首页 > 最新文献

Current Protocols in Protein Science最新文献

英文 中文
De Novo Protein Design Using the Blueprint Builder in Rosetta 在Rosetta中使用蓝图构建器重新设计蛋白质
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-12-15 DOI: 10.1002/cpps.116
Linna An, Gyu Rie Lee

While native proteins cover diverse structural spaces and achieve various biological events, not many of them can directly serve human needs. One reason is that the native proteins usually contain idiosyncrasies evolved for their native functions but disfavoring engineering requirements. To overcome this issue, one strategy is to create de novo proteins which are designed to possess improved stability, high environmental tolerance, and enhanced engineering potential. Compared to other protein engineering strategies, in silico design of de novo proteins has significantly expanded the protein structural and sequence spaces, reduced wet lab workload, and incorporated engineered features in a guided and efficient manner. In the Baker laboratory we have been applying a design pipeline that uses the blueprint builder to design different folds of de novo proteins, and have successfully obtained libraries of de novo proteins with improved stability and engineering potential. In this article, we will use the design of de novo β-barrel proteins as an example to describe the principles and basic procedures of the blueprint builder−based design pipeline. © 2020 Wiley Periodicals LLC.

Basic Protocol 1: The construction of blueprints

Alternate Protocol: Build blueprints based on existing protein .pdb files

Basic Protocol 2: De novo protein design pipeline using the blueprint builder

虽然天然蛋白覆盖了不同的结构空间,实现了不同的生物事件,但能够直接服务于人类需求的天然蛋白并不多。其中一个原因是,天然蛋白质通常含有为其天然功能而进化的特质,但不利于工程需求。为了克服这个问题,一种策略是创造新的蛋白质,这些蛋白质被设计成具有更好的稳定性,高环境耐受性和增强的工程潜力。与其他蛋白质工程策略相比,从头开始的蛋白质的芯片设计显著扩展了蛋白质结构和序列空间,减少了湿实验室工作量,并以一种指导和有效的方式融入了工程特征。在Baker实验室,我们一直在应用一种设计管道,该管道使用蓝图构建器来设计不同折叠的新生蛋白,并成功获得了具有更高稳定性和工程潜力的新生蛋白库。在本文中,我们将以从头开始的β桶蛋白的设计为例,描述基于蓝图构建器的设计管道的原理和基本过程。©2020 Wiley期刊有限公司基本协议1:构建蓝图替代协议:基于现有的蛋白质。pdb文件构建蓝图基本协议2:使用蓝图构建器从头设计蛋白质管道
{"title":"De Novo Protein Design Using the Blueprint Builder in Rosetta","authors":"Linna An,&nbsp;Gyu Rie Lee","doi":"10.1002/cpps.116","DOIUrl":"10.1002/cpps.116","url":null,"abstract":"<p>While native proteins cover diverse structural spaces and achieve various biological events, not many of them can directly serve human needs. One reason is that the native proteins usually contain idiosyncrasies evolved for their native functions but disfavoring engineering requirements. To overcome this issue, one strategy is to create de novo proteins which are designed to possess improved stability, high environmental tolerance, and enhanced engineering potential. Compared to other protein engineering strategies, in silico design of de novo proteins has significantly expanded the protein structural and sequence spaces, reduced wet lab workload, and incorporated engineered features in a guided and efficient manner. In the Baker laboratory we have been applying a design pipeline that uses the blueprint builder to design different folds of de novo proteins, and have successfully obtained libraries of de novo proteins with improved stability and engineering potential. In this article, we will use the design of de novo β-barrel proteins as an example to describe the principles and basic procedures of the blueprint builder−based design pipeline. © 2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol 1</b>: The construction of blueprints</p><p><b>Alternate Protocol</b>: Build blueprints based on existing protein <span>.pdb</span> files</p><p><b>Basic Protocol 2</b>: De novo protein design pipeline using the blueprint builder</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.116","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38723942","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 5
Methods for Expression of Recombinant Proteins Using a Pichia pastoris Cell-Free System 利用毕赤酵母无细胞系统表达重组蛋白的方法
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-10-27 DOI: 10.1002/cpps.115
Rochelle Aw, Alex J. Spice, Karen M. Polizzi

Cell-free protein synthesis is a powerful tool for engineering biology and has been utilized in many diverse applications, from biosensing and protein prototyping to biomanufacturing and the design of metabolic pathways. By exploiting host cellular machinery decoupled from cellular growth, proteins can be produced in vitro both on demand and rapidly. Eukaryotic cell-free platforms are often neglected due to perceived complexity and low yields relative to their prokaryotic counterparts, despite providing a number of advantageous properties. The yeast Pichia pastoris (also known as Komagataella phaffii) is a particularly attractive eukaryotic host from which to generate cell-free extracts, due to its ability to grow to high cell densities with high volumetric productivity, genetic tractability for strain engineering, and ability to perform post-translational modifications. Here, we describe methods for conducting cell-free protein synthesis using P. pastoris as the host, from preparing the cell lysates to protocols for both coupled and linked transcription-translation reactions. By providing these methodologies, we hope to encourage the adoption of the platform by new and experienced users alike. © 2020 The Authors.

Basic Protocol 1: Preparation of Pichia pastoris cell lysate

Basic Protocol 2: Coupled in vitro transcription and translation

Basic Protocol 3: Determining luciferase production from cell-free protein synthesis reactions

Alternate Protocol 1: Linked in vitro transcription and translation

Alternate Protocol 2: Quantifying HSA protein concentration

Support Protocol 1: Preparation of mRNA by in vitro transcription for linked transcription and translation

无细胞蛋白质合成是工程生物学的一个强大工具,已被用于许多不同的应用,从生物传感和蛋白质原型到生物制造和代谢途径的设计。通过利用与细胞生长分离的宿主细胞机制,可以在体外按需快速地生产蛋白质。真核无细胞平台往往被忽视,由于感知的复杂性和低产量相对于他们的原核同行,尽管提供了许多有利的性质。酵母毕赤酵母(也称为Komagataella phaffii)是一种特别有吸引力的真核宿主,可以产生无细胞提取物,因为它能够生长到高细胞密度,高容量生产力,菌株工程的遗传易感性,以及进行翻译后修饰的能力。在这里,我们描述了利用帕斯德酵母作为宿主进行无细胞蛋白合成的方法,从制备细胞裂解物到偶联和连锁转录-翻译反应的方案。通过提供这些方法,我们希望鼓励新用户和有经验的用户都采用这个平台。©2020作者。基本方案1:制备毕赤酵母裂解细胞基本方案2:耦合体外转录和翻译基本方案3:测定无细胞蛋白合成反应中荧光素酶的产生备选方案1:连接体外转录和翻译备选方案2:定量HSA蛋白浓度支持方案1:通过体外转录制备mRNA进行连接转录和翻译
{"title":"Methods for Expression of Recombinant Proteins Using a Pichia pastoris Cell-Free System","authors":"Rochelle Aw,&nbsp;Alex J. Spice,&nbsp;Karen M. Polizzi","doi":"10.1002/cpps.115","DOIUrl":"10.1002/cpps.115","url":null,"abstract":"<p>Cell-free protein synthesis is a powerful tool for engineering biology and has been utilized in many diverse applications, from biosensing and protein prototyping to biomanufacturing and the design of metabolic pathways. By exploiting host cellular machinery decoupled from cellular growth, proteins can be produced in vitro both on demand and rapidly. Eukaryotic cell-free platforms are often neglected due to perceived complexity and low yields relative to their prokaryotic counterparts, despite providing a number of advantageous properties. The yeast <i>Pichia pastoris</i> (also known as <i>Komagataella phaffii</i>) is a particularly attractive eukaryotic host from which to generate cell-free extracts, due to its ability to grow to high cell densities with high volumetric productivity, genetic tractability for strain engineering, and ability to perform post-translational modifications. Here, we describe methods for conducting cell-free protein synthesis using <i>P. pastoris</i> as the host, from preparing the cell lysates to protocols for both coupled and linked transcription-translation reactions. By providing these methodologies, we hope to encourage the adoption of the platform by new and experienced users alike. © 2020 The Authors.</p><p><b>Basic Protocol 1</b>: Preparation of <i>Pichia pastoris</i> cell lysate</p><p><b>Basic Protocol 2</b>: Coupled in vitro transcription and translation</p><p><b>Basic Protocol 3</b>: Determining luciferase production from cell-free protein synthesis reactions</p><p><b>Alternate Protocol 1</b>: Linked in vitro transcription and translation</p><p><b>Alternate Protocol 2</b>: Quantifying HSA protein concentration</p><p><b>Support Protocol 1</b>: Preparation of mRNA by in vitro transcription for linked transcription and translation</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.115","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38531382","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
Histone Purification Combined with High-Resolution Mass Spectrometry to Examine Histone Post-Translational Modifications and Histone Variants in Caenorhabditis elegans 组蛋白纯化结合高分辨率质谱法检测秀丽隐杆线虫组蛋白翻译后修饰和组蛋白变异
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-09-30 DOI: 10.1002/cpps.114
Lluís Millan-Ariño, Zuo-Fei Yuan, Marlies E. Oomen, Simone Brandenburg, Alexey Chernobrovkin, Jérôme Salignon, Lioba Körner, Roman A. Zubarev, Benjamin A. Garcia, Christian G. Riedel
Histones are the major proteinaceous component of chromatin in eukaryotic cells and an important part of the epigenome, affecting most DNA‐related events, including transcription, DNA replication, and chromosome segregation. The properties of histones are greatly influenced by their post‐translational modifications (PTMs), over 200 of which are known today. Given this large number, researchers need sophisticated methods to study histone PTMs comprehensively. In particular, mass spectrometry (MS)−based approaches have gained popularity, allowing for the quantification of dozens of histone PTMs at once. Using these approaches, even the study of co‐occurring PTMs and the discovery of novel PTMs become feasible. The success of MS‐based approaches relies substantially on obtaining pure and well‐preserved histones for analysis, which can be difficult depending on the source material. Caenorhabditis elegans has been a popular model organism to study the epigenome, but isolation of pure histones from these animals has been challenging. Here, we address this issue, presenting a method for efficient isolation of pure histone proteins from C. elegans at good yield. Further, we describe an MS pipeline optimized for accurate relative quantification of histone PTMs from C. elegans. We alkylate and tryptically digest the histones, analyze them by bottom‐up MS, and then evaluate the resulting data by a C. elegans−adapted version of the software EpiProfile 2.0. Finally, we show the utility of this pipeline by determining differences in histone PTMs between C. elegans strains that age at different rates and thereby achieve very different lifespans. © 2020 The Authors.
组蛋白是真核细胞中染色质的主要蛋白质成分,也是表观基因组的重要组成部分,影响大多数DNA相关事件,包括转录、DNA复制和染色体分离。组蛋白的特性很大程度上受其翻译后修饰(PTMs)的影响,目前已知的PTMs有200多种。由于数量庞大,研究人员需要复杂的方法来全面研究组蛋白ptm。特别是,基于质谱(MS)的方法已经得到普及,允许一次定量几十个组蛋白PTMs。使用这些方法,甚至研究共同发生的ptm和发现新的ptm也变得可行。基于质谱的方法的成功主要依赖于获得纯的和保存良好的组蛋白进行分析,这可能是困难的,取决于源材料。秀丽隐杆线虫一直是研究表观基因组的流行模式生物,但从这些动物中分离纯组蛋白一直具有挑战性。在这里,我们解决了这个问题,提出了一种从秀丽隐杆线虫中高效分离纯组蛋白的方法。此外,我们描述了一个MS管道,优化了秀丽隐杆线虫组蛋白PTMs的准确相对定量。我们对组蛋白进行烷基化和胰消化,用自下而上的质谱法对其进行分析,然后用适合线虫的EpiProfile 2.0软件对结果数据进行评估。最后,我们通过确定以不同速度衰老的秀丽隐杆线虫菌株之间组蛋白ptm的差异,从而实现非常不同的寿命,展示了这一管道的实用性。©2020作者。基本方案1:同步秀丽隐杆线虫的大规模生长和收获。基本方案2:核制备、组蛋白提取和组蛋白纯化。基本方案3:组蛋白PTMs和组蛋白变体的自下而上质谱分析
{"title":"Histone Purification Combined with High-Resolution Mass Spectrometry to Examine Histone Post-Translational Modifications and Histone Variants in Caenorhabditis elegans","authors":"Lluís Millan-Ariño,&nbsp;Zuo-Fei Yuan,&nbsp;Marlies E. Oomen,&nbsp;Simone Brandenburg,&nbsp;Alexey Chernobrovkin,&nbsp;Jérôme Salignon,&nbsp;Lioba Körner,&nbsp;Roman A. Zubarev,&nbsp;Benjamin A. Garcia,&nbsp;Christian G. Riedel","doi":"10.1002/cpps.114","DOIUrl":"10.1002/cpps.114","url":null,"abstract":"Histones are the major proteinaceous component of chromatin in eukaryotic cells and an important part of the epigenome, affecting most DNA‐related events, including transcription, DNA replication, and chromosome segregation. The properties of histones are greatly influenced by their post‐translational modifications (PTMs), over 200 of which are known today. Given this large number, researchers need sophisticated methods to study histone PTMs comprehensively. In particular, mass spectrometry (MS)−based approaches have gained popularity, allowing for the quantification of dozens of histone PTMs at once. Using these approaches, even the study of co‐occurring PTMs and the discovery of novel PTMs become feasible. The success of MS‐based approaches relies substantially on obtaining pure and well‐preserved histones for analysis, which can be difficult depending on the source material. Caenorhabditis elegans has been a popular model organism to study the epigenome, but isolation of pure histones from these animals has been challenging. Here, we address this issue, presenting a method for efficient isolation of pure histone proteins from C. elegans at good yield. Further, we describe an MS pipeline optimized for accurate relative quantification of histone PTMs from C. elegans. We alkylate and tryptically digest the histones, analyze them by bottom‐up MS, and then evaluate the resulting data by a C. elegans−adapted version of the software EpiProfile 2.0. Finally, we show the utility of this pipeline by determining differences in histone PTMs between C. elegans strains that age at different rates and thereby achieve very different lifespans. © 2020 The Authors.","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.114","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38439533","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 4
A Rapid and Facile Purification Method for Glycan-Binding Proteins and Glycoproteins 一种快速简便的糖结合蛋白和糖蛋白纯化方法
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-09-03 DOI: 10.1002/cpps.113
Christina J. Welch, Priyanka D. Kadav, Jared L. Edwards, Jessica Krycia, Melanie L. Talaga, Purnima Bandyopadhyay, Tarun K. Dam

Glycosylated proteins, namely glycoproteins and proteoglycans (collectively called glycoconjugates), are indispensable in a variety of biological processes. The functions of many glycoconjugates are regulated by their interactions with another group of proteins known as lectins. In order to understand the biological functions of lectins and their glycosylated binding partners, one must obtain these proteins in pure form. The conventional protein purification methods often require long times, elaborate infrastructure, costly reagents, and large sample volumes. To minimize some of these problems, we recently developed and validated a new method termed capture and release (CaRe). This method is time-saving, precise, inexpensive, and it needs a relatively small sample volume. In this approach, targets (lectins and glycoproteins) are captured in solution by multivalent ligands called target capturing agents (TCAs). The captured targets are then released and separated from their TCAs to obtain purified targets. Application of the CaRe method could play an important role in discovering new lectins and glycoconjugates. © 2020 Wiley Periodicals LLC.

Basic Protocol 1: Preparation of crude extracts containing the target proteins from soybean flour

Alternate Protocol 1: Preparation of crude extracts from Jack bean meal

Alternate Protocol 2: Preparation of crude extracts from the corms of Colocasia esculenta, Xanthosoma sagittifolium, and from the bulbs of Allium sativum

Alternate Protocol 3: Preparation of Escherichia coli cell lysates containing human galectin-3

Alternate Protocol 4: Preparation of crude extracts from chicken egg whites (source of ovalbumin)

Basic Protocol 2: Preparation of 2% (v/v) red blood cell suspension

Basic Protocol 3: Detection of lectin activity of the crude extracts

Basic Protocol 4: Identification of multivalent inhibitors as target capturing agents by hemagglutination inhibition assays

Basic Protocol 5: Testing the capturing abilities of target capturing agents by precipitation/turbidity assays

Basic Protocol 6: Capturing of targets (lectins and glycoproteins) in the crude extracts by target capturing agents and separation of the target-TCA complex from other components of the crude extracts

Basic Protocol 7: Releasing the captured targets (lectins and glycoproteins) by dissolving the complex

Basic Protocol 8: Separation of the targets (lectins and glycoproteins) from their respective target capturing agents

