首页 > 最新文献

Current protocols in chemical biology最新文献

英文 中文
Metabolic Labeling for the Visualization and Identification of Potentially O-GlcNAc-Modified Proteins 代谢标记用于潜在的o - glcnac修饰蛋白的可视化和鉴定
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-04-14 DOI: 10.1002/cpch.81
Nichole J. Pedowitz, Balyn W. Zaro, Matthew R. Pratt

O-GlcNAcylation is a posttranslational modification involving the addition of the single monosaccharide N-acetylglucosamine (GlcNAc) onto serine and threonine residues of intracellular proteins. Though O-GlcNAc is found on ∼1000 proteins in mammals, its specific function on individual substrates remains largely a mystery. To overcome this shortcoming, work has been put toward developing metabolic chemical reporters (MCRs) to label O-GlcNAcylated proteins for subsequent biochemical analysis. Typically, these MCRs are GlcNAc or GalNAc analogs functionalized with azide or alkyne handles. These unnatural sugar moieties can be metabolically incorporated directly on to protein substrates. The protocols outlined in this article describe how to use MCRs as tools for visualizing and identifying potentially O-GlcNAc modified proteins via in-gel fluorescence, Western blotting, and mass spectrometry. Taken together, MCR labeling provides a powerful tool to discover where and when substrates are O-GlcNAc modified. © 2020 by John Wiley & Sons, Inc.

Basic Protocol 1: Treatment of cells and CuAAC

Basic Protocol 2: In-gel fluorescence of labeled cell lysates (1 mg scale)

Basic Protocol 3: Enrichment of labeled proteins, trypsinolysis, and collection of peptides for proteomics

Basic Protocol 4: Proteomic identification of labeled proteins

o - glcn酰化是一种翻译后修饰,涉及将单糖n -乙酰氨基葡萄糖(GlcNAc)添加到细胞内蛋白质的丝氨酸和苏氨酸残基上。尽管O-GlcNAc存在于哺乳动物中约1000种蛋白质中,但其在单个底物上的特定功能在很大程度上仍是一个谜。为了克服这一缺点,研究人员致力于开发代谢化学报告器(mcr)来标记o - glcn酰化蛋白,以便进行后续的生化分析。通常,这些mcr是GlcNAc或GalNAc类似物,具有叠氮化物或炔烃手柄。这些非天然的糖部分可以通过代谢直接结合到蛋白质底物上。本文中概述的方案描述了如何使用mcr作为工具,通过凝胶内荧光,Western blotting和质谱法来可视化和鉴定潜在的O-GlcNAc修饰蛋白。综上所述,MCR标记提供了一个强大的工具来发现底物在何时何地被O-GlcNAc修饰。©2020 by John Wiley &基本方案1:细胞和cuaac处理基本方案2:标记细胞裂解物的凝胶内荧光(1mg级)基本方案3:标记蛋白的富集,胰蛋白酶解,和蛋白质组学肽的收集基本方案4:标记蛋白的蛋白质组学鉴定
{"title":"Metabolic Labeling for the Visualization and Identification of Potentially O-GlcNAc-Modified Proteins","authors":"Nichole J. Pedowitz,&nbsp;Balyn W. Zaro,&nbsp;Matthew R. Pratt","doi":"10.1002/cpch.81","DOIUrl":"10.1002/cpch.81","url":null,"abstract":"<p>O-GlcNAcylation is a posttranslational modification involving the addition of the single monosaccharide <i>N-</i>acetylglucosamine (GlcNAc) onto serine and threonine residues of intracellular proteins. Though O-GlcNAc is found on ∼1000 proteins in mammals, its specific function on individual substrates remains largely a mystery. To overcome this shortcoming, work has been put toward developing metabolic chemical reporters (MCRs) to label O-GlcNAcylated proteins for subsequent biochemical analysis. Typically, these MCRs are GlcNAc or GalNAc analogs functionalized with azide or alkyne handles. These unnatural sugar moieties can be metabolically incorporated directly on to protein substrates. The protocols outlined in this article describe how to use MCRs as tools for visualizing and identifying potentially O-GlcNAc modified proteins via in-gel fluorescence, Western blotting, and mass spectrometry. Taken together, MCR labeling provides a powerful tool to discover where and when substrates are O-GlcNAc modified. © 2020 by John Wiley &amp; Sons, Inc.</p><p><b>Basic Protocol 1</b>: Treatment of cells and CuAAC</p><p><b>Basic Protocol 2</b>: In-gel fluorescence of labeled cell lysates (1 mg scale)</p><p><b>Basic Protocol 3</b>: Enrichment of labeled proteins, trypsinolysis, and collection of peptides for proteomics</p><p><b>Basic Protocol 4</b>: Proteomic identification of labeled proteins</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"12 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2020-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.81","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37833901","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: 9
Detection of Small-Molecule Aggregation with High-Throughput Microplate Biophysical Methods 高通量微孔板生物物理方法检测小分子聚集
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-03-09 DOI: 10.1002/cpch.78
Samantha J. Allen, Corey M. Dower, Annie X. Liu, Kevin J. Lumb

Small-molecule drug discovery can be hindered by the formation of aggregates that act as non-selective inhibitors of drug targets. Such aggregates appear as false positives in high-throughput screening campaigns and can bedevil structure-activity relationships during compound optimization. Protocols are described for resonant waveguide grating (RWG) and dynamic light scattering (DLS) as microplate-based high-throughput approaches to identify compound aggregation. Resonant waveguide grating and dynamic light scattering give equivalent results for the compound test set, as assessed with Bland-Altman analysis. © 2019 The Authors.

