Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 720: 150097, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38754162

ABSTRACT

Inteins are unique single-turnover enzymes that can excise themselves from the precursor protein without the aid of any external cofactors or energy. In most cases, inteins are covalently linked with the extein sequences and protein splicing happens spontaneously. In this study, a novel protein ligation system was developed based on two atypical split inteins without cross reaction, in which the large segments of one S1 and one S11 split intein fusion protein acted as a protein ligase, the small segments (only several amino acids long) was fused to the N-extein and C-extein, respectively. The splicing activity was demonstrated in E. coli and in vitro with different extein sequences, which showed ∼15% splicing efficiency in vitro. The protein trans-splicing in vitro was further optimized, and possible reaction explanations were explored. As a proof of concept, we expect this approach to expand the scope of trans-splicing-based protein engineering and provide new clues for intein based protein ligase.


Subject(s)
Escherichia coli , Inteins , Protein Splicing , Inteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Ligases/metabolism , Ligases/genetics , Ligases/chemistry , Exteins/genetics
2.
Protein Sci ; 32(7): e4699, 2023 07.
Article in English | MEDLINE | ID: mdl-37313648

ABSTRACT

Intein enzymes catalyze the splicing of their flanking polypeptide chains and have found tremendous biotechnological applications. Their terminal residues form the catalytic core and participate in the splicing reaction. Hence, the neighboring N- and C-terminal extein residues influence the catalytic rate. As these extein residues vary depending on the substrate identity, we tested the influence of 20 amino acids at these sites in the Spl DnaX intein and observed significant variation of spliced product as well as N- and C-terminus cleavage product formation. We investigated the dependence of these reactions on the extein residues by molecular dynamics (MD) simulations on eight extein variants, and found that the conformational sampling of the active-site residues of the intein enzyme differed among these extein variants. We found that the extein variants that sample higher population of near-attack conformers (NACs) of the active-site residues undergo higher product formation in our activity assays. Ground state conformers that closely resemble the transition state are referred to as NACs. Very good correlation was observed between the NAC populations from the MD simulations of eight extein variants and the corresponding product formation from our activity assays. Furthermore, this molecular detail enabled us to elucidate the mechanistic roles of several conserved active-site residues in the splicing reaction. Overall, this study shows that the catalytic power of Spl DnaX intein enzyme, and most likely other inteins, depends on the efficiency of formation of NACs in the ground state, which is further modulated by the extein residues.


Subject(s)
Exteins , Inteins , Catalytic Domain , Protein Splicing , Amino Acids
3.
J Am Chem Soc ; 144(41): 19196-19203, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36194550

ABSTRACT

Split intein-mediated protein trans-splicing (PTS) is widely applied in chemical biology and biotechnology to carry out traceless and specific protein ligation. However, the external residues immediately flanking the intein (exteins) can reduce the splicing rate, thereby limiting certain applications of PTS. Splicing by a recently developed intein with atypical split architecture ("Cat") exhibits a stark dependence on the sequence of its N-terminal extein residues. Here, we further developed Cat using error-prone polymerase chain reaction (PCR) and a cell-based selection assay to produce Cat*, which exhibits greatly enhanced PTS activity in the presence of unfavorable N-extein residues. We then applied solution nuclear magnetic resonance spectroscopy and molecular dynamics simulations to explore how the dynamics of a conserved B-block histidine residue (His78) contribute to this extein dependence. The enhanced extein tolerance of Cat* reported here should expand the applicability of atypically split inteins, and the mechanism highlights common principles that contribute to extein dependence.


Subject(s)
Exteins , Inteins , Histidine/metabolism , Protein Splicing , Proteins/metabolism
4.
Biosci Rep ; 42(3)2022 03 31.
Article in English | MEDLINE | ID: mdl-35234249

