi post processor for etching text.∙Enhanced Salvagnini post processor to control head movement when traversing and support laser marking.∙Enhanced CRE post processer for central cuts with 0 offset (rip cut) and view NC control.∙Enhanced Micro Step post process with support for Micro Punch tooling.∙Enhanced Fanuc Plasma post processor with height control.∙Enhanced AMS coil line post with tie rod holes and collate 4 piece and U+ straights.∙Improved support for displaying material and gauge properties in manual nest.∙Enhanced NFP nesting to better support nesting in holes and auto quantity parts.∙Updated oversize dialogue to redisplay the “show” control when hidden.∙Enhanced quick takeoff with attacher arrow control.∙Improved support for 3D viewer printing worksheets.Improvements made in Service Pack 2 build 3.01.094:∙All SP1 enhancements incorporated (see below).∙Attacher Arrow and associated functionality enhanced in 3D Viewer.∙Improved stability using Slice tool in Opus/Profiler.∙Enhanced stability of Write NC.∙Improved support in Amada post processor to prevent short arcs being output as full circles.∙Enhanced Burny10 post processor to support alternate tool On/Off commands M21/M20.∙Updated Cyberstep post processor to support Torch Height Control (THC).∙Improved support in Vicon decoiler post processor when outputting Metric values on an Imperial setup.∙Enhanced Digisaf 620 post processor to support multiple tools (Oxy fuel, Plasma cut and mark).∙Improve support for machines with more than one tool and using more than one setup rule. i.e. Hypertherm Voyager.∙Enhanced support for kerf compensation when used with DuctBoard patterns, i.e. keft left and kerf right supported.∙Enhanced CID 33 for processing multiple branches when one of the diameters is set to 0.∙Enhanced CID 7 to support the appropriate connector fold notches for C1 and C2 ends.∙Improved support for Double Walled items for sizing Insulation panels.Improvements made in Service Pack 1 build 3.01.057:∙Barcode settings, when changed in the main database, are maintained between sessions∙Opus now loads Raster to Vector DLL on 64 bit systems∙Enhanced stability of Opus DXF import∙Catalogue items are no longer created when enhanced editing a template ITM though Folders and selecting develop.∙Windows permissions validation now occurs when Writing NC.∙MAP2ADSK addresses various incidents with regard to migrating from older systems.∙Enable Item Spool, eTag, Zone and Alt fields on item and as print objects.∙Enhance Grooving/Marking options for panels when angle is less user defined angle, switches to marking tool.∙Print object Item Duct Weight adjusted for units used.∙Splitter entries in dialogue editor synced with regard to specification entries∙Plasma Straight, marker notches included from the developments.∙Oval Collars now resolve holes moving to accommodate notch positions.∙Square Elbow – Option provided for Seam Number for Throat supports more effective application of seams.∙Multi Branch on Reducer now allows multiple length splits along the body.∙Branch on Reducer: The option to change seam position on the body, is now in the development.∙Developments adjusted for seam lengths on Elliptical Reducers∙Extension allowances on Straights when working with Square Elbows and Square Tees have been improved.∙Enhanced stability for Setup Processes, Export Data and Select Exports button.∙Enhanced browsing through reports in Item Report BuilderAutodesk, CADmep, CAMduct, and ESTmep are registered trademarks or trademarks of Autodesk, Inc., and/or its subsidiaries and/or affiliates in the USA and/or other coun-tries. All other brand names, product names, or trademarks belong to their respective holders. Autodesk reserves the right to alter product and services offerings, and specifications and pricing at any time without notice, and is not responsible for typographical or graphical errors that may appear in this document. © 2013 Autodesk, Inc. All rights reserved.。

硕士毕业论文-蛋白质生物功能的机器学习方法研究37075

硕士毕业论文-蛋白质生物功能的机器学习方法研究37075

硕士学位论文SHANGHAI UNIVERSITYMASTER DISSERTATION题蛋白质生物功能的机器学习方法研究目摘要近些年来,随着信息技术和生物检测手段的不断发展,生命科学的数据资源急剧膨胀。

实验工作者在产生大量数据的同时,也对理论研究者提出了更多的难题。

利用机器学习这一方法来分析这些数据,我们可以从中找出隐含的规律和模式,从而进一步加深对事物的认识。

本文就是采取这一研究方法,对蛋白质的生物功能进行建模和预报。

在本文的工作中,我们使用了机器学习方法来对蛋白质和小分子的相互作用、蛋白质糖基化位点的识别进行建模和预报。

另外我们还探讨了一系蛋白质列生物功能在线预报系统的建设和优化。

本文的主体工作分为三个部分:1.用集成学习算法对蛋白质和小分子的相互作用进行研究。

我们针对代谢途径下的酶和底物之间的相关作用,建立了相互作用预报模型。

通过对数据集的变量筛选和降维的评价,我们保留了原有的变量集合。

在后续的建模过程中分别用AdaBoost,Bagging, SVM, KNN, 决策树对酶和底物进行建模。

10组交叉验证和独力测试集的结构显示,集成学习方法AdaBoost,Bagging的分类能力最好,都达到了71%以上。

而我们接着又把不同的分类器组合集成后发现,前2个性能最好的集成学习算法和KNN组合后的体系具有最好的推广能力,其独立测试集中正样本的正确率又在原先最好的结果下提高了近4%,而其总体正确率也达到了84.6%。

结果证明,多重集成学习算法可以用来研究蛋白质和小分子相互作用,所得到的模型有很好的预测性能。

此外,我们根据所建立的酶和底物相互作用的预测模型,同时开发了相应的在线预报系统。

2.用CFS-Wrapper筛选变量法结合AdaBoost集成方法对蛋白质O端糖基化位点进行研究。

在许多的生化过程中都需要有O-端糖链的参与。

然而糖基化是一个复杂的过程,迄今为止还未得出一个固定的模式。

我们对收集到的糖基化和非糖基化肽段, 并用肽段中残基的物化参数,以AAIndex库中的数据进行表征。

基于ELMC的蛋白质折叠识别方法

基于ELMC的蛋白质折叠识别方法

基于ELMC的蛋白质折叠识别方法唐立力【摘要】传统的机器学习方法在处理蛋白质折叠类型识别问题时需要花费大量的时间来调节最佳参数,利用一种新的极限学习机(Extreme Learning Machine,ELM)分类优化方法(Extreme Learning Machine for Classification,ELMC)对蛋白质折叠进行识别,仅需调节很少的参数值就可达到很好的测试精度。

与支持向量机(Support Vector Machine,SVM)和推荐相关向量机(Relevance Vector Machine,RVM)相比,ELMC能获得更好的泛化性能,而且在寻找最优解的训练时间比较上,ELMC比SVM平均要快35倍,比RVM要快12倍。

%With traditional machine learning methods, one may spends a lot of time adjusting the optimal parameters in tackling the problem of protein fold recognition. A new optimization method of ELM for classification is used to recognize the protein fold, one can only adjusts few parameters to achieve good enough testing accuracy. Compared to SVM and RVM, better general-ization performance can be obtained by ELMC, in the comparison of training time in finding the optimal solution, ELMC is 35 times faster than SVM averagely and is 12 times faster than RVM averagely.【期刊名称】《计算机工程与应用》【年(卷),期】2013(000)010【总页数】4页(P114-117)【关键词】蛋白质折叠识别;ELM分类优化方法;多类分类【作者】唐立力【作者单位】重庆工商大学融智学院,重庆 400033【正文语种】中文【中图分类】TP315蛋白质的三维空间结构是由它的氨基酸序列决定,而氨基酸序列如何决定空间结构是生物学研究的重要问题之一。

Protein-Protein Interaction Networks, developed by

Protein-Protein Interaction Networks, developed by

ProViz:Protein Interaction Visualization and ExplorationFlorian Iragne (1),Macha Nikolski (1),Bertrand Mathieu (1),David Auber (1)and David Sherman(1)(1)LaBRI UMR 5800,Universit´e Bordeaux 1,351cours de la Lib´e ration,33405Talence Cedex,Franceiragne@labri.fr,macha@labri.fr,mathieu@labri.fr,auber@labri.fr,david@labri.frAbstractSummary :ProViz is a tool for the visualization ofProtein-Protein Interaction Networks,developed by the IntAct european project.It provides facilities for navigating in large graphs and exploring biologically-relevant features,and adopts emerging standards such as GO and PSI-MI.Availability :ProViz is available under the GPL and may be freely downloaded.1Contact :david.sherman@labri.frIntroductionAnalysis of protein-protein interaction (PPI)net-works requires a combination of algorithmic and vi-sualization tools,ideally integrated within a software platform that is itself integrated with access to local and distant data banks.We present a software tool called ProViz that provides highly-interactive visual-ization of large networks of interactions,integrated with the IntAct data model[5].ProViz is similar in purpose to PIMrider,[8],Osprey[2],and other visual-ization or analysis tools[11,7,6,10].Overview of ProVizGraph drawing and interactive graph exploration are active domains in computer science and many tools are available for this task.Adaptation of these tools and techniques to the specific needs of biologists exploring PPI networks is a current effort in bioinfor-matics.The challenge is to add valuable information and functions that enable the user to discover interest-ing biological relations hidden within the data.ProViz improves over existing work by providing a fast,scalable,open tool with extensive plugins,that integrates emerging standards for representing biolog-ical knowledge in a biologist-oriented interface.Intended use ProViz is designed with an under-standing of the ways that biologists prefer to work.It may be used for exploring large graphs in order to identify proteins and interactions of interest,ei-ther through keyword search or through analysis of the combinatorial structure of the network;for comparing graphs from different strains or species over ortholo-gous sets of genes;for extracting views and subgraphsfor further analysis;and for clustering related proteins and interactions (see examples in Supplementary Ma-terial).ProViz is highly interactive,providing screen updates within 50ms on standard workstations while manipulating graphs with a million elements.ProViz can be a content-type helper for interaction database query results in PSI-MI format[3].Name-based or sequence-based queries to the IntAct feder-ated database of protein-protein interactions produce networks that “link out”to ProViz for detailed study,and protein nodes and interaction edges link back in to IntAct web services.User interface The ProViz screen is intentionally uncluttered (figure 1).The right half of the screen dis-plays the current view of the current graph;the differ-ent views available are selected through the use of tabs above the window.Above this window in the tool bar are buttons for changing the layout of the current view.The mouse can be used to select elements or to move elements,and the mouse wheel can be used to zoom in or out and to pan the image.Below are buttons for cloning and for closing the view.Four tabs are avail-able on the left:Views ,for information about existing views;Node Ontology ,for selecting proteins based on GO terms;Edge Ontology ,for selecting interac-tions based on controlled vocabularies;and Proper-ties ,for viewing the complete set of properties associ-ated with a node or edge element.Views Subgraphs produced by selection,filtering,or clustering are automatically organized into views that can be manipulated independently and used to produce subsequent views.Each view has its own lay-out and zoom,and views can be used to compare dif-ferent analyses of the same interaction network.Views are organized in a tree,a quotient graph whose nodes are individual subgraphs.Layout algorithms Of the dozens of layout algo-rithms in the plugin library,three were chosen for direct use based on their capacity to highlight bio-logically pertinent information.GEM [4]is an effi-cient directed force-based graph drawing algorithm.It groups related nodes and can be used to quickly iden-tify proteins with a given role,or for visualizing pro-tein complexes.Hierarchical layout [9]reveals ances-1Sourcecode and binaries are available at bri.fr/eng/proviz.htmBioinfor m atics © Oxford University Press 2004; all rights reserved.Bioinformatics Advance Access published September 3, 2004Figure1.View of a spoke-model PPI graph for yeast.(a)Protein properties(b)Protein selection using GO terms(c)Main window showing part of current view(right)and the cluster tree(left)tral relationships between nodes and is useful when looking for cascade-type interactions or comparison to metabolic pathway data.Circular layout is a neutral choice that does not attribute any semantics to edge relations.Integrating controlled vocabularies ProViz uses GO and PSI-MI controlled vocabularies for describing proteins and ers employ these vocabu-laries when building views of interaction networks by manualfiltering or through the use of clustering plug-ins.Infigure1we see the property list for the node corresponding to yeast Rad16(nucleotide excision re-pair protein),including GO evidence,gene names,and external links.Tulip Development Platform ProViz development is based on the Tulip platform[1],designed for management and three-dimensional display of large graphs.It provides a rich set of operations on graphs: metric computation,node and edge layout,selection, extraction of view and subgraphs,and labeling of nodes and edges with arbitrary sets of attributes.Oper-ations specific to the application domain are provided by means of software plugins.Any program using Tulip can add to the core features by providing its own domain-specific plugins.Tulip is written in C++and uses Qt and OpenGL for enhanced portability. AckowledgementsThis work is supported by EU grant number QLRI-CT-2001-00015under the RDD programme“Quality of Life and Management of Living Resources.”References[1]David Auber.Tulip-a huge graph visualizationframework.In M.Juenger and P.Mutzel,editors,Graph Drawing Software,Mathematics and Vi-sualization,pages80–102,Heidelberg,August 2003.Springer-Verlag.[2]BJ.Breitkreutz,C.Stark,and M.Tyers.Osprey:a network visualization system.Genome Biol-ogy,4(3):R22,2003.[3]H.Hermjakob et al.(39authors).The HUPOPSI’s molecular interaction format–a community standard for the representation of protein interac-tion data.Nat.Biotechnol.,22(2):177–83,Feb.2004.[4]A.Frick,A.Ludwig,and H.Mehldan.A fastadaptive layout algorithm for undirected graphs.In Springer-Verlag,editor,Proc.Workshop on Graph Drawing94,volume LNCS894,pages 389–403,1994.[5]H.Hermjakob,L.Montecchi-Palazzi,C.Lew-ington,S.Mudali,S.Kerrien,S.Orchard, M.Vingron,B.Roechert,P.Roepstorff,A.Va-lencia,H.Margalit,J.Armstrong,A.Bairoch,G.Cesareni, D.Sherman,and R.Apweiler.Intact:an open source molecular interaction database.Nucleic Acids Res.,32:D452–5,Jan.2004.[6]T.Koike and A.Rzhetsky.A graphic editor foranalysing signal-transduction pathways.Gene, 259:235–244,2000.[7]ppe,J.Park,O.Niggermann,and L.Holm.Generating protein interaction maps from in-complete data:application to fold assignment.Bioinformatics,17(1):S149–S156,2001.[8]P.Legrain,J.Wojcik,and JM.Gauthier.Protein-protein interaction maps:a lead towards cellular functions.Trends in Genetics,17,2001.[9]E.B.Messinger,L.A.Rowe,and R.R.Henry.Adivide and conquer algorithm for the automaticlayout of large directed graphs.IEEE Transac-tions on Systems,Man and Cybernetics,SMC-21(1):1–11,Jan/Feb1991.[10]P.Shannon,A.Markiel,O.Ozier,N.Baliga,J.Wang,D.Ramage,N.Amin,B.Schwikowski,and T.Ideker.Cytoscape:a software environ-ment for integrated models of biomolecular in-teraction networks.Genome Res.,13(11):2498–504,Nov.2003.[11]CL.Tucker,JF.Gera,and P.Uetz.Towardsan understanding of complex protein networks.Trends in Cell Biol.,11(3):102–106,2001.。

Protein A, Protein G, and Protein 原理及应用

Protein A, Protein G, and Protein 原理及应用

Chapter24Analysis and Purification of Antibody Fragments Using Protein A,Protein G,and Protein LRemko Griep and John McDougall24.1IntroductionToday,monoclonal antibodies(mAbs)form the largest category of biopharmaceu-ticals in clinical trials,and their number is expanding rapidly(DataMonitor2007a,b). The antibodies or functional antibody fragments are being produced not only in artificial production systems such as mammalian cells,yeast,bacteria,and plant cells but also in transgenic animals such as goats,sheep,and cows.Regardless of the production method,the quality control demand is the same for all of them.Host cell proteins,cell culture media additives,DNA,and endotoxins have to be removed from the mAb preparation to allow the proteins to be safely applied for human therapy.Moreover,antibody aggregates,clipped and low molecular weight species, should also be removed.Several proteins with an inherent affinity for immunoglobulins(Ig)have been isolated from various bacteria.These molecules include protein-A,derived from Staphylococcus aureus(Forsgren and Sjo¨quist1966);protein-G,derived from a group-C Streptococcus(Bjo¨rk and Kronvall1984);andfinally protein-L,derived from Peptostreptococcus magnus(A˚kerstro¨m and Bjo¨rk1989;Housden et al.2003, 2004).They all contain repetitive55–76amino acid residues(Fig.24.1)that mediate the actual Ig binding(Kastern et al.1992).The recombinant protein-L can be produced at a yield of up to3g/L in pilot-scale studies.It yields a highly pure,stable,and active protein-L fraction after purification,which is binding efficiently to most of the human antibodies of the Kappa isotype(Fig.24.2).Protein-G binds not only to the Fc-region but also to the CH1-domain of the human IgG1-isotype.Therefore,it has a broader application compared to protein-A. Some academic groups have also reported the use of genetically fused protein-LG (Kihlberg et al.1996;Harrison et al.2008)or protein-AG(Eliasson et al.1988; R.Griep(*)and J.McDougallAffitech AS,Gaustadalle´en21,Oslo3490,Norwaye-mail:r.griep@affi301 R.Kontermann and S.Du¨bel(eds.),Antibody Engineering Vol.2,DOI10.1007/978-3-642-01147-4_24,#Springer-Verlag Berlin Heidelberg2010Bergmann-Leitner et al.2008)and protein-LA (Svensson et al.1998)for monoclo-nal antibody purification.They indeed obtained broader functional ligands because the binding characteristics of both parental proteins were maintained.The ability of protein-A,-G,or -L to maintain their functionality,on conjugation with fluoro-chromes,enzymes (Fig.24.3a ),or gold particles,makes them highly valuable secondary reagents for the detection of primary antibodies in ELISA,immunohis-tochemistry,flowcytometry,and electronmicroscopy.Protein-A mainly binds to the Fc-region of the IgG from several human isotypes (Table 24.1)but only to a single variable region of the heavy-chain family (Starovasnik et al.1999).In contrast,protein-L binds to most of the human Kappa light-chains of the k I,k III,and k IV families.These comprise 55–60%of all IgA,IgE,and IgM antibodies in the human serum (Solomon 1976)and can thus be used to purify all monoclonal antibodies of those Kappa sub-types (Nilson et al.1992)or fragments derived thereof.This,without the need to genetically engineer affinity-tags onto the protein of interest (Devaux et al.2001;Das et al.2005;Cossins et al.2007).The k antibodies described in Fig 24.3b were originally derived from a large human unbiased antibody phage library (Løset et al.2005)and six out of the ten k antibodies strongly react with protein-L (Fig.24.3b ).These authors also demon-strated that preselection of this particular phage-library for the binding to protein-L can be of use.It yields phage-antibodies with improved functionality,as each phage is actually assayed for its ability to express at least one functional scFv on its surface prior to its selection against an antigen.An alternative approach is to build a highly diverse library,on the basis of certain well-expressing and protein-L binding Kappa light-chain genes (Holt et al.2008).Moreover,protein-L has a clear advantage over protein-A and protein-G,as it does not bind to bovine IgG or to bovine serum albumin.This might be of major importance when one is forced to use bovine serum as additive to the cell culture medium to prevent certain types of mammalian cells from dying.Thus far,protein-L has not been available for theindustrial-scale117824473169575239Fig.24.1Structure of the protein-L molecule comprising 719amino acids.The numbers,indicat-ing the amino acids of the beginning of each domain,are listed below the boxes.Included are the signal peptide (SP),the signal peptide cleavage site is indicated by the arrow,the NH 2-terminal(A),the repeated units with Ig-binding activity (B1–B5),the spacer region (S),the repeats (C),the wall spanning domain (W),and the transmembrane region (M).The recombinant protein-L consists of four Ig-binding domains (B1–B4),which can bind to the Kappa region without interfering with the antigen-binding site of the immunoglobulin302R.Griep and J.McDougallpurification,but recently,a development toward introduction into the bulk market has been initiated.A prerequisite for (cost)-efficient industrial-scale purification of MAbs is that the ligands like protein-A,protein-L,and protein-G can be coupled efficiently to solid matrices like controlled pore glass (Millipore)and to agarose with varying degrees of cross-linking (GE Healthcare).These materials are rigid and can be operated at high flow velocities.Highly porous materials exert a low-pressure drop,a low mass transfer resistance,and a high dynamic capacity (LeVan et al.1997).Unfortunately,these features are nonexclusive to a certain extent.A highly porous medium could have a low equilibrium capacity because of a limited surface area and simulta-neously have good mass transfer characteristics but bad flow properties as a result bm g /m L Protein-L 2341-+ -+ IgG 101520253750kDa751342ac Tim e after induction Fig.24.2(a )A pilot-scale production system has been set up for production of recombinant protein-L in E.coli .The DNA sequence encoding the B1-4domains has been cloned into a pJB-vector (Sletta et al.2004)and the recombinant protein-L was expressed intracellular in high-cell-density-cultivation as shown here for five separate cases.The produced protein-L was extracted from the cytoplasm,purified,and analyzed by SDS-PAGE.(b )SDS-PAGE analysis of the purified protein-L (c )CNBR activated-sepharose beads were conjugated without (À)and with polyclonal human IgG (þ)and incubated with protein-L preparations which were stored for 1month,either at 4 C (lane 1and 2)or at 37 C (lane 3and 4).Subsequently,the obtained supernatants were analyzed by SDS-PAGE for the presence of unbound protein-L.As can be observed from this picture,the majority of the protein-L is specifically binding to the polyclonal IgG,even after storage for 1month at 37 C24Analysis and Purification of Antibody Fragments 303of its softness.In contrast,a resin with a high equilibrium capacity might have increased mass transfer resistance.As the costs of resins are high,the ligands should maintain their selectivity and have good chemical stabilities over a long period of time.Cleaning in place procedures (CIP)with repeated alkaline exposures can be detrimental for ligands like protein-A and protein-G.To facilitate CIP,some of the ligands,such as MabSelect (GE Healthcare)or a protein-A analog Z(F30A)(Linhult et al.2004),could be optimized and are now available as an improved alkaline resistant alter-native for protein-A.Also,for protein-G,an improved mutant was engineered (Gu¨lich et al.2002),while according to the results of Enever (Enever et al.2005),higher affinity variants can also be expected for protein-L.Because of the acidic elution and the high concentration of Mabs on the column,aggregates are easily formed (Shukla et al.2007).In addition,leaching and cleavage of the ligand is observed for protein-A (Carter-Franklin et al.2007)and protein-G.As a consequence,both aggregates and leached ligand have to be removed from the a0.40.81.21.6O D 405Applied antibody0.51.01.52.02.53.0I g G -1I g G -2I g G -3I g G -4I g G -5I g G -6I g G -7I g G -8F ab -1sc F v -1c o n t r o l O D 405bFig.24.3(a )Quality control of the produced recombinant protein-L with the aid of an ELISA.A maxisorb ELISA-plate,coated with human IgG,was preincubated with different concentrations of unconjugated recombinant protein-L (rProtein-L TM ,#101Actigen)prior to incubation with a protein-L/HRP conjugate (rProtein-L TM HRP,#301Actigen).After washing,chromogenic sub-strate was added and the absorbance of the individual wells was measured at OD 405nm.The signal shows clear inhibition by the unconjugated protein-L (b )A maxisorb ELISA-plate,coated with different human IgG(k )antibodies,a human Fab(k )or with a scFv(k )fragment (all at 0.1m g/well)was incubated with a protein-L/HRP conjugate.After washing,chromogenic substrate was added and the absorbance of the individual wells was measured at OD 405nm304R.Griep and J.McDougallTable24.1Binding of immunoglobulin isotypes and some of their smaller derivatives to protein-A,protein-G,protein-L,protein-AG protein-LG,and protein-LA,on the basis of data that were obtained from Pierce;GE healthcare;Bonifacino,and Dell’Angelica1998;Hober et al.2007; Kihlberg et al.1996;de Chaˆteau et al.1993,and Svensson et al.1998.(?:unknown,À¼no binding,Ƽvery low binding,þ¼low binding,þþ¼good binding,þþþ¼high binding, V H3and K k=binding only to these specific human heavy-and light-chain families)Species Subclass Prot-A Prot-GProt-L Prot-AGProt-LGProt-LAHuman IgG1þþþþþþþþ(k)þþþþþþþIgG2þþþþþþþþ(k)þþþþþþþIgG3–þþþþþ(k)þþþþþþIgG4þþþþþþþþ(k)þþþþþþþIgE V H3–þþ(k)V H3þþþ(k)þþþ(k)(V H3)IgA V H3–þþ(k)V H3þþþ(k)þþþ(k)(V H3)IgM V H3–þþ(k)V H3þþþ(k)þþþ(k)(V H3)Human Antibody fragments Lamdda-LC––––––Kappa-LC V H3–þþ(k)V H3þþþ(k)þþþ(k)(V H3) IgG1-Fab V H3þþþþþ(k)V H3þþþ(k)þþþ(k)(V H3) Fv V H3–þþ(k)V H3þþþ(k)þþþ(k)(V H3) scFv V H3–þþ(k)V H3þþþ(k)þþþ(k)(V H3) single domain V H3–þþ(k-LC)V H3þþþ(k)þþþ(k)(V H3)Mouse IgG1þþ35%of total IgGin mouse sera þþþþþIgG2aþþþþþþþþþIgG2bþþþþþþþþþIgG3þþþþþþþGuinea pig IgG1þþþþ<10%of total IgG?þþþþIgG2þþþþ?þþþþBovine IgGþþþþ–þþþþþþþCat IgGþþþþ?þþþþþþChicken IgYÆþ>50%of total IgGÆþ>50%oftotal IgG Dog IgGþþþþþ–þþþþþþþDonkey IgG–þþ??þþ?Hamster IgGþþþþþþþþþþþþþþHorse IgGþþþþþ–þþþþþþþGoat IgGþþþ??þþþþþMonkey IgGþþþþþþ?þþþ??Pig IgGþþþþþþ50%of total IgGþþþþþþþþRabbit Nodistinctionþþþþþþ–þþþþþþþþRat IgGþþþ35%of total IgGþþþþþþSheep IgGþþþ?þþþþþþ24Analysis and Purification of Antibody Fragments305306R.Griep and J.McDougall antibody preparation before it can be applied.Therefore,IgG purification with protein-A,-L,or-G is usually only thefirst step and is usually followed by a series of multiple polishing steps.A combination of anion exchange chromatography in flow through mode and cation exchange chromatography removes host cell proteins, DNA,endotoxins,leached protein,and aggregates efficiently(Tugcu et al.2007).Despite the wide variety within the applied monoclonal antibodies,such as, chimeric,humanized,and fully human IgGs of various isotypes,a general purifica-tion strategy is desirable.To date,several comparative studies are available in the literature(Fuglistaller1989;Fahrner et al.1999;Godfrey et al.1993;Hahn et al. 2003,2006;Ghose et al.2007;Swinnen et al.2007and Katoh et al.2007),but new matrices are available to be introduced on theflourishing antibody market(Boi et al.2008).In addition,a total matrix free purification method has been described (Kim et al.2005),which is on the basis of a reversible temperature triggered precipitation of antibodies with the aid of protein-L,or protein-LG fused to elastin-like proteins.The basic protocols for protein-A,protein-L,and protein-G chromatography are relatively straightforward.Bind the immonoglobulins at a neutal pH and elute at an acidic pH.Salt ions even promote binding of IgG to protein-A.Often a stationary phase is employed for the purification of multiple monoclonal antibodies and although the Fc region is the same,still different binding and elution parameters might have to be established for different variable regions(Ghose et al.2005, 2007).As demonstration,methods are described for the purification of polyclonal human IgG/k from serum IgG,a scFv(k)and a IgG1derived CH1/l Fab fragment from an E.coli extract using protein-L and protein-G,respectively.Despite the described differences in the literature between unique human IgG molecules,the purification methodology described below will yield pure,homogeneous,and highly active antibody preparations for almost any antibody without any major changes to these protocols.24.2Purification of Human IgG/k Antibody Fragmentswith Protein-LFor the isolation of a polyclonal IgG fraction from a human serum or of an scFv fragment from bacterial periplasmic preparation,protein-L is known to be an excellent ligand(Fig.24.4).The isolated IgG and scFv have a high purity and the purification method,as described below,is easy to use.24.2.1Materials–Protein-L agarose slurry(rProtein-L TM–agarose,#201,Actigen)in50%ethanol;maximum binding capacity is10mg IgG per mL beads–Human serumabcFig.24.4Representative examples of the versatile application of protein-L.(a)Purification of Polyclonal antibodies from human serum.The pooled fractions are indicated with the double arrow;the solid lines indicate the optical density at 280nm,whereas the dotted lines reflect the pH.(b)Separation of a protein-A purified human IgG preparation in a Kappa and Lambda fraction via protein-L.(c)Purification of a Kappa scFv from a bacterial extract on a protein-A column24Analysis and Purification of Antibody Fragments 307308R.Griep and J.McDougall –PBS–Elution buffer(0,1M Glycin-HCl,pH2,5)–Neutralizing buffer(1M Tris-HCL,pH9,0)–Polystyrene columns,2mL(Pierce,#29920)–20%Ethanol–Deionised water24.2.2Method1.Set up a2mL column and load with0.5mL Protein L-agarose(thus1mL as in50%volume with ethanol/PBS).2.Wait until the gel is settled and wash with5mL PBS.3.Load5mL IgG-solution.4.Collect the IgGflow through fraction.5.Wash with10mL PBS.6.Collect the wash fraction.7.Add350m L1M Tris-HCl,pH9.0to the tubes in the fraction collector prior toelution to immediately neutralize the sample upon elution.8.Elute with5mL elution buffer.9.Collect the eluate.10.Wash the column with5column volumes of deionised water.11.Wash the column with5column volumes of20%ethanol,and store it at4 C.12.Dilute the eluate,flow through,wash,and eluted fraction1:10with PBS.13.Determine the absorbance at280nm.14.Analyze the purity of the sample by SDS-PAGE.24.3Purification of a Monoclonal Human IgG Fab Fragmentwith Protein-GThe isolation of recombinant Fab fragments from bacterial extracts requires a more demanding purification procedure because the heavy-and light-chain frag-ments are not produced in equal amounts.In general,the light-chain is produced at higher levels and secreted as a contaminating light-chain dimer.Therefore,the isolation procedure has to consist of two subsequent steps.Thefirst step is isolation of all the light-chains via a his-tag,which is located on the C-terminus. This is followed by an affinity purification of the heavy-chain fragment via protein-G,which binds to the CH1-region of human IgG1.As a consequence,24Analysis and Purification of Antibody Fragments309 all light-chain dimers will be removed during the procedure described below, which is easy to use and will yield high quality Fab fragments(Fig.24.5).24.3.1Step1:Ni-IMAC Purification of a Fab Fragment24.3.1.1Materials–A¨kta TM Purifier–A¨kta column HisTrap TM FF,1mL(GE Healthcare)–20%Ethanol–Deionised water–0.8m m,0.45m m and0,20m mfilters–2M Imidazole,pH7.0(Preferably from Fluka,sold by Sigma-Aldrich,ultra-pure,cat.no56749,which has no interfering absorbance at280nm)–Buffer-A:IMAC loading buffer(20mM sodium phosphate,500mM NaCl,pH7.4)–Buffer-B:IMAC Elution buffer(20mM sodium phosphate,150mM NaCl, 500mM Imidazole,10%glycerol,pH7.4)–Dialyzed periplasmic E.coli extracts24.3.1.2Method1.Filter all the buffers through a0.20m mfilter.2.Preferably precool the buffers at4 C.3.Filter the pooled and dialyzed periplasmic fractions through0.8and0.45m mfilters before loading it onto the IMAC column.4.Add500m L of the2M imidazole stock per100mL of thefiltered periplasmicfraction to obtain afinal concentration of10mM.5.Equilibrate the column with5column volumes of buffer-A.6.Load the sample on the column.7.Wash the column with20mM imidazole until the unbound proteins have beenwashed out of the column(5column volumes)and the OD280signal has returned to the baseline.8.Elute with100%Buffer-B.9.Wash the column with5column volumes of deionised water.10.Wash the column with5column volumes of20%ethanol,and store it at4 C.11.Optional:analyze the isolated fractions by SDS-PAGE before pooling.12.Avoid freezing samples with imidazole,as it has been observed that this canseverely decrease the activity of the purified antibody fragments.310R.Griep and J.McDougall abcFig.24.5Representative example of the protein-G purification of a monoclonal human IgG1/l Fab fragment from the eluent of a nickel-NTA column.(a)The Fab fragments were isolated from a bacterial extract through the interaction of the His-tag of the light-chain with nickel-NTA beads.24.3.2Step2:Protein-G Purification of a Fab Fragment24.3.2.1Materials–A¨kta TM Purifier–Nickel-NTA prepurified Fab fragments–HiTrap TM_ProteinG_HP_1mL FF,(GE HEALTHCARE)–20%Ethanol–Deionised water–1M Tris-HCl,pH9,0–Loading buffer:20mM sodium phosphate with500mM NaCl,pH7.4–Elution buffer:0,1M Glycine-HCl,pH2.524.3.2.2Method1.Pool the fractions,preferably obtained from a Fab preparation,which wereprepurified on a nickel-NTA column.2.Wash the general system of the A¨kta Purifier TM as well as the10mL sampleloop with Loading buffer.3.Add300m L Tris-HCl,pH9.0to the tubes in the fraction collector prior toelution to immediately neutralize the samples upon elution.4.Load the dialyzed sample onto the column.5.Wash the column with minimal5column volumes of loading buffer until theunbound proteins have been washed out of the column and the OD280signal has returned to the baseline.6.Elute the captured Fab fragments via elution with100%of the elution buffer.7.Wash the column with5column volumes of deionised water.8.Wash the column with5column volumes of20%ethanol,and store it at4 C.9.To obtain Fab fragments of the highest quality an SDS-PAGE analysis can beperformed before deciding which of the fractions should be pooled.10.Dialyze against PBS containing5%glycerol,preferably at a Fab concentrationbelow1mg/mL;this is to prevent precipitation.<Fig.24.5(continued)The pooled fractions are indicated with the double arrow,and the solid lines indicate the optical density at280nm,whereas the dotted lines reflect the pH.(b)The excess of light-chain dimers was removed with the protein-G purification step.The pooled fractions are indicated with the double arrow.(c)Analysis by SDS-PAGE under nonreducing conditions,at the left of the marker(M)and under reducing conditions at the right side(lane-1,first periplasmic extract1;lane-2second periplasmic extract;lane-3,effluent from the IMAC column;lane4,eluent from IMAC column,also used to load the protein-G column;lane5,effluent from the protein-G column;lane6eluent from protein-G column and lane7,thefinal obtained Fab fragment after up concentration and dialyses against PBS).The analysis clearly showed that the light-chain dimer is efficiently removed during the protein-G step(lane-5versus lane-6under reducing conditions)and the high purity of the obtained Fab fragment in thefinal product11.Determine the protein concentration with a spectrophotometer at OD280.12.Store the samples at4 C(1day)or atÀ20 C for longer periods of time,butstorage atÀ80 C is recommended to guarantee long lasting quality of the purified Fab fragments.24.4Trouble ShootingIt might be valuable to monitor the binding efficiency for each specific antibody with techniques such as ELISA,SDS-PAGE,and Western blotting.Optimization of the binding properties of,for instance,rProtein L can result in a tenfold higher yield for a particular antibody.Similar optimizations have been reported for protein-G and protein-A with the application of salts such as sodium chloride and sodium sulfate,which favor increases in hydrophobic interactions.In addition, the pH of the loading buffer can be increased from neutral to more basic(pH9)to maximize the yield.In addition,the concentration of the feedstock should be altered for each antibody during the optimization process to gain maximum binding and elution characteristics.In case of problems with serum derived impurities,protein-L performs specifically in the presence of a large background (up to tenfold)of bovine immunoglobulins.This is particularly valuable when isolating antibodies from culture media containing bovine serum or from the milk of transgenic animals.24.5Concluding RemarksBefore purifying an antibody,regardless the source,consideration should be given to thefinal use of the product.For many applications,both monoclonal and polyclonal antibodies may be used in an impure form.However,for conjugation tofluorochromes or enzymes,simple ligand-based purification is sufficient,but for cell-based assays,a higher level of purification is an absolute requirement.In addition,it all depends on the nature of the antibody fragment combined with the method used for its production whether protein A,protein G,protein L or even a combination of these should be used to obtain optimal results.Whichever method is chosen,care should be taken not to expose the antibodies for an extended time to either strong acidic or basic conditions.This can be avoided by adding a neutraliz-ing buffer in the collection tubes prior to the elution step.In addition,buffer conditions with a pH around the isoelectric point might favor precipitation.A general formulation buffer(10mM Na-citrate/pH6containing:300mM sucrose, 0.9%NaCl,50mM glycine,3.5mM methionine,and0.05%polysorbate-80)can be recommended,which prevents precipitation,aggregation,and oxidation of the purified antibody fragments.Finally,antibody purification can be performed withfancy equipment,but this is not at all an absolute requirement to obtain excellent results.Simple gravityflow always works,even in the case of power failure. ReferencesA˚kerstro¨m B,Bjo¨rk L(1989)Protein L:an immunoglobulin light chain binding bacterial protein.J Biol Chem264:19740–19746Bergmann-Leitner ES,Mease RM,Duncan EH,Khan F,Waitumbi J,Angov E(2008)Evaluation of immunoglobulin purification methods and their impact on quality and yield of antigen-specific antibodies.Malar J7:129–139Bjo¨rk L,Kronvall G(1984)Purification and some properties of Streptococcal protein G a novel IgG-binding reagent.J Immunol133:969–974Boi C,Dimartino S,Sarti GC(2008)Performance of a new protein A affinity membrane for the primary recovery of antibodies.Biotech Prog24:640–647Bonifacino JS,Dell’Angelica EC(1998)Immunoprecipitation.Curr Protoc Cell Biol Chapter 7:7.2.1–7.2.21Carter-Franklin JN,Victa C,McDonald P,Fahrner R(2007)Fragments of protein A eluted during protein A chromatography.J Chromatog A1163:105–111Cossins AJ,Harrison S,Popplewell AG,Gore MG(2007)Recombinant production of a V L single domain antibody in Escherichia coli and analysis of its interaction with peptostreptococcal protein L.Protein Expr Purif51:253–259Das D,Allen TM,Suresh MR(2005)Comparative evaluation of two purification methods of anti-CD19-c-myc-His6-Cys scFv.Protein Expr Purif39:199–208DataMonitor(2007)Monoclonal Antibodies Report Market Model–Detailed analysis of the monoclonal antibody segment,encompassing market dynamics,key therapy areas,technology and target types through to2012,evaluating the strategies companies are using to capitalize on this lucrative market.Reference Code:IMHC0090,June2007DataMonitor(2007)Monoclonal Antibodies Report Part 1.Reference Code:DMHC2291, June2007De Chaˆteau M,Nilson BH,Erntell M,Myhre E,Magnusson CG,Akerstro¨m B,Bjo¨rck L(1993) On the interaction between protein L and immunoglobulins of various mammalian species.Scand J Immunol37:339–405Devaux C,Moreau E,Goyffon M,Rochat H,Billiald P(2001)Construction and functional evaluation of a single-chain antibody fragment that neutralizes toxin AahI from the venom of the scorpion Androctonus australis hector.Eur J Biochem268:694–702Eliasson M,Olsson A,Palmcrantz E,Wiberg K,Ingana¨s M,Guss B,Lindberg M,Uhle´n M(1988) Chimeric IgG-binding receptors engineered from staphylococcal protein A and streptococcal protein G.J Biol Chem263:4323Enever C,Tomlinson IA,Lund J,Levens M,Holliger P(2005)Engineering high affinity super-antigens by phage display.J Mol Biol347:107–120Fahrner RL,Whitney DH,Vanderlaan M,Blank GS(1999)Performance comparison of protein A affinity-chromatography sorbents for purifying recombinant monoclonal antibodies.Biotech-nol Appl Biochem30:121–128Forsgren A,Sjo¨quist J(1966)Protein A from staphylococcus Aureus I Pseudoimmune reaction with human gamma-globulin.J Immunol97:822–827Fuglistaller P(1989)Comparison of immunoglobulin binding capacities and ligand leakage using eight different protein A affinity chromatography matrices.J Immunol Methods124:171–177 Ghose S,Allen M,Hubbard B,Brooks C,Cramer SM(2005)Antibody variable region interactions with protein A:Implications for the development of generic purification process.Biotechnol Bioeng92:665–673。

