Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Mol Ther Methods Clin Dev ; 18: 145-158, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32637446

ABSTRACT

Despite efforts to develop effective treatments for eradicating HIV-1, a cure has not yet been achieved. Whereas antiretroviral drugs target an actively replicating virus, latent, nonreplicative forms persist during treatment. Pharmacological strategies that reactivate latent HIV-1 and expose cellular reservoirs to antiretroviral therapy and the host immune system have, so far, been unsuccessful, often triggering severe side effects, mainly due to systemic immune activation. Here, we present an alternative approach for stimulating latent HIV-1 expression via direct protein delivery of cell-penetrating zinc-finger activators (ZFAs). Cys2-His2 zinc-fingers, fused to a transcription activation domain, were engineered to recognize the HIV-1 promoter and induce targeted viral transcription. Following conjugation with multiple positively charged nuclear localization signal (NLS) repeats, protein delivery of a single ZFA (3NLS-PBS1-VP64) efficiently internalized HIV-1 latently infected T-lymphocytes and specifically stimulated viral expression. We show that short-term treatment with this ZFA protein induces higher levels of viral reactivation in cell line models of HIV-1 latency than those observed with gene delivery. Our work establishes protein delivery of ZFA as a novel and safe approach toward eradication of HIV-1 reservoirs.

2.
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
3.
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
4.
PLoS One ; 11(3): e0150037, 2016.
Article in English | MEDLINE | ID: mdl-26933881

ABSTRACT

The presence of replication-competent HIV-1 -which resides mainly in resting CD4+ T cells--is a major hurdle to its eradication. While pharmacological approaches have been useful for inducing the expression of this latent population of virus, they have been unable to purge HIV-1 from all its reservoirs. Additionally, many of these strategies have been associated with adverse effects, underscoring the need for alternative approaches capable of reactivating viral expression. Here we show that engineered transcriptional modulators based on customizable transcription activator-like effector (TALE) proteins can induce gene expression from the HIV-1 long terminal repeat promoter, and that combinations of TALE transcription factors can synergistically reactivate latent viral expression in cell line models of HIV-1 latency. We further show that complementing TALE transcription factors with Vorinostat, a histone deacetylase inhibitor, enhances HIV-1 expression in latency models. Collectively, these findings demonstrate that TALE transcription factors are a potentially effective alternative to current pharmacological routes for reactivating latent virus and that combining synthetic transcriptional activators with histone deacetylase inhibitors could lead to the development of improved therapies for latent HIV-1 infection.


Subject(s)
Activating Transcription Factors/genetics , HIV Infections/genetics , HIV-1/genetics , Virus Latency/genetics , Virus Replication/genetics , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Line , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/genetics , HEK293 Cells , HIV Infections/drug therapy , HIV Long Terminal Repeat/genetics , HIV-1/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Virus Latency/drug effects , Virus Replication/drug effects , Vorinostat
5.
Nat Protoc ; 10(11): 1842-59, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26492140

ABSTRACT

Targeted nucleases, including zinc-finger nucleases (ZFNs), transcription activator-like (TAL) effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9), have provided researchers with the ability to manipulate nearly any genomic sequence in human cells and model organisms. However, realizing the full potential of these genome-modifying technologies requires their safe and efficient delivery into relevant cell types. Unlike methods that rely on expression from nucleic acids, the direct delivery of nuclease proteins to cells provides rapid action and fast turnover, leading to fewer off-target effects while maintaining high rates of targeted modification. These features make nuclease protein delivery particularly well suited for precision genome engineering. Here we describe procedures for implementing protein-based genome editing in human embryonic stem cells and primary cells. Protocols for the expression, purification and delivery of ZFN proteins, which are intrinsically cell-permeable; TALEN proteins, which can be internalized via conjugation with cell-penetrating peptide moieties; and Cas9 ribonucleoprotein, whose nucleofection into cells facilitates rapid induction of multiplexed modifications, are described, along with procedures for evaluating nuclease protein activity. Once they are constructed, nuclease proteins can be expressed and purified within 6 d, and they can be used to induce genomic modifications in human cells within 2 d.


Subject(s)
Gene Targeting/methods , Molecular Biology/methods , Recombinases/metabolism , Cells, Cultured , Humans , Protein Transport , Stem Cells
6.
PLoS One ; 10(9): e0139123, 2015.
Article in English | MEDLINE | ID: mdl-26414179

ABSTRACT

Site-specific recombinases (SSRs) are valuable tools for genetic engineering due to their ability to manipulate DNA in a highly specific manner. Engineered zinc-finger and TAL effector recombinases, in particular, are two classes of SSRs composed of custom-designed DNA-binding domains fused to a catalytic domain derived from the resolvase/invertase family of serine recombinases. While TAL effector and zinc-finger proteins can be assembled to recognize a wide range of possible DNA sequences, recombinase catalytic specificity has been constrained by inherent base requirements present within each enzyme. In order to further expand the targeted recombinase repertoire, we used a genetic screen to isolate enhanced mutants of the Bin and Tn21 recombinases that recognize target sites outside the scope of other engineered recombinases. We determined the specific base requirements for recombination by these enzymes and demonstrate their potential for genome engineering by selecting for variants capable of specifically recombining target sites present in the human CCR5 gene and the AAVS1 safe harbor locus. Taken together, these findings demonstrate that complementing functional characterization with protein engineering is a potentially powerful approach for generating recombinases with expanded targeting capabilities.


