Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38915575

ABSTRACT

We introduce an adaptor-based strategy for regulating fluorescein-binding synthetic Notch (SynNotch) receptors using ligands based on conjugates of fluorescein isomers and analogs. To develop a versatile system, we evaluated the surface expression and activities of multiple constructs containing distinct extracellular fluorescein-binding domains. Using an optimized receptor, we devised ways to regulate signaling via fluorescein-based chemical transformations, including an approach based on a bio-orthogonal chemical ligation and a spatially controllable strategy via the photo-patterned uncaging of an o -nitrobenzyl-caged fluorescein conjugate. We further demonstrate that fluorescein-conjugated extracellular matrix (ECM)-binding peptides can regulate SynNotch activity depending on the folding state of collagen-based ECM networks. Treatment with these conjugates enabled cells to distinguish between folded versus denatured collagen proteins and enact dose-dependent gene expression responses depending on the nature of the signaling adaptors presented. To demonstrate the utility of these tools, we applied them to control the myogenic conversion of fibroblasts into myocytes with spatial and temporal precision and in response to denatured collagen-I, a biomarker of multiple pathological states. Overall, we introduce an optimized fluorescein-binding SynNotch as a versatile tool for regulating transcriptional responses to extracellular ligands based on the widely used and clinically-approved fluorescein dye.

2.
bioRxiv ; 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37662194

ABSTRACT

We introduce Fe-TAML, a small molecule-based peroxidase as a versatile new member of the correlated fluorescence and electron microscopy toolkit. The utility of the probe is demonstrated by high resolution imaging of newly synthesized DNA (through biorthogonal labeling), genetically tagged proteins (using HaloTag), and untagged endogenous proteins (via immunostaining). EM visualization in these applications is facilitated by exploiting Fe-TAML's catalytic activity for the deposition of localized osmiophilic precipitates based on polymerized 3,3'-diaminobenzidine. Optimized conditions for synthesizing and implementing Fe-TAML based probes are also described. Overall, Fe-TAML is a new chemical biology tool that can be used to visualize diverse biomolecular species along nanometer and micron scales within cells.

3.
ACS Chem Biol ; 18(5): 1228-1236, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37140437

ABSTRACT

Chemical control of protein activity is a powerful tool for scientific study, synthetic biology, and cell therapy; however, for broad use, effective chemical inducer systems must minimally crosstalk with endogenous processes and exhibit desirable drug delivery properties. Accordingly, the drug-controllable proteolytic activity of hepatitis C cis-protease NS3 and its associated antiviral drugs have been used to regulate protein activity and gene modulation. These tools advantageously exploit non-eukaryotic and non-prokaryotic proteins and clinically approved inhibitors. Here, we expand the toolkit by utilizing catalytically inactive NS3 protease as a high affinity binder to genetically encoded, antiviral peptides. Through our approach, we create NS3-peptide complexes that can be displaced by FDA-approved drugs to modulate transcription, cell signaling, and split-protein complementation. With our developed system, we invented a new mechanism to allosterically regulate Cre recombinase. Allosteric Cre regulation with NS3 ligands enables orthogonal recombination tools in eukaryotic cells and functions in divergent organisms to control prokaryotic recombinase activity.


Subject(s)
Antiviral Agents , Viral Proteases , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Hepacivirus , Peptide Hydrolases , Peptides/pharmacology , Peptides/chemistry , Protease Inhibitors/chemistry , Viral Nonstructural Proteins/metabolism
4.
bioRxiv ; 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36909459

ABSTRACT

Chemical control of protein activity is a powerful tool for scientific study, synthetic biology, and cell therapy; however, for broad use, effective chemical inducer systems must minimally crosstalk with endogenous processes and exhibit desirable drug delivery properties. Accordingly, the drug-controllable proteolytic activity of hepatitis C cis- protease NS3 and its associated antiviral drugs have been used to regulate protein activity and gene modulation. These tools advantageously exploit non-eukaryotic/prokaryotic proteins and clinically approved inhibitors. Here we expand the toolkit by utilizing catalytically inactive NS3 protease as a high affinity binder to genetically encoded, antiviral peptides. Through our approach, we create NS3-peptide complexes that can be displaced by FDA-approved drugs to modulate transcription, cell signaling, split-protein complementation. With our developed system, we discover a new mechanism to allosterically regulate Cre recombinase. Allosteric Cre regulation with NS3 ligands enables orthogonal recombination tools in eukaryotic cells and functions in divergent organisms to control prokaryotic recombinase activity.

