Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 180(4): 717-728.e19, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32084341

ABSTRACT

Consumption of glucosinolates, pro-drug-like metabolites abundant in Brassica vegetables, has been associated with decreased risk of certain cancers. Gut microbiota have the ability to metabolize glucosinolates, generating chemopreventive isothiocyanates. Here, we identify a genetic and biochemical basis for activation of glucosinolates to isothiocyanates by Bacteroides thetaiotaomicron, a prominent gut commensal species. Using a genome-wide transposon insertion screen, we identified an operon required for glucosinolate metabolism in B. thetaiotaomicron. Expression of BT2159-BT2156 in a non-metabolizing relative, Bacteroides fragilis, resulted in gain of glucosinolate metabolism. We show that isothiocyanate formation requires the action of BT2158 and either BT2156 or BT2157 in vitro. Monocolonization of mice with mutant BtΔ2157 showed reduced isothiocyanate production in the gastrointestinal tract. These data provide insight into the mechanisms by which a common gut bacterium processes an important dietary nutrient.


Subject(s)
Bacteroides thetaiotaomicron/metabolism , Dietary Carbohydrates/metabolism , Glucosinolates/metabolism , Intestines/microbiology , Animals , Bacteroides thetaiotaomicron/genetics , Bacteroides thetaiotaomicron/pathogenicity , Gene Expression Regulation, Bacterial , Humans , Male , Mice , Operon , Symbiosis
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
bioRxiv ; 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38168418

ABSTRACT

Bacteroides species are abundant and prevalent stably colonizing members of the human gut microbiota, making them a promising chassis for developing long-term interventions for chronic diseases. Engineering these bacteria as on-site production and delivery vehicles for biologic drugs or diagnostics, however, requires efficient heterologous protein secretion tools, which are currently lacking. To address this limitation, we systematically investigated methods to enable heterologous protein secretion in Bacteroides using both endogenous and exogenous secretion systems. Here, we report a collection of secretion carriers that can export functional proteins across multiple Bacteroides species at high titers. To understand the mechanistic drivers of Bacteroides secretion, we characterized signal peptide sequence features as well as post-secretion extracellular fate and cargo size limit of protein cargo. To increase titers and enable flexible control of protein secretion, we developed a strong, self-contained, inducible expression circuit. Finally, we validated the functionality of our secretion carriers in vivo in a mouse model. This toolkit should enable expanded development of long-term living therapeutic interventions for chronic gastrointestinal disease.

9.
Tumour Biol ; 33(3): 669-77, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22392499

ABSTRACT

Combining the specificity of tumor-targeting antibodies with the sensitivity and quantification offered by positron emission tomography (PET) provides tremendous opportunities for molecular characterization of tumors in vivo. Until recently, significant challenges have been faced when attempting to combine antibodies which show long biological half-lives and positron-emitting radionuclides with comparably short physical half-lives, in particular (18)F (half-life, 109 min). A fast and simple microwave-assisted method of generating N-succinimidyl-4-[(18)F]fluorobenzoate has been developed and employed for radiolabeling a small, rapidly targeting HER2-specific engineered antibody fragment, the cys-diabody. Using this tracer, HER2-positive tumor xenografts in mice were detected at 1-4 h post-injection by microPET. This confirms the rapid kinetics of [(18)F]fluorobenzoyl cys-diabody localization, and demonstrates the feasibility of same-day immunoPET imaging. This approach can be broadly applied to antibodies targeting cell surface biomarkers for molecular imaging of tumors and should be highly translatable for clinical use.


Subject(s)
Fluorine Radioisotopes , Immunoconjugates , Multimodal Imaging , Positron-Emission Tomography , Single-Chain Antibodies , Tomography, X-Ray Computed , Animals , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Female , Fluorine Radioisotopes/chemistry , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Mice , Mice, Nude , Molecular Imaging , Receptor, ErbB-2/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Transplantation, Heterologous
10.
ACS Chem Biol ; 17(2): 404-413, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35050570

ABSTRACT

Therapeutic antibodies have become one of the most widely used classes of biotherapeutics due to their unique antigen specificity and their ability to be engineered against diverse disease targets. There is significant interest in utilizing truncated antibody fragments as therapeutics, as their small size affords favorable properties such as increased tumor penetration as well as the ability to utilize lower-cost prokaryotic production methods. Their small size and simple architecture, however, also lead to rapid blood clearance, limiting the efficacy of these potentially powerful therapeutics. A common approach to circumvent these limitations is to enable engagement with the half-life extending neonatal Fc receptor (FcRn). This is usually achieved via fusion with a large Fc domain, which negates the benefits of the antibody fragment's small size. In this work, we show that modifying antibody fragments with short FcRn-binding peptide domains that mimic native IgG engagement with FcRn enables binding and FcRn-mediated recycling and transmembrane transcytosis in cell-based assays. Further, we show that rational, single amino acid mutations to the peptide sequence have a significant impact on the receptor-mediated function and investigate the underlying structural basis for this effect using computational modeling. Finally, we report the identification of a short peptide from human serum albumin that enables FcRn-mediated function when grafted onto a single-chain variable fragment (scFv) scaffold, establishing an approach for the rational selection of short-peptide domains from full-length proteins that could enable the transfer of non-native functions to small recombinant proteins without significantly impacting their size or structure.