Basic Protocol 1: Resonant waveguide grating (RWG) in 384-well or 1536-well plate format to detect compound aggregation

Basic Protocol 2: Dynamic light scattering (DLS) in 384-well plate format to detect compound aggregation

小分子药物的发现可能会被聚集物的形成所阻碍,这些聚集物作为药物靶点的非选择性抑制剂。这种聚合体在高通量筛选活动中出现假阳性,并可能在化合物优化过程中困扰结构-活性关系。描述了共振波导光栅(RWG)和动态光散射(DLS)作为基于微板的高通量方法来识别化合物聚集的协议。谐振波导光栅和动态光散射对复合测试集给出了等效的结果,用Bland-Altman分析进行了评估。©2019作者。基本协议1:384孔或1536孔板格式的谐振波导光栅(RWG)检测化合物聚集基本协议2:384孔板格式的动态光散射(DLS)检测化合物聚集
{"title":"Detection of Small-Molecule Aggregation with High-Throughput Microplate Biophysical Methods","authors":"Samantha J. Allen,&nbsp;Corey M. Dower,&nbsp;Annie X. Liu,&nbsp;Kevin J. Lumb","doi":"10.1002/cpch.78","DOIUrl":"10.1002/cpch.78","url":null,"abstract":"<p>Small-molecule drug discovery can be hindered by the formation of aggregates that act as non-selective inhibitors of drug targets. Such aggregates appear as false positives in high-throughput screening campaigns and can bedevil structure-activity relationships during compound optimization. Protocols are described for resonant waveguide grating (RWG) and dynamic light scattering (DLS) as microplate-based high-throughput approaches to identify compound aggregation. Resonant waveguide grating and dynamic light scattering give equivalent results for the compound test set, as assessed with Bland-Altman analysis. © 2019 The Authors.</p><p><b>Basic Protocol 1</b>: Resonant waveguide grating (RWG) in 384-well or 1536-well plate format to detect compound aggregation</p><p><b>Basic Protocol 2</b>: Dynamic light scattering (DLS) in 384-well plate format to detect compound aggregation</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"12 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2020-03-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.78","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37719503","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: 3
Ratiometric Single-Molecule FRET Measurements to Probe Conformational Subpopulations of Intrinsically Disordered Proteins. 比率单分子FRET测量探针构象亚群的内在无序的蛋白质。
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-03-01 DOI: 10.1002/cpch.80
Irem Nasir, Emily P Bentley, Ashok A Deniz

Over the past few decades, numerous examples have demonstrated that intrinsic disorder in proteins lies at the heart of many vital processes, including transcriptional regulation, stress response, cellular signaling, and most recently protein liquid-liquid phase separation. The so-called intrinsically disordered proteins (IDPs) involved in these processes have presented a challenge to the classic protein "structure-function paradigm," as their functions do not necessarily involve well-defined structures. Understanding the mechanisms of IDP function is likewise challenging because traditional structure determination methods often fail with such proteins or provide little information about the diverse array of structures that can be related to different functions of a single IDP. Single-molecule fluorescence methods can overcome this ensemble-average masking, allowing the resolution of subpopulations and dynamics and thus providing invaluable insights into IDPs and their function. In this protocol, we describe a ratiometric single-molecule Förster resonance energy transfer (smFRET) routine that permits the investigation of IDP conformational subpopulations and dynamics. We note that this is a basic protocol, and we provide brief information and references for more complex analysis schemes available for in-depth characterization. This protocol covers optical setup preparation and protein handling and provides insights into experimental design and outcomes, together with background information about theory and a brief discussion of troubleshooting. © 2020 by John Wiley & Sons, Inc. Basic Protocol: Ratiometric smFRET detection and analysis of IDPs Support Protocol 1: Fluorophore labeling of a protein through maleimide chemistry Support Protocol 2: Sample chamber preparation Support Protocol 3: Determination of direct excitation of acceptor by donor excitation and leakage of donor emission to acceptor emission channel.