5.
Nat Biotechnol ; 41(9): 1287-1295, 2023 09.
Article in English | MEDLINE | ID: mdl-36646932

ABSTRACT

Cells interpret mechanical stimuli from their environments and neighbors, but the ability to engineer customized mechanosensing capabilities has remained a synthetic and mechanobiology challenge. Here we introduce tension-tuned synthetic Notch (SynNotch) receptors to convert extracellular and intercellular forces into specifiable gene expression changes. By elevating the tension requirements of SynNotch activation, in combination with structure-guided mutagenesis, we designed a set of receptors with mechanical sensitivities spanning the physiologically relevant picoNewton range. Cells expressing these receptors can distinguish between varying tensile forces and respond by enacting customizable transcriptional programs. We applied these tools to design a decision-making circuit, through which fibroblasts differentiate into myoblasts upon stimulation with distinct tension magnitudes. We also characterize cell-generated forces transmitted between cells during Notch signaling. Overall, this work provides insight into how mechanically induced changes in protein structure can be used to transduce physical forces into biochemical signals. The system should facilitate the further programming and dissection of force-related phenomena in biological systems.


Subject(s)
Mechanotransduction, Cellular , Signal Transduction , Proteins
6.
Cancer Cell ; 40(11): 1294-1305.e4, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36084652

ABSTRACT

Chimeric antigen receptor (CAR) T cells can revolutionize cancer medicine. However, overactivation, lack of tumor-specific surface markers, and antigen escape have hampered CAR T cell development. A multi-antigen targeting CAR system regulated by clinically approved pharmaceutical agents is needed. Here, we present VIPER CARs (versatile protease regulatable CARs), a collection of inducible ON and OFF switch CAR circuits engineered with a viral protease domain. We established their controllability using FDA-approved antiviral protease inhibitors in a xenograft tumor and a cytokine release syndrome mouse model. Furthermore, we benchmarked VIPER CARs against other drug-gated systems and demonstrated best-in-class performance. We showed their orthogonality in vivo using the ON VIPER CAR and OFF lenalidomide-CAR systems. Finally, we engineered several VIPER CAR circuits by combining various CAR technologies. Our multiplexed, drug-gated CAR circuits represent the next progression in CAR design capable of advanced logic and regulation for enhancing the safety of CAR T cell therapy.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Mice , Animals , Humans , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Immunotherapy, Adoptive , Lenalidomide , Receptors, Antigen, T-Cell/genetics
7.
8.
Anat Rec (Hoboken) ; 303(6): 1630-1641, 2020 06.
Article in English | MEDLINE | ID: mdl-30851151

ABSTRACT

The circadian clock, which consists of endogenous self-sustained and cell-autonomous oscillations in mammalian cells, is known to regulate a wide range of peripheral tissues. The unique upregulation of a clock gene, neuronal PAS domain protein 2 (Npas2), observed along with fibroblast aging prompted us to investigate the role of Npas2 in the homeostasis of dermal structure using in vivo and in vitro wound healing models. Time-course healing of a full-thickness skin punched wound exhibited significantly faster wound closure in Npas2-/- mice than wild-type (WT) C57Bl/6J mice. Dorsal skin fibroblasts isolated from WT, Npas2+/-, and Npas2-/- mice exhibited consistent profiles of core clock gene expression except for Npas2 and Per2. In vitro behavioral characterizations of dermal fibroblasts revealed that Npas2-/- mutation was associated with increased proliferation, migration, and cell contraction measured by floating collagen gel contraction and single-cell force contraction assays. Npas2 knockout fibroblasts carrying sustained the high expression level of type XII and XIV FAICT collagens and synthesized dermis-like thick collagen fibers in vitro. Confocal laser scanning microscopy demonstrated the reconstruction of dermis-like collagen architecture in the wound healing area of Npas2-/- mice. This study indicates that the induced Npas2 expression in fibroblasts may interfere with skin homeostasis, wound healing, and dermal tissue reconstruction, providing a basis for novel therapeutic target and strategy. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Collagen/metabolism , Fibroblasts/metabolism , Nerve Tissue Proteins/metabolism , Skin/metabolism , Wound Healing/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Movement/physiology , Cell Proliferation/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
9.
Cell Chem Biol ; 26(10): 1407-1416.e5, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31378710

ABSTRACT

A protein-fragment complementation assay (PCA) for detecting and localizing intracellular protein-protein interactions (PPIs) was built by bisection of miniSOG, a fluorescent flavoprotein derived from the light, oxygen, voltage (LOV)-2 domain of Arabidopsis phototropin. When brought together by interacting proteins, the fragments reconstitute a functional reporter that permits tagged protein complexes to be visualized by fluorescence light microscopy (LM), and then by standard as well as "multicolor" electron microscopy (EM) via the photooxidation of 3-3'-diaminobenzidine and its derivatives.


Subject(s)
Arabidopsis Proteins/chemistry , Flavoproteins/chemistry , Luminescent Proteins/chemistry , 3,3'-Diaminobenzidine/chemistry , Arabidopsis/chemistry , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans , Microscopy, Electron , Microscopy, Fluorescence , Oxidation-Reduction , Photochemical Processes , Protein Binding
10.
Nat Methods ; 15(7): 519-522, 2018 07.
Article in English | MEDLINE | ID: mdl-29967495

ABSTRACT

We developed a method in which the NS3 cis-protease from hepatitis C virus can be used as a ligand-inducible connection to control the function and localization of engineered proteins in mammalian cells. To demonstrate the versatility of this approach, we designed drug-sensitive transcription factors and transmembrane signaling proteins, the activities of which can be tightly and reversibly controlled through the use of clinically tested antiviral protease inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Gene Expression Regulation/drug effects , Animals , CHO Cells , Cricetulus , DNA/genetics , DNA/metabolism , Signal Transduction , Viral Nonstructural Proteins/metabolism
11.
Nat Chem Biol ; 12(6): 459-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27110681

ABSTRACT

EM has long been the main technique for imaging cell structures with nanometer resolution but has lagged behind light microscopy in the crucial ability to make specific molecules stand out. Here we introduce click-EM, a labeling technique for correlative light microscopy and EM imaging of nonprotein biomolecules. In this approach, metabolic labeling substrates containing bioorthogonal functional groups are provided to cells for incorporation into biopolymers by endogenous biosynthetic machinery. The unique chemical functionality of these analogs is exploited for selective attachment of singlet oxygen-generating fluorescent dyes via bioorthogonal 'click chemistry' ligations. Illumination of dye-labeled structures generates singlet oxygen to locally catalyze the polymerization of diaminobenzidine into an osmiophilic reaction product that is readily imaged by EM. We describe the application of click-EM in imaging metabolically tagged DNA, RNA and lipids in cultured cells and neurons and highlight its use in tracking peptidoglycan synthesis in the Gram-positive bacterium Listeria monocytogenes.


Subject(s)
Click Chemistry , DNA/analysis , Lipids/analysis , Microscopy, Electron/methods , Peptidoglycan/analysis , RNA/analysis , Aminobutyrates/chemistry , DNA/chemistry , DNA/metabolism , Fluorescent Dyes/chemistry , HEK293 Cells , HeLa Cells , Humans , Lipids/chemistry , Listeria monocytogenes/metabolism , Molecular Structure , Neurons/chemistry , Neurons/metabolism , Peptidoglycan/biosynthesis , RNA/chemistry , RNA/metabolism , Singlet Oxygen/chemistry
12.
J Neurosci ; 35(20): 7736-49, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995463

ABSTRACT

Synthesizing, localizing, and stabilizing new protein copies at synapses are crucial factors in maintaining the synaptic changes required for storing long-term memories. PKMζ recently emerged as a molecule putatively responsible for maintaining encoded memories over time because its presence correlates with late LTP and because its inhibition disrupts LTP in vitro and long-term memory storage in vivo. Here we investigated PKMζ stability in rat neurons to better understand its role during information encoding and storage. We used TimeSTAMP reporters to track the synthesis and degradation of PKMζ as well as a related atypical PKC, PKCλ. These reporters revealed that both PKMζ and PKCλ were upregulated after chemical LTP induction; however, these new PKMζ copies exhibited more rapid turnover than basally produced PKMζ, particularly in dendritic spines. In contrast to PKMζ, new PKCλ copies exhibited elevated stability. Stable information storage over long periods of time is more challenging the shorter the metabolic lifetime of the candidate molecules.