Subject(s)
Single-Chain Antibodies , Antibody Specificity , Half-Life , Histocompatibility Antigens Class I/metabolism , Humans , Infant, Newborn , Receptors, Fc/genetics , Receptors, Fc/metabolism , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/metabolism , Transcytosis
11.
Mol Imaging ; 10(3): 168-76, 1-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21496447

ABSTRACT

Methods for tagging biomolecules with fluorine 18 as immuno-positron emission tomography (immunoPET) tracers require tedious optimization of radiolabeling conditions and can consume large amounts of scarce biomolecules. We describe an improved method using a digital microfluidic droplet generation (DMDG) chip, which provides computer-controlled metering and mixing of 18F tag, biomolecule, and buffer in defined ratios, allowing rapid scouting of reaction conditions in nanoliter volumes. The identified optimized conditions were then translated to bench-scale 18F labeling of a cancer-specific engineered antibody fragments, enabling microPET imaging of tumors in xenografted mice at 0.5 to 4 hours postinjection.


Subject(s)
Fluorine Radioisotopes , Isotope Labeling/methods , Microfluidics/methods , Positron-Emission Tomography/methods , Prostate-Specific Antigen/immunology , Animals , Fluorine Radioisotopes/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Male , Mice , Mice, SCID , Tissue Distribution , Tomography, X-Ray Computed
12.
ACS Synth Biol ; 9(12): 3184-3201, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33205966

ABSTRACT

Modern medicine has long studied the mechanism and impact of pathogenic microbes on human hosts, but has only recently shifted attention toward the complex and vital roles that commensal and probiotic microbes play in both health and dysbiosis. Fueled by an enhanced appreciation of the human-microbe holobiont, the past decade has yielded countless insights and established many new avenues of investigation in this area. In this review, we discuss advances, limitations, and emerging frontiers for microbes as agents of health maintenance, disease prevention, and cure. We highlight the flexibility of microbial therapeutics across disease states, with special consideration for the rational engineering of microbes toward precision medicine outcomes. As the field advances, we anticipate that tools of synthetic biology will be increasingly employed to engineer functional living therapeutics with the potential to address longstanding limitations of traditional drugs.


Subject(s)
Precision Medicine , Antigens/genetics , Antigens/metabolism , Bacteria/genetics , Bacteria/metabolism , Dysbiosis , Gastrointestinal Microbiome , Genetic Engineering , Humans , Metabolic Diseases/therapy , Metabolic Engineering , Probiotics/administration & dosage
13.
Bioconjug Chem ; 20(8): 1474-81, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19642689

ABSTRACT

The present work demonstrates the use of small bivalent engineered antibody fragments, cys-diabodies, for biological modification of nanoscale particles such as quantum dots (Qdots) for detection of target antigens. Novel bioconjugated quantum dots known as immunoQdots (iQdots) were developed by thiol-specific oriented coupling of tumor specific cys-diabodies, at a position away from the antigen binding site to amino PEG CdSe/ZnS Qdots. Initially, amino PEG Qdot 655 were coupled with reduced anti-HER2 cys-diabody by amine-sulfhydryl-reactive linker [N-ε-maleimidocaproyloxy] succinimide ester (EMCS) to produce anti-HER2 iQdot 655. Spectral characterization of the conjugate revealed that the spectrum was symmetrical and essentially identical to unconjugated Qdot. Specific receptor binding activity of anti-HER2 iQdot 655 was confirmed by flow cytometry on HER2 positive and negative cells. Immunofluorescence results showed homogeneous surface labeling of the cell membrane with Qdot 655 conjugate. In addition, cys-diabodies specific for HER2, as well as prostate stem cell antigen (PSCA), were conjugated successfully with amino PEG Qdot 800. All of these iQdots retain the photoluminescence properties of the unconjugated Qdot 800 as well as the antigen binding specificity of the cys-diabody as demonstrated by flow cytometry. Simultaneous detection of two tumor antigens on LNCaP/PSCA prostate cancer cells (which express PSCA and HER2) in culture was possible using two iQdots, anti-HER2 iQdot 655 and anti-PSCA iQdot 800. Thus, these iQdots are potentially useful as optical probes for sensitive, multiplexed detection of surface markers on tumor cells. The present thiol-specific conjugation method demonstrates a general approach for site-specific oriented coupling of cys-diabodies to a wide variety of nanoparticles without disturbing the antigen binding site and maintaining small size compared to intact antibody.