在过去的几十年里,大量的例子表明,蛋白质的内在紊乱是许多重要过程的核心,包括转录调节、应激反应、细胞信号传导,以及最近的蛋白质液-液相分离。参与这些过程的所谓内在无序蛋白(IDPs)对经典的蛋白质“结构-功能范式”提出了挑战,因为它们的功能不一定涉及定义良好的结构。了解IDP功能的机制同样具有挑战性,因为传统的结构确定方法通常无法确定此类蛋白质,或者无法提供与单个IDP不同功能相关的多种结构的信息。单分子荧光方法可以克服这种集合平均掩蔽,允许亚种群和动态的分辨率,从而提供宝贵的见解国内流离失所者和他们的功能。在本协议中,我们描述了一个比例单分子Förster共振能量转移(smFRET)程序,允许调查IDP构象亚群和动力学。我们注意到这是一个基本的协议,我们为更复杂的分析方案提供了简要的信息和参考,可用于深入表征。该协议涵盖光学装置制备和蛋白质处理,并提供对实验设计和结果的见解,以及有关理论的背景信息和故障排除的简要讨论。©2020 by John Wiley & Sons, Inc。基本方案:IDPs的比率smFRET检测和分析支持方案1:通过马来酰亚胺化学对蛋白质进行荧光团标记支持方案2:样品室制备支持方案3:通过供体激发和供体发射向受体发射通道泄漏来确定受体的直接激发。
{"title":"Ratiometric Single-Molecule FRET Measurements to Probe Conformational Subpopulations of Intrinsically Disordered Proteins.","authors":"Irem Nasir,&nbsp;Emily P Bentley,&nbsp;Ashok A Deniz","doi":"10.1002/cpch.80","DOIUrl":"https://doi.org/10.1002/cpch.80","url":null,"abstract":"<p><p>Over the past few decades, numerous examples have demonstrated that intrinsic disorder in proteins lies at the heart of many vital processes, including transcriptional regulation, stress response, cellular signaling, and most recently protein liquid-liquid phase separation. The so-called intrinsically disordered proteins (IDPs) involved in these processes have presented a challenge to the classic protein \"structure-function paradigm,\" as their functions do not necessarily involve well-defined structures. Understanding the mechanisms of IDP function is likewise challenging because traditional structure determination methods often fail with such proteins or provide little information about the diverse array of structures that can be related to different functions of a single IDP. Single-molecule fluorescence methods can overcome this ensemble-average masking, allowing the resolution of subpopulations and dynamics and thus providing invaluable insights into IDPs and their function. In this protocol, we describe a ratiometric single-molecule Förster resonance energy transfer (smFRET) routine that permits the investigation of IDP conformational subpopulations and dynamics. We note that this is a basic protocol, and we provide brief information and references for more complex analysis schemes available for in-depth characterization. This protocol covers optical setup preparation and protein handling and provides insights into experimental design and outcomes, together with background information about theory and a brief discussion of troubleshooting. © 2020 by John Wiley & Sons, Inc. Basic Protocol: Ratiometric smFRET detection and analysis of IDPs Support Protocol 1: Fluorophore labeling of a protein through maleimide chemistry Support Protocol 2: Sample chamber preparation Support Protocol 3: Determination of direct excitation of acceptor by donor excitation and leakage of donor emission to acceptor emission channel.</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"12 1","pages":"e80"},"PeriodicalIF":0.0,"publicationDate":"2020-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.80","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37728209","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: 5
N-Terminal Modification of Proteins with Subtiligase Specificity Variants 枯草酶特异性变异蛋白的n端修饰
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2020-02-19 DOI: 10.1002/cpch.79
Amy M. Weeks, James A. Wells

Subtiligase is a powerful enzymatic tool for N-terminal modification of proteins and peptides. In a typical subtiligase-catalyzed N-terminal modification reaction, a peptide ester donor substrate is ligated onto the unblocked N terminus of a protein, resulting in the exchange of the ester bond in the donor substrate for an amide bond between the donor substrate and protein N terminus. Using this strategy, new chemical probes and payloads, such as fluorophores, affinity handles, cytotoxic drugs, and reactive functional groups, can be introduced site-specifically into proteins. While the efficiency of this reaction depends on the sequences to be ligated, a panel of mutants was recently developed that expands the scope of substrate sequences that are suitable for subtiligase modification. This article outlines the steps for applying subtiligase or specificity variants for both site-specific bioconjugation of purified proteins and for global modification of cellular N termini to enable their sequencing by tandem mass spectrometry. © 2020 by John Wiley & Sons, Inc.

Basic Protocol 1: Subtiligase-catalyzed site-specific protein bioconjugation

Support Protocol 1: Expression and purification of subtiligase-His6

Support Protocol 2: Subtiligase substrate synthesis

Basic Protocol 2: Subtiligase N terminomics using a cocktail of subtiligase specificity mutants

枯草酶是一种功能强大的酶工具,用于蛋白质和肽的n端修饰。在典型的枯草酶催化的N端修饰反应中,肽酯供体底物连接到蛋白质的未阻断N端,导致供体底物中的酯键交换为供体底物与蛋白质N端之间的酰胺键。利用这一策略,新的化学探针和有效载荷,如荧光团、亲和柄、细胞毒性药物和反应性官能团,可以被特异地引入蛋白质中。虽然这种反应的效率取决于要连接的序列,但最近开发的一组突变体扩大了适合枯草酶修饰的底物序列的范围。本文概述了应用枯草化酶或特异性变体的步骤,用于纯化蛋白的位点特异性生物偶联和细胞N末端的全局修饰,以便通过串联质谱法进行测序。©2020 by John Wiley &基本方案1:枯草草酶催化的位点特异性蛋白生物偶联;支持方案1:枯草草酶his6的表达和纯化;支持方案2:枯草草酶底物合成;基本方案2:使用枯草草酶特异性突变体鸡尾酒的枯草草酶N终端组学
{"title":"N-Terminal Modification of Proteins with Subtiligase Specificity Variants","authors":"Amy M. Weeks,&nbsp;James A. Wells","doi":"10.1002/cpch.79","DOIUrl":"10.1002/cpch.79","url":null,"abstract":"<p>Subtiligase is a powerful enzymatic tool for N-terminal modification of proteins and peptides. In a typical subtiligase-catalyzed N-terminal modification reaction, a peptide ester donor substrate is ligated onto the unblocked N terminus of a protein, resulting in the exchange of the ester bond in the donor substrate for an amide bond between the donor substrate and protein N terminus. Using this strategy, new chemical probes and payloads, such as fluorophores, affinity handles, cytotoxic drugs, and reactive functional groups, can be introduced site-specifically into proteins. While the efficiency of this reaction depends on the sequences to be ligated, a panel of mutants was recently developed that expands the scope of substrate sequences that are suitable for subtiligase modification. This article outlines the steps for applying subtiligase or specificity variants for both site-specific bioconjugation of purified proteins and for global modification of cellular N termini to enable their sequencing by tandem mass spectrometry. © 2020 by John Wiley &amp; Sons, Inc.</p><p><b>Basic Protocol 1</b>: Subtiligase-catalyzed site-specific protein bioconjugation</p><p><b>Support Protocol 1</b>: Expression and purification of subtiligase-His<sub>6</sub></p><p><b>Support Protocol 2</b>: Subtiligase substrate synthesis</p><p><b>Basic Protocol 2</b>: Subtiligase N terminomics using a cocktail of subtiligase specificity mutants</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"12 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2020-02-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.79","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37657906","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: 10
Issue Information TOC 发布信息TOC
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2019-12-19 DOI: 10.1002/cpch.57

