Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Virol ; 97(11): e0116323, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37843374

ABSTRACT

IMPORTANCE: The use of adeno-associated viruses (AAVs) as gene delivery vectors has vast potential for the treatment of many severe human diseases. Over one hundred naturally existing AAV capsid variants have been described and classified into phylogenetic clades based on their sequences. AAV8, AAV9, AAVrh.10, and other intensively studied capsids have been propelled into pre-clinical and clinical use, and more recently, marketed products; however, less-studied capsids may also have desirable properties (e.g., potency differences, tissue tropism, reduced immunogenicity, etc.) that have yet to be thoroughly described. These data will help build a broader structure-function knowledge base in the field, present capsid engineering opportunities, and enable the use of novel capsids with unique properties.


Subject(s)
Dependovirus , Genetic Therapy , Genetic Vectors , Humans , Capsid , Capsid Proteins/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Phylogeny , Tissue Distribution
3.
Front Neurol ; 13: 1051559, 2022.
Article in English | MEDLINE | ID: mdl-36452163

ABSTRACT

Understanding the kinetics and durability of AAV-mediated transgene expression in the brain is essential for conducting basic neuroscience studies as well as for developing gene therapy approaches for CNS diseases. Here, we characterize and compare the temporal profile of transgene expression after bilateral injections into the mouse striatum of rAAV9 encoding GFP under the control of either a ubiquitous promoter (CAG), or the neuron-specific human synapsin (hSyn) and CamKII promoters. GFP protein expression with the CAG promoter was highest at 3 weeks, and then decreased to stable levels at 3 and 6 months. Surprisingly, GFP mRNA levels continued to increase from 3 weeks to 3 months, despite GFP protein expression decreasing during this time. GFP protein expression with hSyn increased more slowly, reaching a maximum at 3 months, which was equivalent to protein expression levels from CAG at that time point. Importantly, transgene expression driven by the hSyn promoter at 6 months was not silenced as previously reported, and GFP mRNA was continuing to rise even at the final 6-month time point. Thus, hSyn as a promoter for transgene expression demonstrates long-term durability but may require more time after vector administration to achieve steady-state levels. Because CAG had the highest GFP protein expression in our comparison, which was at 3 weeks post administration, the early kinetics of transgene expression from CAG was examined (1, 2, 5, and 10 days after injection). This analysis showed that GFP protein expression and GFP mRNA increased during the first 3 weeks after administration. Interestingly, vector DNA rapidly decreased 10-fold over the first 3 weeks following injection as it assembled into stable circular episomes and concatemers. Surprisingly, the processing of vector genomes into circular episomes and concatemers was continually dynamic up to 3 months after injection. These results provide novel insight into the dynamic processing of vector genomes and promoter-specific temporal patterns of transgene expression in the brain.

4.
Biotechnol Bioeng ; 118(5): 2001-2015, 2021 05.
Article in English | MEDLINE | ID: mdl-33580508

ABSTRACT

Age-related macular degeneration (AMD) associated with dysfunction of retinal pigment epithelial (RPE) cells is the most common cause of untreatable blindness. To advance gene therapy as a viable treatment for AMD there is a need for technologies that enable controlled, RPE-specific expression of therapeutic genes. Here we describe design, construction and testing of compact synthetic promoters with a pre-defined transcriptional activity and RPE cell specificity. Initial comparative informatic analyses of RPE and photoreceptor (PR) cell transcriptomic data identified conserved and overrepresented transcription factor regulatory elements (TFREs, 8-19 bp) specifically associated with transcriptionally active RPE genes. Both RPE-specific TFREs and those derived from the generically active cytomegalovirus-immediate early (CMV-IE) promoter were then screened in vitro to identify sequence elements able to control recombinant gene transcription in model induced pluripotent stem (iPS)-derived and primary human RPE cells. Two libraries of heterotypic synthetic promoters varying in predicted RPE specificity and transcriptional activity were designed de novo using combinations of up to 20 discrete TFREs in series (323-602 bp) and their transcriptional activity in model RPE cells was compared to that of the endogenous BEST1 promoter (661 bp, plus an engineered derivative) and the highly active generic CMV-IE promoter (650 bp). Synthetic promoters with a highpredicted specificity, comprised predominantly of endogenous TFREs exhibited a range of activities up to 8-fold that of the RPE-specific BEST1 gene promoter. Moreover, albeit at a lower predicted specificity, synthetic promoter transcriptional activity in model RPE cells was enhanced beyond that of the CMV-IE promoter when viral elements were utilized in combination with endogenous RPE-specific TFREs, with a reduction in promoter size of 15%. Taken together, while our data reveal an inverse relationship between synthetic promoter activity and cell-type specificity, cell context-specific control of recombinant gene transcriptional activity may be achievable.