Basic Protocol 9: Verification of the purity of the isolated targets (lectins or glycoproteins)

糖基化蛋白,即糖蛋白和蛋白聚糖(统称为糖缀合物),在各种生物过程中是不可或缺的。许多糖缀合物的功能是通过它们与另一组称为凝集素的蛋白质的相互作用来调节的。为了了解凝集素及其糖基化结合伙伴的生物学功能,必须以纯形式获得这些蛋白质。传统的蛋白质纯化方法通常需要很长时间、复杂的基础设施、昂贵的试剂和大量的样品。为了尽量减少这些问题,我们最近开发并验证了一种称为捕获和释放(CaRe)的新方法。该方法具有省时、精确、廉价、样本量相对较小等优点。在这种方法中,靶标(凝集素和糖蛋白)在溶液中被称为靶标捕获剂(TCAs)的多价配体捕获。然后释放捕获的目标并将其与tca分离以获得纯化的目标。该方法的应用将在发现新的凝集素和糖缀合物方面发挥重要作用。©2020 Wiley Periodicals llc .基本方案1:从豆粉中制备含有目标蛋白的粗提物备用方案1:从豆粕中制备粗提物备用方案2:从大白菜球茎、矢状叶黄索菌和葱球茎中制备粗提物备用方案3:制备含有人半乳糖凝集素-3的大肠杆菌细胞裂解物备用方案4:基础方案2:制备2% (v/v)红细胞悬浮液基础方案3:检测粗提取物的凝集素活性基础方案4:通过血凝抑制试验鉴定多价抑制剂作为靶捕集剂基础方案5:通过沉淀/浊度试验测试靶捕集剂的捕集能力基础方案6:用目标捕获剂捕获粗提取物中的目标(凝集素和糖蛋白),并将目标- tca复合物与粗提取物的其他组分分离基本方案7:通过溶解络合物释放捕获的目标(凝集素和糖蛋白)基本方案8:从各自的目标捕获剂中分离目标(凝集素和糖蛋白)基本方案9:验证分离的目标(凝集素或糖蛋白)的纯度
{"title":"A Rapid and Facile Purification Method for Glycan-Binding Proteins and Glycoproteins","authors":"Christina J. Welch,&nbsp;Priyanka D. Kadav,&nbsp;Jared L. Edwards,&nbsp;Jessica Krycia,&nbsp;Melanie L. Talaga,&nbsp;Purnima Bandyopadhyay,&nbsp;Tarun K. Dam","doi":"10.1002/cpps.113","DOIUrl":"10.1002/cpps.113","url":null,"abstract":"<p>Glycosylated proteins, namely glycoproteins and proteoglycans (collectively called glycoconjugates), are indispensable in a variety of biological processes. The functions of many glycoconjugates are regulated by their interactions with another group of proteins known as lectins. In order to understand the biological functions of lectins and their glycosylated binding partners, one must obtain these proteins in pure form. The conventional protein purification methods often require long times, elaborate infrastructure, costly reagents, and large sample volumes. To minimize some of these problems, we recently developed and validated a new method termed capture and release (CaRe). This method is time-saving, precise, inexpensive, and it needs a relatively small sample volume. In this approach, targets (lectins and glycoproteins) are captured in solution by multivalent ligands called target capturing agents (TCAs). The captured targets are then released and separated from their TCAs to obtain purified targets. Application of the CaRe method could play an important role in discovering new lectins and glycoconjugates. © 2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol 1</b>: Preparation of crude extracts containing the target proteins from soybean flour</p><p><b>Alternate Protocol 1</b>: Preparation of crude extracts from Jack bean meal</p><p><b>Alternate Protocol 2</b>: Preparation of crude extracts from the corms of <i>Colocasia esculenta</i>, <i>Xanthosoma sagittifolium</i>, and from the bulbs of <i>Allium sativum</i></p><p><b>Alternate Protocol 3</b>: Preparation of <i>Escherichia coli</i> cell lysates containing human galectin-3</p><p><b>Alternate Protocol 4</b>: Preparation of crude extracts from chicken egg whites (source of ovalbumin)</p><p><b>Basic Protocol 2</b>: Preparation of 2% (v/v) red blood cell suspension</p><p><b>Basic Protocol 3</b>: Detection of lectin activity of the crude extracts</p><p><b>Basic Protocol 4</b>: Identification of multivalent inhibitors as target capturing agents by hemagglutination inhibition assays</p><p><b>Basic Protocol 5</b>: Testing the capturing abilities of target capturing agents by precipitation/turbidity assays</p><p><b>Basic Protocol 6</b>: Capturing of targets (lectins and glycoproteins) in the crude extracts by target capturing agents and separation of the target-TCA complex from other components of the crude extracts</p><p><b>Basic Protocol 7</b>: Releasing the captured targets (lectins and glycoproteins) by dissolving the complex</p><p><b>Basic Protocol 8</b>: Separation of the targets (lectins and glycoproteins) from their respective target capturing agents</p><p><b>Basic Protocol 9</b>: Verification of the purity of the isolated targets (lectins or glycoproteins)</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.113","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38341341","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Synthesis of Recombinant Human Hemoglobin With NH2-Terminal Acetylation in Escherichia coli NH2末端乙酰化在大肠杆菌中合成重组人血红蛋白
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-07-20 DOI: 10.1002/cpps.112
Chandrasekhar Natarajan, Anthony V. Signore, Vikas Kumar, Jay F. Storz

The development of new technologies for the efficient expression of recombinant hemoglobin (rHb) is of interest for experimental studies of protein biochemistry and the development of cell-free blood substitutes in transfusion medicine. Expression of rHb in Escherichia coli host cells has numerous advantages, but one disadvantage of using prokaryotic systems to express eukaryotic proteins is that they are incapable of performing post-translational modifications such as NH2-terminal acetylation. One possible solution is to coexpress additional enzymes that can perform the necessary modifications in the host cells. Here, we report a new method for synthesizing human rHb with proper NH2-terminal acetylation. Mass spectrometry experiments involving native and recombinant human Hb confirmed the efficacy of the new technique in producing correctly acetylated globin chains. Finally, functional experiments provided insights into the effects of NH2-terminal acetylation on O2 binding properties. © 2020 Wiley Periodicals LLC.