TIE2配体寡肽的设计、筛选及其在基因治疗中的靶向导入作用

TIE2配体寡肽的设计、筛选及其在基因治疗中的靶向导入作用

TlE2配体寡肽的设计、筛选及其在基因治疗中的靶向导入作用博十研究生:导师:复旦人学医学院吴向华顾健人教授上海市肿痛研究所中文摘要肿瘤基因治疗将成为除手术、放疗和化疗等方法外又‘新的抗癌策略。

尤其在抗肿瘤转移和复发方面将起重要作用。

目前还缺乏将基因导入人体细胞的高效靶向性载体系统,这是基冈治疗至今尚未成为临床常规治疗措施的关键因素之。

因此研发~种高效靶盼陛基冈导入系统成为当务之急。

本研究旨在建立一种靶向Tie2受体的非病毒摹因导入系统并检验其何效性,为今后肿瘤的基因治疗提供可靠的理沦依据与实践指导。

第一部分Tie2配体寡肽的设计与筛选【目的】掌握特定受体的配体寡肽的设计与筛选方法;获得Tie2受体的候选配体寡敝;【方法】(1)应用基于Tie2受体天然配体Angiopoietin一2的同源序列比较/二级结构分析及疏水性分析方法没计Tie2受体的配体寡肽并化学合成;(2)RT-PCR和WestemBloting筛选Tie2阴性表达细胞株;构建pCDNA3.0一ExTie2质粒并转染Tie2表达阴性细胞,G418筛选mTie2稳定表达细胞系;分别以重组人Tie2融合蛋白(rh—Tie2/Fc)与稳定表达Tie2受体的细胞为筛选靶,用噬菌体展示随机12肽库避行筛选。

经过5轮筛选的噬菌体,经测序、ELISA、免疫组化及噬菌体回收试验鉴定出高度富集的阳性噬葡体克隆;然后化学合成高亲和力阳性噬菌体克隆展示肽。

【结果】基于Tie2受体的配体AnDopoietin一2设计的22肽命名为GA3(WTIIQRREDGSVDFQRTWKEYK);筛选到Tie2阴性表达细胞株SMMC7721;建立了Tie2{急定表达细胞系SMMC7721一ExTie2;以rh—Tie2/Fc与SMMC7721-ExTie2为筛选靶获得的高亲和力富集噬菌体展示肽序列羧基端各加一个酪氨酸后分别命名为GA4(HATGTHGLsLsHY),}FIGA5(NsLsNAsEFRAPY)。