Subject(s)
Genes, Synthetic/genetics , Genetic Therapy/methods , Promoter Regions, Genetic/genetics , Retinal Pigment Epithelium/cytology , Synthetic Biology/methods , Cells, Cultured , Epithelial Cells/cytology , Humans , Organ Specificity/genetics , Transcriptome/genetics
5.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 2): 58-64, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32039886

ABSTRACT

Adeno-associated viruses (AAVs) are used as in vivo gene-delivery vectors in gene-therapy products and have been heavily investigated for numerous indications. Over 100 naturally occurring AAV serotypes and variants have been isolated from primate samples. Many reports have described unique properties of these variants (for instance, differences in potency, target cell or evasion of the immune response), despite high amino-acid sequence conservation. AAVhu.37 is of interest for clinical applications owing to its proficient transduction of the liver and central nervous system. The sequence identity of the AAVhu.37 VP1 to the well characterized AAVrh.10 serotype, for which no structure is available, is greater than 98%. Here, the structure of the AAVhu.37 capsid at 2.56 Šresolution obtained via single-particle cryo-electron microscopy is presented.


Subject(s)
Capsid/ultrastructure , Cryoelectron Microscopy/methods , Dependovirus/chemistry , Genetic Vectors/chemistry , Mutation , Protein Conformation , Crystallography, X-Ray , Dependovirus/genetics , Genetic Vectors/genetics , Humans , Models, Molecular
6.
J Am Chem Soc ; 136(13): 5047-56, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24611715

ABSTRACT

Despite recent advances in genome engineering made possible by the emergence of site-specific endonucleases, there remains a need for tools capable of specifically delivering genetic payloads into the human genome. Hybrid recombinases based on activated catalytic domains derived from the resolvase/invertase family of serine recombinases fused to Cys2-His2 zinc-finger or TAL effector DNA-binding domains are a class of reagents capable of achieving this. The utility of these enzymes, however, has been constrained by their low overall targeting specificity, largely due to the formation of side-product homodimers capable of inducing off-target modifications. Here, we combine rational design and directed evolution to re-engineer the serine recombinase dimerization interface and generate a recombinase architecture that reduces formation of these undesirable homodimers by >500-fold. We show that these enhanced recombinases demonstrate substantially improved targeting specificity in mammalian cells and achieve rates of site-specific integration similar to those previously reported for site-specific nucleases. Additionally, we show that enhanced recombinases exhibit low toxicity and promote the delivery of the human coagulation factor IX and α-galactosidase genes into endogenous genomic loci with high specificity. These results provide a general means for improving hybrid recombinase specificity by protein engineering and illustrate the potential of these enzymes for basic research and therapeutic applications.


Subject(s)
Protein Engineering/methods , Recombinases/chemistry , Recombinases/genetics , Recombination, Genetic , Zinc Fingers , Amino Acid Sequence , Catalytic Domain , DNA/genetics , Directed Molecular Evolution/methods , Factor IX/genetics , Genome, Human , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinases/metabolism , alpha-Galactosidase/genetics
7.
Nucleic Acids Res ; 42(7): 4755-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24452803

ABSTRACT

The serine recombinases are a diverse family of modular enzymes that promote high-fidelity DNA rearrangements between specific target sites. Replacement of their native DNA-binding domains with custom-designed Cys2-His2 zinc-finger proteins results in the creation of engineered zinc-finger recombinases (ZFRs) capable of achieving targeted genetic modifications. The flexibility afforded by zinc-finger domains enables the design of hybrid recombinases that recognize a wide variety of potential target sites; however, this technology remains constrained by the strict recognition specificities imposed by the ZFR catalytic domains. In particular, the ability to fully reprogram serine recombinase catalytic specificity has been impeded by conserved base requirements within each recombinase target site and an incomplete understanding of the factors governing DNA recognition. Here we describe an approach to complement the targeting capacity of ZFRs. Using directed evolution, we isolated mutants of the ß and Sin recombinases that specifically recognize target sites previously outside the scope of ZFRs. Additionally, we developed a genetic screen to determine the specific base requirements for site-specific recombination and showed that specificity profiling enables the discovery of unique genomic ZFR substrates. Finally, we conducted an extensive and family-wide mutational analysis of the serine recombinase DNA-binding arm region and uncovered a diverse network of residues that confer target specificity. These results demonstrate that the ZFR repertoire is extensible and highlights the potential of ZFRs as a class of flexible tools for targeted genome engineering.