Subject(s)
Dendritic Spines/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Proteolysis , Synapses/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dendritic Spines/physiology , Enzyme Stability , HEK293 Cells , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Long-Term Potentiation , Molecular Sequence Data , Protein Kinase C/genetics , Rats , Rats, Sprague-Dawley , Synapses/physiology , Up-Regulation
13.
Proc Natl Acad Sci U S A ; 112(9): 2705-10, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25691744

ABSTRACT

Proteomic analysis of rare cells in heterogeneous environments presents difficult challenges. Systematic methods are needed to enrich, identify, and quantify proteins expressed in specific cells in complex biological systems including multicellular plants and animals. Here, we have engineered a Caenorhabditis elegans phenylalanyl-tRNA synthetase capable of tagging proteins with the reactive noncanonical amino acid p-azido-L-phenylalanine. We achieved spatiotemporal selectivity in the labeling of C. elegans proteins by controlling expression of the mutant synthetase using cell-selective (body wall muscles, intestinal epithelial cells, neurons, and pharyngeal muscle) or state-selective (heat-shock) promoters in several transgenic lines. Tagged proteins are distinguished from the rest of the protein pool through bioorthogonal conjugation of the azide side chain to probes that permit visualization and isolation of labeled proteins. By coupling our methodology with stable-isotope labeling of amino acids in cell culture (SILAC), we successfully profiled proteins expressed in pharyngeal muscle cells, and in the process, identified proteins not previously known to be expressed in these cells. Our results show that tagging proteins with spatiotemporal selectivity can be achieved in C. elegans and illustrate a convenient and effective approach for unbiased discovery of proteins expressed in targeted subsets of cells.


Subject(s)
Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Gene Expression Regulation/physiology , Proteome/biosynthesis , Proteomics/methods , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Isotope Labeling/methods , Mutation , Proteome/genetics
14.
Proc Natl Acad Sci U S A ; 111(1): 433-8, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24347637

ABSTRACT

Pathogenic microbes have evolved complex secretion systems to deliver virulence factors into host cells. Identification of these factors is critical for understanding the infection process. We report a powerful and versatile approach to the selective labeling and identification of secreted pathogen proteins. Selective labeling of microbial proteins is accomplished via translational incorporation of azidonorleucine (Anl), a methionine surrogate that requires a mutant form of the methionyl-tRNA synthetase for activation. Secreted pathogen proteins containing Anl can be tagged by azide-alkyne cycloaddition and enriched by affinity purification. Application of the method to analysis of the type III secretion system of the human pathogen Yersinia enterocolitica enabled efficient identification of secreted proteins, identification of distinct secretion profiles for intracellular and extracellular bacteria, and determination of the order of substrate injection into host cells. This approach should be widely useful for the identification of virulence factors in microbial pathogens and the development of potential new targets for antimicrobial therapy.


Subject(s)
Amino Acids/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microbiological Techniques , Anti-Bacterial Agents/chemistry , HeLa Cells , Humans , Mass Spectrometry , Methionine-tRNA Ligase/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Mutation , Norleucine/chemistry , Proteome , Proteomics/methods , Time Factors , Virulence Factors , Yersinia enterocolitica/metabolism
15.
Proc Natl Acad Sci U S A ; 110(13): 4992-7, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23479642

ABSTRACT

Newly synthesized cellular proteins can be tagged with a variety of metabolic labels that distinguish them from preexisting proteins and allow them to be identified and tracked. Many such labels are incorporated into proteins via the endogenous cellular machinery and can be used in numerous cell types and organisms. Though broad applicability has advantages, we aimed to develop a strategy to restrict protein labeling to specified mammalian cells that express a transgene. Here we report that heterologous expression of a mutant methionyl-tRNA synthetase from Escherichia coli permits incorporation of azidonorleucine (Anl) into proteins made in mammalian (HEK293) cells. Anl is incorporated site-selectively at N-terminal positions (in competition with initiator methionines) and is not found at internal sites. Site selectivity is enabled by the fact that the bacterial synthetase aminoacylates mammalian initiator tRNA, but not elongator tRNA. N-terminally labeled proteins can be selectively conjugated to a variety of useful probes; here we demonstrate use of this system in enrichment and visualization of proteins made during various stages of the cell cycle. N-terminal incorporation of Anl may also be used to engineer modified proteins for therapeutic and other applications.


Subject(s)
Escherichia coli Proteins/biosynthesis , Escherichia coli/enzymology , Leucine/metabolism , Methionine-tRNA Ligase/biosynthesis , Peptide Chain Initiation, Translational , RNA, Transfer, Met/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , HEK293 Cells , Humans , Leucine/analogs & derivatives , Leucine/genetics , Methionine-tRNA Ligase/genetics , RNA, Transfer, Met/genetics
16.
ACS Chem Biol ; 7(8): 1326-30, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22692961

ABSTRACT

Transcriptional activity from a specified promoter can provide a useful marker for the physiological state of a cell. Here we introduce a method for selective tagging of proteins made in cells in which specified promoters are active. Tagged proteins can be modified with affinity reagents for enrichment or with fluorescent dyes for visualization. The method allows state-selective analysis of the proteome, whereby proteins synthesized in predetermined physiological states can be identified. The approach is demonstrated by proteome-wide labeling of bacterial proteins upon activation of the P(BAD) promoter and the SoxRS regulon and provides a basis for analysis of more complex systems including spatially heterogeneous microbial cultures and biofilms.


Subject(s)
Transcription, Genetic , Arabinose/genetics , Bacterial Proteins/chemistry , Biofilms , Cell Survival , Cloning, Molecular , Escherichia coli/metabolism , Fluorescent Dyes/chemistry , Gene Expression Regulation , Genetic Techniques , Ligands , Magnetic Resonance Spectroscopy/methods , Models, Biological , Models, Genetic , Promoter Regions, Genetic , Proteomics/methods , Regulon
17.
Acc Chem Res ; 44(9): 677-85, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21815659

ABSTRACT

Proteins in living cells can be made receptive to bioorthogonal chemistries through metabolic labeling with appropriately designed noncanonical amino acids (ncAAs). In the simplest approach to metabolic labeling, an amino acid analog replaces one of the natural amino acids specified by the protein's gene (or genes) of interest. Through manipulation of experimental conditions, the extent of the replacement can be adjusted. This approach, often termed residue-specific incorporation, allows the ncAA to be incorporated in controlled proportions into positions normally occupied by the natural amino acid residue. For a protein to be labeled in this way with an ncAA, it must fulfill just two requirements: (i) the corresponding natural amino acid must be encoded within the sequence of the protein at the genetic level, and (ii) the protein must be expressed while the ncAA is in the cell. Because this approach permits labeling of proteins throughout the cell, it has enabled us to develop strategies to track cellular protein synthesis by tagging proteins with reactive ncAAs. In procedures similar to isotopic labeling, translationally active ncAAs are incorporated into proteins during a "pulse" in which newly synthesized proteins are tagged. The set of tagged proteins can be distinguished from those made before the pulse by bioorthogonally ligating the ncAA side chain to probes that permit detection, isolation, and visualization of the labeled proteins. Noncanonical amino acids with side chains containing azide, alkyne, or alkene groups have been especially useful in experiments of this kind. They have been incorporated into proteins in the form of methionine analogs that are substrates for the natural translational machinery. The selectivity of the method can be enhanced through the use of mutant aminoacyl tRNA synthetases (aaRSs) that permit incorporation of ncAAs not used by the endogenous biomachinery. Through expression of mutant aaRSs, proteins can be tagged with other useful ncAAs, including analogs that contain ketones or aryl halides. High-throughput screening strategies can identify aaRS variants that activate a wide range of ncAAs. Controlled expression of mutant synthetases has been combined with ncAA tagging to permit cell-selective metabolic labeling of proteins. Expression of a mutant synthetase in a portion of cells within a complex cellular mixture restricts labeling to that subset of cells. Proteins synthesized in cells not expressing the synthetase are neither labeled nor detected. In multicellular environments, this approach permits the identification of the cellular origins of labeled proteins. In this Account, we summarize the tools and strategies that have been developed for interrogating cellular protein synthesis through residue-specific tagging with ncAAs. We describe the chemical and genetic components of ncAA-tagging strategies and discuss how these methods are being used in chemical biology.


Subject(s)
Amino Acids/metabolism , Protein Biosynthesis , Proteins/metabolism , Alkenes/chemistry , Alkynes/chemistry , Alkynes/metabolism , Amino Acids/chemistry , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Azides/chemistry , Catalysis , Copper/chemistry , Fluorescent Dyes/chemistry , Glycine/analogs & derivatives , Glycine/chemistry , Glycine/metabolism , Mass Spectrometry , Microscopy, Fluorescence , Mutation , Proteins/chemistry
18.
J Am Chem Soc ; 132(51): 18351-60, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21141861

ABSTRACT

The azide-alkyne cycloaddition provides a powerful tool for bio-orthogonal labeling of proteins, nucleic acids, glycans, and lipids. In some labeling experiments, e.g., in proteomic studies involving affinity purification and mass spectrometry, it is convenient to use cleavable probes that allow release of labeled biomolecules under mild conditions. Five cleavable biotin probes are described for use in labeling of proteins and other biomolecules via azide-alkyne cycloaddition. Subsequent to conjugation with metabolically labeled protein, these probes are subject to cleavage with either 50 mM Na(2)S(2)O(4), 2% HOCH(2)CH(2)SH, 10% HCO(2)H, 95% CF(3)CO(2)H, or irradiation at 365 nm. Most strikingly, a probe constructed around a dialkoxydiphenylsilane (DADPS) linker was found to be cleaved efficiently when treated with 10% HCO(2)H for 0.5 h. A model green fluorescent protein was used to demonstrate that the DADPS probe undergoes highly selective conjugation and leaves a small (143 Da) mass tag on the labeled protein after cleavage. These features make the DADPS probe especially attractive for use in biomolecular labeling and proteomic studies.


Subject(s)
Alkynes/chemistry , Azides/chemistry , Biotin/chemistry , Molecular Probes/chemistry , Proteins/chemistry , Cyclization , Dapsone , Green Fluorescent Proteins/chemistry , Molecular Probes/chemical synthesis , Protein Structure, Secondary
19.
Nat Neurosci ; 13(7): 897-905, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20543841

ABSTRACT

Protein translation has been implicated in different forms of synaptic plasticity, but direct in situ visualization of new proteins is limited to one or two proteins at a time. Here we describe a metabolic labeling approach based on incorporation of noncanonical amino acids into proteins followed by chemoselective fluorescence tagging by means of 'click chemistry'. After a brief incubation with azidohomoalanine or homopropargylglycine, a robust fluorescent signal was detected in somata and dendrites. Pulse-chase application of azidohomoalanine and homopropargylglycine allowed visualization of proteins synthesized in two sequential time periods. This technique can be used to detect changes in protein synthesis and to evaluate the fate of proteins synthesized in different cellular compartments. Moreover, using strain-promoted cycloaddition, we explored the dynamics of newly synthesized membrane proteins using single-particle tracking and quantum dots. The newly synthesized proteins showed a broad range of diffusive behaviors, as would be expected for a pool of labeled proteins that is heterogeneous.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Protein Biosynthesis/physiology , Proteomics/methods , Staining and Labeling/methods , Alanine/analogs & derivatives , Alanine/metabolism , Alkynes/metabolism , Amino Acids/metabolism , Animals , Cells, Cultured , Fluorescent Dyes/metabolism , Glycine/analogs & derivatives , Glycine/metabolism , Hippocampus/cytology , Immunohistochemistry , Microdialysis , Quantum Dots , Rats , Rats, Sprague-Dawley
20.
Nat Chem Biol ; 5(10): 715-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19668194

ABSTRACT

Metabolic labeling of proteins with the methionine surrogate azidonorleucine can be targeted exclusively to specified cells through expression of a mutant methionyl-tRNA synthetase (MetRS). In complex cellular mixtures, proteins made in cells that express the mutant synthetase can be tagged with affinity reagents (for detection or enrichment) or fluorescent dyes (for imaging). Proteins made in cells that do not express the mutant synthetase are neither labeled nor detected.


Subject(s)
Affinity Labels/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Methionine-tRNA Ligase/metabolism , Proteins/metabolism , Alanine/analogs & derivatives , Alanine/metabolism , Amino Acyl-tRNA Synthetases/genetics , Animals , Cell Line , Coculture Techniques , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Methionine-tRNA Ligase/genetics , Mice , Mutagenesis, Site-Directed , Mutation , Norleucine/analogs & derivatives , Norleucine/metabolism , Protein Biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...