Cover: In DeMeester et al. (https://doi.org/10.1002/cpch.74), PG biosynthesis begins with formation of UDP-NAM through MurA/B and UDP-NAG. Recycling enzymes AmgK/MurU provide another route to synthesize UDP-NAM with NAM as the building block. UDP-NAM is converted into Park's nucleotide through enzymes MurC-F. MraY links Park's nucleotide to the cell membrane, where MurG then glycosylates this Lipid I fragment to form Lipid II. MurJ transports Lipid II into the periplasmic space, where transglycosylases (TGase) and transpeptidases (TPase) further crosslink these molecules to form mature PG. NAM probes (blue) with bioorthogonal functionality at the 2-N position (X) or 3-lactic acid position (Y) are metabolically incorporated into PG through both recycling and biosynthetic machineries.

封面:在DeMeester等人(https://doi.org/10.1002/cpch.74)中,PG的生物合成始于通过MurA/B和UDP-NAG形成UDP-NAM。再循环酶AmgK/MurU为以NAM为主体合成UDP-NAM提供了另一种途径。UDP-NAM通过酶MurC-F转化为Park的核苷酸。MraY将Park的核苷酸连接到细胞膜上,然后MurG将脂质I片段糖基化形成脂质II。MurJ将脂质II转运到质周空间,在那里,转糖基酶(TGase)和转肽酶(TPase)进一步将这些分子交联,形成成熟的PG。在2-N位置(X)或3-乳酸位置(Y)具有生物正交功能的NAM探针(蓝色)通过循环和生物合成机制代谢纳入PG。
{"title":"Issue Information TOC","authors":"","doi":"10.1002/cpch.57","DOIUrl":"10.1002/cpch.57","url":null,"abstract":"<p><b>Cover</b>: In DeMeester et al. (https://doi.org/10.1002/cpch.74), PG biosynthesis begins with formation of UDP-NAM through MurA/B and UDP-NAG. Recycling enzymes AmgK/MurU provide another route to synthesize UDP-NAM with NAM as the building block. UDP-NAM is converted into Park's nucleotide through enzymes MurC-F. MraY links Park's nucleotide to the cell membrane, where MurG then glycosylates this Lipid I fragment to form Lipid II. MurJ transports Lipid II into the periplasmic space, where transglycosylases (TGase) and transpeptidases (TPase) further crosslink these molecules to form mature PG. NAM probes (blue) with bioorthogonal functionality at the 2-<i>N</i> position (X) or 3-lactic acid position (Y) are metabolically incorporated into PG through both recycling and biosynthetic machineries.\u0000\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"11 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.57","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42689304","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: 0
Proteomic Detection and Validation of Translated Small Open Reading Frames 翻译小开放阅读框的蛋白质组学检测与验证
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2019-11-21 DOI: 10.1002/cpch.77
Alexandra Khitun, Sarah A. Slavoff

Small open reading frames (smORFs) encode previously unannotated polypeptides or short proteins that regulate translation in cis (eukaryotes) and/or are independently functional (prokaryotes and eukaryotes). Ongoing efforts for complete annotation and functional characterization of smORF-encoded proteins have yielded novel regulators and therapeutic targets. However, because they are excluded from protein databases, initiate at non-AUG start codons, and produce few unique tryptic peptides, unannotated small proteins cannot be detected with standard proteomic methods. Here,, we outline a procedure for mass spectrometry-based detection of translated smORFs in cultured human cells from protein extraction, digestion, and LC-MS/MS, to database preparation and data analysis. Following proteomic detection, translation from a unique smORF may be validated via siRNA-based silencing or overexpression and epitope tagging. This is necessary to unambiguously assign a peptide to a smORF within a specific transcript isoform or genomic locus. Provided that sufficient starting material is available, this workflow can be applied to any cell type/organism and adjusted to study specific (patho)physiological contexts including, but not limited to, development, stress, and disease. © 2019 by John Wiley & Sons, Inc.

Basic Protocol 1: Protein extraction, size selection, and trypsin digestion

Alternate Protocol 1: In-solution C8 column size selection

Support Protocol 1: Chloroform/methanol precipitation

Support Protocol 2: Reduction, alkylation, and in-solution protease digestion

Support Protocol 3: Peptide de-salting

Basic Protocol 2: Two-dimensional LC-MS/MS with ERLIC fractionation

Basic Protocol 3: Transcriptomic database construction

Alternate Protocol 2: Transcriptomics database generation with gffread

Basic Protocol 4: Non-annotated peptide identification from LC-MS/MS data

Basic Protocol 5: Validation using isotopically labeled synthetic peptide standards and siRNA