Basic Protocol 1: Gene synthesis and cloning the cassette to the expression plasmid

Basic Protocol 2: Selection of E. coli expression strains for coexpression

Basic Protocol 3: Large-scale recombinant hemoglobin expression and purification

Support Protocol 1: Measuring O2 equilibration curves

Support Protocol 2: Mass spectrometry to confirm NH2-terminal acetylation

重组血红蛋白(rHb)高效表达新技术的开发对蛋白质生物化学的实验研究和输血医学中无细胞血液替代品的开发具有重要意义。rHb在大肠杆菌宿主细胞中的表达具有许多优点,但使用原核系统表达真核蛋白质的一个缺点是它们不能进行翻译后修饰,如NH2末端乙酰化。一种可能的解决方案是共表达额外的酶,这些酶可以在宿主细胞中进行必要的修饰。在此,我们报道了一种通过适当的NH2末端乙酰化合成人rHb的新方法。涉及天然和重组人Hb的质谱实验证实了新技术在产生正确乙酰化珠蛋白链方面的有效性。最后,功能实验为NH2末端乙酰化对O2结合特性的影响提供了见解。©2020 Wiley Periodicals LLC基本方案1:基因合成和克隆表达质粒盒基本方案2:选择共表达的大肠杆菌表达菌株基本方案3:大规模重组血红蛋白表达和纯化支持方案1:测量O2平衡曲线支持方案2:质谱法确认NH2末端乙酰化
{"title":"Synthesis of Recombinant Human Hemoglobin With NH2-Terminal Acetylation in Escherichia coli","authors":"Chandrasekhar Natarajan,&nbsp;Anthony V. Signore,&nbsp;Vikas Kumar,&nbsp;Jay F. Storz","doi":"10.1002/cpps.112","DOIUrl":"https://doi.org/10.1002/cpps.112","url":null,"abstract":"<p>The development of new technologies for the efficient expression of recombinant hemoglobin (rHb) is of interest for experimental studies of protein biochemistry and the development of cell-free blood substitutes in transfusion medicine. Expression of rHb in <i>Escherichia coli</i> host cells has numerous advantages, but one disadvantage of using prokaryotic systems to express eukaryotic proteins is that they are incapable of performing post-translational modifications such as NH<sub>2</sub>-terminal acetylation. One possible solution is to coexpress additional enzymes that can perform the necessary modifications in the host cells. Here, we report a new method for synthesizing human rHb with proper NH<sub>2</sub>-terminal acetylation. Mass spectrometry experiments involving native and recombinant human Hb confirmed the efficacy of the new technique in producing correctly acetylated globin chains. Finally, functional experiments provided insights into the effects of NH<sub>2</sub>-terminal acetylation on O<sub>2</sub> binding properties. © 2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol 1</b>: Gene synthesis and cloning the cassette to the expression plasmid</p><p><b>Basic Protocol 2</b>: Selection of <i>E. coli</i> expression strains for coexpression</p><p><b>Basic Protocol 3</b>: Large-scale recombinant hemoglobin expression and purification</p><p><b>Support Protocol 1</b>: Measuring O<sub>2</sub> equilibration curves</p><p><b>Support Protocol 2</b>: Mass spectrometry to confirm NH<sub>2</sub>-terminal acetylation</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-07-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.112","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72159192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
A Fluorescence-Based Assay to Monitor SUMOylation in Real-Time 实时监测SUMOylation的荧光检测方法
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-07-07 DOI: 10.1002/cpps.111
Vasvi Tripathi, Ranabir Das

The small ubiquitin-like modifier (SUMO) is an important post-translational modifier that regulates various cellular processes. Extensive investigations have been made to comprehend the enzymatic process and consequence of SUMOylation. In vitro SUMOylation assays are invaluable for understanding the fundamental mechanisms of SUMOylation. A majority of these assays monitor changes in the size of the substrate upon SUMO conjugation. Current methods typically detect the size difference through SDS-PAGE and western blots, which makes these methods cumbersome, error-prone, and time-consuming. Here, we describe a fluorescence-based assay for real-time detection of SUMOylation. In the method, a fluorophore-tagged substrate is used in the SUMOylation reaction. Upon SUMOylation, the size and correlation time (τc) of the substrate increases, and so does its anisotropy. The rate of change in anisotropy with time reflects the efficiency of the SUMOylation machinery. The real-time SUMOylation assay protocol is elegant, time-saving, and less prone to errors. © 2020 Wiley Periodicals LLC.

Basic Protocol: Fluorescent anisotropy-based in vitro SUMOylation assay

小泛素样修饰物(SUMO)是一种重要的翻译后修饰物,可调节多种细胞过程。广泛的研究已经完成,以了解酶的过程和后果的SUMOylation。体外SUMOylation分析对于理解SUMOylation的基本机制是非常宝贵的。大多数这些检测监测在SUMO偶联的底物的大小变化。目前的方法通常通过SDS-PAGE和western blots检测大小差异,这使得这些方法繁琐、容易出错且耗时。在这里,我们描述了一种基于荧光的实时检测SUMOylation的方法。在该方法中,在SUMOylation反应中使用荧光团标记的底物。在SUMOylation后,底物的尺寸和相关时间(τc)增加,其各向异性也增加。各向异性随时间的变化率反映了SUMOylation机制的效率。实时SUMOylation分析方案是优雅的,节省时间,不容易出错。©2020 Wiley期刊公司基本方案:基于荧光各向异性的体外sumo酰化测定
{"title":"A Fluorescence-Based Assay to Monitor SUMOylation in Real-Time","authors":"Vasvi Tripathi,&nbsp;Ranabir Das","doi":"10.1002/cpps.111","DOIUrl":"10.1002/cpps.111","url":null,"abstract":"<p>The small ubiquitin-like modifier (SUMO) is an important post-translational modifier that regulates various cellular processes. Extensive investigations have been made to comprehend the enzymatic process and consequence of SUMOylation. In vitro SUMOylation assays are invaluable for understanding the fundamental mechanisms of SUMOylation. A majority of these assays monitor changes in the size of the substrate upon SUMO conjugation. Current methods typically detect the size difference through SDS-PAGE and western blots, which makes these methods cumbersome, error-prone, and time-consuming. Here, we describe a fluorescence-based assay for real-time detection of SUMOylation. In the method, a fluorophore-tagged substrate is used in the SUMOylation reaction. Upon SUMOylation, the size and correlation time (τ<sub>c</sub>) of the substrate increases, and so does its anisotropy. The rate of change in anisotropy with time reflects the efficiency of the SUMOylation machinery. The real-time SUMOylation assay protocol is elegant, time-saving, and less prone to errors. © 2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol</b>: Fluorescent anisotropy-based in vitro SUMOylation assay</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-07-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.111","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38126252","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Quantitative Analysis of Protein Self-Association by Sedimentation Velocity 用沉降速度定量分析蛋白质的自结合
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-07-02 DOI: 10.1002/cpps.109
Huaying Zhao, Wenqi Li, Wendan Chu, Mary Bollard, Regina Adão, Peter Schuck

Sedimentation velocity analytical ultracentrifugation is a powerful classical method to study protein self-association processes in solution based on the size-dependent macromolecular migration in the centrifugal field. This technique can elucidate the assembly scheme, measure affinities ranging from picomolar to millimolar Kd, and in favorable cases provide information on oligomer lifetimes and hydrodynamic shape. The present step-by-step protocols detail the essential steps of instrument calibration, experimental setup, and data analysis. Using a widely available commercial protein as a model system, the protocols invite replication and comparison with our results. A commentary discusses principles for modifications in the protocols that may be necessary to optimize application of sedimentation velocity analysis to other self-associating proteins. ©2020 Wiley Periodicals LLC.

Basic Protocol 1: Measurement of external calibration factors

Basic Protocol 2: Sedimentation velocity experiment for protein self-association

Basic Protocol 3: Sedimentation coefficient distribution analysis in SEDFIT and isotherm analysis in SEDPHAT

沉降速度分析超离心是一种基于离心场中大小依赖的大分子迁移来研究蛋白质在溶液中自结合过程的有力的经典方法。该技术可以阐明组装方案,测量从皮摩尔到毫摩尔Kd的亲和度,并在有利的情况下提供有关低聚物寿命和流体动力学形状的信息。目前一步一步的协议详细说明仪器校准,实验设置和数据分析的基本步骤。使用广泛可用的商业蛋白质作为模型系统,该方案可以与我们的结果进行复制和比较。评论讨论了可能需要优化沉降速度分析在其他自结合蛋白中的应用的协议修改原则。©2020 Wiley期刊公司基本方案1:外部校准因子的测量基本方案2:蛋白质自结合的沉降速度实验基本方案3:SEDFIT中的沉降系数分布分析和SEDPHAT中的等温线分析
{"title":"Quantitative Analysis of Protein Self-Association by Sedimentation Velocity","authors":"Huaying Zhao,&nbsp;Wenqi Li,&nbsp;Wendan Chu,&nbsp;Mary Bollard,&nbsp;Regina Adão,&nbsp;Peter Schuck","doi":"10.1002/cpps.109","DOIUrl":"10.1002/cpps.109","url":null,"abstract":"<p>Sedimentation velocity analytical ultracentrifugation is a powerful classical method to study protein self-association processes in solution based on the size-dependent macromolecular migration in the centrifugal field. This technique can elucidate the assembly scheme, measure affinities ranging from picomolar to millimolar <i>K</i><sub>d</sub>, and in favorable cases provide information on oligomer lifetimes and hydrodynamic shape. The present step-by-step protocols detail the essential steps of instrument calibration, experimental setup, and data analysis. Using a widely available commercial protein as a model system, the protocols invite replication and comparison with our results. A commentary discusses principles for modifications in the protocols that may be necessary to optimize application of sedimentation velocity analysis to other self-associating proteins. ©2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol 1</b>: Measurement of external calibration factors</p><p><b>Basic Protocol 2</b>: Sedimentation velocity experiment for protein self-association</p><p><b>Basic Protocol 3</b>: Sedimentation coefficient distribution analysis in SEDFIT and isotherm analysis in SEDPHAT</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.109","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38109604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 6
PLiMAP: Proximity-Based Labeling of Membrane-Associated Proteins PLiMAP:基于接近度的膜相关蛋白标记
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-06-30 DOI: 10.1002/cpps.110
Gregor P. Jose, Thomas J. Pucadyil

Peripheral membrane proteins participate in numerous biological pathways. Thus, methods to analyze their membrane-binding characteristics have become important. In this report, we detail protocols for the synthesis and utilization of a photoactivable fluorescent lipid as a reporter to monitor membrane binding of proteins. The assay, referred to as proximity-based labeling of membrane-associated proteins (PLiMAP), is based on UV activation of a fluorescent lipid reporter, which in turn crosslinks with proteins bound to membranes and renders them fluorescent. © 2020 Wiley Periodicals LLC.

Basic Protocol 1: Synthesis of BODIPY-diazirine phosphatidylethanolamine (BDPE)

Basic Protocol 2: Preparation of BDPE-containing liposomes

Basic Protocol 3: Performing PLiMAP with a candidate protein

Basic Protocol 4: Quantitation of liposome-binding properties of the candidate protein from analyzing in-gel fluorescence

Support Protocol: Purification of GST-2×P4M domain of SidM protein

外周膜蛋白参与多种生物学途径。因此,分析其膜结合特性的方法变得非常重要。在本报告中,我们详细介绍了一种光活化荧光脂质的合成和利用方案,作为监测蛋白质膜结合的报告。该检测被称为基于接近度的膜相关蛋白标记(PLiMAP),是基于紫外线激活的荧光脂质报告蛋白,该报告蛋白反过来与结合在膜上的蛋白质交联并使其荧光。©2020 Wiley期刊公司。基本方案1:合成bodipy -二嗪磷脂酰乙醇胺(BDPE)基本方案2:制备含BDPE的脂质体基本方案3:用候选蛋白进行PLiMAP基本方案4:通过分析凝胶内荧光定量候选蛋白的脂质体结合特性支持方案:纯化SidM蛋白的GST-2×P4M结构域
{"title":"PLiMAP: Proximity-Based Labeling of Membrane-Associated Proteins","authors":"Gregor P. Jose,&nbsp;Thomas J. Pucadyil","doi":"10.1002/cpps.110","DOIUrl":"10.1002/cpps.110","url":null,"abstract":"<p>Peripheral membrane proteins participate in numerous biological pathways. Thus, methods to analyze their membrane-binding characteristics have become important. In this report, we detail protocols for the synthesis and utilization of a photoactivable fluorescent lipid as a reporter to monitor membrane binding of proteins. The assay, referred to as proximity-based labeling of membrane-associated proteins (PLiMAP), is based on UV activation of a fluorescent lipid reporter, which in turn crosslinks with proteins bound to membranes and renders them fluorescent. © 2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol 1</b>: Synthesis of BODIPY-diazirine phosphatidylethanolamine (BDPE)</p><p><b>Basic Protocol 2</b>: Preparation of BDPE-containing liposomes</p><p><b>Basic Protocol 3</b>: Performing PLiMAP with a candidate protein</p><p><b>Basic Protocol 4</b>: Quantitation of liposome-binding properties of the candidate protein from analyzing in-gel fluorescence</p><p><b>Support Protocol</b>: Purification of GST-2×P4M domain of SidM protein</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.110","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38100434","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 6
Target Protein Identification on Photocatalyst-Functionalized Magnetic Affinity Beads 光催化功能化磁亲和珠靶蛋白的鉴定
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-06-30 DOI: 10.1002/cpps.108
Michihiko Tsushima, Shinichi Sato, Keita Nakane, Hiroyuki Nakamura

Although various affinity chromatography and photoaffinity labeling methods have been developed for target protein identification of bioactive molecules, it is often difficult to detect proteins that bind the ligand with weak transient affinity using these techniques. We have developed single electron transfer–mediated tyrosine labeling using ruthenium photocatalysts. Proximity labeling using 1-methyl-4-aryl-urazole (MAUra) labels proteins in close proximity to the photocatalyst with high efficiency and selectivity. Performing this labeling reaction on affinity beads makes it possible to label proteins that bind the ligand with weak transient affinity. In this article, novel protocols are described for target protein identification using photocatalyst proximity labeling on ruthenium photocatalyst-functionalized magnetic affinity beads. © 2020 Wiley Periodicals LLC.

Basic Protocol 1: Synthesis of ruthenium photocatalyst

Basic Protocol 2: Synthesis of azide- or desthiobiotin-conjugated labeling reagents

Basic Protocol 3: Preparation of photocatalyst and ligand-functionalized affinity beads

Basic Protocol 4: Target protein labeling in cell lysate

Basic Protocol 5: Enrichment of labeled proteins with MAUra-DTB for LC-MS/MS analysis

