Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioorg Med Chem ; 25(21): 5952-5961, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28988625

ABSTRACT

Vaccination is a reliable method of prophylaxis and a crucial measure for public health. However, the majority of vaccines cannot be administered orally due to their degradation in the harsh gut environment or inability to cross the GI tract. In this study, we report the first proof-of-concept study of orally producible chemically programmed antibodies via specific conjugation of adaptor ligands to endogenous antibodies, in vivo. Pre-immuniztion with 2,4-dinitrophenyl (DNP), or the reactive hapten, 1,3-diketone (DK), or a novel reactive hapten, vinyl sulfone (VS) in mice, followed by oral administration of adaptor ligands composed of the hapten and biotin to the pre-immunized mice resulted in successful in vivo formation of the biotin-hapten-antibody complexes within 2h. Pharmacokinetic evaluations revealed that apparent serum concentrations of programmed antibodies were up to 144nM and that the serum half-lives reached up to 34.4h. These findings show promise for the future development of orally bioavailable drug-hapten-antibody complexes asa strategy to quickly and easily modulate immune targets for aggressive pathogens as well as cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Biotin/immunology , Haptens/immunology , Ketones/immunology , Administration, Oral , Animals , Antibodies, Monoclonal/pharmacokinetics , Antigen-Antibody Reactions/drug effects , Biotin/administration & dosage , Haptens/administration & dosage , Ketones/administration & dosage , Ligands , Mice , Mice, Inbred BALB C , Molecular Structure
2.
Bioconjug Chem ; 27(10): 2271-2275, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27666414

ABSTRACT

Bioorthogonal labeling of antibodies enables the conjugation of compounds, such as small molecules or peptides, which expand targeting capacity or enhance cytotoxicity. Taking advantage of a cyclohexene sulfonamide compound that site-selectively labels Lys64 in human serum albumin (HSA), we demonstrate that domain I of HSA can be used as a fusion protein for the preparation of antibody conjugates. Trastuzumab fusions were expressed at the N-terminus of the light chain or the C-terminus of the heavy chain enabling conjugation to small molecules. Moreover, these conjugates retained HER2 binding and proved to be highly stable in human plasma. Antibody conjugation via HSA domain I fusion should therefore have broad utility for making serum-stable antibody conjugates, particularly for antibody-drug conjugates.


Subject(s)
Immunoconjugates/chemistry , Recombinant Fusion Proteins/chemistry , Serum Albumin/chemistry , Antibodies/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunoconjugates/blood , Immunoconjugates/metabolism , Lysine/chemistry , Protein Domains , Protein Engineering/methods , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/metabolism , Rhodamines/chemistry , Trastuzumab/chemistry
3.
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
4.
Nat Methods ; 9(8): 805-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22751204

ABSTRACT

Zinc-finger nucleases (ZFNs) are versatile reagents that have redefined genome engineering. Realizing the full potential of this technology requires the development of safe and effective methods for delivering ZFNs into cells. We demonstrate the intrinsic cell-penetrating capabilities of the standard ZFN architecture and show that direct delivery of ZFNs as proteins leads to efficient endogenous gene disruption in various mammalian cell types with minimal off-target effects.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Knockout Techniques/methods , Genetic Engineering/methods , Recombinant Fusion Proteins/metabolism , Zinc Fingers/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , CHO Cells , Catalytic Domain , Cell Membrane Permeability , Cricetinae , Deoxyribonucleases, Type II Site-Specific/chemistry , Humans , Molecular Sequence Data , Peptides/chemistry , Receptors, CCR5/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Zinc Fingers/genetics , tat Gene Products, Human Immunodeficiency Virus/chemistry
5.
Acc Chem Res ; 47(8): 2309-18, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-24877793