Subject(s)
Genome, Human , Recombinases/metabolism , Base Sequence , Catalytic Domain , Genetic Loci , Humans , Molecular Sequence Data , Mutation/genetics , Receptors, CCR5/genetics , Recombinases/chemistry , Recombination, Genetic , Serine/chemistry , Serine/metabolism , Substrate Specificity
7.
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
8.
Mol Ther Nucleic Acids ; 4: e232, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25756962

ABSTRACT

Safe, efficient, and broadly applicable methods for delivering site-specific nucleases into cells are needed in order for targeted genome editing to reach its full potential for basic research and medicine. We previously reported that zinc-finger nuclease (ZFN) proteins have the innate capacity to cross cell membranes and induce genome modification via their direct application to human cells. Here, we show that incorporation of tandem nuclear localization signal (NLS) repeats into the ZFN protein backbone enhances cell permeability nearly 13-fold and that single administration of multi-NLS ZFN proteins leads to genome modification rates of up to 26% in CD4(+) T cells and 17% in CD34(+) hematopoietic stem/progenitor cells. In addition, we show that multi-NLS ZFN proteins attenuate off-target effects and that codelivery of ZFN protein pairs facilitates dual gene modification frequencies of 20-30% in CD4(+) T cells. These results illustrate the applicability of ZFN protein delivery for precision genome engineering.

9.
Chem Commun (Camb) ; 51(15): 3196-9, 2015 Feb 21.
Article in English | MEDLINE | ID: mdl-25604598

ABSTRACT

A routine thioketal protecting group reacts rapidly and selectively with singlet oxygen to reveal ketone products in good (aryl 1,3-dithiolane) to excellent (aryl 1,3-oxathiolane) yields. Arylthiolanes are stable to biologically relevant reactive oxygen species and can be used as a light-activated gating mechanism for activating fluorescent sensors or small molecule prodrugs.


Subject(s)
Heterocyclic Compounds/chemistry , Ketones/chemistry , Reactive Oxygen Species/chemistry , Thiophenes/chemistry , Doxorubicin/chemistry , Light , Methylene Blue/chemistry , Photosensitizing Agents/chemistry , Prodrugs , Rose Bengal/chemistry
10.
Methods Enzymol ; 546: 79-91, 2014.
Article in English | MEDLINE | ID: mdl-25398336

ABSTRACT

Site-specific recombinases are valuable tools for myriad basic research and genome engineering applications. In particular, hybrid recombinases consisting of catalytic domains from the resolvase/invertase family of serine recombinases fused to Cys2-His2 zinc-finger or TAL effector DNA-binding domains are capable of introducing targeted modifications into mammalian cells. Due to their inherent modularity, new recombinases with distinct targeting specificities can readily be generated and utilized in a "plug-and-play" manner. In this protocol, we provide detailed, step-by-step instructions for generating new hybrid recombinases with user-defined specificity, as well as methods for achieving site-specific integration into targeted genomic loci using these systems.


Subject(s)
DNA Nucleotidyltransferases/chemistry , DNA Nucleotidyltransferases/metabolism , Protein Engineering/methods , Amino Acid Sequence , Animals , DNA Nucleotidyltransferases/genetics , Gene Targeting , Genome, Human , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Zinc Fingers
11.
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
12.
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
13.
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
14.
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
15.
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
16.
ACS Med Chem Lett ; 5(2): 133-137, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24563723

ABSTRACT

CCR5 antagonists are among the most advanced approaches in HIV therapy and may also be relevant to treatment of graft-versus-host disease and Staphylococcus aureus infection. To expand the potential of the only approved CCR5 antagonist, Maraviroc, we studied derivatives that would enable functional linkage of Maraviroc to long-lived carriers. Through targeted synthesis, we discovered an effective linkage site on Maraviroc and demonstrate the potential of these derivatives to prepare potent chemically programmed antibodies and PEGylated derivatives. The resulting compounds effectively neutralized a variety of HIV-1 isolates. Both chemically programmed antibody and PEGylation approaches extend the neutralization activity of serum circulating Maraviroc. Derivation of a successful conjugation strategy for Maraviroc should further enable its use in chemically programmed vaccines, novel bispecific antibodies, and topical microbicides.

17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...