跨膜蛋白抽提试剂盒说明书_Merck 英文版

跨膜蛋白抽提试剂盒说明书_Merck 英文版

NovagenUSA and Canada Europe All Other CountriesTel (800) 526-7319 novatech@FranceFreephone0800 126 461GermanyFreecall0800 100 3496IrelandToll Free1800 409 445United KingdomFreephone0800 622 935All otherEuropean Countries+44 115 943 0840Contact Your Local Distributornovatech@ProteoExtract® Transmembrane Protein Extraction Kit Table of ContentsAbout the Kits (2)Description 2Components 2Storage 2Equipment and materials required but not supplied 3ProteoExtract®Transmembrane Protein Extraction Protocol (3)Extraction of membrane proteins from adherent cultured cells 3Extraction of membrane proteins from suspension cells or frozen cell pellets 4Extraction of membrane proteins from tissue 6Frequently Asked Questions (8)Appendix (8)Example extractions 8Examples of total protein yields using TM-PEK 9© 2009 EMD Chemicals Inc., an affiliate of Merck KGaA, Darmstadt, Germany. All rights reserved. The Novagen® logo and Novagen® name are registered trademarks of EMD Chemicals Inc. in the United States and in certain other jurisdictions. ProteoExtract® is a registered trademark of Merck KGaA, Darmstadt, Germany. TRITON® is a registered trademark of Dow Chemical Company.About the KitsProteoExtract® Transmembrane Protein Extraction Kit 1 kit 71772-3DescriptionThe ProteoExtract® Transmembrane Protein Extraction Kit (TM-PEK) uses a novel, detergent-freechemistry for extraction of peripherally-associated and integral membrane proteins, such asG-Protein Coupled Receptors (GPCRs), from mammalian cells and tissues. The membrane protein fractionis directly compatible with enzyme assays, native and denaturing gel electrophoresis, Western blotting,immunoprecipitation, and (following in-gel tryptic digestion) mass spectrometry.The TM-PEK method comprises a two-step protocol for the enrichment of transmembrane (TM) proteins. Inthe first step, cells or homogenized tissues are permeablized using Extraction Buffer 1 and their soluble(cytoplasmic) fraction separated from the insoluble (membrane) fraction by centrifugation. In the secondstep, membrane proteins are extracted from the lipid bilayer using one of two novel extraction buffers.These buffers, Extraction Buffer 2A and Extraction Buffer 2B, are prepared by diluting TM-PEK Reagent Aor TM-PEK Reagent B into Extraction Buffer 2. The optimal extraction reagent must be determinedempirically, as it will depend on characteristics of the protein(s) of interest and intended downstreamapplications. Extraction Buffer 2A is a very mild extraction agent, allowing for recovery of fragile proteincomplexes. Extraction Buffer 2B, by comparison, is a highly efficient extraction agent and facilitatesrecovery of difficult-to-extract transmembrane proteins, including those with multiple transmembranesegments.If using the TM-PEK kit for the first time, prepare duplicate samples. Extract the first set of replicates withTM-PEK Reagent A, diluted 2-fold with Extraction Buffer 2. Extract the second set of replicates with TM-PEK Reagent B, diluted 2-fold with Extraction Buffer 2. Extraction conditions can be optimized further byvarying the dilution range of each reagent into Extraction Buffer, from undiluted to a 10-fold dilution.Unlike alternative membrane protein extraction methods, the TM-PEK kit does not require sonication,rigorous vortexing, time-consuming ultracentrifugation, or incubation at elevated temperatures. The absenceof such harsh treatments minimizes potential damage or changes to the target protein.Each TM-PEK kit provides sufficient reagents to process a total of 40 samples (20 samples using ExtractionBuffer 2A at the recommended dilution, and 20 samples using Extraction Buffer 2B at the recommendeddilution). Each sample may be comprised of 1–5 x 107 cultured cells or 25–50 mg of tissueComponents•••••40 ml Extraction Buffer 150 ml Extraction Buffer 22.0 ml TM-PEK Reagent A2.0 ml TM-PEK Reagent B0.4 ml Protease Inhibitor Cocktail Set IIIStorageStore all components at 4°C for up to 6 months. For prolonged storage, dispense the components in working aliquots and store at –20°C. Before performing extractions, thaw all kit components at room temperature and mix completely. Avoid repeated freezing and thawing.Note that the protease inhibitors are provided in DMSO and must be kept at room temperature during the extraction procedure to prevent freezing.Equipment and materials required but not supplied•••••Rocking platform or elliptical mixer (required at room temperature and 4°C; the device can be transferred from 4°C to room temperature during the course of the experiment)Homogenizer (e.g., Dounce or Potter-Elvehjem) or mortar and pestle (for tissue samples)Refrigerated centrifuge with rotor accommodating a 15 ml and/or a 50 ml tubeRefrigerated centrifuge with rotor accommodating a 2 ml tube and generating 16,000 × gPhosphate Buffered Saline (PBS) (137 mM NaCl, 10 mM Na2HPO4, 2.7 mM KCl, 1.8 mM KH2PO4,pH 7.4)ProteoExtract®Transmembrane Protein Extraction ProtocolTable 1. Buffer volumes required for membrane protein extraction from cultured cells or tissue.Source Material Cultured cells* Fresh or frozen tissueAmount 1.0–5.0 x 10725–50 mg 100–200 mg 200–1000 mgExtraction Buffer 1 (ml) 1.0 1.0 1.0 2.0Extraction Buffer 2A or 2B (ml) 0.2 0.20.51.0Protease Inhibitor (µl) 5 5 20 40*Adherent, suspension, or frozen pelletExtraction of membrane proteins from adherent cultured cellsConsiderations Before You BeginThe following protocol is optimized for extraction of membrane proteins from adherent cells grown in oneT-75 culture flask. Cells should be of high viability (>90%) and 70–90% confluent(1.0–2.0 x 107 cells). Different cell types yield considerably different amounts of protein in the membrane protein fraction (see Table 2 on p 9 in Appendix). If low yield is anticipated, the total number of cells can be increased to 5.0 x 107 without increasing reagent volumes (Table 1). For membrane protein extraction from larger cell numbers, we recommend performing replicate extractions from aliquots of 1.0–5.0 x 107 cells. Alternatively, buffer volumes may be scaled up appropriately.The kit is supplied with two transmembrane solubilization agents, TM-PEK Reagents A and B(see Description on p 2). Depending on the unique characteristics of the target membrane protein, Extraction Buffer 2A or Extraction Buffer 2B may prove to be more efficient. As a starting point, we recommend testing both membrane extraction buffers. If performing trial extractions using both reagents, carry out Steps 2–9 in parallel on each of two sets of sample replicates.Extraction Buffer 2A is a mild extraction reagent which can facilitate recovery of protein complexes. Thus, protein yields obtained using Extraction Buffer 2A may be lower than those obtained using Extraction Buffer 2B (see Table 2 on p 9). Extracts can be concentrated with a Vivaspin 2 or Vivaspin 500 Ultrafiltration Centrifugal Device.Large volumes of TM-PEK 2B reagent can interfere with protein migration in SDS-PAGE. If high resolution is desired, Extraction Buffer 2B samples for SDS-PAGE may be prepared in 2X SDS sample buffer. Alternatively, employ a buffer exchange step.For most transmembrane proteins, a 2-fold dilution of TM-PEK Reagent A or B in Extraction Buffer 2 results in efficient extraction. For some transmembrane proteins, extraction efficiency may be improved by using TM-PEK Reagent A or B at a different concentration, from undiluted to a 10-fold dilution in Extraction Buffer 2.Protocol1.Prepare Extraction Buffer(s) 2A and/or 2B by diluting the appropriate TM-PEK Reagent 2-fold withExtraction Buffer 2. For membrane protein extraction from 1.0–5.0 x 107 cells, 0.2 ml of ExtractionBuffer 2A or 0.2 ml of Extraction Buffer 2B is required.- To prepare 0.2 ml of Extraction Buffer 2A, mix 0.1 ml Extraction Buffer 2 and 0.1 mlTM-PEK Reagent A.- To prepare 0.2 ml of Extraction Buffer 2B, mix 0.1 ml Extraction Buffer 2 and 0.1 mlTM-PEK Reagent B.Notes: We recommend testing both Extraction Buffer 2A and Extraction Buffer 2B to determine which is optimal for your protein of interest.Samples prepared with Extraction Buffer 2A may require concentration, depending ondownstream application.High levels of Reagent B can interfere with protein migration on SDS-PAGE. Buffer exchange ordilution in sample buffer may be required.A 2-fold dilution of TM-PEK Reagent A orB in Extraction Buffer 2 results in an efficient extractionof most proteins, but reagent dilutions may be optimized (from undiluted to a 10-fold dilution).2.Discard medium from the culture flask.3.Wash cells two times with PBS at 4°C.4.Add 3 ml PBS to the culture vessel. Using a cell scraper or a rubber policeman, free cells from theculture vessel. Transfer cells to a 15 ml conical tube.5.Centrifuge cells at 1000 × g for 5 min at 4°C. (Alternatively, collect cells by centrifuging at 500 × g for10 min at 4°C.)6.Resuspend cells in 1 ml Extraction Buffer 1 + 5 µl Protease Inhibitor Cocktail Set III.7.Incubate 10 min at 4°C with gentle agitation to avoid formation of cell clumps.8.Centrifuge at 1000 × g for 5 min at 4°C.9.Carefully remove supernatant. Store on ice. This is referred to as ‘cytosolic (soluble)’ protein fraction.10.Resuspend pellet in 0.2 ml Extraction Buffer 2A + 5 µl of Protease Inhibitor Cocktail Set IIIor 0.2 ml Extraction Buffer 2B + 5 µl of Protease Inhibitor Cocktail Set III.11.Incubate for 45 min at room temperature with gentle agitation.Note: The length and temperature of the incubation at Step 11 can be varied to improve extraction efficiency or to preserve target protein activity. Increasing the incubation time (up to 120 min) canincrease the protein recovery, but may result in decreased target protein activity. Conversely,incubation at low temperature (4°C) may better preserve activity, but may lower the extractionefficiency.12.Centrifuge at 16,000 × g for 15 min at 4°C.13.Transfer the supernatant, which is enriched in integral membrane proteins, to a fresh tube.Note: Store cytosolic and membrane fractions on ice if they will be analyzed on the same day. For long-term storage, dispense into aliquots and store at –20°C.14.Determine total protein concentration of the cytosolic and membrane protein fractions with the BCAassay.Note: For some cell types, total protein concentration of the membrane protein fraction (Step 13 above) will be >1.0 mg/ml. A two- to four-fold dilution in sterile, deionized water may be required to bringthe concentration of these samples within the linear region of the BCA standard curve.Extraction of membrane proteins from suspension cells or frozen cell pelletsConsiderations Before You BeginThe following protocol is optimized for extraction of membrane proteins from 1.0–2.0 x 107 cells cultured insuspension. Cells should be of high viability (>90%). Different cell types yield considerably differentamounts of protein in the membrane protein fraction (see Table 2 on p 9 in Appendix). If low yield isanticipated, increase the total number of cells to 5.0 x 107 without increasing reagent volumes (see Table 1on p 3). For extracting membrane proteins from larger cell numbers, perform replicate extractions fromaliquots of 1.0–5.0 x 107 cells. Alternatively, scale up buffer volumes.The TM-PEK kit is also compatible with frozen cell pellets (1.0–5.0 x 107 cells per extraction). Cells shouldbe washed a minimum of two times with an appropriate buffer (e.g., PBS) prior to freezing in liquidnitrogen. If using frozen cell pellets, begin at Step 7 below.The kit is supplied with two transmembrane solubilization agents, TM-PEK Reagents A and B(see Description on p 2). Depending on the unique characteristics of the target membrane protein, ExtractionBuffer 2A or Extraction Buffer 2B may prove to be more efficient. As a starting point, we recommendtesting both membrane extraction buffers. If performing trial extractions using both reagents, carry out Steps2–9 in parallel on each of two sets of sample replicates.Extraction Buffer 2A is a mild extraction reagent which can facilitate recovery of protein complexes. Thus,protein yields obtained using Extraction Buffer 2A may be lower than those obtained using Extraction Buffer2B (see Table 2 on p 9). Extracts can be concentrated with a Vivaspin 2 or Vivaspin 500 UltrafiltrationCentrifugal Device.Large volumes of TM-PEK 2B reagent can interfere with protein migration in SDS-PAGE. If high resolutionis desired, Extraction Buffer 2B samples for SDS-PAGE may be prepared in 2X SDS sample buffer.Alternatively, employ a buffer exchange step.For most transmembrane proteins, a 2-fold dilution of TM-PEK Reagent A or B in Extraction Buffer 2results in efficient extraction. For some transmembrane proteins, extraction efficiency may be improved byusing TM-PEK Reagent A or B at a different concentration, from undiluted to a 10-fold dilution inExtraction Buffer 2.Protocol1.Prepare Extraction Buffer(s) 2A and/or 2B by diluting the appropriate TM-PEK Reagent 2-fold withExtraction Buffer 2. For membrane protein extraction from 1.0–5.0 x 107 cells, 0.2 ml of ExtractionBuffer 2A or 0.2 ml of Extraction Buffer 2B is required.- To prepare 0.2 ml of Extraction Buffer 2A, mix 0.1 ml Extraction Buffer 2 and 0.1 mlTM-PEK Reagent A.- To prepare 0.2 ml of Extraction Buffer 2B, mix 0.1 ml Extraction Buffer 2 and 0.1 mlTM-PEK Reagent B.Notes: We recommend testing both Extraction Buffer 2A and Extraction Buffer 2B to determine which is optimal for your protein of interest.Samples prepared with Extraction Buffer 2A may require concentration, depending ondownstream application.High levels of Reagent B can interfere with protein migration on SDS-PAGE. Buffer exchange ordilution in sample buffer may be required.A 2-fold dilution of TM-PEK Reagent A orB in Extraction Buffer 2 results in an efficient extractionof most proteins, but reagent dilutions may be optimized (from undiluted to a 10-fold dilution).1.Transfer 1.0–5.0 x 107 cells to a centrifuge tube.2.Centrifuge cells at 1000 × g for 5 min at 4°C. (Alternatively, collect cells by centrifuging at500 × g for 10 min at 4°C.)3.Discard supernatant and gently resuspend cells in 5 ml PBS (4°C).4.Centrifuge cells at 1000 × g for 5 min at 4°C.5.Repeat Steps 3 and 4 two times, for a total of three washes.6.Centrifuge cells at 1000 × g for 5 min at 4°C.Note: At this point, cells may be frozen in liquid nitrogen and stored at –70°C.7.Resuspend cells in 1 ml Extraction Buffer 1 + 5 µl Protease Inhibitor Cocktail Set III.8.Incubate 10 min at 4°C with gentle agitation to avoid formation of cell clumps.9.Centrifuge at 1000 × g for 5 min at 4°C.10.Carefully remove supernatant. Store on ice. This is referred to as the ‘cytosolic (soluble)’ proteinfraction.11.Resuspend pellet in 0.2 ml Extraction Buffer 2A + 5 µl Protease Inhibitor Cocktail Set IIIor 0.2 ml Extraction Buffer 2B + 5 µl Protease Inhibitor Cocktail Set III.12.Incubate for 45 min at room temperature with gentle agitation.Note: The length and temperature of the incubation at Step 12 can be varied to improve extraction efficiency or to preserve target protein activity. Increasing the incubation time (up to 120 min) canincrease the protein recovery, but may result in decreased target protein activity. Conversely,incubation at low temperature (4°C) may better preserve activity, but may lower the extractionefficiency.13.Centrifuge at 16,000 × g for 15 min at 4°C.14.Transfer supernatant, which is enriched in integral membrane proteins, to a fresh tube.Note: Store cytosolic and membrane fractions on ice if they will be analyzed on the same day. For long-term storage, dispense into aliquots and store at –20°C.15.Determine the total protein concentration of the cytosolic and membrane protein fractions with the BCAassay.Note: For some cell types, total protein concentration of the membrane protein fraction (Step 15 above) will be >1.0 mg/ml. A two- to four-fold dilution in sterile, deionized water may be required to bringthe concentration of these samples within the linear region of the BCA standard curve.Extraction of membrane proteins from tissueConsiderations Before You BeginThe following protocol is optimized for extracting membrane proteins from 25–50 mg of fresh or frozentissue. If tissue is not limiting, start with 100–1000 mg of tissue to offset sample loss during homogenization.When using > 50 mg of tissue, increase volumes of the extraction buffers (see Table 1 on p 3 for buffervolume guidelines). As an example, ~2 mg total protein can be extracted from 35 mg bovine liver (see Table3 on p 9 in the Appendix for representative yields from other tissue sources and types). Yields from varioustissue types can vary considerably, however. Certain transmembrane proteins are expressed at very lowlevels in various tissues, and thus may be below the lower limit of detection by immunological methods. Inthis circumstance, analysis by mass spectrometry may prove beneficial.Protocol1.Prepare membrane Extraction Buffer(s) 2A and/or 2B by diluting the appropriate TM-PEK Reagent 2-fold with Extraction Buffer 2. For membrane protein extraction from 25–50 mg of tissue, 0.2 ml ofExtraction Buffer 2A or 0.2 ml of Extraction Buffer 2B is required.- To prepare 0.2 ml of Extraction Buffer 2A, mix 0.1 ml Extraction Buffer 2 and 0.1 mlTM-PEK Reagent A.- To prepare 0.2 ml of Extraction Buffer 2B, mix 0.1 ml Extraction Buffer 2 and 0.1 mlTM-PEK Reagent B.Note: We recommend testing both Extraction Buffer 2A and Extraction Buffer 2B to determine which is optimal for your protein of interest.Samples prepared with Extraction Buffer 2A may require concentration, depending ondownstream application.High levels of Reagent B can interfere with protein migration on SDS-PAGE. Buffer exchange ordilution in sample buffer may be required.A 2-fold dilution of TM-PEK Reagent A orB in Extraction Buffer 2 results in an efficient extractionof most proteins, but reagent dilutions may be optimized (from undiluted to a 10-fold dilution).2.Ensure that all buffers are thawed and well mixed. Keep Extraction Buffers 1, 2A, and/or 2B on iceduring the extraction procedure. Keep the Protease Inhibitor Cocktail Set III at room temperature toprevent DMSO from freezing.3.Following dissection of the tissue of interest, quickly remove unwanted materials(e.g., connective tissue, fat, blood vessels, etc). To slow proteolysis, keep tissue at 4°C while refiningthe dissection.4.Quickly slice the tissue into ~2 mm3 pieces. Add tissue slices to a tube containing 2 ml ice-cold PBS.5.Gently flick tube to dislodge blood cells and other loosely attached material.6.Collect tissue pieces by centrifuging at 100 × g for 2 min at 4°C. Remove and discard the supernatant.7.Repeat Steps 5 and 6 for a total of two washes. After the second wash, ensure that all PBS has beenremoved completely.Notes: At this point, the tissue can be frozen on liquid nitrogen and stored at –70°C.During tissue extraction, it is important to work quickly, but carefully. Keep the sample cool (<4°C)and store buffers on ice throughout the extraction procedure.8.Transfer the tissue (fresh or frozen) to a pre-cooled homogenizer. We recommend ideally, a glassPotter-Elvehjem or Dounce homogenizer.9.Add 5 µl Protease Inhibitor Cocktail Set III to the wall of the homogenizer.10.Add 2 ml ice-cold Extraction Buffer 1 to the homogenizer.11.Carefully homogenize until tissue is completely homogenized and intact pieces are no longer visible.Use as few strokes as possible (e.g., ~10 strokes for 50 mg mouse liver). The number of strokes willdepend on the type of tissue used. If desired, homogenization efficiency can be monitored by phasecontrast microscopy. An efficient homogenization should generate small cell clumps rather thatfragmentation of individual cells.Note: Some tissues (e.g., heart, muscle, brain) may be difficult to completely dissociate by mechanical homogenization. As an alternative, the ProteoExtract®Tissue Dissociation Buffer Kit(Cat. No. 539720) including collagenase may be used. Other tissue-specific protocols may becompatible with the TM-PEK kit.12.Incubate 10 min at 4°C with gentle agitation.13.Centrifuge at 1000 × g for 5 min at 4°C.14.Carefully remove supernatant. Store on ice. This is referred to as the ‘cytosolic (soluble)’ proteinfraction.15.Add 5 ml ice-cold PBS. Gently resuspend pellet. Collect membranes by centrifuging at 1000 × g for 5min at 4°C. Carefully remove supernatant.Note: When using > 200 mg tissue, perform an extra wash at this point to remove additional cytosolic proteins. Repeat Step 15 for a total of two washes.pletely and carefully resuspend pellet in 0.2 ml Extraction Buffer 2A + 5 µl of Protease InhibitorCocktail Set IIIor 0.2 ml Extraction Buffer 2B + 5 µl Protease Inhibitor Cocktail Set III.17.Incubate 15 min at 4°C with gentle agitation to avoid formation of cell clumps.18.Centrifuge at 16,000 × g for 15 min at 4°C.19.Transfer supernatant, which is enriched in integral membrane proteins, to a fresh tube.Note: Store cytosolic and membrane fractions on ice if they will be analyzed on the same day.For long-term storage, dispense into aliquots and store at –20°C.20.Determine the total protein concentration of the cytosolic and membrane fractions with the BCA assay. Note: For some tissue types, total protein concentration of the membrane protein fraction (Step 19 above) will be >1.0 mg/ml. A two- to ten-fold dilution in sterile, deionized water may be required tobring the concentration of these samples within the linear region of the BCA standard curve.Frequently Asked QuestionsQuestion AnswerHow do I determine the proteinconcentration of the membrane proteinextract? The components in the extraction buffers are directly compatible with common protein assays. We recommend using the BCA Protein Assay Kit (Cat. No. 71285-3). Note that the Extraction Buffer 2B fraction may require a 2-4-fold dilution to bring the total protein concentration within the linearrange of the BCA assay.How do I prepare the TM-PEK fraction for one-dimensional SDS-PAGE? The TM-PEK fractions can be analyzed directly by one-dimensional SDS-PAGE. For samplesextracted with Reagent A, add SDS-PAGE sample buffer to a 1X final concentration and loaddirectly on to the gel. Samples extracted with Reagent B should be prepared using a 2Xconcentration of SDS-PAGE sample buffer.How can I concentrate the TM-PEK extracted proteins? It is possible to reduce the volume of Extraction Buffer 2A or 2B used, but this may decrease thetotal protein yield. We recommend using the ProteExtract ® Protein Precipitation Kit (Cat. No.539180) to concentrate samples for use in downstream applications not requiring native protein.For applications that do require native protein, we recommend using an ultrafiltration device suchas Vivaspin 2 or Vivaspin 500.How should the membrane fractions be treated prior to mass spectrometry analysis?The membrane extracts should be applied to a one-dimensional or two-dimensional gel, spots orbands cut from the gel, and then digested with trypsin.Appendix Example extractionsIn the examples below, the SDS extract (total cell lysate) serves as a positive control. The Extraction Buffer 2A recovers EGFR, which has a single transmembrane span, with efficiency comparable to TRITON ® X-100. However, Extraction Buffer 2A recovers Frizzled-4 and CELSR-3, both of which contain seven transmembrane domains, with far greater efficiency than does TRITON X-100.A: Figure 1. Ex ction of transmembrane prote EGFRB: Frizzled-4 C: CELSR-3tra ins from MDA-MB-468 breast adenocarcinoma cultured ells. Transmembrane proteins were extracted from MDA-MB 468 cells using the TM-PEK kit. In the first two identical pools of 1 x 107 cells were treated with Extraction Buffer 1, which recovers proteins from the cytosolic fraction. The insoluble material was then treated with TM-PEK Extraction Buffer 2A or 0.5% TRITON ® X-100. Lane 1 shows the expected size of the target proteins and is not a quantitative control. Lanes 2–4 contain the extracted protein from 1 x 106 cells. Fractions were separated using a 10% SDS-PAGE gel and transferred to a nitrocellulose membrane. Membranes were blocked and incubated with primary antibody to EGFR (panel A), Frizzled-4 (panel B) or CELSR-3 (panel C). Blots were developed using an HRP-conjugated secondary antibody and a chemiluminescent substrate. Lane 1, 0.5% SDS (total cell lysate); Lane 2, cytoplasmic fraction; Lane 3, membrane fraction (TRITON X-100); Lane 4, membrane fraction (Extraction Buffer 2A). Arrows indicate size of the full-length version of each protein.c step,Examples of total protein yields using TM-PEKThe values presented in Tables 2 and 3 below are intended to serve as a guide to estimate the required amount of starting material. For each cultured cell type, proteins were extracted according to the TM-PEK protocol from cell monolayers grown to 80% confluency in a T-75 flask. All tissues were disrupted mechanically using a Dounce homogenizer. Table 2. Protein yields for cytosolic and membrane fractions recovered from cultured cells using the TM-PEK kit.Total Protein (mg/107 cells)*Cell TypeBuffer 1(cytosolic) Buffer 2A (membrane) Buffer 2B (membrane) ExtractionExtraction Extraction MDA-MB-468 (breast adenocarcinoma)0.62 0.15 0.78 MCF 7 (breast adenocarcinoma)0.68 0.26 1.27 A-431 (epidermoid carcinoma)0.55 0.12 0.75 CHO-K1 (Chinese hamster ovary)0.52 0.22 0.94 NCI-H292 (mucoepidermoid carcinoma)0.06 0.04 0.47 HEP-G2 (hepatocellular carcinoma)0.97 76 0.13 0.Mia PaCa-2 (pancreatic carcinoma)0.23 0.05 0.52 HCT 116 (colon carcinoma) 0.60 0.10 0.76 *Protein concentrations were determined using the BCA .Table 3. olic and membrane fractions recovered from mouse tissue Total Protein g/mg tissue)*assayProtein yields for cytos TM-PEK kit.s using the (μTissue TypeExtractionB(cytos Extraction Bu A (me ne) Extraction Bu B (memb uffer 1olic)ffer 2mbra ffer 2rane)Liver §55.6 1.7 11.4 Heart28.2 2.9 17.3 Brain18.4 0.9 5.8 Spleen 31.6 8.2 8.8 §Skeletal Muscle13.6 2.1 6.3 Kidney 33.6.9 5.4 16*Protein concentrations were determined using the B §Average of two tr CA assay.ials.。

Identification of Binding Proteins by NestLink说明书

Identification of Binding Proteins by NestLink说明书

Supplementary ProtocolIdentification of Binding Proteins by NestLinkIntroductionNestLink enables the analysis of individual library members within protein ensembles by the establishment of an in silico genotype-phenotype linkage. This protocol describes the application of NestLink for the identification and characterization of binding proteins of approximately 16 kDa. The input of this workflow is a pool of binding protein candidates that is typically generated via in vitro display or via animal immunization. Initial remarksA Steps 1 - 3 can be performed in parallel for several binder pools. The pools are labelled with differentindices during Illumina MiSeq adaptor joining, such that they can be deep-sequenced together, but are still distinguishable during sequencing data analysis.B Binder selection (step 6) is typically performed sequentially for different pools, as selection pressurescan vary depending on the purpose of the experiment. Examples of selection pressures can be found in the associated publication.C Flycode isolation (step 7) can be parallelized for different pools or selections, whereas LC-MS/MS(step 8) and LC-MS/MS data analysis (step 9) are typically sequential processes.Prerequisites and custom reagents1. T he pre-enriched binder pool must be encoded on a plasmid conferring resistance to an antibiotic otherthan chloramphenicol when propagated in E. coli. This plasmid is referred to as input plasmid henceforth.2. The binder pool must be excisable from the input plasmid via SapI restriction digest leading to three-base pair single-stranded overhangs on either side of the pool in-frame with the binder open reading frame. The sticky ends at the 5’-ending are: AGT (positive strand) and TGC (negative strand).3. A flycoded control binder must be available in purified form. The control binder must be linked 30times to different unique flycodes of known sequence.4. Double-stranded MiSeq adaptor oligonucleotides (see Supplementary table 2) must be available.5. The flycode library encoded in pNLx (see Supplementary Fig. 13b, top) must be available.Protocol ContentStep 1: Diversity restriction of input poolStep 2: Library NestingStep 3: Illumina Adaptor LigationStep 4: Illumina MiSeq and Data ProcessingStep 5: Binder Selection▪Expression (selection of well-expressing nested library members)▪Purification (selection of monomeric nested library members)▪Custom selection experimentStep 6: Flycode IsolationStep 7: LC-MS/MSStep 8: LC-MS/MS Data AnalysisStep 1: Diversity restriction of input poolPurpose: Setting the diversity limit (in this case 3,000 library members) to the input pool.Reason: The diversity limit ensures high read redundancy in deep-sequencing and it is essential to achieve redundant labelling of pool members with flycodes.- Evenly plate E. coli cells that harbour the input plasmid encoding the pre-enriched input library members on antibiotic-selective agar plates and incubate overnight at 37°C.-Pool approximately 3’000 cfu by scratching colonies collectively from the agar plate using an appropriate volume of LB medium for resuspension (depending on the area of the plate(s)) and a plastic spreader.- Inoculate 5 ml LB containing the appropriate antibiotic and grow to saturation overnight at 37°C. Minimize basal binder expression to reduce growth bias (e.g. by the addition of a promotor repressor). -Isolate the diversity restricted input plasmid by the Nucleobond PC20 kit (Macherie-Nagel #740571.100).Step 2: Library nestingPurpose: Redundant attachment of unique flycodes (20-30 flycodes/binder) by FX-cloning.Reason: Redundant flycoding is essential to overcome selection and/or detection biases that might be associated with individual flycodes.- Prepare the following restriction digest reaction mix in a PCR tube and incubate for 10 min at 37°C.- Add 2 µl of 10 mM ATP, 1 µl of T4 ligase (Thermo Scientific #L0011) and incubate for 1h at 37 °C - Heat inactivate for 10 min at 80°C.- Distribute each reaction into 4 x 50 µl of chemically competent E. coli MC1061 cells and incubate on ice for 30 min.- Heat shock by incubation at 42°C for 45 sec, followed by incubation on ice for 5 min. - Add 1 ml LB per transformation reaction and incubate for 30 min at 37°C. - Combine the 4 recoveries in one tube and chill on ice.- Prepare an analytical dilution series (using a small fraction of the recovered cells) and plate on chloramphenicol-selective agar plates.- Inoculate 3 flasks, each containing 150 ml LB (25 µg/ml chloramphenicol), by 0.5 ml, 1 ml or 2.5 ml of the recovered cells, respectively.- Incubate the analytical dilution series (plates) and the flasks overnight at 37°C.-Count colonies on the analytical dilution series agar plates to determine the cfu number used to inoculate the 3 flasks. The inoculated cfu corresponds to the number of flycodes of the nested library.Diversity restricted input plasmid (175 ng/µl)2.6 µl pNLx (100 ng/µl) 1.5 µl CutSmart buffer 2.0 µl H 2O12.1 µl SapI (NEB #R0569S) 1.8 µl20 µl-Choose a flask containing 60,000 – 90,000 cfu for further processing. In case cultures need to be combined to achieve the correct diversity range, mixed culture volumes must be proportional to their respective flycode diversities.- Generate a glycerol stock of the culture, which comprises the desired flycode diversity. To this end, mix 700 µl of 50 % (v/v) glycerol to 700 µl of cultured cells and freeze at -80°C.- Isolate the plasmids encoding the nested library using the Nucleobond Xtra Midi Plus kit (Macherie-Nagel #740412.10).Step 3: Illumina Adaptor LigationPurpose: Preparation of the nested library for MiSeq deep-sequencing by the ligation of double-stranded adaptor oligonucleotides on either end of the nested library.- Prepare the following restriction digest reaction mix and incubate for 1.5h at 50°C.- Stop reaction by addition of 8 µl EDTA (0.5 M) - Run a preparative 2 % (w/v) agarose gel.- Isolate the fragment corresponding to the nested library by the NucleoSpin Gel and PCR Clean-up kit (Machery-Nagel #740609.250).-Ligate 600 ng insert to a 3-fold molar excess of double-stranded MiSeq adaptors according to the following scheme and incubate for 1 h at 37°C.- Heat inactivate for 10 min at 65°C-Run a preparative 2 % (w/v) agarose gel and cut out the fragment corresponding to adaptor-joined nested library (i.e. the highest molecular weight band).Nested library (200 ng/µl) 125 µl CutSmart buffer 20 µl H 2O35 µl SfiI (NEB, 20U/µl) 20 µl200 µlT4 buffer3.3 µl Purified nested library (60 ng/µl) 10.0 µl dsNGS_adaptor 1 (10 µM) 1.3 µl dsNGS_adaptor 2 (10 µM) 1.3 µl H 2O (nuclease-free!)16.1 µl T4 ligase ( Thermo Scientific #L0011) 1.0 µl33.0 µlRemark: Use adaptors that result in a unique index combination for each nested library within the same MiSeq run (see initial remark (A)). Adaptor sequences can be found in Supplementary table 2.-Isolate the adaptor-joined nested library by the NucleoSpin Gel and PCR Clean-up kit (Machery-Nagel #740609.250).-Determine DNA concentration of each adaptor-joined nested library (see initial remark (A)) by absorbance measurement at 260 nm.-Pool adaptor-joined nested libraries if several are to be deep-sequenced at once (see initial remark (A)).In this case, combine the adaptor-joined nested libraries using quantities that are proportional to their respective, expected flycode diversities. Up to at least 300,000 flycodes may be combined in one MiSeq run.Step 4: Illumina MiSeq and Data ProcessingPurpose: Assignment of flycodes to binding protein candidates.Reason: The assignment is central to link phenotype (binder properties) and genotype in silico.-Confirm the molarity of the adaptor joined nested library using Tapestation 2200 (Agilent).-Dilute the sample to 8 pM using hybridization buffer HT1, which is provided in the MiSeq reagent kit (Illumina #MS-102-3003).-Mix 420 µl of the diluted sample with 180 µl of PHiX v3 (Illumina #FC-110-3001) at 12.5 pM, which is provided in the MiSeq reagent kit.-Perform an Illumina MiSeq run using a 600cycle v3 Miseq reagent kit for 2 x 300 bp paired-end reads on a MiSeq sequencer (Illumina #MS-102-3003).-Process the raw reads by Trimmomatic (v0.33, parameters: AVGQUAL:20 MINLEN:100) and by Flexbar (v2.5, parameters: --pre-trim-left 4 --pre-trim-right 4).- Combine read pairs by Flash (v1.2.11, default parameters).- Run the runNGSAnalysis script of the R-NestLink-package provided at https:///cpanse/NestLink using the parameters from the NGS_filteringWorkflow vignette in the package. The same package also contains scripts to compile the mascot search database (https:///cpanse/NestLink/tree/master/exec). Standardized documentation and tools are in preparation for publication via Bioconductor at the time of writing this protocol (release no earlier than Bioconductor 3.9).Step 5: Binder SelectionPurpose: Physical separation of desired/favoured and undesired/unfavoured binding protein candidates.Expression (selection of well-expressing nested library members)-Inoculate a 50 ml pre-culture (LB, 25 µg/ml chloramphenicol, 1% (w/v) glucose) by 30 µl of a glycerol stock generated after library nesting (see step 2) and incubate overnight at 37°C.-Inoculate 2 x 600 ml cultures (TB, 25 µg/ml chloramphenicol) in 2 L baffled flasks by 12 ml pre-culture each. Incubate at 37°C to OD600 = 0.3, followed by temperature reduction to 20°C and induction at OD600 = 0.7 by 0.05% (w/v) arabinose overnight.-Pellet cells at 5,000 g for 15 min and resuspend in 60 ml buffer (20 mM Tris-HCl pH 7.5, 150 mM NaCl, 10 mM imidazole pH 8.0, DNaseI).-The cell suspension may be stored at -20°C at this stage.Purification (selection of monomeric nested library members)-Lyse cells by microfluidizer processor (Microfluidics) at 30,000 lb/in2.-Pellet debris at 4,400 g for 30 min.-Using gravity-flow, apply the supernatant to 2 columns, each containing 1.5 ml superflow Ni-NTA resin (3 ml slurry, Quiagen: #1018142).-Wash each column by 30 ml wash buffer (20 mM Tris-HCl pH 7.5, 150 mM NaCl, 30 mM imidazole pH 8).-Elute each column by 6 ml elution buffer (20 mM Tris-HCl pH 7.5, 150 mM NaCl, 300 mM imidazole pH 8). Do not cool elution below room temperature to minimize precipitation.-Take a 50 µl sample of the elution and add a flycoded control binder as a reference (10 µl at A280=0.05). Freeze the sample at -20°C. Remark: Previous to this experiment, the control binder must be linked to a known set of 30 flycodes and it must be purified (see online methods).-Filter the elution by a 0.2 µm syringe filter and separate by size-exclusion chromatography (several runs may be required depending on the sample volume and column type).-Pool monomeric binder candidates and concentrate to A280 = 4 using an Amicon Ultra-15 concentrator with a 10 kDa cut-off.-Produce aliquots à 625 µl and snap-freeze in liquid N2 for storage at -20°C.-To analyze the nested input pool prior to custom binder selections by LC-MS/MS, take a 50 µl sample of the concentrated sample and add flycoded control binder as a reference (e.g. 10 µl at A280=0.05).Freeze the sample at -20°C.Custom selection experimentThis stage of the selection process is highly variable, as it strongly depends on the overall goal of the NestLink experiment (see associated publication for three examples). We recommend adhering to the following guidelines:1. A NestLink selection pressure must lead to a physical separation of desired and undesiredlibrary members.2.An individual nested library member linked to 10 – 30 flycodes is typically well detectable atquantities above 100 ng prior to flycode isolation.3.Ideally, an individual binding protein linked to a known set of flycodes is added as a referenceto each sample after selection but prior to flycode isolation (flycoded control binder).4.For the subsequent steps of this protocol, nested library members must reside in aqueoussolution in a buffer, which is compatible with immobilized metal-chelate affinity purification(IMAC) after selection. Please note that it is also possible to isolate flycodes from proteinsimmobilized on solid phases such as from a streptavidin-sepharose resin (see application I inmanuscript).Step 6: Flycode IsolationPurpose: Removal of contaminants and proteolytic isolation of the flycodes for LC-MS/MS.-Add 100 µl slurry Ni-NTA to each sample and incubate for 2.5 h at 4°C.-Pellet the resin at 1,500 g for 30 min in a swinging-bucket centrifuge.-Remove the supernatant and transfer the resin to a Mini Bio-Spin® Chromatography Column (Bio-Rad #732-6207).Remark: All subsequent washing steps of the Mini Bio-Spin® Chromatography Columns are performed by centrifugation at 50 g for 10 sec.-Drain resin and wash by 3 x 500 µl buffer 1 (20 mM Tris-HCl pH 7.5, 150 mM NaCl, 10 mM imidazole pH 8.0, 4.8 M GdmCl).-Wash by 3 x 500 µl buffer TH-Im (20 mM Triethylammonium bicarbonate (TEAB) pH 8.0, 150 mM NaCl, 2.5 mM CaCl2, 30 mM imidazole pH 8.0).-Wash by 2 x 500 µl buffer TH (20 mM TEAB pH 8.0, 150 mM NaCl, 2.5 mM CaCl2).-Close the column and add 100 µl buffer TH containing 2.4 U of thrombin (Novagen, #69671-3).Incubate over night at 20°C. This is to cleave the flycode fused to His6-tag from the binding protein.-Drain resin and wash by 5 x 500 µl buffer TH-Im. This is to wash off cleaved binding proteins and to retain the His-tagged flycodes.-Elute by 2 x 50 µl buffer TRY-Im (20 mM TEAB pH 8.0, 50 mM NaCl, 2.5 mM CaCl2, 250 mM imidazole pH 8.0). This is to elute the His-tagged flycodes.-Filter by a pre-washed (H2O) Ultracell YM-10 (Millipore #42407) at 14,000 g for 20 min and retain flow-through. This is to remove background proteins larger than 10 kDa from the His-tagged flycodes.-Repeat elution and filtering and retain flow-through.-Wash filter by 50 µl of buffer TRY-Im and retain flow-through.-Add 1 µg of Trypsin (Promega, #V5113) to the combined retained flow-through and incubate at 37°C overnight. In this step the His-tag is severed from the flycodes.-Add 5% (v/v) of trifluoroacetic acid (TFA) and double volume by solution A (3 % (v/v) acetonitrile (ACN), 0.1 % (v/v) TFA).-Load onto a ZipTip (Millipore, #ZTC185960), which was pre-washed by 200 µl methanol, 200 µl of solution B (60% (v/v) ACN, 0.1 % (v/v) TFA), 200 µl of solution A.-Wash the ZipTip by 200 µl of solution A.-Elute the ZipTip by 2 x 40 µl of solution B.-Snap-freeze eluate in liquid N2 and lyophilize.-Resuspend in 15 µl of 3 % (v/v) ACN, 0.1 % (v/v) formic acid (FA).Step 7: LC-MS/MSPurpose: Detection of flycodes and quantification of flycodes.-Apply 2 – 5 µl of the sample to an Easy-nLC 1000 HPLC system coupled to an Orbitrap Fusion mass spectrometer (Thermo Scientific).-Reverse-phase chromatography specifications are:o Column material: ReproSil-Pur 120 C18-AQ, 1.9 μmo Column dimensions: 150 mm x 0.075 mmo Column temperature: 50 Celsiuso Solvent A (0.1 % FA in water)o Solvent B (0.1 % FA in ACN)o Flow-rate: 0.3 µl/mino Gradient: 5 - 20 % solvent B in 60 min20 - 97 % solvent B in 10 min-Orbitrap Fusion mass spectrometer specifications are:o Scan range: 300 – 1,500 m/zo AGC-target: 5e5o Resolution: 120,000 (at m/z 200)o Maximum injection time: 100 mso MS/MS recording: Data-dependent, rapid scan mode, linear ion trap using quadrupole isolation (1.6 m/z window), AGC target of 1e4, 35 ms maximum injection time, HCD-fragmentation with 30 % collision energy, maximum cycle time = 3 sec. Enable all availableparallelizable time. Select mono isotopic precursor signals for MS/MS with charge statesbetween 2 and 6 and a minimum signal intensity of 5e4. Set dynamic exclusion to 25 sec andthe exclusion window to 10 ppm.Step 8: LC-MS/MS data analysisPurpose: Determination of relative binder abundances within samples.Reason: Differences in relative binder abundances across selection pressures allow conclusions on the characteristics of individual binders.-Progenesis QI:o Align LC-MS/MS runs in Progenesis QI (Nonlinear Dynamics). In case alignment scores are below 90 %, process LC-MS/MS runs individually.o Perform precursor ion peak picking with an allowed ion charge of +2 to +5.o Export fragment mass spectra with a feature rank-threshold of <5 using deisotoping, charge deconvolution and an ion fragment count limit of 1,000.-Analyse the exported fragment mass spectra for flycode identification with Mascot 2.5 (Matrix Science) using the decoyed Swissprot database (release 2014) concatenated with the previouslygenerated database (see step 5) and the following search parametero Precursor tolerance: 10ppm; MS/MS tolerance: 0.6 Da; Enzyme: Trypsin; Variable modification: Deamidation (N,Q); Allowed missed cleavages: 1-Scaffold (Proteome Software Inc.):o Import Mascot search resulto Filter peptide identifications for FDR less than 0.1 % by the peptide prophet algorithm.o Filter protein identifications for FDR less than 1 % by the protein prophet algorithm.-Progenesis QI:o Import the scaffold spectrum report.o Normalize aligned LC-MS/MS runs via the flycoded control binder.o Sum the integrals of all non-conflicting flycode MS1 features per binder candidate (binder abundance).-Calculate the relative abundance of each binder in each sample (the sum of all binder abundances in a sample corresponds to 100 %).。

第六届全国生物信息学与系统生物学学术大会

第六届全国生物信息学与系统生物学学术大会

第六届全国生物信息学与系统生物学学术大会
暨国际生物信息学前沿研讨会
会议时间:2014年10月6日— 9日
会议地点:南京市中山东路307号钟山宾馆(江苏省会议中心)
会议日程概要
会议期间联系:侯越,谢建明,孙啸
会议日程
3
四、主题报告、专题报告(三)10月7日下午,309会议室
五、10月8日上午, 三楼大会堂
六、主题报告、专题报告(四)10月8日下午,307会议室
5
八、主题报告、专题报告(六)10月8日下午,309会议室
九、会议墙报交流10月8日晚上,20:00开始,金陵厅
十、青年沙龙10月8日晚上,20:00开始,307会议室
欢迎青年科研工作者参加。

7
十一、10月9日上午, 三楼大会堂。

高考英语一轮总复习课时质量评价13Unit2MoralsandVirtues新人教版

高考英语一轮总复习课时质量评价13Unit2MoralsandVirtues新人教版

课时质量评价(十三)必修第三册UNIT 2主题语境:优秀品行,正确的人生态度,公民义务与社会责任Ⅰ.阅读理解Jose Alberto Gutierrez's life would never be the same again after finding a copy of AnnaKarenina by Leo Tolstoy in the trash 20 years ago.It happened while he was driving his garbage truck through wealthier neighbourhoods at night and seeing books that were thrown away.It lit his desire to start rescuing books from the trash.He used to take home between 50 and 60 books every morning after his nine­hour shift that ran from 9 p.m.to 6 a.m.Eventually, he turned his book collection into a munity library for children from low­ine families.Colombia's capital city of Bogota has 13 million residents and 19 public libraries.However, these libraries tend to be far away from where rural and poorer munities live.The option of buying new books is non­existent for families struggling to make ends meet.Gutierrez's library is true representation of how one man's trash can be another's treasure.Having access to a library of books and being taken away instantly to another world while absorbed in a book is a luxury for the kids.“I think we are simply a bridge between people who have books and those who don't have anything,”he said humbly about his remarkable attempt.Gutierrez grew up poor, and his family could not afford to educate him beyond primary school.Nevertheless, his mother was an eager reader and read stories to him every night.Her love for books left a deep impression on Gutierrez, who never let a lack of formal education stop him from reading classics.He once said, “There's nothing more beautiful than having a book in your pocket, in your bag or inside your car.”Today, his library, titled “The Strength of Words”, occupies most of his home, and is piled from floor to ceiling with fiction and non­fiction titles.He has also roped in his family to expand this mission.Mrs Gutierrez, for example, selects and repairs the books her husband finds.As word began to spread about his amazing project, fifty­six­year­old Gutierrez earned himself the nickname “Lord of the Books”.In spite of all this, Gutierrez is not yet content to call it a day.He continues to search bins for reading material.1.What is Gutierrez by occupation?A.A public librarian.B.A professional driver.C.A trash collector.D.A shift worker in a plant.2.What is a luxury for the kids in Gutierrez's munity?A.Enjoying pleasure from reading.B.Having their own real library.C.The distance from public libraries.D.The choices of buying new books.3.What really motivated Gutierrez to start his library for underprivileged children? A.An assignment to rescue rejected books.B.A passion for reading since childhood.C.A pity on the underprivileged.D.A desire to make a difference.4.What is the best title of the passage?A.Lord of the BooksB.From Trash to TreasureC.The Strength of WordsD.A Bridge between PeopleⅡ.完形填空(2022·青岛一模)Eradajere Oleita thinks she may have got something about the 1 to two of her country's problems: garbage and poverty.It is called the Chip (薯片) Bag Project.The 26­year­old student an d environmentalist is asking a 2 of local snack lovers: Rather than throw empty chip bags into garbage cans, 3 them so she can turn them into sleeping bags for the 4 .Chip eaters 5 their empty bags at two locations in Detroit: a print shop and a clothing store, where Oleita and her volunteer helpers 6 them.After they disinfect (消毒) the chip bags in soapy hot water, they slice them open, 7 them flat, and iron them together.Then they use cotton and liners (衬层) from old coats to line the insides.It takes about four hours to 8 a sleeping bag, and each takes around 150 to 300 chip bags, 9 on whether they're single­serve or family size.Since its start in 2020, the Chip Bag Project has 10 110 sleeping bags.Sure, it would be 11 to raise the money to buy new sleeping bags.However, that's onlyhalf the 12 for Oleita.“We aim to make a(n) 13 not only socially, but environmentally,”said Oleita.It is worth 14 chip bags and using them to help the homeless. 15 , they would land in the garbage.1.A.reaction B.solutionC.response D.thought2.A.suggestion B.permissionC.favour D.promise3.A.donate B.reserveC.sort D.change4.A.disabled B.elderlyC.homeless D.sick5.A.deliver B.drop offC.hand out D.reuse6.A.guard B.promoteC.place D.collect7.A.fold B.layC.cut D.hang8.A.sew B.designC.order D.clean9.A.concentrating B.insistingC.depending D.agreeing10.A.sold B.createdC.decorated D.received11.A.simpler B.coolerC.cleverer D.more formal12.A.project B.issueC.battle D.goal13.A.impression B.announcementC.impact D.decision14.A.recycling B.maintainingC.improving D.producing15.A.However B.BesidesC.Therefore D.Otherwise课时质量评价(十三)Ⅰ.【语篇解读】本文是一篇记叙文。

双绕蛋白质的分类与识别

双绕蛋白质的分类与识别

双绕蛋白质的分类与识别刘岳;徐海松;乔辉;李晓琴【摘要】蛋白质折叠识别是蛋白质结构研究的重要内容.双绕是α/β蛋白质中结构典型的常见折叠类型.选取22个家族中序列一致性小于25%的79个典型双绕蛋白质作为训练集,以RMSD为指标进行系统聚类,并对各类建立基于结构比对的概形隐马尔科夫模型(profile-HMM).将Astral1.65中序列一致性小于95%的9 505个样本作为检验集,整体识别敏感性为 93.9%,特异性为82.1%,MCC值为0.876.结果表明:对于成员较多,无法建立统一模型的折叠类型,分类建模可以实现较高准确率的识别.【期刊名称】《生物信息学》【年(卷),期】2010(008)001【总页数】6页(P1-6)【关键词】双绕蛋白质;RMSD;系统聚类;隐马尔科夫模型;折叠类型识别【作者】刘岳;徐海松;乔辉;李晓琴【作者单位】北京工业大学生命科学与生物工程学院,北京,100124;北京工业大学生命科学与生物工程学院,北京,100124;北京工业大学生命科学与生物工程学院,北京,100124;北京工业大学生命科学与生物工程学院,北京,100124【正文语种】中文【中图分类】Q523蛋白质的氨基酸序列如何决定空间结构是生命科学研究中的核心问题之一,被称为第二遗传密码。

目前,国际上由蛋白质的氨基酸序列预测其空间结构的方法有以下三种:即同源模建法、折叠识别法和从头计算法[1]。

同源模建受到序列相似度的限制,从头计算运算量太大,介于两者之间的折叠识别被认为是最有潜力的方法。

大量实验和理论研究表明[2],蛋白质的三级结构是非常复杂而不规则的,但其整体折叠类型却十分有限。

折叠类型反映了蛋白质核心结构的拓扑模式[3,4],一般认为只有数百到数千种[5],远小于蛋白质所具有的自由度数。

对自然界存在的数百到数千种折叠类型进行系统分类和识别,将有助于揭示蛋白质的折叠规律。

蛋白质折叠类型分类基本上都是靠专家来完成的,不同的库分类颇不相同[6,7],鉴于此,通过对蛋白质折叠类型的研究,以结构核心的拓扑连接和空间排布为依据,建立了统一原理的蛋白质折叠类型数据库[8,9],为进一步的蛋白质折叠类型分类及识别奠定基础。

蛋白质序列分析

蛋白质序列分析

蛋白质二级结构预测1
预测蛋白质二级结构的算法大多以已知三维 分为三类: 结构和二级结构的蛋白质为依据 ,分为三类: 统计/经验算法 Chou-Fasman法 GOR法 经验算法: 统计/经验算法:Chou-Fasman法、GOR法 物理化学法: 物理化学法:Lim法 法 机器学习法:结合上2种方法的优点 种方法的优点。 机器学习法:结合上 种方法的优点。
X-ray Crystallography
X-ray Crystallography..
From small molecules to viruses Information about the positions of individual atoms Limited information about dynamics Requires crystals
1.同源建模法 2.折叠子识别 3.从头预测法
蛋 白 质 三 级 结 构 分 析 流 程
/people/rob/CCP11BBS/
nnPredict
用神经网络方法预测二级结构,蛋白质 神经网络方法预测二级结构, 方法预测二级结构 结构类型分为全α蛋白、 结构类型分为全α蛋白、全β蛋白和 α/β蛋白 输出结果包括“ (螺旋) 蛋白, α/β蛋白,输出结果包括“H”(螺旋)、 “E”(折叠)和“-”(转角)。这个方法对 (折叠) (转角) 蛋白能达到79%的准确率。 79%的准确率 全α蛋白能达到79%的准确率。 nnPredict网址 网址: nnPredict网址: /~nomi/nn predict.html
PROSEARCH网址: 网址: 网址
http://www.embl-heidelberg.de/prs.html 程序也可以完成。 用Bioedit程序也可以完成。 程序也可以完成

蛋白质分析和蛋白质组学ppt课件

蛋白质分析和蛋白质组学ppt课件

生物学途径是由分子功能有序地组成的,具有多个 步骤的一个过程。(细胞生长和维持、信号传导 、 嘧啶代谢或α-配糖基的运输 )。
cell division
gluconeogenesis
19
Biological Process
20
lipocalin
21
以树状图形式 显示的GO词汇 之间的关系
22
6
本体(ontology)



计算机科学对自然世界认知的形式化的表示,既是 可被计算机表示,解释和利用的知识的形式化的研 究—即本体 。本体是结构化的领域知识,并可以被 计算机解释和利用 。 实现对生命世界中这些概念理解上的共享,包括从 不同的视角,不同的术语分类, 不同的主体( 人和机 器)共享概念 --概念化的规范 Gene Ontology(GO)协会致力于这样一项工程: 编辑一组动态的而又可控的词汇来描述基因和基 因产物(主要是蛋白质)不同方面的性质。
酶的数目
1003 — — 1076 — 1125 356 156 126
子类的例子
作用于CH-OH基团 作用于醛类或氧络集团 转移—碳基团
38
Functional assignment of proteins: Clusters of Orthologous Groups (COGs)
39
Proteomics: High throughput protein analysis
蛋白质分析和蛋白质组学
DNA
RNA
protein
1
[1] Molecular biology
[3] Protein localization
protein [4] Protein function

基于生物素连接酶的邻近标记技术在蛋白质组学中的研究进展

基于生物素连接酶的邻近标记技术在蛋白质组学中的研究进展

Journal of China Pharmaceutical University2022,53(1):18-24学报基于生物素连接酶的邻近标记技术在蛋白质组学中的研究进展张雨欣1,丁明2*,柳军1**(1中国药科大学新药筛选中心,南京210009;2中国药科大学生命科学与技术学院,南京210009)摘要基于生物素酶的蛋白质邻近标记技术是利用融合在感兴趣蛋白上的生物素连接酶对邻近的蛋白质生物素化,通过生物素与链酶亲和素之间的亲和力进行分离,再结合质谱分析鉴定出生物素化蛋白。

该技术能够用于检测弱而短暂的蛋白质相互作用,也给在无膜细胞器和其他不易分离或纯化的亚细胞结构的上的蛋白互作研究提供了新的选择,很好地补充了传统研究蛋白质互作方法的空白。

本文对近几年出现的基于生物素酶的蛋白质邻近标记的技术发展及其应用进行了综述。

关键词邻近标记技术;生物素化;蛋白质相互作用;BioID;BioID2;TurboID;进展中图分类号Q816文献标志码A文章编号1000-5048(2022)01-0018-07doi:10.11665/j.issn.1000-5048.20220103引用本文张雨欣,丁明,柳军.基于生物素连接酶的邻近标记技术在蛋白质组学中的研究进展[J].中国药科大学学报,2022,53(1):18–24.Cite this article as:ZHANG Yuxin,DING Ming,LIU Jun.Research progress of proximity labeling technology based on biotin ligase in proteomics[J].J China Pharm Univ,2022,53(1):18–24.Research progress of proximity labeling technology based on biotin ligase in proteomicsZHANG Yuxin1,DING Ming2*,LIU Jun1**1Center for New Drug Screening,China Pharmaceutical University,Nanjing210009;2School of Life Science&Technology,China Pharmaceutical University,Nanjing210009,ChinaAbstract Proximity-dependent biotinylation(PDB)uses biotin ligase fused to the protein of interest to bioti⁃nylate adjacent proteins,purify them with streptavidin beads,and then identify the biotinylated protein by mass spectrometry.This technology can be used to detect transient and/or low affinity interactions,provide a chance to learn more about membrane-less organelles and other subcellular structures that cannot be easily isolated or puri⁃fied,and fill the gap in traditional methods.This article summarizes the technological development and applica⁃tion of PDB in recent years.Key words adjacent protein markers;biotinylation;protein-protein interaction;BioID;BioID2;TurboID;advances蛋白质是生命体中真正发挥作用的物质,大多数蛋白质不能单独发挥作用,它们与其他蛋白质的相互作用决定了细胞的功能。

我的很凶的英语老师男作文400字左右

我的很凶的英语老师男作文400字左右

我的很凶的英语老师男作文400字左右全文共6篇示例,供读者参考篇1My Strict English Teacher, Mr. JamesonMr. Jameson was the most feared teacher at Oakwood High. As an English teacher, he had a legendary reputation for being incredibly demanding, giving out harsh criticism, and struck terror into the hearts of students with his intense glares and booming voice. I'll never forget my first encounter with him.It was my freshman year, and I had him for English Literature. On the first day, he strode into the classroom looking like an old-school disciplinarian. He was tall and slim with a neatly trimmed greying beard. His dark brown eyes seemed to bore right through you."Welcome to English Literature," he announced in his deep baritone voice. "I'm Mr. Jameson, and I don't tolerate any nonsense in this class. You will work hard, you will hand in assignments on time, and you will show respect. Fail to meet my standards, and you'll face the consequences."The whole class seemed to hold its breath. This was a man you didn't want to cross.True to his word, Mr. Jameson's classes were grueling. We analyzed works of classic literature down to the smallest details and metaphors. He demanded flawless grammar, sophisticated vocabulary, and critical thinking in our essays. Mere summary wasn't good enough - he wanted original analysis and insights.Whenever someone spoke out of turn or came unprepared, he'd fix them with a withering look. "Miss Thompson, do you have something more important to say than the literary commentary I've prepared?" His voice could slice like a blade.I worked harder in his class than any other, spending hours poring over books and crafting essays late into the night. Whenever I thought I nailed an assignment, he'd return it cramped with his scathing red ink - circling mistakes, questions challenging my arguments, and demands to "explore deeper."Mr. Jameson's cutting feedback used to make me want to cry. A few kids accused him of being overly harsh andpower-tripping. But I realized his lofty standards pushed us to achieve more than we thought possible. He refused to coddle us or let us skate by - he genuinely wanted to make us better readers, writers, and thinkers.Earning praise from Mr. Jameson was immensely satisfying because you knew you had to truly earn it. An "excellent work" scrawled at the top of an essay filled me with pride. When he complimented my insightful analysis or clever wording in class, I glowed from his rare approval.As tough as he was, I could sense Mr. Jameson's commitment to nurturing our minds. Maybe his teaching style seemed harsh, but it came from a good place - a desire to impart knowledge and push us to fulfill our potential. He refused to settle for less.My favorite Mr. Jameson moment came during my junior year poetry unit. I have vivid memories of him standing at the front of the class, enthusiastically reciting verses with incredible theatrical flair. His whole demeanor transformed. The stern disciplinarian melted away as he became completely enraptured by the beauty and rhythm of the words. It was mesmerizing to watch.In those moments, I realized Mr. Jameson's gruff exterior masked a true passion for literature. He wasn't just an enforcer of rules - he lived and breathed the poetry, novels, and plays he taught. His love for the written word inspired me to appreciate it on a deeper level too.Mr. Jameson's reputation for being a harsh taskmaster was well-earned. But his unwavering high expectations made me a dramatically better student and person. I honed critical thinking abilities, improved my writing tenfold, and developed a greater respect for great works of literature. His classes were genuinely rigorous in the best way.People often say High school English was one of the biggest academic challenges of their life. For me, that rings especially true - courtesy of my very strict, very demanding, yet ultimately inspiring English teacher, Mr. Jameson. Getting through his class was grueling, but graduating with his approval felt like a badge of honor. His tough love approach has stuck with me long after leaving Oakwood High. I'm forever grateful for his commitment to enriching young minds, even if heritalized a few souls along the way!篇2My Strict English Teacher Mr. WilliamsMr. Williams was the kind of teacher that struck fear into the hearts of students. From the moment he stepped into the classroom, you could feel the shift in the atmosphere. It was as ifall the air had been sucked out, leaving us gasping for breath under his intense gaze.At first glance, Mr. Williams didn't seem like someone you'd want to cross paths with. He was tall, broad-shouldered, and had a perpetual frown etched onto his face. His salt-and-pepper hair was always neatly combed, and he wore crisp button-down shirts that seemed to crease in all the right places. But it was his eyes that really got to you – they were sharp and piercing, as if they could see right through you.Despite his intimidating demeanor, Mr. Williams was an excellent English teacher. He had a way of breaking down complex concepts and making them easily understandable. His lessons were always well-structured and engaging, and he had a knack for keeping even the most easily distracted students focused.However, Mr. Williams was also known for his strict discipline. He ran a tight ship in his classroom, and any disruptions or misbehavior were swiftly dealt with. He had a zero-tolerance policy for tardiness, and if you were even a minute late, you could expect a stern reprimand and possibly even a detention slip.One of the things that made Mr. Williams so formidable was his unwavering consistency. He treated every student the same, regardless of their academic performance or social standing. It didn't matter if you were the class valedictorian or the class clown – if you stepped out of line, you could expect the same consequences.I remember one incident in particular that really exemplified Mr. Williams' no-nonsense approach. It was during our sophomore year, and a group of students thought it would be funny to pass around a note during his class. The note made its way across the room, and eventually landed on my desk. Just as I was about to read it, Mr. Williams swooped in like a hawk and snatched it from my hands.The look on his face was one of pure fury. He demanded to know who had started the note, but no one was willing to own up to it. That's when he made an example of me. Despite my protests of innocence, he gave me a week's worth of detention for "participating in the disruption."At the time, I was livid. I felt like I had been unfairly punished for something I hadn't even done. But looking back, I can appreciate the life lesson that Mr. Williams was trying to impart. He was teaching us about accountability and the importance oftaking responsibility for our actions, even if it meant facing consequences that seemed unjust.As much as we feared and resented Mr. Williams at times, there was no denying that he was an excellent teacher. He pushed us to our limits, challenged us to think critically, and instilled in us a strong work ethic. Many of us went on to succeed in our academic and professional pursuits, and I truly believe that Mr. Williams played a significant role in shaping us into the individuals we became.In the end, Mr. Williams taught us far more than just grammar rules and literary analysis. He taught us discipline, perseverance, and the value of hard work. And while his methods may have seemed harsh at times, they ultimately prepared us for the challenges that lay ahead in the real world.So, while I may have dreaded walking into his classroom back then, I now look back on Mr. Williams with a sense of gratitude and respect. He was the kind of teacher that left an indelible mark on his students, and for that, we owe him a debt of thanks.篇3My Strict English Teacher, Mr. JohnsonMr. Johnson was the strictest teacher I've ever had. From the moment I stepped into his English class on the first day of 9th grade, I knew he meant business. He was a tall, imposing figure with a stern expression that made even the boldest students shrink back in their seats.On that first day, he wasted no time in laying out his expectations. "In this class, there is no fooling around," he bellowed in his deep, commanding voice. "You will come to class prepared, you will participate, and you will turn in all assignments on time, no exceptions." He fixed us all with a piercing gaze, daring anyone to challenge him.True to his word, Mr. Johnson ran a tight ship. He had a zero-tolerance policy for tardiness, and if you were even a minute late, you could expect a tongue-lashing in front of the entire class. Talking out of turn or passing notes was an absolute no-no, and he had eyes like a hawk, able to catch even the slightest whisper or movement.His assignments were notoriously difficult, with lengthy reading passages followed by a barrage of comprehension questions that delved into the minutest of details. Essays had to adhere to his strict formatting guidelines, with margins precisely set and not a single citation out of place. Woe betide the studentwho tried to fluff their word count with filler sentences – Mr. Johnson could sniff out that kind of trickery from a mile away.Despite his fearsome reputation, I soon realized that beneath that gruff exterior beat the heart of a true educator. Mr. Johnson pushed us hard because he wanted us to succeed. He had an uncanny ability to pinpoint each student's weaknesses and find ways to shore them up. For me, it was my tendency to ramble and lose focus in my writing. Through his meticulous feedback and one-on-one conferences, he taught me the value of clarity and concision.As the year progressed, I started to see the method behind Mr. Johnson's madness. His unwavering standards and refusal to accept anything less than our best work instilled in us a sense of discipline and pride in our accomplishments. When I received a hard-earned 'A' on a paper, it felt like a true badge of honor, knowing the rigor it had taken to meet his exacting criteria.And despite his reputation as a intimidating disciplinarian, Mr. Johnson did have a softer side. Occasionally, he would crack a wry joke during a lesson, his eyes crinkling at the corners with mirth. Or he would share a personal anecdote that revealed glimpses of his life outside the classroom, reminding us that he was human, too.By the end of the year, I had developed a profound respect for Mr. Johnson. His unwavering commitment to upholding high standards had pushed me to heights I didn't know I could reach. And while I may have grumbled about his stringent policies at the time, I now recognize that they were preparing me for the rigors of higher education and the professional world beyond.These days, whenever I find myself facing a daunting writing task or a challenge that demands my utmost effort, I think back to Mr. Johnson's class. I remember the satisfaction of turning in a pristine essay, knowing that I had given it my all. And I'm reminded that with hard work, discipline, and a refusal to accept mediocrity, I can conquer any obstacle that stands in my way.So thank you, Mr. Johnson, for being the strict teacher I needed, even if I didn't realize it at the time. Your lessons have stayed with me long after I left your classroom, shaping me into the diligent, detail-oriented person I am today. And while I may have cursed your name under my breath more times than I can count, I wouldn't have had it any other way.篇4My Very Strict English Teacher, Mr. JohnsonMr. Johnson, my English teacher, is a man who commands respect and strikes fear into the hearts of students with his stern demeanor and uncompromising standards. From the moment he strides into the classroom, his presence alone is enough to silence even the most unruly of pupils.At first glance, Mr. Johnson might appear intimidating, with his tall frame, piercing gaze, and a countenance that rarely betrays a smile. However, beneath that formidable exterior lies a dedicated educator who genuinely cares about his students' growth and academic success.One of the defining characteristics of Mr. Johnson's teaching style is his unwavering commitment to proper grammar and impeccable writing skills. He has an uncanny ability to spot even the most minute errors, whether it's a misplaced comma or a dangling participle. Woe betides the student who submits a poorly written essay or fails to adhere to the prescribed formatting guidelines."Sloppy work is unacceptable in my class," Mr. Johnson would declare, his voice booming across the room, sending shivers down our spines. "If you cannot master the fundamentals of language, how can you expect to communicate effectively in the real world?"Despite his intimidating manner, Mr. Johnson's lessons are always meticulously planned and delivered with a level of clarity that leaves no room for misunderstanding. He has a knack for breaking down complex concepts into digestible chunks, ensuring that even the most challenging topics are comprehensible to his students.Yet, it is his dedication to fostering critical thinking skills that truly sets Mr. Johnson apart. He encourages us to question, analyze, and form our own opinions, challenging us to defend our viewpoints with sound reasoning and evidence. Discussions in his class are never dull, as he skillfully guides us through thought-provoking debates and intellectual discourse.One particular memory that stands out vividly is the time when I mustered the courage to challenge Mr. Johnson's interpretation of a literary work. Bracing myself for his wrath, I presented my counterargument with trembling hands. To my surprise, he listened intently, his eyes narrowing as he considered my perspective. After a moment of silence, he offered a thoughtful rebuttal, engaging me in a lively exchange that left me both enlightened and emboldened.While Mr. Johnson's strict approach may seem daunting to some, it is undeniable that his methods yield remarkable results.Students who emerge from his tutelage possess not only a solid grasp of the English language but also the critical thinking skills and self-discipline necessary for success in any academic or professional endeavor.As I reflect on my time in Mr. Johnson's class, I realize that his unwavering standards and uncompromising demeanor were not born out of a desire to intimidate or belittle his students. Rather, they stemmed from a profound belief in our potential and a commitment to helping us achieve excellence. Though his methods may have seemed harsh at times, they ultimately instilled in us a sense of resilience, determination, and a deep respect for the power of language.In the years to come, as I navigate the challenges of higher education and the professional world, I will carry with me the invaluable lessons learned from Mr. Johnson's class. His unwavering dedication to academic rigor and his unwavering belief in our abilities have left an indelible mark on my character and will continue to guide me on my journey toward personal and intellectual growth.篇5My Strict English Teacher Mr. SmithI can still vividly remember the first day I walked into Mr. Smith's English class. The classroom was eerily quiet, and the atmosphere was tense. Mr. Smith stood at the front, his piercing gaze scanning the room as if daring any of us to step out of line. With his stern expression and commanding presence, he seemed more like a drill sergeant than an English teacher.From the moment he opened his mouth, it was clear that Mr. Smith was not one to be trifled with. His deep, booming voice reverberated off the walls, and his words carried an unmistakable authority. "In this class, we will focus on mastering the English language," he declared. "There will be no room for slackers or those unwilling to put in the effort. My expectations are high, and I will accept nothing less than your best."As the weeks went by, we quickly learned that Mr. Smith was a man of his word. His lessons were rigorous, and he pushed us to our limits. He would meticulously dissect our essays, pointing out every grammatical error, every awkward phrase, and every instance where our writing fell short of its potential. His critiques were brutal, but they were also invaluable in helping us hone our skills.Mr. Smith's teaching methods were unconventional, to say the least. He had a knack for keeping us on our toes, often callingon students at random to answer questions or recite passages from literary works. If we stumbled or gave an inadequate response, he would fix us with a withering glare that could make even the most confident among us shrink in our seats.Despite his stern demeanor, Mr. Smith was not without a sense of humor. Occasionally, he would crack a wry joke or share an amusing anecdote, but these moments were fleeting, and we quickly learned not to let our guard down. His focus was always on pushing us to achieve excellence, and he would not tolerate any distractions or lapses in concentration.One incident that has stuck with me occurred during a class discussion on Shakespeare's "Hamlet." I had been struggling to grasp the nuances of the play, and my frustration must have shown on my face. Without warning, Mr. Smith called on me to analyze a particularly complex soliloquy. As I fumbled through my response, he watched me with an inscrutable expression.When I finally finished, he let the silence linger for what felt like an eternity. Then, in a voice that was equal parts disappointment and challenge, he said, "Mr. Johnson, I expected better from you. This work is a masterpiece, and your analysis has done it a disservice. I suggest you spend some time trulyunderstanding the depth of Shakespeare's genius, or you will continue to flounder."Those words stung, but they also lit a fire within me. I redoubled my efforts, poring over the text and seeking a deeper understanding of its themes and nuances. The next time Mr. Smith called on me, I was prepared, and my analysis was met with a rare nod of approval.As the year progressed, I came to appreciate Mr. Smith's unwavering commitment to excellence. His demanding nature pushed me to limits I didn't know I could reach, and his constructive criticism helped me become a better writer and thinker.In the end, Mr. Smith's class was one of the most challenging experiences of my academic career, but it was also one of the most rewarding. His unyielding standards and uncompromising approach instilled in me a deep respect for the English language and a drive to continually improve my skills.篇6My Very Scary English TeacherMr. Jenkins was the most frightening teacher I ever had. He taught 10th grade English, and I shuddered in fear every time I walked into his classroom. At over six feet tall with a booming voice and intense glare, he commanded respect and struck terror into the hearts of students. I tried my best to never draw his wrath.On the first day of class, he laid out his strict expectations in a tone that allowed no argument. "There will be no talking, no tardiness, no sleeping, no phones, no gum, no getting out of your seat without permission," he barked. We sat up straight, intimidated into absolute obedience.His teaching style matched his fierce demeanor. He would call on students at random, demanding we analyze passages of literature with deep insights. If someone stammered or froze up, he would unleash his scathing ridicule. "Utterly pathetic analysis! Are you even awake? Maybe you need a bucket of ice water dumped on your head!" We all feared being scalded by his scorching criticism.Despite the overwhelming anxiety, I worked harder in his class than any other to avoid his rebukes. I studied the reading assignments thoroughly, prepared diligent analyses, and always had my homework completed flawlessly. Even then, he wouldzero in on tiny mistakes with relentless scrutiny. Once he spent 15 agonizing minutes berating me for misusing a semi-colon.If students dared to talk out of turn or disturb the class even slightly, he would erupt like an angry volcano. "EXCUSE ME! What is so Important that you feel the need to disrupt my lesson and waste everyone's time with your incessant yammering?" Entire classes would be subjected to his full-volume tirades until we shrunk down trembling in our seats.The punishments he doled out only added to the dread we felt. He loved assigning multi-page essays, forcing us to labor over literary analysis for hours upon hours. Detention was his other favored weapon, which meant suffering through 60 tortuous minutes trapped alone with him after school. During those detentions, the intensity of his glares and disappointed scowls made our souls want to shrivel up.Even outside the classroom, Mr. Jenkins inspired fear and awe. I'll never forgot the time he broke up a hallway fight between two students with stunning force. Sprinting down the hall, he bellowed at the combatants with a voice of thunder, "HEY! KNOCK IT OFF RIGHT NOW BEFORE I MAKE YOU SORRY!" The two fighting teens froze, their eyes widening in sheer panic at the sound of his roar. He then grabbed them both by the collar andwrangled them into the principal's office like disobedient toddlers. The sight was both terrifying and strangely impressive.In spite of the constant stress, I came to respect Mr. Jenkins' high standards and his evident determination to push us to our full potential. His commitment to academic excellence was undeniable, even if his methods caused inordinate anxiety. My writing improved markedly that year thanks to his merciless critiques and demanding workload.Even years later, whenever I think back to 10th grade English, my heart rate still increases as I'm flooded with memories of that snarling, intimidating presence at the front of the classroom. He was undoubtedly the stuff of student nightmares. But I'll also always appreciate how he toughened me up and instilled a vigorous work ethic. Mr. Jenkins truly was the most scary teacher I ever had...but also one of the most impactful.。

老师点评孩子作文家长回复

老师点评孩子作文家长回复

老师点评孩子作文家长回复英文回答:Thank you for your feedback on my child's composition.I appreciate your effort in providing constructive criticism. I understand the importance of incorporating both English and Chinese in my response, so I will address your points accordingly.Firstly, I agree with your comment about my child's need to improve their vocabulary. To address this, I will encourage my child to read more books in both English and Chinese. By exposing them to a wide range of words, they will be able to expand their vocabulary and use more diverse language in their writing. For example, I can encourage my child to read English classics like "To Kill a Mockingbird" and Chinese classics like "红楼梦" (Dream of the Red Chamber). This way, they will not only learn new words but also gain insights into different cultures.Secondly, I appreciate your suggestion to focus on sentence structure and grammar. To help my child improve in this aspect, I will encourage them to practice writing regularly. By doing so, they will become more familiar with sentence structures and grammar rules. Additionally, I can also suggest that my child reads English and Chinese newspapers or magazines. This will expose them to different writing styles and help them understand how to construct well-formed sentences.Furthermore, I understand the importance of using idioms and colloquial expressions to make writing more engaging. To assist my child in this area, I will introduce them to popular idioms and expressions in both English and Chinese. For instance, I can teach them English idioms like "the ball is in your court" and Chinese idioms like "亡羊补牢" (mend the fold after a sheep is lost). By incorporating these idioms and expressions into their writing, my child can add depth and authenticity to their compositions.Lastly, I want to assure you that I will not disclose your prompt in my response. I understand the importance ofmaintaining the integrity of the assignment and ensuringthat my child's work is original. I will continue tosupport my child in their writing journey and encouragethem to express their thoughts and ideas independently.中文回答:非常感谢您对我孩子作文的点评。

Western印迹中蛋白质定量分析的替代方法

Western印迹中蛋白质定量分析的替代方法

切下, 置入盛有 200 µl/ 孔二甲基亚砜(DMSO)的 96 孔圆底微孔板, 静置或轻微振荡15 min, 萃取红色产 物直至PVDF膜上无着色为止, 以酶联免疫仪(Thermo) 测波长为 492 nm 的吸收值(A492)定量。
图2 W e s t e r n 印迹定量分析
A: 蛋白质样品稀释为0.5 ̄16 µg, 在20 µl 上样缓冲液里加热变性, 经 SDS-PAGE、转膜、抗体杂交, 底物反应显带呈红色; B: 将相 应的特异蛋白质带切下, DMSO 萃取后酶联免疫仪测 A492 定量; C: A492 与样本蛋白质量关系曲线( γ=0.999)。
收稿日期: 2006-07-13 接受日期: 2006-12-01 清华 - 裕元基金资助项目 *通讯作者。Tel: 010-64308204, Fax: 010-64361322, E-mail: lois222@163.com
与世界接轨,用宝尔超纯水系统 Tel: 021-64040161 www.baolor.com
王尧尧等: Western 印迹中蛋白质定量分析的替代方法
抗体标记物 底物 探测信号 定量法 所需材料 需样本量
表1 Western 印迹不同蛋白质定量分析法比较
显像密度计量法(image densitometry)
DMSO萃取法(colorimetric assay)
碱性磷酸酶
碱性磷酸酶
化学发光剂
Abstract Image densitometry has been widely used to quantify specific cellular protein expression in Western blotting analysis, but still there exists an unresolved problem of sensitivity. Furthermore, the use of expensive reagents and complex equipment limits its application. Here, we establish an improved method for quantitative examination by using alkaline phosphatase-labeled secondary antibody in combination with substrate compound naphthol AS-MX phosphate and chromagen 4-chloro-2-methylbenzenediazonium salt (fast red TR). The final colorization on blotted membrane is eluted using an organic solvent dimethylsulfoxide (DMSO) and quantified by reading absorptance at 492 nm. Our results prove that this technique is more simplified, fast and economic, without loss of its sensitivity.
  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)。

Int. J. Bioinformatics Research and Applications, Vol. 1, No. 3, 2006 319Copyright © 2006 Inderscience Enterprises Ltd. Improved protein fold assignment using support vector machines Robert E. Langlois, Alice Diec, Ognjen Perisic, Yang Dai and Hui Lu* Department of Bioengineering, University of Illinois at Chicago, 60607 Illinois, USA Fax: 312 413 2018 E-mail: rlangl1@ E-mail: adiec1@ E-mail: operis1@ E-mail: yangdai@ E-mail: huilu@ *Corresponding author Abstract: Because of the relatively large gap of knowledge between number of protein sequences and protein structures, the ability to construct a computational model predicting structure from sequence information has become an important area of research. The knowledge of a protein’s structure is crucial in understanding its biological role. In this work, we present a support vector machine based method for recognising a protein’s fold from sequence information alone, where this sequence has less similarity with sequences of known structures. We have focused on improving multi-class classification, parameter tuning, descriptor design, and feature selection. The current implementation demonstrates better prediction accuracy than previous similar approaches, and has similar performance when compared with straightforward threading. Keywords: fold recognition; support vector machines; machine learning; proteomics; structure prediction. Reference to this paper should be made as follows: Langlois, R.E., Diec, A., Perisic, O., Dai, Y. and Lu, H. (2006) ‘Improved protein fold assignment using support vector machines’, Int. J. Bioinformatics Research and Applications , Vol. 1, No. 3, pp.319–334. Biographical notes: Robert Ezra Langlois is a second year PhD student of Bioinformatics in Department of Bioengineering at University of Illinois at Chicago. He earned BS Degree in Bioengineering at UIC, May 2003. Currently he is supported by a NIH training grant: Cellular Signaling in Cardiovascular System. His research interests include machine learning, protein folding, structure prediction, protein function prediction, and binding prediction of signaling proteins. Alice Diec earned her Masters Degree in Bioinformatics from the Department of Bioengineering at UIC, October 2004. Currently, she is working in the Washington University Genome Center. Ognjen Perisic is a third year PhD student of Bioinformatics in Department of Bioengineering at UIC. His research interests are in computational biophysics, free energy calculation, non-equilibrium statistical physics in biology, and protein structure prediction.320 R.E. Langlois, A. Diec, O. Perisic, Y. Dai and H. LuYang Dai received a PhD Degree in Management Science and Engineeringfrom the University of Tsukuba, Japan, in 1991. She was a Research Associateand Assistant Professor at the Department of Management Science of KobeUniversity of Commerce (1991–1997) and at the Department of Mathematicaland Computing Sciences of Tokyo Institute of Technology (1997–2001), bothin Japan. Since 2001, she has had Faculty position in the Department ofBioengineering, University of Illinois at Chicago. Her current research focuseson bioinformatics, computational biology, machine learning, data mining, aswell as in algorithm design associated with network optimisation,combinatorial optimisation, and global optimisation.Hui Lu is an Assistant Professor in the Department of Bioengineering atUniversity of Illinois at Chicago. He earned his PhD from University of Illinoisat Urbana-Champaign, and BS from Beijing University. His research interestsin bioinformatics include: machine learning, protein folding, protein structureprediction, protein-protein and protein-DNA interactions, molecular dynamicsand Monte Carlo simulations, protein function annotation, microarray and geneexpression and gene regulation networks.1 IntroductionWith the completion of the human genome project, the accumulation of sequence information has grown and continues to grow at an exponential pace. The growth in the number of structures in the Protein Data Bank (PDB) is enough to illustrate the importance of protein structure prediction vs. more costly and time-consuming experimental methods such as X-ray crystallography and Nuclear Magnetic Resonance (NMR) spectroscopy. Having a computational method to accurately assign a structure to a specific protein sequence will provide insights into its function and evolutionary origins. There are four main strategies to solve this problem. First, homology modelling matches sequences to a particular structure using sequence similarity; common tools include HMMer (Karplus et al., 1998) and MODELLER (Sonchez and Sali, 1997; Jones, 1999) and others. Second, fold recognition in which threading is the main method. Threading detects structural similarities between sequences of little similarity; typical current programs include GenTHREADER (McGuffin and Jones 2003), PROSPECT (Xu and Xu 2000), PROSPECTOR (Skolnick and Kihara, 2001) and many others. Comparing a library of structures to a sequence, threading slides a sequence along the template structure and evaluates the match using a combination of scoring functions. Third, de novo approaches start from the assumption that native protein state is at global free energy minimum. The conformational space is searched with molecular dynamics or Monte Carlo simulations using empirical or physical based potentials, examples include Rosetta (Bonneau et al., 2001) and Touchstone (Kihara et al., 2001) among others. In the past few years, a fourth strategy, machine-learning techniques have shown promise in identifying the three-dimension fold of a protein from sequence alone where no significant identity exists to proteins with known structure using classification.Improved protein fold assignment using support vector machines 321 Two popular approaches in machine learning are neural networks and support vector machines (Cortes and Vapnik, 1995). Although each method has its own suitable application areas, support vector machines (SVM) have demonstrated better performance than other machine learning methods in a number of tasks: classifying microarray data (Brown et al., 2000), text classification (Joachims, 1998), and fold recognition (Dubchak et al., 1999; Ding and Dubchak, 2001; Yu et al., 2003). Likewise, SVM has been combined with Hidden Markov Models (HMM) for remote homology detection (Jaakkola et al., 1999). However, as a relatively new technique, there are many open research issues on how to correctly implement SVM for a given task. In this paper, we present our approach of building a multi-class SVM classifier in the context of protein fold recognition and the advances made in designing and selecting feature vectors.A wide range of techniques has been used to solve the protein fold recognition problem. The most successful one is threading. Threading attempts to assign a sequence to a set of known structures based on a set of empirical potentials (Xu and Xu, 2000). Limiting our scope to discriminative techniques still leaves us with a wide variety of approaches. One of the most accurate approaches in protein family/superfamily recognition is called SVM pairwise (Liao and Noble, 2003). This approach maps the sequence to be featured using the Smith – Waterman similarity score (SW score); the target feature vector is created using the SW score for each sequence in the training set and SVM is used to discriminate classes. Another technique, called SVM – Fisher (Jaakkola et al., 2000), uses HMM to create a feature vector of the protein sequence using the gradient of the log probability with respect to each parameter. Likewise, SVM I-sites (Hou et al., 2004) takes a different approach to vectorising a protein sequence. Here, the sequence is broken into subsequences with variable length and scored against a library of structural fragments. In other words, the feature vector consists of sequence fragment correlations for each fragment in the library. This approach is about as accurate as SVM pairwise with greater efficiency. Turning the problem on its head, the string kernel (Leslie et al., 2002) approach saves a step by implicitly representing the sequence as a feature vector. That is, the sequences are represented as vectors in the high dimensional feature space via a string-base feature map. Using kernel tricks, this can be done efficiently with accuracy similar to SVM – Fisher. Finally, Ding and Dubchak (2001) reduce a protein sequence into its structural and physical-chemical properties. The advantage of this approach lies in its efficiency and expressiveness, and it can be applied to fold recognition.The first key factor in the use of machine-learning algorithms is how to efficiently build a compact yet informative descriptor set. In this paper, we demonstrate how a carefully designed secondary structure descriptor could improve the performance of SVM based classification. Employing SVM, we have tested the effectiveness on a dataset constructed from ASTRAL40 database (Brenner et al., 2000). Our dataset is comprised of 53 folds spanning six classes with at least 20 examples in each fold. Our SVM procedure achieves an accuracy of about 20% when using amino acid composition alone and 40% for our own secondary structure descriptor. When all features were combined, we have achieved 48% (fine-grained, see Table 1) accuracy and 53% confidence over a randomised dataset of the 53 Structural Classification of Proteins (SCOP) fold dataset mentioned previously and an accuracy of 85% for class level (coarse-grained, see Table 2) classification.322 R.E. Langlois, A. Diec, O. Perisic, Y. Dai and H. LuTable 1SCOP fold-level results broken-down by SCOP classSummary: fine-grained average resultsComposition New SS Composition and new SSClass Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)a 29.09 39.73 49.09 54.33 58.18 64.93b 22.00 24.44 33.33 29.19 38.67 39.42c 15.00 23.65 49.29 47.50 52.86 54.11d 13.33 27.81 37.78 56.45 38.89 50.15f 30.00 75.00 40.00 44.44 70.00 87.50g 63.33 77.78 33.33 43.21 63.33 82.14 Average 22.64 31.95 41.70 44.96 48.49 53.74Table 2SCOP class-level resultsCoarse-grained resultsCount Composition New SS Composition and NewSSClass Fold Sequence Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)a 11 429 56.36 68.13 91.82 90.18 96.36 90.60b 15 728 64.67 65.54 90.67 87.74 92.67 93.29c 14 855 82.14 47.33 85.00 77.78 89.29 78.62d 9 353 6.67 30.00 57.78 67.53 60.00 72.00f 1 27 30.00 100.00 30.00 100.00 50.00 100.00g 3 151 80.00 96.00 83.33 83.33 76.67 92.00 Weighted 57.92 57.92 82.26 82.26 85.28 85.28 Average 53.31 67.83 73.10 84.43 77.50 87.75 The other key factor addressed here is the feature selection. There are many feature vectors that may or may not help in fold recognition from our current understanding of protein structure prediction, thus a feature selection process can be very useful. We have implemented and tested feature selection protocol that may improve the results and/or decrease the running time. This protocol will be crucial when the feature vectors increase to a larger number.We describe our implementation of SVM and feature selection protocols in ‘Methods’, and the performance of our protocol in ‘Results’. Summary and possible future improvement are presented in ‘Discussions’.Improved protein fold assignment using support vector machines 3232 Methods 2.1 Support vector machines SVM is a binary classification method, using a non-linear transformation, which maps the data to a high dimensional feature space where a linear classification is performed. It is equivalent to solving the quadratic optimisation problem: ,,1min 2i i w b i w w C ξξ⋅+∑ (1) subject to (())1,1,...,,0,1,...,,i i i i y x w b i m i m φξξ⋅+≥−=≥= where x i is a feature vector labelled by y i ∈ {+1, –1}(x i , y j ) i = 1, …, m and C is a parameter. More precisely, the given model summarises the so-called soft-margin SVM which tolerates noise within the data. The above model generates a separating plane using the equation ()()0f x x w b φ=⋅+=. Through the representation of ()j j j w a x φ=∑, we obtain ()()()i j i j j x w x x φαφφ⋅=⋅∑. This gives us an efficient approach to solve SVM without the explicit use of the non-linear transformation (Cristianini and Shawe-Taylor, 1999). 2.2 Extending SVM to multiple classes Efficiently, extending SVM to handle multiple labels has taken many forms. Some implementations (Chang and Lin, 2001) alter the above problem to handle multiple classes implicitly while others use standard machine learning techniques to extend the basic binary classifier explicitly (Ding and Dubchak, 2001). Given the maturity of the later methods and their flexibility, we implemented three such techniques: one-vs-one, one-vs-others and Decision Directed Acyclic Graph (DDAG) (Platt et al., 2000). Training DDAG and one- vs.-one is performed on every pairwise classifier or 1/2n (n – 1) classifiers where n is the number of classes. For one-vs.-others, only n classifiers are trained. Note counter-intuitively, the training time is not much different for one-vs.-one and one-vs.-others (Platt et al., 2000). Predicting the label for the ‘versus’ methods is as follows: sum the predictions for each classifier and take the label with the highest output (see Table 3). As for DDAG, the prediction creates a list of every label, and then starting with the labels at opposite ends of the list (label 1 and label n for list of size n ), predict using the corresponding classifier. The ‘winning’ class is kept and the next (previous) from the losing class indicates the corresponding classifier is to be predicted against next. Figure 1 graphically illustrates the DDAG decision process. The DDAG classifier is significantly faster (only makes n – 1 predictions, where n is the number of classes) than the previous two methods whose speeds are comparable with one another.324 R.E. Langlois, A. Diec, O. Perisic, Y. Dai and H. LuTable 3 Illustrates the voting systems in the different multiple class extensions Method Class voteAVA OVA 1 2 3 1v2 1 1(1) 0(0) 0(0) 1v3 2 1(1) 0(0) 0(1) 2v3 3 0(1) 0(1) 0(0) – – 3(3) 0(1) 0(1) This table illustrates the voting system used to combine classifiers into a single output. AVA (OVO). Figure 1 A graphical representation of the DDAG voting system The previously described feature vector (x i ) is a fixed set of attributes describing the state. Our problem is to take each state, in this case a protein sequence, and assign it a label (a SCOP fold level classification). The labels are comprised of a fixed set; the assumption being that any given input (sequence) will fall into one of these categories. It follows that finding a good mapping between feature and state lies at the crux of our work. In the following sections, we elucidate the state, labels, and features used to describe the state. 2.3 Parameter tuning The first step in training an SVM classifier is to choose the kernel. As suggested in literature (Chang and Lin, 2001), we choose the Gaussian kernel. A multi-scale grid search was used to find the best combination of two parameters, C for the soft-margin SVM and γ for the Gaussian kernel. The optimal parameters were selected by the best weighted-accuracy averaged over all classifiers (pairwise for one-vs.-one and DDAG) for five-fold cross-validation. This procedure proved optimal when compared to other techniques where each classifier maintains its own set of parameters (results not shown). However, without careful separation between training and testing, this technique may fail rather spectacularly in real application. The training accuracy value reported here reflects the average of cross-validation accuracy of each individual two-class classifier. Thus, the high percentage reported here does not indicate over-training.Improved protein fold assignment using support vector machines 3252.4 Feature selection We applied feature selection to a set of features in the dataset created by Ding and Dubchak (2001). That dataset is similar to ours but with less folds. It consists of 27 folds with no two sequences having greater than 35% sequence identity. Ding and Dubchak (2001) developed six feature vector sets utilising structural and physical-chemical properties extracted from the protein sequences. The first feature set, composition (C ), is just a simple percent composition vector of the 20 amino acids. Predicted secondary structure (S ), hydrophobicity (H ), normalised van der Waals volume (V ), polarity (P ) and polarisibility (Z ) were constructed differently. Details of the feature construction can be found in literature (Ding and Dubchak, 2001). Here, we summarise their final vector with all six independent feature vectors in Table 4, which represents a total of 20 + 21 × 5 = 125 features. Table 4 Summary of feature vector dimensions for protein fold recognition Symbol Parameter Dimension C Amino acid composition 20 S Predicted secondary structure 21 H Hydrophobicity 21 P Polarity 21 V Normalised van der Waals volume 21 Z Polarizibility 21 A ll All six feature vectors combined 125 In many classification problems, input vectors may be of high dimension. For computational efficiency, discarding irrelevant features prior to training may be favourable, particularly when the number of available features significantly outnumbers the number of examples. This is especially the case for many bioinformatic applications and the protein fold recognition problem mentioned here. Feature selection is performed for each binary classifier. The Fisher score was used as the feature selection ranking value. It is defined as 22()r r r r F r µµσσ+−+−−=+ (2) where r µ± is the mean value for the r th feature in the positive/negative class and σ is the standard deviation. To find the optimal subset of features, the protocol starts by using the top 5% of the newly ranked feature list, then adding 5% in each round, until the best performance in training set is reached. The optimal percentage of features is then used on the independent test set.326 R.E. Langlois, A. Diec, O. Perisic, Y. Dai and H. Lu2.5 DatasetThe state in our problem is comprised of a set of sequences, which are classified into different categories according to the SCOP system (Murzin et al., 1995). SCOP classifies a protein according to a hierarchical system breaking down into class, fold, superfamily and family. In previous work, (Dubchak et al., 1999), it has been shown that class level classification can already be achieved with high accuracy. Similarly, using homology-modelling techniques, superfamily level and family level recognition has not proven hard for current methods. To this end, we have focused on assigning protein sequences on the fold level. Given the difficulties in reconstructing Dubchak’s 27-fold dataset (missing identifiers and reclassified sequences), we opted to create our own, more complete dataset. Note, while Dubchak does provide an online dataset, this is composed of the features but not the original sequences. The ASTRAL40 (Brenner et al., 2000) database was used to ensure no example had more than 40% identity to another. Moreover, only folds with no less than 20 examples are taken to ensure large enough testing and training sets for accurate and significant results, respectively. The final dataset consists of 53 folds. The training and testing sets consist of 2,013 and 530 examples, respectively. That is, for each of the 53 folds, the test set has exactly ten examples whereas the training set has no less than ten examples.2.6 Accuracy scoresThe following describes the standard accuracy and confidence scores used in this paper. Note an interesting review of extending these binary accuracy scores to multi-class problems is given in Baldi et al. (2000).AccuracyTPTP FN=+(3)ConfidenceTPTP FP=+. (4)3 Results3.1 Descriptor designsOur state, a protein sequence, does not make a very good input into most standard machine-learning algorithms. These sequences vary in length and because of insertions and deletions exhibit some positional dependence. Here, our task is to map the sequences to a unique set of fixed length features and attempt to remove this positional dependence. Given that the protein sequences consist of a fixed alphabet of residues, our first attempt was to look at the relative frequencies of each residue in a protein sequence. However, as described in previous literature, this is not a very expressive descriptor. Indeed, for the 27 fold test case published before, the discriminative power of amino acid composition is around 50%. The performance in our blind 53-fold experiment is about 20% (Figure 2).Improved protein fold assignment using support vector machines 327Figure 2 Overall accuracy of the various descriptors tested on the harder dataset Next, we designed a descriptor based on the secondary structure assignment of each residue in a sequence using PSI-PRED (McGuffin et al., 2000) (not the actual DSSP (Kabsch and Sander, 1983) assignments). The idea behind this descriptor is to capture the main secondary structure elements and the corresponding topology. Here, we analysed the number of structural units and number of element patterns. For instance, we count the number of alpha helices, and then alpha helices followed by beta sheets, and so on. This is performed without preference up to some predefined limit, i.e., four-element patterns (see Figure 3). Additionally, this descriptor was extended using bins to count the sizes of individual secondary structure elements. Finally, we combine this descriptor with amino acid composition. Note this descriptor is different from other secondary structure descriptors published in the literature (Ding and Dubchak, 2001; Yu et al., 2003). Figure 3 An example of the secondary structure descriptor 3.2 Secondary structure descriptor performance Figure 2 and Table 5 show the performance of secondary structure descriptors. The accuracy, 40%, is much better than that of composition, 20%. This is a considerable improvement when compared to the already published work showing that their secondary structure descriptor as having a slightly unsatisfactory performance than the amino acid composition (Ding and Dubcheck, 2001).328 R.E. Langlois, A. Diec, O. Perisic, Y. Dai and H. LuTable 5SCOP class level accuracy for secondary structure descriptor using the gaussiankernel (C = 8, γ = 0.262)SCOP class Accuracy (%) Confidence(%)Number of foldsAll alpha 49.96 45.45 11All beta 26.64 29.33 15Alpha\beta 39.15 44.29 14 Alpha + beta 55.89 36.67 9Membrane 44.44 40.00 1 Small 49.9645.45 3As anticipated, using structural and topological information derived from sequence information provides a strong signal to classify folds. However, this approach is limitedby the accuracy of secondary structure prediction. As seen in Table 5, the best performance is from folds that consist primarily of alpha helices. This is because of the relatively accurate prediction of alpha helical secondary structures. An interesting problem arising from the SCOP method for classification is that quite a few of the alphahelical folds are wrongly predicted as folds in the alpha + beta class. This is because ofthe misnomer that the all alpha helical folds consist of only alpha helices. In those former instances, a small or insignificant beta sheet is found in a particular group of folds; thus,such folds have a greater chance of falling into the wrong category.Although the accuracy of secondary structure descriptors is better than compositionby a large amount, the best performance comes when we combine these two.The accuracy is increased to 48.5% while the confidence level is 53.7% (Figure 2).This accuracy is lower than previous publications on SVM based fold recognition, owingto the reason we are dealing with prediction of 53 folds, rather than 27 folds.From the fold-level results in Table 6, we found there is no obvious correlation between fold class and prediction accuracy (as the coarse level results might indicate).In more than a few cases the failings in secondary structure prediction account forthe misclassification.Table 6 A summary of the fold level classification displaying the accuracy for each foldFine-grained resultsComp New SS Comp and New SSFold # Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)a.1 31 50.00 71.43 100.00 76.92 90.00 69.23 a.102 25 10.00 20.00 20.00 66.67 60.00 85.71 a.118 52 40.00 40.00 90.00 69.23 80.00 72.73 a.2 22 50.00 55.56 10.00 33.33 50.00 100.00 a.24 31 30.00 42.86 60.00 46.15 50.00 55.56 a.26 26 40.00 57.14 40.00 80.00 40.00 80.00 a.3 34 30.00 30.00 50.00 55.56 60.00 54.55 a.39 42 10.00 08.33 30.00 60.00 60.00 66.67 a.4 117 40.00 11.76 90.00 26.47 90.00 26.47 a.45 20 20.00 100.00 40.00 50.00 50.00 83.33 a.60 29 00.00 00.00 10.00 33.33 10.00 20.00Improved protein fold assignment using support vector machines 329 Table 6 A summary of the fold level classification displaying the accuracy for each fold (continued)Fine-grained resultsComp New SS Comp and New SSFold # Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)Accuracy(%)Confidence(%)b.1 241 70.00 12.28 90.00 21.95 60.00 17.65 b.10 41 40.00 50.00 50.00 29.41 50.00 35.71 b.18 25 00.00 00.00 00.00 00.00 00.00 00.00 b.2 27 10.00 50.00 00.00 00.00 10.00 20.00 b.29 33 20.00 33.33 30.00 37.50 20.00 28.57 b.34 53 50.00 38.46 40.00 40.00 60.00 37.50 b.40 87 30.00 17.65 50.00 17.86 70.00 29.17 b.42 24 10.00 14.29 00.00 00.00 40.00 44.44 b.43 27 00.00 00.00 10.00 33.33 20.00 50.00 b.47 31 50.00 41.67 80.00 57.14 80.00 61.54 b.55 28 20.00 22.22 50.00 50.00 50.00 71.43 b.6 38 10.00 20.00 20.00 40.00 40.00 66.67 b.60 21 20.00 66.67 60.00 85.71 60.00 100.00 b.71 21 00.00 00.00 00.00 00.00 00.00 00.00b.82 31 00.00 00.00 20.00 25.00 20.00 28.57c.1 182 50.00 05.38 90.00 25.71 80.00 28.57 c.2 100 40.00 25.00 60.00 50.00 60.00 46.15 c.23 66 10.00 12.50 40.00 40.00 40.00 36.36 c.26 42 00.00 00.00 30.00 60.00 30.00 33.33 c.3 46 20.00 28.57 70.00 53.85 80.00 57.14 c.37 122 10.00 03.33 50.00 19.23 60.00 27.27 c.47 51 10.00 07.69 70.00 58.33 80.00 53.33 c.52 23 00.00 00.00 20.00 50.00 20.00 50.00 c.55 53 00.00 00.00 10.00 20.00 10.00 14.29 c.56 24 00.00 00.00 00.00 00.00 20.00 66.67 c.66 35 20.00 28.57 40.00 66.67 40.00 66.67 c.67 35 10.00 100.00 90.00 100.00 90.00 100.00 c.69 51 30.00 20.00 60.00 54.55 70.00 77.78c.94 25 10.00 100.00 60.00 66.67 60.00 100.00d.142 20 00.00 00.00 10.00 33.33 20.00 66.67 d.144 26 30.00 100.00 50.00 83.33 70.00 77.78 d.15 56 30.00 42.86 80.00 57.14 70.00 58.33 d.153 25 20.00 50.00 70.00 100.00 70.00 77.78 d.169 25 20.00 50.00 50.00 62.50 40.00 50.00 d.17 24 00.00 00.00 10.00 100.00 00.00 00.00 d.3 20 00.00 00.00 00.00 00.00 10.00 50.00 d.58 133 20.00 07.41 50.00 21.74 50.00 20.83 d.92 24 00.00 00.00 20.00 50.00 20.00 50.00 Weighted 22.64 22.64 41.70 41.70 48.49 48.49 Average 22.64 31.95 41.70 44.96 48.49 53.74330 R.E. Langlois, A. Diec, O. Perisic, Y. Dai and H. Lu3.3 Compare with a threading programTo evaluate our SVM setup, we compared the current performance with a threading protocol PROSPECT (Xu and Xu, 2000). The program is downloaded from Ying Xu’s webpage in University of Georgia. We ran the threading program on each of our test sequences using a fold library comprised of our training set. The program was not manually tuned, but uses the default parameters. In the end, we calculated the accuracy in the same way as the analysis of the SVM performance. The overall accuracy from threading is 56.2% which is higher than the 48.5% from SVM. The performance on individual class is: 72%, 52% 52%, 47%, 30%, and 77%. Notice that threading used much more information from the sequences and structures than SVM, such as structural contacts, statistical potentials, and sequence similarities, it is expected SVM performance will increase when such information is included. On the other hand, we want to emphasise that the current threading setup doesn’t reflect the true performance of state of art in threading when the template structure database is properly set-up and human knowledge is included. This comparison between SVM and threading is for the purpose of providing a quick evaluation of SVM to insure that we are on the right track.3.4 Feature selectionFeature selection is done for two reasons. First, removing unnecessary or ‘bad’ features can improve the accuracy of most machine learning algorithms. While some work claimed feature selection would improve the accuracy of SVM experiments (Weston et al., 2000), this is not the case in some other applications (Liu, 2004). Second, feature selection does provide insights into the quality and productivity of each feature. The features selected by the Fisher score equation (2) in the 75th percentile across all OVO classifiers were tallied and the frequencies presented in Figure 4. Note that the order of features reflects the order of C, S, H, P, V and Z (using Ding and Dubchak dataset). As shown in Figures 4 and 5, composition (C) and secondary structure (S) generate the strongest signals. Further evidence is provided in Figure 4 where the combination of C and S performs quite well. Hydrophobicity also induces a moderate signal (Figure 4), as further supported in Figure 5 demonstrating that this combination yields the best results.requency of features ranked in the 75th percentileFigure 4F。

相关文档
最新文档