Basic Protocol 6: Transcript validation using transient overexpression

小的开放阅读框(smorf)编码以前未注释的多肽或短蛋白,这些蛋白在顺式(真核生物)和/或独立功能(原核生物和真核生物)中调节翻译。对smorf编码蛋白的完整注释和功能表征的持续努力已经产生了新的调节因子和治疗靶点。然而,由于它们被排除在蛋白质数据库之外,在非aug启动密码子处启动,并且产生很少独特的色氨酸,因此无法用标准的蛋白质组学方法检测到未注释的小蛋白。在这里,我们概述了一种基于质谱的检测方法,从蛋白质提取、消化、LC-MS/MS到数据库准备和数据分析,在培养的人类细胞中检测翻译的smorf。在进行蛋白质组学检测后,可以通过基于sirna的沉默或过表达和表位标记来验证独特smORF的翻译。这对于明确地将肽分配到特定转录异构体或基因组位点内的smORF是必要的。只要有足够的起始材料,该工作流程可以应用于任何细胞类型/生物体,并调整以研究特定(病理)生理环境,包括但不限于发育、压力和疾病。©2019 by John Wiley &基本方案1:蛋白质提取,大小选择,胰蛋白酶消化备用方案1:溶液中C8柱大小选择支持方案1:氯仿/甲醇沉淀支持方案2:还原,烷基化,和溶液中蛋白酶消化支持方案3:肽脱盐基本方案2:二维LC-MS/MS与ERLIC分离基本方案3:转录组数据库构建备用方案2:转录组学数据库的生成与gffreadBasic协议4:从LC-MS/MS中鉴定无注释的肽。数据库协议5:使用同位素标记的合成肽标准和sirnabbasic协议6:使用瞬时过表达进行转录验证
{"title":"Proteomic Detection and Validation of Translated Small Open Reading Frames","authors":"Alexandra Khitun,&nbsp;Sarah A. Slavoff","doi":"10.1002/cpch.77","DOIUrl":"10.1002/cpch.77","url":null,"abstract":"<p>Small open reading frames (smORFs) encode previously unannotated polypeptides or short proteins that regulate translation in <i>cis</i> (eukaryotes) and/or are independently functional (prokaryotes and eukaryotes). Ongoing efforts for complete annotation and functional characterization of smORF-encoded proteins have yielded novel regulators and therapeutic targets. However, because they are excluded from protein databases, initiate at non-AUG start codons, and produce few unique tryptic peptides, unannotated small proteins cannot be detected with standard proteomic methods. Here,, we outline a procedure for mass spectrometry-based detection of translated smORFs in cultured human cells from protein extraction, digestion, and LC-MS/MS, to database preparation and data analysis. Following proteomic detection, translation from a unique smORF may be validated via siRNA-based silencing or overexpression and epitope tagging. This is necessary to unambiguously assign a peptide to a smORF within a specific transcript isoform or genomic locus. Provided that sufficient starting material is available, this workflow can be applied to any cell type/organism and adjusted to study specific (patho)physiological contexts including, but not limited to, development, stress, and disease. © 2019 by John Wiley &amp; Sons, Inc.</p><p><b>Basic Protocol 1</b>: Protein extraction, size selection, and trypsin digestion</p><p><b>Alternate Protocol 1</b>: In-solution C8 column size selection</p><p><b>Support Protocol 1</b>: Chloroform/methanol precipitation</p><p><b>Support Protocol 2</b>: Reduction, alkylation, and in-solution protease digestion</p><p><b>Support Protocol 3</b>: Peptide de-salting</p><p><b>Basic Protocol 2</b>: Two-dimensional LC-MS/MS with ERLIC fractionation</p><p><b>Basic Protocol 3</b>: Transcriptomic database construction</p><p><b>Alternate Protocol 2</b>: Transcriptomics database generation with gffread</p><p><b>Basic Protocol 4</b>: Non-annotated peptide identification from LC-MS/MS data</p><p><b>Basic Protocol 5</b>: Validation using isotopically labeled synthetic peptide standards and siRNA</p><p><b>Basic Protocol 6</b>: Transcript validation using transient overexpression</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"11 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.77","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47088110","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: 19
Synthesis and Application of an Activity-Based Peptide-Peptoid Hybrid Probe for the Immunoproteasome 免疫蛋白酶体肽-类肽杂交探针的合成及应用
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2019-11-12 DOI: 10.1002/cpch.76
Breanna L. Zerfas, Darci J. Trader

The immunoproteasome (iCP), a specific isoform of the proteasome's catalytic particle, is becoming an important protein complex of interest in various diseases. However, there is still much left to be learned about its activity in cells and how this can be altered by various endogenous conditions or with treatment with small molecules. Current strategies to investigate the iCP lack in their ability to be used in live, intact cells, limiting them to use in endpoint experiments. The iCP-selective probe presented here has been shown to be compatible with various live-cell assays, including monitoring iCP activity kinetically in a plate reader–based assay and observing single cells with confocal microscopy. A well-studied iCP-selective inhibitor, ONX-0914, has also been demonstrated to decrease the fluorescence signal of the iCP probe in both of these assays, showing its potential function in investigating small-molecule modulators of the iCP. © 2019 by John Wiley & Sons, Inc.