Subject(s)
Recombinases/chemistry , Recombinases/genetics , Zinc Fingers , Catalytic Domain , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Directed Molecular Evolution , Genome, Human , Humans , Mutagenesis , Recombinases/metabolism , Recombination, Genetic , Substrate Specificity
8.
ACS Synth Biol ; 3(10): 723-30, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-24251925

ABSTRACT

The construction of increasingly sophisticated synthetic biological circuits is dependent on the development of extensible tools capable of providing specific control of gene expression in eukaryotic cells. Here, we describe a new class of synthetic transcription factors that activate gene expression in response to extracellular chemical stimuli. These inducible activators consist of customizable transcription activator-like effector (TALE) proteins combined with steroid hormone receptor ligand-binding domains. We demonstrate that these ligand-responsive TALE transcription factors allow for tunable and conditional control of gene activation and can be used to regulate the expression of endogenous genes in human cells. Since TALEs can be designed to recognize any contiguous DNA sequence, the conditional gene regulatory system described herein will enable the design of advanced synthetic gene networks.


Subject(s)
Trans-Activators/metabolism , Base Sequence , DNA/genetics , DNA/metabolism , Gene Expression Regulation , Gene Regulatory Networks , HEK293 Cells , HeLa Cells , Humans , Ligands , Protein Engineering , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synthetic Biology , Trans-Activators/genetics
9.
Nucleic Acids Res ; 41(21): 9779-85, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23980031

ABSTRACT

Transcription activator-like effector (TALE) proteins can be designed to bind virtually any DNA sequence. General guidelines for design of TALE DNA-binding domains suggest that the 5'-most base of the DNA sequence bound by the TALE (the N0 base) should be a thymine. We quantified the N0 requirement by analysis of the activities of TALE transcription factors (TALE-TF), TALE recombinases (TALE-R) and TALE nucleases (TALENs) with each DNA base at this position. In the absence of a 5' T, we observed decreases in TALE activity up to >1000-fold in TALE-TF activity, up to 100-fold in TALE-R activity and up to 10-fold reduction in TALEN activity compared with target sequences containing a 5' T. To develop TALE architectures that recognize all possible N0 bases, we used structure-guided library design coupled with TALE-R activity selections to evolve novel TALE N-terminal domains to accommodate any N0 base. A G-selective domain and broadly reactive domains were isolated and characterized. The engineered TALE domains selected in the TALE-R format demonstrated modularity and were active in TALE-TF and TALEN architectures. Evolved N-terminal domains provide effective and unconstrained TALE-based targeting of any DNA sequence as TALE binding proteins and designer enzymes.


Subject(s)
DNA-Binding Proteins/chemistry , Deoxyribonucleases/chemistry , Recombinases/chemistry , Transcription Factors/chemistry , Base Sequence , Binding Sites , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/metabolism , Deoxyribonucleases/metabolism , Directed Molecular Evolution , Protein Binding , Protein Structure, Tertiary , Recombinases/metabolism , Transcription Factors/metabolism
10.
Nucleic Acids Res ; 41(6): 3937-46, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23393187

ABSTRACT

Zinc-finger recombinases (ZFRs) represent a potentially powerful class of tools for targeted genetic engineering. These chimeric enzymes are composed of an activated catalytic domain derived from the resolvase/invertase family of serine recombinases and a custom-designed zinc-finger DNA-binding domain. The use of ZFRs, however, has been restricted by sequence requirements imposed by the recombinase catalytic domain. Here, we combine substrate specificity analysis and directed evolution to develop a diverse collection of Gin recombinase catalytic domains capable of recognizing an estimated 3.77 × 10(7) unique DNA sequences. We show that ZFRs assembled from these engineered catalytic domains recombine user-defined DNA targets with high specificity, and that designed ZFRs integrate DNA into targeted endogenous loci in human cells. This study demonstrates the feasibility of generating customized ZFRs and the potential of ZFR technology for a diverse range of applications, including genome engineering, synthetic biology and gene therapy.