Subject(s)
Antibodies/chemistry , Biomarkers, Tumor/analysis , Immunoconjugates/chemistry , Neoplasms/diagnosis , Quantum Dots , Animals , Antibodies/immunology , Antibodies/isolation & purification , Cadmium Compounds/chemistry , Cell Line, Tumor , Humans , Immunoconjugates/immunology , Luminescence , Mice , Polyethylene Glycols/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Selenium Compounds/chemistry , Sulfides/chemistry , Zinc Compounds/chemistry
14.
Bioconjug Chem ; 19(12): 2527-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19053310

ABSTRACT

Small, engineered antibody fragments such as diabodies (50 kDa noncovalent dimers of single-chain Fv fragments) are useful alternatives to their larger antibody counterparts. However, due to their size, they are more susceptible to disruption of their antigen binding sites when modified using random conjugation techniques. Previous work has demonstrated the utility of a C-terminal cysteine modification for site-specific radiolabeling of an anti-CEA diabody, resulting in the creation of a cys-diabody (CysDb). In the present work, the adaptability of the CysDb system was explored by creating two additional CysDbs: one specific for CD20 and one for HER2. Purified CysDbs of both specificities demonstrated behavior consistent with stable, covalent dimers harboring a readily reducible disulfide bond. Each CysDb was site-specifically conjugated to three different fluorophores for optical detection: the large fluorescent proteins phycoerythrin (PE) and allophycocyanin (APC), and the small fluorescent molecule Alexa Fluor488. Fluorophore-conjugated CysDbs bound specifically to their targets in both antigen systems and with each different fluorescent tag as determined by flow cytometry. In vitro specific antigen binding was observed in the presence of a mixture of specific and nonspecifically conjugated CysDbs. Conjugates retained both specificity and fluorescence, demonstrating the successful expansion of the CysDb repertoire to new targets and to new site-specific conjugation possibilities.


Subject(s)
Antigens, CD20/immunology , Cysteine/metabolism , Fluorescent Dyes/chemistry , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Receptor, ErbB-2/immunology , Sulfhydryl Compounds/chemistry , Animals , Binding Sites , Cell Line, Tumor , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Sensitivity and Specificity
15.
Methods Mol Biol ; 1867: 89-102, 2018.
Article in English | MEDLINE | ID: mdl-30155817

ABSTRACT

Over the past several years, genome engineering has become an established component of basic research endeavors, and is emerging as a vital element of clinical research applications. Site-specific recombinases are one of the several tools that can facilitate genome modification by catalyzing rearrangements between specific DNA targets. Of particular interest are the small serine recombinases, which are modular in both form and function. This unique structure permits replacement of the native DNA-binding domain with designer targeting modules such as zinc fingers, TALEs, or catalytically inactivated Cas9, enabling modification of investigator-defined genomic loci. Importantly, the catalytic domain of these enzymes also contributes to target specificity, and can be reprogrammed to recognize custom sequences for genomic targeting. Here we describe the steps required to construct, select, and validate hybrid recombinase catalytic domains for targeted genome engineering.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Directed Molecular Evolution , Genome, Human , Protein Engineering/methods , Catalytic Domain , DNA Nucleotidyltransferases/genetics , HEK293 Cells , Humans , Recombination, Genetic , Zinc Fingers
16.
Article in English | MEDLINE | ID: mdl-27908936

ABSTRACT

Targeted nucleases have provided researchers with the ability to manipulate virtually any genomic sequence, enabling the facile creation of isogenic cell lines and animal models for the study of human disease, and promoting exciting new possibilities for human gene therapy. Here we review three foundational technologies-clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9), transcription activator-like effector nucleases (TALENs), and zinc-finger nucleases (ZFNs). We discuss the engineering advances that facilitated their development and highlight several achievements in genome engineering that were made possible by these tools. We also consider artificial transcription factors, illustrating how this technology can complement targeted nucleases for synthetic biology and gene therapy.