Basic Protocol 1: Synthesis of an immunoproteasome-selective peptide-peptoid hybrid probe

Basic Protocol 2: Expression of the immunoproteasome in A549 cells

Basic Protocol 3: Using the immunoproteasome probe to monitor activity in live cells with a fluorescence plate reader

Basic Protocol 4: Using the immunoproteasome probe to monitor activity in live cells with confocal microscopy

免疫蛋白酶体(iCP)是蛋白酶体催化颗粒的特异性异构体,正在成为多种疾病中重要的蛋白质复合物。然而,关于其在细胞中的活性,以及如何通过各种内源性条件或小分子治疗来改变这种活性,仍有许多有待了解的地方。目前研究iCP的策略缺乏在活的、完整的细胞中使用的能力,限制了它们在终点实验中的使用。本文介绍的iCP选择性探针已被证明与各种活细胞测定相兼容,包括在基于平板阅读器的测定中动态监测iCP活性,以及用共聚焦显微镜观察单细胞。一种经过充分研究的iCP选择性抑制剂ONX-0914也被证明可以在这两种检测中降低iCP探针的荧光信号,显示其在研究iCP的小分子调节剂方面的潜在功能。©2019 by John Wiley &基本方案1:合成一种免疫蛋白酶体-选择性肽-类肽杂交探针基本方案2:在A549细胞中表达免疫蛋白酶体基本方案3:使用荧光板阅读器使用免疫蛋白酶体探针监测活细胞活性基本方案4:使用共聚焦显微镜使用免疫蛋白酶体探针监测活细胞活性
{"title":"Synthesis and Application of an Activity-Based Peptide-Peptoid Hybrid Probe for the Immunoproteasome","authors":"Breanna L. Zerfas,&nbsp;Darci J. Trader","doi":"10.1002/cpch.76","DOIUrl":"10.1002/cpch.76","url":null,"abstract":"<p>The immunoproteasome (iCP), a specific isoform of the proteasome's catalytic particle, is becoming an important protein complex of interest in various diseases. However, there is still much left to be learned about its activity in cells and how this can be altered by various endogenous conditions or with treatment with small molecules. Current strategies to investigate the iCP lack in their ability to be used in live, intact cells, limiting them to use in endpoint experiments. The iCP-selective probe presented here has been shown to be compatible with various live-cell assays, including monitoring iCP activity kinetically in a plate reader–based assay and observing single cells with confocal microscopy. A well-studied iCP-selective inhibitor, ONX-0914, has also been demonstrated to decrease the fluorescence signal of the iCP probe in both of these assays, showing its potential function in investigating small-molecule modulators of the iCP. © 2019 by John Wiley &amp; Sons, Inc.</p><p><b>Basic Protocol 1</b>: Synthesis of an immunoproteasome-selective peptide-peptoid hybrid probe</p><p><b>Basic Protocol 2</b>: Expression of the immunoproteasome in A549 cells</p><p><b>Basic Protocol 3</b>: Using the immunoproteasome probe to monitor activity in live cells with a fluorescence plate reader</p><p><b>Basic Protocol 4</b>: Using the immunoproteasome probe to monitor activity in live cells with confocal microscopy</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"11 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.76","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37477667","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: 2
Metabolic Incorporation of N-Acetyl Muramic Acid Probes into Bacterial Peptidoglycan n -乙酰基陶瓷酸探针在细菌肽聚糖中的代谢结合
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2019-10-10 DOI: 10.1002/cpch.74
Kristen E. DeMeester, Hai Liang, Junhui Zhou, Kimberly A. Wodzanowski, Benjamin L. Prather, Cintia C. Santiago, Catherine L. Grimes

Bacterial cells utilize small carbohydrate building blocks to construct peptidoglycan (PG), a highly conserved mesh-like polymer that serves as a protective coat for the cell. PG production has long been a target for antibiotics, and its breakdown is a source for human immune recognition. A key component of bacterial PG, N-acetyl muramic acid (NAM), is a vital element in many synthetically derived immunostimulatory compounds. However, the exact molecular details of these structures and how they are generated remain unknown due to a lack of chemical probes surrounding the NAM core. A robust synthetic strategy to generate bioorthogonally tagged NAM carbohydrate units is implemented. These molecules serve as precursors for PG biosynthesis and recycling. Escherichia coli cells are metabolically engineered to incorporate the bioorthogonal NAM probes into their PG network. The probes are subsequently modified using copper-catalyzed azide-alkyne cycloaddition to install fluorophores directly into the bacterial PG, as confirmed by super-resolution microscopy and high-resolution mass spectrometry. Here, synthetic notes for key elements of this process to generate the sugar probes as well as streamlined user-friendly metabolic labeling strategies for both microbiology and immunological applications are described. © 2019 by John Wiley & Sons, Inc.

Basic Protocol 1: Synthesis of peracetylated 2-azido glucosamine

Basic Protocol 2: Synthesis of 2-azido and 2-alkyne NAM

Basic Protocol 3: Synthesis of 3-azido NAM methyl ester

Basic Protocol 4: Incorporation of NAM probes into bacterial peptidoglycan

Basic Protocol 5: Confirmation of bacterial cell wall remodeling by mass spectrometry

细菌细胞利用小碳水化合物构建肽聚糖(PG),这是一种高度保守的网状聚合物,可作为细胞的保护层。PG的生产长期以来一直是抗生素的目标,其分解是人类免疫识别的来源。细菌PG的关键成分n -乙酰氨基乙酸(NAM)是许多合成衍生的免疫刺激化合物的重要元素。然而,由于缺乏围绕不结盟运动核的化学探针,这些结构的确切分子细节以及它们是如何产生的仍然未知。一个强大的合成策略,以产生生物正交标记NAM碳水化合物单位实施。这些分子作为PG生物合成和再循环的前体。大肠杆菌细胞通过代谢工程将生物正交NAM探针纳入其PG网络。随后使用铜催化叠氮化物-炔环加成修饰探针,将荧光团直接安装到细菌PG中,超分辨率显微镜和高分辨率质谱法证实了这一点。本文描述了该过程中生成糖探针的关键要素的合成笔记,以及用于微生物学和免疫学应用的简化用户友好的代谢标记策略。©2019 by John Wiley &基础方案1:合成过乙酰化2-叠氮基氨基葡萄糖;基础方案2:合成2-叠氮基氨基葡萄糖;基础方案3:合成3-叠氮基氨基甲酯;基础方案4:将NAM探针掺入细菌肽聚糖;基础方案5:用质谱法确认细菌细胞壁重塑
{"title":"Metabolic Incorporation of N-Acetyl Muramic Acid Probes into Bacterial Peptidoglycan","authors":"Kristen E. DeMeester,&nbsp;Hai Liang,&nbsp;Junhui Zhou,&nbsp;Kimberly A. Wodzanowski,&nbsp;Benjamin L. Prather,&nbsp;Cintia C. Santiago,&nbsp;Catherine L. Grimes","doi":"10.1002/cpch.74","DOIUrl":"10.1002/cpch.74","url":null,"abstract":"<p>Bacterial cells utilize small carbohydrate building blocks to construct peptidoglycan (PG), a highly conserved mesh-like polymer that serves as a protective coat for the cell. PG production has long been a target for antibiotics, and its breakdown is a source for human immune recognition. A key component of bacterial PG, <i>N</i>-acetyl muramic acid (NAM), is a vital element in many synthetically derived immunostimulatory compounds. However, the exact molecular details of these structures and how they are generated remain unknown due to a lack of chemical probes surrounding the NAM core. A robust synthetic strategy to generate bioorthogonally tagged NAM carbohydrate units is implemented. These molecules serve as precursors for PG biosynthesis and recycling. <i>Escherichia coli</i> cells are metabolically engineered to incorporate the bioorthogonal NAM probes into their PG network. The probes are subsequently modified using copper-catalyzed azide-alkyne cycloaddition to install fluorophores directly into the bacterial PG, as confirmed by super-resolution microscopy and high-resolution mass spectrometry. Here, synthetic notes for key elements of this process to generate the sugar probes as well as streamlined user-friendly metabolic labeling strategies for both microbiology and immunological applications are described. © 2019 by John Wiley &amp; Sons, Inc.</p><p><b>Basic Protocol 1</b>: Synthesis of peracetylated 2-azido glucosamine</p><p><b>Basic Protocol 2</b>: Synthesis of 2-azido and 2-alkyne NAM</p><p><b>Basic Protocol 3</b>: Synthesis of 3-azido NAM methyl ester</p><p><b>Basic Protocol 4</b>: Incorporation of NAM probes into bacterial peptidoglycan</p><p><b>Basic Protocol 5</b>: Confirmation of bacterial cell wall remodeling by mass spectrometry</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"11 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.74","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44889692","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: 10
Small Molecule Interactome Mapping by Photo-Affinity Labeling (SIM-PAL) to Identify Binding Sites of Small Molecules on a Proteome-Wide Scale 利用光亲和标记(SIM-PAL)在蛋白质组范围内鉴定小分子的结合位点
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2019-10-03 DOI: 10.1002/cpch.75
Hope A. Flaxman, David K. Miyamoto, Christina M. Woo

Identification and characterization of small molecule–protein interactions is critical to understanding the mechanism of action of bioactive small molecules. Photo-affinity labeling (PAL) enables the capture of noncovalent interactions for enrichment and unbiased analysis by mass spectrometry (MS). Quantitative proteomics of the enriched proteome reveals potential interactions, and MS characterization of binding sites provides validation and structural insight into the interactions. Here, we describe the identification of the protein targets and binding sites of a small molecule using small molecule interactome mapping by PAL (SIM-PAL). Cells are exposed to a diazirine-alkyne-functionalized small molecule, and binding interactions are covalently captured upon UV irradiation. An isotopically coded, acid-cleavable biotin azide handle is attached to the conjugated proteins using copper-catalyzed azide-alkyne cycloaddition. Biotin-labeled proteins are enriched for on-bead digestion and quantitative proteomics. Acid cleavage of the handle releases the bead-bound conjugated peptides for MS analysis and isotope-directed assignment of the binding site. © 2019 by John Wiley & Sons, Inc.

Basic Protocol 1: Generation of a small molecule–conjugated protein sample following treatment of live cells

Alternate Protocol: Generation of a small molecule–conjugated protein sample following treatment of cell lysate

Basic Protocol 2: Copper-catalyzed azide-alkyne cycloaddition functionalization and enrichment of labeled peptides

Support Protocol 1: Synthesis of acid-cleavable, isotopically coded biotin picolyl azide handle

Support Protocol 2: Monitoring enrichment by immunoblotting

Basic Protocol 3: Mass spectrometry analysis to identify interacting proteins and conjugation sites