Basic Protocol 6: 2D-DIGE analysis of fluorescence-labeled proteins

尽管各种亲和层析和光亲和标记方法已经被开发出来用于生物活性分子的靶蛋白鉴定,但使用这些技术通常很难检测到与配体结合的具有弱瞬时亲和力的蛋白质。我们利用钌光催化剂开发了单电子转移介导的酪氨酸标记。利用1-甲基-4-芳基-乌拉唑(MAUra)对光催化剂附近的蛋白质进行近距离标记,具有高效率和选择性。在亲和珠上进行这种标记反应使得标记与配体结合的具有弱瞬时亲和力的蛋白质成为可能。在这篇文章中,描述了利用光催化剂接近标记钌光催化剂功能化磁亲和珠靶蛋白鉴定的新方案。©2020 Wiley期刊有限责任公司基本方案1:钌光催化剂的合成基本方案2:叠氮化物或去硫代生物素共轭标记试剂的合成基本方案3:光催化剂和配体功能化亲和珠的制备基本方案4:细胞裂解中的靶蛋白标记基本方案5:用MAUra-DTB富集标记蛋白用于LC-MS/MS分析基本方案6:荧光标记蛋白的2D-DIGE分析
{"title":"Target Protein Identification on Photocatalyst-Functionalized Magnetic Affinity Beads","authors":"Michihiko Tsushima,&nbsp;Shinichi Sato,&nbsp;Keita Nakane,&nbsp;Hiroyuki Nakamura","doi":"10.1002/cpps.108","DOIUrl":"10.1002/cpps.108","url":null,"abstract":"<p>Although various affinity chromatography and photoaffinity labeling methods have been developed for target protein identification of bioactive molecules, it is often difficult to detect proteins that bind the ligand with weak transient affinity using these techniques. We have developed single electron transfer–mediated tyrosine labeling using ruthenium photocatalysts. Proximity labeling using 1-methyl-4-aryl-urazole (MAUra) labels proteins in close proximity to the photocatalyst with high efficiency and selectivity. Performing this labeling reaction on affinity beads makes it possible to label proteins that bind the ligand with weak transient affinity. In this article, novel protocols are described for target protein identification using photocatalyst proximity labeling on ruthenium photocatalyst-functionalized magnetic affinity beads. © 2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol 1</b>: Synthesis of ruthenium photocatalyst</p><p><b>Basic Protocol 2</b>: Synthesis of azide- or desthiobiotin-conjugated labeling reagents</p><p><b>Basic Protocol 3</b>: Preparation of photocatalyst and ligand-functionalized affinity beads</p><p><b>Basic Protocol 4</b>: Target protein labeling in cell lysate</p><p><b>Basic Protocol 5</b>: Enrichment of labeled proteins with MAUra-DTB for LC-MS/MS analysis</p><p><b>Basic Protocol 6</b>: 2D-DIGE analysis of fluorescence-labeled proteins</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.108","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38099869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Purification of Ciliary Tubulin from Chlamydomonas reinhardtii 莱茵衣藻纤毛小管蛋白的纯化
Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-06-22 DOI: 10.1002/cpps.107
Ron Orbach, Jonathon Howard

Cilia and flagella play essential roles in environmental sensing, cell locomotion, and development. These organelles possess a central microtubule–based structure known as the axoneme, which serves as a scaffold and is crucial for the function of cilia. Despite their key roles, the biochemical and biophysical properties of the ciliary proteins are poorly understood. To address this issue, we have developed a novel method to purify functional tubulins from different parts of the axoneme, namely the central pair and B-tubule. We use the biflagellate green alga Chlamydomonas reinhardtii, a model organism for studying cilia due to the conserved structure of this organelle, availability of genetic tools and a large collection of mutant strains. Our method yields highly purified functional axonemal tubulins in sufficient quantities to be used for in vitro biochemical and biophysical studies, such as microtubule dynamic assays. It takes 7 to 8 days to grow enough cells; the isolation of the flagella and the purification of the axonemal tubulins require an additional two full days.© 2020 Wiley Periodicals LLC.

Basic Protocol 1: Growth and harvest of large volume of cell culture

Support Protocol: Assembly of homemade concentration apparatus

Basic Protocol 2: Isolation of flagella

Basic Protocol 3: Tubulin extraction and purification

纤毛和鞭毛在环境感知、细胞运动和发育中起着重要作用。这些细胞器具有一个以微管为基础的中心结构,称为轴素,它作为一个支架,对纤毛的功能至关重要。尽管纤毛蛋白具有重要的作用,但人们对其生物化学和生物物理特性的了解甚少。为了解决这个问题,我们开发了一种新的方法来从轴素的不同部分纯化功能性小管,即中央对和b小管。我们使用双鞭毛藻莱茵衣藻作为研究纤毛的模式生物,这是由于这种细胞器的保守结构,遗传工具的可用性以及大量突变菌株的收集。我们的方法产生高纯度的功能性轴突小管,其数量足以用于体外生化和生物物理研究,如微管动力学分析。培养足够的细胞需要7到8天;鞭毛的分离和轴突小管的纯化还需要整整两天的时间。©2020 Wiley期刊有限责任公司基本方案1:大容量细胞培养的生长和收获支持方案:自制浓度仪的组装基本方案2:鞭毛的分离基本方案3:微管蛋白的提取和纯化
{"title":"Purification of Ciliary Tubulin from Chlamydomonas reinhardtii","authors":"Ron Orbach,&nbsp;Jonathon Howard","doi":"10.1002/cpps.107","DOIUrl":"10.1002/cpps.107","url":null,"abstract":"<p>Cilia and flagella play essential roles in environmental sensing, cell locomotion, and development. These organelles possess a central microtubule–based structure known as the axoneme, which serves as a scaffold and is crucial for the function of cilia. Despite their key roles, the biochemical and biophysical properties of the ciliary proteins are poorly understood. To address this issue, we have developed a novel method to purify functional tubulins from different parts of the axoneme, namely the central pair and B-tubule. We use the biflagellate green alga <i>Chlamydomonas reinhardtii</i>, a model organism for studying cilia due to the conserved structure of this organelle, availability of genetic tools and a large collection of mutant strains. Our method yields highly purified functional axonemal tubulins in sufficient quantities to be used for <i>in vitro</i> biochemical and biophysical studies, such as microtubule dynamic assays. It takes 7 to 8 days to grow enough cells; the isolation of the flagella and the purification of the axonemal tubulins require an additional two full days.© 2020 Wiley Periodicals LLC.</p><p><b>Basic Protocol 1</b>: Growth and harvest of large volume of cell culture</p><p><b>Support Protocol</b>: Assembly of homemade concentration apparatus</p><p><b>Basic Protocol 2</b>: Isolation of flagella</p><p><b>Basic Protocol 3</b>: Tubulin extraction and purification</p>","PeriodicalId":10866,"journal":{"name":"Current Protocols in Protein Science","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-06-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpps.107","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38070193","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
期刊
Current Protocols in Protein Science
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1