Subject(s)
Gene Editing , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/metabolism , Humans
17.
PLoS One ; 9(1): e85755, 2014.
Article in English | MEDLINE | ID: mdl-24465685

ABSTRACT

Transcription activator-like (TAL) effector nucleases (TALENs) have enabled the introduction of targeted genetic alterations into a broad range of cell lines and organisms. These customizable nucleases are comprised of programmable sequence-specific DNA-binding modules derived from TAL effector proteins fused to the non-specific FokI cleavage domain. Delivery of these nucleases into cells has proven challenging as the large size and highly repetitive nature of the TAL effector DNA-binding domain precludes their incorporation into many types of viral vectors. Furthermore, viral and non-viral gene delivery methods carry the risk of insertional mutagenesis and have been shown to increase the off-target activity of site-specific nucleases. We previously demonstrated that direct delivery of zinc-finger nuclease proteins enables highly efficient gene knockout in a variety of mammalian cell types with reduced off-target effects. Here we show that conjugation of cell-penetrating poly-Arg peptides to a surface-exposed Cys residue present on each TAL effector repeat imparted cell-penetrating activity to purified TALEN proteins. These modifications are reversible under reducing conditions and enabled TALEN-mediated gene knockout of the human CCR5 and BMPR1A genes at rates comparable to those achieved with transient transfection of TALEN expression vectors. These findings demonstrate that direct protein delivery, facilitated by conjugation of chemical functionalities onto the TALEN protein surface, is a promising alternative to current non-viral and viral-based methods for TALEN delivery into mammalian cells.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Endonucleases/administration & dosage , Gene Targeting/methods , Genetic Engineering/methods , Cell Proliferation , Cell-Penetrating Peptides/genetics , Endonucleases/genetics , HEK293 Cells , HeLa Cells , Humans , Mutagenesis, Site-Directed , Receptors, CCR5/genetics , Receptors, CCR5/metabolism
18.
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
19.
ACS Chem Biol ; 9(8): 1662-7, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24936957

ABSTRACT

The development of new methods for delivering proteins into cells is a central challenge for advancing both basic research and therapeutic applications. We previously reported that zinc-finger nuclease proteins are intrinsically cell-permeable due to the cell-penetrating activity of the Cys2-His2 zinc-finger domain. Here, we demonstrate that genetically fused zinc-finger motifs can transport proteins and enzymes into a wide range of primary and transformed mammalian cell types. We show that zinc-finger domains mediate protein uptake at efficiencies that exceed conventional protein transduction systems and do so without compromising enzyme activity. In addition, we demonstrate that zinc-finger proteins enter cells primarily through macropinocytosis and facilitate high levels of cytosolic delivery. These findings establish zinc-finger proteins as not only useful tools for targeted genome engineering but also effective reagents for protein delivery.


Subject(s)
Cysteine/chemistry , Histidine/chemistry , Proteins/administration & dosage , Zinc Fingers , Amino Acid Sequence , Cell Line , Humans , Models, Molecular , Molecular Sequence Data , Proteins/chemistry
20.
Protein Eng Des Sel ; 23(4): 243-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20053640

ABSTRACT

Rapid clearing engineered antibody fragments for immunoPET promise high sensitivity at early time points. Here, tumor targeting of anti-CD20 diabodies (scFv dimers) for detection of low-grade B-cell lymphomas were evaluated. In addition, the effect of linker length on oligomerization of the diabody was investigated. Four rituximab scFv variants in the V(L)-V(H) orientation with different linker lengths between the V domains (scFv-1, scFv-3, scFv-5, scFv-8), plus the scFv-5 with a C-terminal cysteine (Cys-Db) for site-specific modification were generated. The scFv-8 and Cys-Db were radioiodinated with (124)I for PET imaging, and biodistribution of (131)I-Cys-Db was carried out at 2, 4 10 and 20 h. The five anti-CD20 scFv variants were expressed as fully functional dimers. Shortening the linker to three or one residue did not produce higher order of multimers. Both (124)I-labeled scFv-8 and Cys-Db exhibited similar tumor targeting at 8 h post injection, with significantly higher uptakes than in control tumors (P < 0.05). At 20 h, less than 1% ID/g of (131)I-labeled Cys-Db was present in tumors and tissues. Specific tumor targeting and high contrast images were achieved with the anti-CD20 diabodies. These agents extend the repertoire of reagents that can potentially be used to improve detection of low-grade lymphomas.


Subject(s)
Antibodies, Monoclonal/chemistry , Lymphoma, B-Cell/diagnostic imaging , Positron-Emission Tomography , Single-Chain Antibodies/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , Dimerization , Female , Humans , Immunologic Factors/chemistry , Immunologic Factors/immunology , Iodine Radioisotopes , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred Strains , Rituximab , Single-Chain Antibodies/immunology
SELECTION OF CITATIONS
SEARCH DETAIL