ABSTRACT

The understanding of gene regulation and the structure and function of the human genome increased dramatically at the end of the 20th century. Yet the technologies for manipulating the genome have been slower to develop. For instance, the field of gene therapy has been focused on correcting genetic diseases and augmenting tissue repair for more than 40 years. However, with the exception of a few very low efficiency approaches, conventional genetic engineering methods have only been able to add auxiliary genes to cells. This has been a substantial obstacle to the clinical success of gene therapies and has also led to severe unintended consequences in several cases. Therefore, technologies that facilitate the precise modification of cellular genomes have diverse and significant implications in many facets of research and are essential for translating the products of the Genomic Revolution into tangible benefits for medicine and biotechnology. To address this need, in the 1990s, we embarked on a mission to develop technologies for engineering protein-DNA interactions with the aim of creating custom tools capable of targeting any DNA sequence. Our goal has been to allow researchers to reach into genomes to specifically regulate, knock out, or replace any gene. To realize these goals, we initially focused on understanding and manipulating zinc finger proteins. In particular, we sought to create a simple and straightforward method that enables unspecialized laboratories to engineer custom DNA-modifying proteins using only defined modular components, a web-based utility, and standard recombinant DNA technology. Two significant challenges we faced were (i) the development of zinc finger domains that target sequences not recognized by naturally occurring zinc finger proteins and (ii) determining how individual zinc finger domains could be tethered together as polydactyl proteins to recognize unique locations within complex genomes. We and others have since used this modular assembly method to engineer artificial proteins and enzymes that activate, repress, or create defined changes to user-specified genes in human cells, plants, and other organisms. We have also engineered novel methods for externally controlling protein activity and delivery, as well as developed new strategies for the directed evolution of protein and enzyme function. This Account summarizes our work in these areas and highlights independent studies that have successfully used the modular assembly approach to create proteins with novel function. We also discuss emerging alternative methods for genomic targeting, including transcription activator-like effectors (TALEs) and CRISPR/Cas systems, and how they complement the synthetic zinc finger protein technology.


Subject(s)
DNA-Binding Proteins/metabolism , Genome, Human , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Early Growth Response Protein 1/chemistry , Early Growth Response Protein 1/metabolism , Gene Expression Regulation , Humans , Protein Structure, Tertiary , Recombinases/chemistry , Recombinases/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Zinc Fingers
6.
Bioorg Med Chem Lett ; 25(8): 1684-1687, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25791455

ABSTRACT

Utilization of chemically programmed antibodies (cpAbs) is regarded to be one of the most efficient methods for the development of therapeutic systems. cpAbs can extend the half-life of programming reagents, activate immune systems via the Fc region of antibodies and achieve universal vaccination by attaching varieties of small, programmed molecules. In the current study, we aimed to develop a novel labeling reagent for the preparation of cpAbs and found that N-sulfonyl-ß-lactams (NSBLs) were optimal. NSBL can be synthesized from readily available 4-(bromomethyl)benzenesulfonyl chloride via few simple manipulations and can label the aldolase monoclonal antibody (mAb) 84G3, which could not be labeled effectively by the conventional labeling reagent, N-acyl-ß-lactam (NABL). We also demonstrated that the conjugate, which consists of mAb 84G3 and an NSBL bearing a biotin moiety, maintained strong binding activity to streptavidin. In addition, the stability assay of NSBL revealed that NSBLs can tolerate aqueous media without significant decomposition over 24h.


Subject(s)
Antibodies, Monoclonal/chemistry , Fructose-Bisphosphate Aldolase/chemistry , Haptens/chemistry , Immunoglobulin Fab Fragments/chemistry , beta-Lactams/chemistry , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Biotin/chemistry , Biotin/metabolism , Fructose-Bisphosphate Aldolase/immunology , Immunoglobulin Fab Fragments/immunology , Protein Binding , Protein Stability , Streptavidin/chemistry , Streptavidin/metabolism
7.
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
8.
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
9.
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
10.
J Virol ; 87(9): 4985-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23427154

ABSTRACT

Broadly neutralizing antibodies PG9 and PG16 effectively neutralize 70 to 80% of circulating HIV-1 isolates. In this study, the neutralization abilities of PG9 and PG16 were further enhanced by bioconjugation with aplaviroc, a small-molecule inhibitor of virus entry into host cells. A novel air-stable diazonium hexafluorophosphate reagent that allows for rapid, tyrosine-selective functionalization of proteins and antibodies under mild conditions was used to prepare a series of aplaviroc-conjugated antibodies, including b12, 2G12, PG9, PG16, and CD4-IgG. The conjugated antibodies blocked HIV-1 entry through two mechanisms: by binding to the virus itself and by blocking the CCR5 receptor on host cells. Chemical modification did not significantly alter the potency of the parent antibodies against nonresistant HIV-1 strains. Conjugation did not alter the pharmacokinetics of a model IgG in blood. The PG9-aplaviroc conjugate was tested against a panel of 117 HIV-1 strains and was found to neutralize 100% of the viruses. PG9-aplaviroc conjugate IC50s were lower than those of PG9 in neutralization studies of 36 of the 117 HIV-1 strains. These results support this new approach to bispecific antibodies and offer a potential new strategy for combining HIV-1 therapies.