Subject(s)
DNA Nucleotidyltransferases/chemistry , Gene Targeting , Recombinases/chemistry , Zinc Fingers , Amino Acid Sequence , Catalytic Domain , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Directed Molecular Evolution , Genome, Human , HEK293 Cells , Humans , Molecular Sequence Data , Protein Engineering , Recombinases/genetics , Recombinases/metabolism , Recombination, Genetic , Substrate Specificity
11.
Nucleic Acids Res ; 40(21): 11163-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23019222

ABSTRACT

Site-specific recombinases are powerful tools for genome engineering. Hyperactivated variants of the resolvase/invertase family of serine recombinases function without accessory factors, and thus can be re-targeted to sequences of interest by replacing native DNA-binding domains (DBDs) with engineered zinc-finger proteins (ZFPs). However, imperfect modularity with particular domains, lack of high-affinity binding to all DNA triplets, and difficulty in construction has hindered the widespread adoption of ZFPs in unspecialized laboratories. The discovery of a novel type of DBD in transcription activator-like effector (TALE) proteins from Xanthomonas provides an alternative to ZFPs. Here we describe chimeric TALE recombinases (TALERs): engineered fusions between a hyperactivated catalytic domain from the DNA invertase Gin and an optimized TALE architecture. We use a library of incrementally truncated TALE variants to identify TALER fusions that modify DNA with efficiency and specificity comparable to zinc-finger recombinases in bacterial cells. We also show that TALERs recombine DNA in mammalian cells. The TALER architecture described herein provides a platform for insertion of customized TALE domains, thus significantly expanding the targeting capacity of engineered recombinases and their potential applications in biotechnology and medicine.


Subject(s)
DNA Nucleotidyltransferases/chemistry , DNA-Binding Proteins/chemistry , Protein Engineering , Recombinases/chemistry , Base Sequence , Catalytic Domain , DNA/chemistry , DNA/metabolism , DNA Nucleotidyltransferases/genetics , DNA-Binding Proteins/genetics , Gene Library , HEK293 Cells , Humans , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinases/metabolism , Repetitive Sequences, Amino Acid
12.
Nucleic Acids Res ; 39(17): 7868-78, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21653554

ABSTRACT

The development of new methods for gene addition to mammalian genomes is necessary to overcome the limitations of conventional genetic engineering strategies. Although a variety of DNA-modifying enzymes have been used to directly catalyze the integration of plasmid DNA into mammalian genomes, there is still an unmet need for enzymes that target a single specific chromosomal site. We recently engineered zinc-finger recombinase (ZFR) fusion proteins that integrate plasmid DNA into a synthetic target site in the human genome with exceptional specificity. In this study, we present a two-step method for utilizing these enzymes in any cell type at randomly-distributed target site locations. The piggyBac transposase was used to insert recombinase target sites throughout the genomes of human and mouse cell lines. The ZFR efficiently and specifically integrated a transfected plasmid into these genomic target sites and into multiple transposons within a single cell. Plasmid integration was dependent on recombinase activity and the presence of recombinase target sites. This work demonstrates the potential for broad applicability of the ZFR technology in genome engineering, synthetic biology and gene therapy.


Subject(s)
Gene Targeting , Genome, Human , Plasmids/genetics , Recombinases/metabolism , Zinc Fingers , Animals , Cell Line , Humans , Mice , Protein Engineering , Recombinant Fusion Proteins , Recombinases/genetics , Transposases/genetics , Transposases/metabolism , Zinc Fingers/genetics
13.
Proc Natl Acad Sci U S A ; 108(2): 498-503, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187418