ABSTRACT

Inteins are auto-processing domains that implement a multistep biochemical reaction termed protein splicing, marked by cleavage and formation of peptide bonds. They excise from a precursor protein, generating a functional protein via covalent bonding of flanking exteins. We report the kinetic study of splicing and cleavage reaction in [Fe-S] cluster assembly protein SufB from Mycobacterium tuberculosis (Mtu). Although it follows a canonical intein splicing pathway, distinct features are added by extein residues present in the active site. Sequence analysis identified two conserved histidines in the N-extein region; His-5 and His-38. Kinetic analyses of His-5Ala and His-38Ala SufB mutants exhibited significant reductions in splicing and cleavage rates relative to the SufB wildtype (WT) precursor protein. Structural analysis and molecular dynamics (MD) simulations suggested that Mtu SufB displays a unique mechanism where two remote histidines work concurrently to facilitate N-terminal cleavage reaction. His-38 is stabilized by the solvent-exposed His-5, and can impact N-S acyl shift by direct interaction with the catalytic Cys1. Development of inteins as biotechnological tools or as pathogen-specific novel antimicrobial targets requires a more complete understanding of such unexpected roles of conserved extein residues in protein splicing.


Subject(s)
Exteins , Mycobacterium tuberculosis , Histidine/genetics , Inteins/genetics , Mycobacterium tuberculosis/genetics , Protein Splicing
5.
Biochemistry ; 61(4): 294-302, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35073064

ABSTRACT

Protein splicing is a post-translational process by which an intervening protein, or an intein, catalyzes its own excision from flanking polypeptides, or exteins, coupled to extein ligation. Four inteins interrupt the MCM helicase of the halophile Haloquadratum walsbyi, two of which are mini-inteins that lack a homing endonuclease. Both inteins can be overexpressed in Escherichia coli and purified as unspliced precursors; splicing can be induced in vitro by incubation with salt. However, one intein can splice in 0.5 M NaCl in vitro, whereas the other splices efficiently only in buffer containing over 2 M NaCl; the organism also requires high salt to grow, with the standard growth media containing over 3 M NaCl and about 0.75 M magnesium salts. Consistent with this difference in salt-dependent activity, an intein-containing precursor protein with both inteins promotes conditional alternative protein splicing (CAPS) to yield different spliced products dependent on the salt concentration. Native Trp fluorescence of the inteins suggests that the difference in activity may be due to partial unfolding of the inteins at lower salt concentrations. This differential salt sensitivity of intein activity may provide a useful mechanism for halophiles to respond to environmental changes.


Subject(s)
Archaeal Proteins/metabolism , Halobacteriaceae/metabolism , Inteins , Minichromosome Maintenance Proteins/metabolism , Protein Splicing , Escherichia coli/metabolism , Exteins , Peptides/metabolism , Protein Precursors/metabolism
6.
Methods Mol Biol ; 2133: 31-54, 2020.
Article in English | MEDLINE | ID: mdl-32144662

ABSTRACT

In recent years, split inteins have seen widespread use as molecular platforms for the design of a variety of peptide and protein chemistry technologies, most notably protein ligation. The development of these approaches is dependent on the identification and/or design of split inteins with robust activity, stability, and solubility. Here, we describe two approaches to characterize and compare the activities of newly identified or engineered split inteins. The first assay employs an E. coli-based selection system to rapidly screen the activities of many inteins and can be repurposed for directed evolution. The second assay utilizes reverse-phase high-performance liquid chromatography (RP-HPLC) to provide insights into individual chemical steps in the protein splicing reaction, information that can guide further engineering efforts. These techniques provide useful alternatives to common assays that utilize SDS-PAGE to analyze splicing reaction progress.


Subject(s)
Cloning, Molecular/methods , Inteins , Protein Engineering/methods , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/methods , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Exteins , Gene Expression , Inteins/genetics , Kanamycin Resistance , Protein Splicing , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Solid-Phase Synthesis Techniques/methods , Trans-Splicing
7.
Methods Mol Biol ; 2133: 55-73, 2020.
Article in English | MEDLINE | ID: mdl-32144663

ABSTRACT

The autocatalytic process of protein splicing is facilitated by an intein, which interrupts flanking polypeptides called exteins. The mechanism of protein splicing has been studied by overexpression in E. coli of intein fusion proteins with nonnative exteins. Inteins can be used to generate reactive α-thioesters, as well as proteins with N-terminal Cys residues, to facilitate expressed protein ligation. As such, a more detailed understanding of the function of inteins can have significant impact for biotechnology applications. Here, we provide biochemical methods to study splicing activity and NMR methods to study intein structure and the catalytic mechanism.


Subject(s)
Biocatalysis , Biochemistry/methods , Inteins , Protein Splicing , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Aspartic Acid/chemistry , Carbon Isotopes , Cysteine/chemistry , Electrophoresis, Polyacrylamide Gel/methods , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Exteins , Gene Expression , Genetic Vectors/genetics , Histidine/chemistry , Hydrogen-Ion Concentration , Maltose-Binding Proteins/chemistry , Maltose-Binding Proteins/genetics , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular/methods , Recombinant Fusion Proteins/biosynthesis
8.
Nucleic Acids Res ; 47(21): 11452-11460, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31667500

ABSTRACT

Serine integrases are emerging as core tools in synthetic biology and have applications in biotechnology and genome engineering. We have designed a split-intein serine integrase-based system with potential for regulation of site-specific recombination events at the protein level in vivo. The ϕC31 integrase was split into two extein domains, and intein sequences (Npu DnaEN and Ssp DnaEC) were attached to the two termini to be fused. Expression of these two components followed by post-translational protein trans-splicing in Escherichia coli generated a fully functional ϕC31 integrase. We showed that protein splicing is necessary for recombination activity; deletion of intein domains or mutation of key intein residues inactivated recombination. We used an invertible promoter reporter system to demonstrate a potential application of the split intein-regulated site-specific recombination system in building reversible genetic switches. We used the same split inteins to control the reconstitution of a split Integrase-Recombination Directionality Factor fusion (Integrase-RDF) that efficiently catalysed the reverse attR x attL recombination. This demonstrates the potential for split-intein regulation of the forward and reverse reactions using the integrase and the integrase-RDF fusion, respectively. The split-intein integrase is a potentially versatile, regulatable component for building synthetic genetic circuits and devices.


Subject(s)
Integrases/physiology , Protein Splicing/genetics , Recombination, Genetic , Trans-Splicing/genetics , Amino Acid Sequence , Cloning, Molecular/methods , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Exteins/genetics , Integrases/metabolism , Inteins/genetics , Organisms, Genetically Modified , Protein Engineering , Serine/metabolism , Substrate Specificity/genetics
9.
Anal Chem ; 90(16): 9779-9786, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30028129

ABSTRACT

Live-cell-based biosensors have emerged as a useful tool for biotechnology and chemical biology. Genetically encoded sensor cells often use bimolecular fluorescence complementation or fluorescence resonance energy transfer to build a reporter unit that suffers from nonspecific signal activation at high concentrations. Here, we designed genetically encoded sensor cells that can report the presence of biologically active molecules via fluorescence-translocation based on split intein-mediated conditional protein trans-splicing (PTS) and conditional protein trans-cleavage (PTC) reactions. In this work, the target molecules or the external stimuli activated intein-mediated reactions, which resulted in activation of the fluorophore-conjugated signal peptide. This approach fully valued the bond-making and bond-breaking features of intein-mediated reactions in sensor construction and thus eliminated the interference of false-positive signals resulting from the mere binding of fragmented reporters. We could also avoid the necessity of designing split reporters to refold into active structures upon reconstitution. These live-cell-based sensors were able to detect biologically active signaling molecules, such as Ca2+ and cortisol, as well as relevant biological stimuli, such as histamine-induced Ca2+ stimuli and the glucocorticoid receptor agonist, dexamethasone. These live-cell-based sensing systems hold large potential for applications such as drug screening and toxicology studies, which require functional information about targets.


Subject(s)
Biosensing Techniques/methods , Calcium/analysis , Hormones/analysis , Inteins/physiology , Protein Splicing , Amino Acid Sequence , Calmodulin/genetics , Cell Engineering/methods , Exteins/genetics , Exteins/physiology , HeLa Cells , Humans , Inteins/genetics , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Protein Engineering/methods , Protein Sorting Signals/genetics , Red Fluorescent Protein
10.
mBio ; 9(1)2018 01 30.
Article in English | MEDLINE | ID: mdl-29382734

ABSTRACT

Inteins are intervening proteins that undergo an autocatalytic splicing reaction that ligates flanking host protein sequences termed exteins. Some intein-containing proteins have evolved to couple splicing to environmental signals; this represents a new form of posttranslational regulation. Of particular interest is RadA from the archaeon Pyrococcus horikoshii, for which long-range intein-extein interactions block splicing, requiring temperature and single-stranded DNA (ssDNA) substrate to splice rapidly and accurately. Here, we report that splicing of the intein-containing RadA from another archaeon, Thermococcus sibericus, is activated by significantly lower temperatures than is P. horikoshii RadA, consistent with differences in their growth environments. Investigation into variations between T. sibericus and P. horikoshii RadA inteins led to the discovery that a nonconserved region (NCR) of the intein, a flexible loop where a homing endonuclease previously resided, is critical to splicing. Deletion of the NCR leads to a substantial loss in the rate and accuracy of P. horikoshii RadA splicing only within native exteins. The influence of the NCR deletion can be largely overcome by ssDNA, demonstrating that the splicing-competent conformation can be achieved. We present a model whereby the NCR is a flexible hinge which acts as a switch by controlling distant intein-extein interactions that inhibit active site assembly. These results speak to the repurposing of the vestigial endonuclease loop to control an intein-extein partnership, which ultimately allows exquisite adaptation of protein splicing upon changes in the environment.IMPORTANCE Inteins are mobile genetic elements that interrupt coding sequences (exteins) and are removed by protein splicing. They are abundant elements in microbes, and recent work has demonstrated that protein splicing can be controlled by environmental cues, including the substrate of the intein-containing protein. Here, we describe an intein-extein collaboration that controls temperature-induced splicing of RadA from two archaea and how variation in this intein-extein partnership results in fine-tuning of splicing to closely match the environment. Specifically, we found that a small sequence difference between the two inteins, a flexible loop that likely once housed a homing endonuclease used for intein mobility, acts as a switch to control intein-extein interactions that block splicing. Our results argue strongly that some inteins have evolved away from a purely parasitic lifestyle to control the activity of host proteins, representing a new form of posttranslational regulation that is potentially widespread in the microbial world.


Subject(s)
Archaeal Proteins/metabolism , DNA-Binding Proteins/metabolism , Exteins , Inteins , Protein Splicing , Thermococcus/metabolism , Thermococcus/radiation effects , Archaeal Proteins/genetics , DNA-Binding Proteins/genetics , Models, Biological , Models, Molecular , Pyrococcus horikoshii/metabolism , Pyrococcus horikoshii/radiation effects , Sequence Deletion , Temperature
11.
Proc Natl Acad Sci U S A ; 114(32): 8538-8543, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739907

ABSTRACT

The protein trans-splicing (PTS) activity of naturally split inteins has found widespread use in chemical biology and biotechnology. However, currently used naturally split inteins suffer from an "extein dependence," whereby residues surrounding the splice junction strongly affect splicing efficiency, limiting the general applicability of many PTS-based methods. To address this, we describe a mechanism-guided protein engineering approach that imbues ultrafast DnaE split inteins with minimal extein dependence. The resulting "promiscuous" inteins are shown to be superior reagents for protein cyclization and protein semisynthesis, with the latter illustrated through the modification of native cellular chromatin. The promiscuous inteins reported here thus improve the applicability of existing PTS methods and should enable future efforts to engineer promiscuity into other naturally split inteins.


Subject(s)
Exteins/genetics , Inteins/genetics , Protein Engineering/methods , Bacterial Proteins/metabolism , Biotechnology , DNA Polymerase III/metabolism , Exteins/physiology , Inteins/physiology , Models, Molecular , Nostoc/genetics , Nostoc/metabolism , Protein Splicing/genetics , Synechocystis/metabolism
12.
Curr Biol ; 27(6): R204-R206, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-28324730

ABSTRACT

Lennon and Belfort introduce inteins - protein introns - and describe how they escape host proteins, their uses in biotechnology, where they are found in nature, and their role in post-translational regulation.


Subject(s)
Inteins/physiology , Animals , Biotechnology , Exteins/genetics , Exteins/physiology , Humans , Inteins/genetics
13.
Nucleic Acids Res ; 43(13): 6631-48, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26101259

ABSTRACT

Post-translational control based on an environmentally sensitive intervening intein sequence is described. Inteins are invasive genetic elements that self-splice at the protein level from the flanking host protein, the exteins. Here we show in Escherichia coli and in vitro that splicing of the RadA intein located in the ATPase domain of the hyperthermophilic archaeon Pyrococcus horikoshii is strongly regulated by the native exteins, which lock the intein in an inactive state. High temperature or solution conditions can unlock the intein for full activity, as can remote extein point mutations. Notably, this splicing trap occurs through interactions between distant residues in the native exteins and the intein, in three-dimensional space. The exteins might thereby serve as an environmental sensor, releasing the intein for full activity only at optimal growth conditions for the native organism, while sparing ATP consumption under conditions of cold-shock. This partnership between the intein and its exteins, which implies coevolution of the parasitic intein and its host protein may provide a novel means of post-translational control.


Subject(s)
Archaeal Proteins/chemistry , DNA-Binding Proteins/chemistry , Exteins , Inteins , Protein Splicing , Archaeal Proteins/metabolism , Bacterial Proteins/chemistry , DNA-Binding Proteins/metabolism , Models, Molecular , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Pyrococcus horikoshii/genetics , Rec A Recombinases/chemistry , Temperature
14.
Biomol NMR Assign ; 9(2): 235-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25281002

ABSTRACT

The backbone resonance assignments of an engineered splicing-inactive mini-RecA intein based on triple resonance experiments with [(13)C,(15)N]-labeled protein are reported. The construct contains inactivating mutations specifically designed to retain most catalytic residues, especially those that are potentially metal-coordinating. The assignments are essential for protein structure determination of a precursor with an active N-terminal catalytic cysteine and for investigation of the atomic details of splicing.


Subject(s)
Cysteine/chemistry , Exteins , Inteins , Nuclear Magnetic Resonance, Biomolecular , Rec A Recombinases/chemistry , Biocatalysis , Protein Engineering , Proton Magnetic Resonance Spectroscopy
15.
J Mol Biol ; 426(24): 4018-4029, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25451033

ABSTRACT

Inteins self-catalytically cleave out of precursor proteins while ligating the surrounding extein fragments with a native peptide bond. Much attention has been lavished on these molecular marvels with the hope of understanding and harnessing their chemistry for novel biochemical transformations including coupling peptides from synthetic or biological origins and controlling protein function. Despite an abundance of powerful applications, the use of inteins is still hampered by limitations in our understanding of their specificity (defined as flanking sequences that permit splicing) and the challenge of inserting inteins into target proteins. We examined the frequently used Nostoc punctiforme Npu DnaE intein after the C-extein cysteine nucleophile (Cys+1) was mutated to serine or threonine. Previous studies demonstrated reduced rates and/or splicing yields with the Npu DnaE intein after mutation of Cys+1 to Ser+1. In this study, genetic selection identified extein sequences with Ser+1 that enabled the Npu DnaE intein to splice with only a 5-fold reduction in rate compared to the wild-type Cys+1 intein and without mutation of the intein itself to activate Ser+1 as a nucleophile. Three different proteins spliced efficiently after insertion of the intein flanked by the selected sequences. We then used this selected specificity to achieve traceless splicing in a targeted enzyme at a location predicted by primary sequence similarity to only the selected C-extein sequence. This study highlights the latent catalytic potential of the Npu DnaE intein to splice with an alternative nucleophile and enables broader intein utility by increasing insertion site choices.


Subject(s)
Bacterial Proteins/genetics , DNA Polymerase III/genetics , Inteins/genetics , Mutation, Missense , Nostoc/genetics , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Blotting, Western , Catalytic Domain/genetics , Cysteine/genetics , DNA Polymerase III/chemistry , DNA Polymerase III/metabolism , Enzyme Activation , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Exteins/genetics , Molecular Sequence Data , Nostoc/enzymology , Protein Splicing , Serine/genetics , Substrate Specificity , Threonine/genetics
17.
J Biol Chem ; 289(21): 14498-505, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24695729

ABSTRACT

Inteins are nature's escape artists; they facilitate their excision from flanking polypeptides (exteins) concomitant with extein ligation to produce a mature host protein. Splicing requires sequential nucleophilic displacement reactions catalyzed by strategies similar to proteases and asparagine lyases. Inteins require precise reaction coordination rather than rapid turnover or tight substrate binding because they are single turnover enzymes with covalently linked substrates. This has allowed inteins to explore alternative mechanisms with different steps or to use different methods for activation and coordination of the steps. Pressing issues include understanding the underlying details of catalysis and how the splicing steps are controlled.


Subject(s)
Inteins/genetics , Models, Genetic , Protein Precursors/genetics , Protein Splicing/genetics , Amino Acids/chemistry , Amino Acids/genetics , Exteins/genetics , Molecular Structure , Protein Precursors/chemistry , Proteins/chemistry , Proteins/genetics
18.
Proc Natl Acad Sci U S A ; 111(23): 8422-7, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24778214

ABSTRACT

Inteins are autoprocessing domains that cut themselves out of host proteins in a traceless manner. This process, known as protein splicing, involves multiple chemical steps that must be coordinated to ensure fidelity in the process. The committed step in splicing involves attack of a conserved Asn side-chain amide on the adjacent backbone amide, leading to an intein-succinimide product and scission of that peptide bond. This cleavage reaction is stimulated by formation of a branched intermediate in the splicing process. The mechanism by which the Asn side-chain becomes activated as a nucleophile is not understood. Here we solve the crystal structure of an intein trapped in the branched intermediate step in protein splicing. Guided by this structure, we use protein-engineering approaches to show that intein-succinimide formation is critically dependent on a backbone-to-side-chain hydrogen-bond. We propose that this interaction serves to both position the side-chain amide for attack and to activate its nitrogen as a nucleophile. Collectively, these data provide an unprecedented view of an intein poised to carry out the rate-limiting step in protein splicing, shedding light on how a nominally nonnucleophilic group, a primary amide, can become activated in a protein active site.


Subject(s)
Exteins/genetics , Inteins/genetics , Protein Splicing , Proteins/genetics , Amides/chemistry , Amides/metabolism , Amino Acid Sequence , Asparagine/chemistry , Asparagine/genetics , Asparagine/metabolism , Catalytic Domain , DNA Gyrase/chemistry , DNA Gyrase/genetics , DNA Gyrase/metabolism , Electrophoresis, Polyacrylamide Gel , Hydrogen Bonding , Kinetics , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization
19.
Biochemistry ; 52(34): 5920-7, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23906287

ABSTRACT

Inteins are intervening polypeptides that catalyze their own removal from flanking exteins, concomitant to the ligation of the exteins. The intein that interrupts the DP2 (large) subunit of DNA polymerase II from Methanoculleus marisnigri (Mma) can promote protein splicing. However, protein splicing can be prevented or reduced by overexpression under nonreducing conditions because of the formation of a disulfide bond between two internal intein Cys residues. This redox sensitivity leads to differential activity in different strains of E. coli as well as in different cell compartments. The redox-dependent control of in vivo protein splicing in an intein derived from an anaerobe that can occupy multiple environments hints at a possible physiological role for protein splicing.


Subject(s)
Disulfides/pharmacology , Inteins/genetics , Protein Splicing/genetics , Cysteine/chemistry , DNA Polymerase II/genetics , Electrophoresis, Polyacrylamide Gel , Exteins/genetics , Oxidation-Reduction , Protein Splicing/drug effects , Tandem Mass Spectrometry
20.
J Am Chem Soc ; 135(20): 7713-9, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23621664

ABSTRACT

Conditional protein splicing is a powerful biotechnological tool that can be used to rapidly and post-translationally control the activity of a given protein. Here we demonstrate a novel conditional splicing system in which a genetically encoded protein scaffold induces the splicing and activation of an enzyme in mammalian cells. In this system the protein scaffold binds to two inactive split intein/enzyme extein protein fragments leading to intein fragment complementation, splicing, and activation of the firefly luciferase enzyme. We first demonstrate the ability of antiparallel coiled-coils (CCs) to mediate splicing between two intein fragments, effectively creating two new split inteins. We then generate and test two versions of the scaffold-induced splicing system using two pairs of CCs. Finally, we optimize the linker lengths of the proteins in the system and demonstrate 13-fold activation of luciferase by the scaffold compared to the activity of negative controls. Our protein scaffold-triggered conditional splicing system is an effective strategy to control enzyme activity using a protein input, enabling enhanced genetic control over protein splicing and the potential creation of splicing-based protein sensors and autoregulatory systems.


Subject(s)
Protein Splicing , Proteins/metabolism , Cells, Cultured , Enzyme Activation , Exteins , Humans , Inteins , Luciferases/chemistry , Luciferases/metabolism , Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...