Subject(s)
Antibodies, Neutralizing/immunology , CD4 Immunoadhesins/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Immunoconjugates/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/pharmacology , Benzoates/chemistry , Benzoates/pharmacology , CD4 Immunoadhesins/chemistry , CD4 Immunoadhesins/pharmacology , Cell Line , Diketopiperazines , HIV Antibodies/chemistry , HIV Antibodies/pharmacology , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Neutralization Tests , Piperazines/chemistry , Piperazines/pharmacology , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Virus Internalization/drug effects
11.
Bioconjug Chem ; 25(8): 1402-7, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25099687

ABSTRACT

Current routes for synthesizing antibody-drug conjugates commonly rely on maleimide linkers to react with cysteine thiols. However, thioether exchange with metabolites and serum proteins can compromise conjugate stability and diminish in vivo efficacy. We report the application of a phenyloxadiazole sulfone linker for the preparation of trastuzumab conjugates. This sulfone linker site-specifically labeled engineered cysteine residues in THIOMABs and improved antibody conjugate stability in human plasma at sites previously shown to be labile for maleimide conjugates. Similarly, sulfone conjugation with selenocysteine in an anti-ROR1 scFv-Fc improved human plasma stability relative to maleimide conjugation. Kinetically controlled labeling of a THIOMAB containing two cysteine substitutions was also achieved, offering a strategy for producing antibody conjugates with expanded valency.


Subject(s)
Immunoconjugates/chemistry , Sulfones/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Binding Sites , Cell Line , Humans , Immunoconjugates/blood , Immunoglobulin Fc Fragments/chemistry , Models, Molecular , Oxadiazoles/chemistry , Protein Conformation , Protein Stability , Single-Chain Antibodies/chemistry , Substrate Specificity , Trastuzumab
12.
Biotechnol Bioeng ; 111(1): 1-15, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23982993

ABSTRACT

Site-specific recombinases are tremendously valuable tools for basic research and genetic engineering. By promoting high-fidelity DNA modifications, site-specific recombination systems have empowered researchers with unprecedented control over diverse biological functions, enabling countless insights into cellular structure and function. The rigid target specificities of many sites-specific recombinases, however, have limited their adoption in fields that require highly flexible recognition abilities. As a result, intense effort has been directed toward altering the properties of site-specific recombination systems by protein engineering. Here, we review key developments in the rational design and directed molecular evolution of site-specific recombinases, highlighting the numerous applications of these enzymes across diverse fields of study.


Subject(s)
Genetic Engineering , Metabolic Engineering , Recombinases , Amino Acid Sequence , Animals , Base Sequence , Humans , Mice , Models, Molecular , Molecular Sequence Data
13.
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
14.
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
15.
Angew Chem Int Ed Engl ; 53(44): 11783-6, 2014 Oct 27.
Article in English | MEDLINE | ID: mdl-25196737

ABSTRACT

Conjugation to human serum albumin (HSA) has emerged as a powerful approach for extending the in vivo half-life of many small molecule and peptide/protein drugs. Current HSA conjugation strategies, however, can often yield heterogeneous mixtures with inadequate pharmacokinetics, low efficacies, and variable safety profiles. Here, we designed and synthesized analogues of TAK-242, a small molecule inhibitor of Toll-like receptor 4, that primarily reacted with a single lysine residue of HSA. These TAK-242-based cyclohexene compounds demonstrated robust reactivity, and Lys64 was identified as the primary conjugation site. A bivalent HSA conjugate was also prepared in a site-specific manner. Additionally, HSA-cyclohexene conjugates maintained higher levels of stability both in human plasma and in mice than the corresponding maleimide conjugates. This new conjugation strategy promises to broadly enhance the performance of HSA conjugates for numerous applications.


Subject(s)
Lysine/chemistry , Serum Albumin/chemical synthesis , Humans
16.
Bioconjug Chem ; 24(4): 520-32, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23534985