ABSTRACT

Routine manipulation of cellular genomes is contingent upon the development of proteins and enzymes with programmable DNA sequence specificity. Here we describe the structure-guided reprogramming of the DNA sequence specificity of the invertase Gin from bacteriophage Mu and Tn3 resolvase from Escherichia coli. Structure-guided and comparative sequence analyses were used to predict a network of amino acid residues that mediate resolvase and invertase DNA sequence specificity. Using saturation mutagenesis and iterative rounds of positive antibiotic selection, we identified extensively redesigned and highly convergent resolvase and invertase populations in the context of engineered zinc-finger recombinase (ZFR) fusion proteins. Reprogrammed variants selectively catalyzed recombination of nonnative DNA sequences > 10,000-fold more effectively than their parental enzymes. Alanine-scanning mutagenesis revealed the molecular basis of resolvase and invertase DNA sequence specificity. When used as rationally designed ZFR heterodimers, the reprogrammed enzyme variants site-specifically modified unnatural and asymmetric DNA sequences. Early studies on the directed evolution of serine recombinase DNA sequence specificity produced enzymes with relaxed substrate specificity as a result of randomly incorporated mutations. In the current study, we focused our mutagenesis exclusively on DNA determinants, leading to redesigned enzymes that remained highly specific and directed transgene integration into the human genome with > 80% accuracy. These results demonstrate that unique resolvase and invertase derivatives can be developed to site-specifically modify the human genome in the context of zinc-finger recombinase fusion proteins.


Subject(s)
DNA Nucleotidyltransferases/genetics , Recombinases/genetics , Serine/chemistry , Transposon Resolvases/genetics , Amino Acid Sequence , Bacteriophage mu/metabolism , Dimerization , Escherichia coli/enzymology , Gene Targeting , Genome, Human , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Conformation , Protein Engineering/methods , Protein Structure, Secondary , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Transgenes
14.
Chembiochem ; 10(6): 1091-100, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19308927

ABSTRACT

Carrier proteins are central to the biosynthesis of primary and secondary metabolites in all organisms. Here we describe metabolic labeling and manipulation of native acyl carrier proteins in both type I and II fatty acid synthases. By utilizing natural promiscuity in the CoA biosynthetic pathway in combination with synthetic pantetheine analogues, we demonstrate metabolic labeling of endogenous carrier proteins with reporter tags in Gram-positive and Gram-negative bacteria and in a human carcinoma cell line. The highly specific nature of the post-translational modification that was utilized for tagging allows for simple visualization of labeled carrier proteins, either by direct fluorescence imaging or after chemical conjugation to a fluorescent reporter. In addition, we demonstrate the utility of this approach for the isolation and enrichment of carrier proteins by affinity purification. Finally, we use these techniques to identify a carrier protein from an unsequenced organism, a finding that validates this proteomic approach to natural product biosynthetic enzyme discovery.


Subject(s)
Acyl Carrier Protein/chemistry , Acyl Carrier Protein/metabolism , Acyl Carrier Protein/analysis , Acyl Carrier Protein/isolation & purification , Affinity Labels/metabolism , Amino Acid Sequence , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Base Sequence , Cell Line, Tumor , Cell Survival , Fatty Acids/biosynthesis , Gene Knockdown Techniques , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Protein Transport , Proteomics , Sequence Analysis, DNA , Staining and Labeling , fas Receptor/metabolism
15.
Bioorg Med Chem Lett ; 18(22): 5991-4, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18701278

ABSTRACT

Pantothenamides have been the subject of much study as potential inhibitors of CoA and carrier protein dependent biosynthetic pathways. Based on an initial observation of growth inhibition in Escherichia coli by 3, we have synthesized a small panel of pantetheine analogues and re-examined the inhibitory properties of this class of antibiotics with an emphasis on understanding the ability of these compounds to act as substrates of native CoA and carrier protein utilizing biosynthetic pathways. Our findings suggest that a secondary structure-activity relationship is an important factor in the antibiotic activity of these compounds.


Subject(s)
Antimetabolites/metabolism , Coenzyme A/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Pantetheine/analogs & derivatives , Aldehyde-Lyases/analysis , Aldehyde-Lyases/metabolism , Anti-Bacterial Agents/metabolism , Combinatorial Chemistry Techniques , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/metabolism , Molecular Structure , Pantetheine/chemical synthesis , Pantetheine/chemistry , Pantetheine/metabolism , Structure-Activity Relationship
16.
J Am Chem Soc ; 130(16): 5443-5, 2008 Apr 23.
Article in English | MEDLINE | ID: mdl-18376827

ABSTRACT

The study of the enzymes responsible for natural product biosynthesis has proven a valuable source of new enzymatic activities and been applied to a number of biotechnology applications. Protein profiling could prove highly complementary to genetics based approaches by allowing us to understand the activity, transcriptional control, and post-translational modification of these enzymes in their native and dynamic proteomic environments. Here we present a method for the fluorescent profiling of PKS, NRPS, and FAS multidomain modular synthases in their whole proteomes using complementary metabolic and activity based probes. After first examining the reactivity of these activity based probes with a variety of purified recombinant PKS, NRPS, and FAS enzymes in vitro, we apply this duel labeling strategy to the analysis of modular synthases in a human breast cancer cell line and two strains of the natural product producer Bacillus subtilis. Collectively, these studies demonstrate that complementary protein profiling approaches can prove highly useful in the identification and assignment of inhibitor specificity and domain structure of these modular biosynthetic enzymes.


Subject(s)
Affinity Labels/chemistry , Biotechnology/methods , Fatty Acid Synthases/biosynthesis , Fluorescent Dyes/chemistry , Peptide Synthases/biosynthesis , Polyketide Synthases/biosynthesis , Proteomics/methods , Affinity Labels/metabolism , Bacillus subtilis/enzymology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Fatty Acid Synthases/genetics , Female , Fluorescent Dyes/metabolism , Humans , Peptide Synthases/genetics , Polyketide Synthases/genetics , Substrate Specificity , Tumor Cells, Cultured
17.
Nat Prod Rep ; 24(4): 750-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17653358

ABSTRACT

Nature has developed a remarkable strategy to isolate metabolites from the milieu of the cell for chemical modification through the use of carrier proteins. Common to both primary and secondary metabolic pathways, acyl-carrier proteins constitute a conserved protein architecture which mediate the biosynthesis of a variety of metabolic products. Analogies have been made between the carrier protein and solid phase resin for chemical synthesis, as both entities provide a mechanism to separate compounds of interest from complex mixtures for selective chemical modification. However, there is significantly more to the carrier protein than an attachment point. In this review, we aim to systematically characterize the role of carrier proteins in various metabolic pathways and outline their utility in biosynthesis and biotechnology; 185 references are cited.


Subject(s)
Carrier Proteins , Bacteria/enzymology , Bacteria/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Carrier Proteins/physiology , Fungi/enzymology , Fungi/metabolism , Molecular Structure , Plants/enzymology , Plants/metabolism
18.
J Am Chem Soc ; 128(37): 12174-84, 2006 Sep 20.
Article in English | MEDLINE | ID: mdl-16967968

ABSTRACT

In vivo carrier protein tagging has recently become an attractive target for the site-specific modification of fusion systems and new approaches to natural product proteomics. A detailed study of pantetheine analogues was performed in order to identify suitable partners for covalent protein labeling inside living cells. A rapid synthesis of pantothenamide analogues was developed and used to produce a panel which was evaluated for in vitro and in vivo protein labeling. Kinetic comparisons allowed the construction of a structure-activity relationship to pinpoint the linker, dye, and bioorthogonal reporter of choice for carrier protein labeling. Finally bioorthogonal pantetheine analogues were shown to target carrier proteins with high specificity in vivo and undergo chemoselective ligation to reporters in crude cell lysate. The methods demonstrated here allow carrier proteins to be visualized and isolated for the first time without the need for antibody techniques and set the stage for the future use of carrier protein fusions in chemical biology.


Subject(s)
Carrier Proteins/metabolism , Pantetheine/analogs & derivatives , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Coenzyme A/chemistry , Coenzyme A/metabolism , Escherichia coli/metabolism , Kinetics , Pantetheine/chemical synthesis , Pantetheine/chemistry , Pantetheine/metabolism , Structure-Activity Relationship , Substrate Specificity , Transferases (Other Substituted Phosphate Groups)/chemistry , Transferases (Other Substituted Phosphate Groups)/metabolism
20.
J Am Chem Soc ; 127(32): 11234-5, 2005 Aug 17.
Article in English | MEDLINE | ID: mdl-16089439

ABSTRACT

We demonstrate site-specific reporter labeling of proteins within live cells. By accessing the coenzyme A (CoA) metabolic pathway with a cell-permeable pantetheine analogue, we deliver CoA-bound reporter molecules for post-translational protein modification in vivo. These methods may be applied to natural product pathway manipulation as well as applications in conventional molecular and cellular biology.


Subject(s)
Carrier Proteins/metabolism , Coenzyme A/metabolism , Fluorescent Dyes/metabolism , Pantetheine/metabolism , Carrier Proteins/chemistry , Coenzyme A/chemistry , Escherichia coli/growth & development , Escherichia coli/metabolism , Fluorescent Dyes/chemistry , Pantetheine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...