小分子-蛋白相互作用的鉴定和表征对于理解生物活性小分子的作用机制至关重要。光亲和标记(PAL)可以捕获非共价相互作用,通过质谱(MS)进行富集和无偏分析。富集蛋白质组的定量蛋白质组学揭示了潜在的相互作用,结合位点的质谱表征提供了相互作用的验证和结构洞察。在这里,我们描述了利用PAL (SIM-PAL)小分子相互作用组作图来鉴定小分子的蛋白质靶点和结合位点。细胞暴露于二氮嘧啶-炔功能化的小分子中,结合相互作用在紫外线照射下被共价捕获。通过铜催化叠氮化物-炔环加成,将同位素编码、酸可切割的生物素叠氮化物手柄连接到偶联蛋白上。生物素标记的蛋白质被富集用于头上消化和定量蛋白质组学。把手的酸裂解释放出头部结合的共轭肽,用于质谱分析和结合位点的同位素定向分配。©2019 by John Wiley &基本方案1:活细胞处理后小分子偶联蛋白样品的生成替代方案:细胞裂解后小分子偶联蛋白样品的生成基本方案2:铜催化叠氮化物-炔环加成功能化和标记肽的富集支持方案1:酸可切割、同位素编码的生物素吡啶叠氮化物处理的合成支持方案2:免疫印迹法监测富集基本方案3:质谱分析鉴定相互作用蛋白和偶联位点
{"title":"Small Molecule Interactome Mapping by Photo-Affinity Labeling (SIM-PAL) to Identify Binding Sites of Small Molecules on a Proteome-Wide Scale","authors":"Hope A. Flaxman,&nbsp;David K. Miyamoto,&nbsp;Christina M. Woo","doi":"10.1002/cpch.75","DOIUrl":"10.1002/cpch.75","url":null,"abstract":"<p>Identification and characterization of small molecule–protein interactions is critical to understanding the mechanism of action of bioactive small molecules. Photo-affinity labeling (PAL) enables the capture of noncovalent interactions for enrichment and unbiased analysis by mass spectrometry (MS). Quantitative proteomics of the enriched proteome reveals potential interactions, and MS characterization of binding sites provides validation and structural insight into the interactions. Here, we describe the identification of the protein targets and binding sites of a small molecule using small molecule interactome mapping by PAL (SIM-PAL). Cells are exposed to a diazirine-alkyne-functionalized small molecule, and binding interactions are covalently captured upon UV irradiation. An isotopically coded, acid-cleavable biotin azide handle is attached to the conjugated proteins using copper-catalyzed azide-alkyne cycloaddition. Biotin-labeled proteins are enriched for on-bead digestion and quantitative proteomics. Acid cleavage of the handle releases the bead-bound conjugated peptides for MS analysis and isotope-directed assignment of the binding site. © 2019 by John Wiley &amp; Sons, Inc.</p><p><b>Basic Protocol 1</b>: Generation of a small molecule–conjugated protein sample following treatment of live cells</p><p><b>Alternate Protocol</b>: Generation of a small molecule–conjugated protein sample following treatment of cell lysate</p><p><b>Basic Protocol 2</b>: Copper-catalyzed azide-alkyne cycloaddition functionalization and enrichment of labeled peptides</p><p><b>Support Protocol 1</b>: Synthesis of acid-cleavable, isotopically coded biotin picolyl azide handle</p><p><b>Support Protocol 2</b>: Monitoring enrichment by immunoblotting</p><p><b>Basic Protocol 3</b>: Mass spectrometry analysis to identify interacting proteins and conjugation sites</p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"11 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.75","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46925830","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: 10
Issue Information TOC 发布信息TOC
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2019-09-03 DOI: 10.1002/cpch.56

Cover: In Papa and Shoulders (https://doi.org/10.1002/cpch.70), Schematic of DIRex recombineering. (A) In DIRex recombineering, intermediates containing the conditionally lethal gene (ccdB) are first selected for using kanamycin. The modification (Mod) is directly repeated in the homology arms. Thus, the direct and inverted repeats (IR) promote spontaneous excision to yield the final recombinant. Successful recombinants are selected for by removing arabinose. In the absence of arabinose, the ccdA antitoxin is no longer expressed, and ccdB then kills unmodified cells. (B) DIRex is hypothesized to promote spontaneous excision through hybridization between the two inverted repeats to form a hairpin during replication that brings the direct repeats into close proximity (Bzymek & Lovett, 2001; Näsvall, 2017). The direct repeats can then promote strand slippage during synthesis, which results in excision of one of the direct repeats and everything between the direct repeats. See e70.

封面:Papa and Shoulders (https://doi.org/10.1002/cpch.70), DIRex重组示意图。(A)在DIRex重组中,首先选择含有条件致死基因(ccdB)的中间体使用卡那霉素。修饰(Mod)直接在同源臂上重复。因此,直接和反向重复(IR)促进自发切除产生最终的重组。通过去除阿拉伯糖来选择成功的重组体。在缺乏阿拉伯糖的情况下,ccdA抗毒素不再表达,然后ccdB杀死未修饰的细胞。(B)假设DIRex通过两个反向重复序列之间的杂交促进自发切除,在复制过程中形成一个发夹,使直接重复序列靠近(Bzymek &洛维特,2001;Nasvall, 2017)。直接重复可以在合成过程中促进链滑动,导致直接重复中的一个和直接重复之间的所有内容被切除。看到e70。
{"title":"Issue Information TOC","authors":"","doi":"10.1002/cpch.56","DOIUrl":"https://doi.org/10.1002/cpch.56","url":null,"abstract":"<p><b>Cover</b>: In Papa and Shoulders (https://doi.org/10.1002/cpch.70), Schematic of DIRex recombineering. (<b>A</b>) In DIRex recombineering, intermediates containing the conditionally lethal gene (ccdB) are first selected for using kanamycin. The modification (Mod) is directly repeated in the homology arms. Thus, the direct and inverted repeats (IR) promote spontaneous excision to yield the final recombinant. Successful recombinants are selected for by removing arabinose. In the absence of arabinose, the ccdA antitoxin is no longer expressed, and ccdB then kills unmodified cells. (<b>B</b>) DIRex is hypothesized to promote spontaneous excision through hybridization between the two inverted repeats to form a hairpin during replication that brings the direct repeats into close proximity (Bzymek &amp; Lovett, 2001; Näsvall, 2017). The direct repeats can then promote strand slippage during synthesis, which results in excision of one of the direct repeats and everything between the direct repeats. See e70.\u0000\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":38051,"journal":{"name":"Current protocols in chemical biology","volume":"11 3","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpch.56","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"137794849","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: 0
期刊
Current protocols in chemical biology
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