ABSTRACT

The scope, chemoselectivity, and utility of the click-like tyrosine labeling reaction with 4-phenyl-3H-1,2,4-triazoline-3,5(4H)-diones (PTADs) is reported. To study the utility and chemoselectivity of PTAD derivatives in peptide and protein chemistry, we synthesized PTAD derivatives possessing azide, alkyne, and ketone groups and studied their reactions with amino acid derivatives and peptides of increasing complexity. With proteins we studied the compatibility of the tyrosine click reaction with cysteine and lysine-targeted labeling approaches and demonstrate that chemoselective trifunctionalization of proteins is readily achieved. In particular cases, we noted that PTAD decomposition resulted in formation of a putative isocyanate byproduct that was promiscuous in labeling. This side reaction product, however, was readily scavenged by the addition of a small amount of 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) to the reaction medium. To study the potential of the tyrosine click reaction to introduce poly(ethylene glycol) chains onto proteins (PEGylation), we demonstrate that this novel reagent provides for the selective PEGylation of chymotrypsinogen, whereas traditional succinimide-based PEGylation targeting lysine residues provided a more diverse range of PEGylated products. Finally, we applied the tyrosine click reaction to create a novel antibody-drug conjugate. For this purpose, we synthesized a PTAD derivative linked to the HIV entry inhibitor aplaviroc. Labeling of the antibody trastuzumab with this reagent provided a labeled antibody conjugate that demonstrated potent HIV-1 neutralization activity demonstrating the potential of this reaction in creating protein conjugates with small molecules. The tyrosine click linkage demonstrated stability to extremes of pH, temperature, and exposure to human blood plasma indicating that this linkage is significantly more robust than maleimide-type linkages that are commonly employed in bioconjugations. These studies support the broad utility of this reaction in the chemoselective modification of small molecules, peptides, and proteins under mild aqueous conditions over a broad pH range using a wide variety of biologically acceptable buffers such as phosphate buffered saline (PBS) and 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) buffers as well as others and mixed buffered compositions.


Subject(s)
Click Chemistry , Triazoles/chemistry , Tyrosine/chemistry , Humans , Hydrogen-Ion Concentration , Models, Molecular , Molecular Structure , Polyethylene Glycols/chemistry , Temperature
17.
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
18.
Proc Natl Acad Sci U S A ; 107(48): 20672-7, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-20639468

ABSTRACT

Given the significance of carbohydrates in life, medicine, and industry, the development of simple and efficient de novo methods to synthesize carbohydrates are highly desirable. Organocatalytic asymmetric assembly reactions are powerful tools to rapidly construct molecules with stereochemical complexity from simple precursors. Here, we present a simple and robust methodology for the asymmetric synthesis of pyranose derivatives with talo- and manno- configurations from simple achiral precursors through organocatalytic asymmetric intermolecular Michael-Henry reaction sequences. In this process, (tert-butyldimethylsilyloxy)acetaldehyde 1 was successfully utilized in two ways: as a donor in a highly selective anti-Michael reaction and as an acceptor in a consecutive Henry reaction. Varied nitroolefins served as Michael acceptors and varied aldehydes substituted for 1 as Henry acceptors providing for the construction of a wide range of carbohydrates with up to 5 stereocenters. In these reactions, a catalyst-controlled Michael reaction followed by a substrate-controlled Henry reaction provided 3,4-dideoxytalose derivatives 6 in a highly stereoselective manner. The Henry reaction was affected by addition of a simple base such as triethylamine: A complex chiral base was not necessary. 3,4-Dideoxymannose derivatives 7 were produced by simply changing the base from triethylamine to 1,8-diazabicyclo[5.4.0]undec-7-ene. Extension of this methodology to a syn-Michael initiated sequence was also successful. A mechanistic discussion is provided to explain the unusual substrate-induced stereoselectivity of the Henry reaction.


Subject(s)
Carbohydrates/chemistry , Carbohydrates/chemical synthesis , Models, Chemical , Catalysis , Crystallography, X-Ray , Lactones/chemistry , Mannose/chemistry
20.
Bioconjug Chem ; 23(12): 2321-8, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23181702

ABSTRACT

4-Formylbenzene diazonium hexafluorophosphate (FBDP) is a novel bench-stable crystalline diazonium salt that reacts selectively with tyrosine to install a bioorthogonal aldehyde functionality. Model studies with N-acyl-tyrosine methylamide allowed us to identify conditions optimal for tyrosine ligation reactions with small peptides and proteins. FBDP-based conjugation was used for the facile introduction of small molecule tags, poly(ethylene glycol) chains (PEGylation), and functional small molecules onto model proteins and to label the surface of living cells.


Subject(s)
Diazonium Compounds/chemistry , Indicators and Reagents/chemistry , Oligopeptides/chemistry , Phosphates/chemistry , Proteins/chemistry , Staining and Labeling/methods , Tyrosine/chemistry , Aldehydes/chemistry , Amides/chemistry , Animals , Biotinylation , Cattle , HeLa Cells , Humans , Hydrogen-Ion Concentration , Models, Molecular , Oligopeptides/analysis , Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL