Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
1.
Pharmaceutics ; 13(10)2021 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-34683891

RESUMO

A major bottleneck in the successful development of central nervous system (CNS) drugs is the discovery and design of molecules that can cross the blood-brain barrier (BBB). Nano-delivery strategies are a promising approach that take advantage of natural portals of entry into the brain such as monoclonal antibodies (mAbs) targeting endogenous BBB receptors. However, the main selected mAbs rely on targeting broadly expressed receptors, such as the transferrin and insulin receptors, and in selection processes that do not fully mimic the native receptor conformation, leading to mistargeting and a low fraction of the administered dose effectively reaching the brain. Thus, there is an urgent need to identify new BBB receptors and explore novel antibody selection approaches that can allow a more selective delivery into the brain. Considering that in vitro models fail to completely mimic brain structure complexity, we explored an in vivo cell immunization approach to construct a rabbit derived single-domain antibody (sdAb) library towards BBB endothelial cell receptors. The sdAb antibody library was used in an in vivo phage display screening as a functional selection of novel BBB targeting antibodies. Following three rounds of selections, next generation sequencing analysis, in vitro brain endothelial barrier (BEB) model screenings and in vivo biodistribution studies, five potential sdAbs were identified, three of which reaching >0.6% ID/g in the brain. To validate the brain drug delivery proof-of-concept, the most promising sdAb, namely RG3, was conjugated at the surface of liposomes encapsulated with a model drug, the pan-histone deacetylase inhibitor panobinostat (PAN). The translocation efficiency and activity of the conjugate liposome was determined in a dual functional in vitro BEB-glioblastoma model. The RG3 conjugated PAN liposomes enabled an efficient BEB translocation and presented a potent antitumoral activity against LN229 glioblastoma cells without influencing BEB integrity. In conclusion, our in vivo screening approach allowed the selection of highly specific nano-antibody scaffolds with promising properties for brain targeting and drug delivery.

2.
Mol Ther Methods Clin Dev ; 18: 145-158, 2020 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-32637446

RESUMO

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.

3.
Sci Rep ; 9(1): 19765, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31874995

RESUMO

We report the development of episomal vectors for the specific γ-globin transcription activation in its native position by activator Zif-VP64, based on the Scaffold/Matrix Attachment Region (S/MAR) for episomal retention and the ß-globin Replicator, the DNA replication-Initiation Region from the ß-globin locus. Vector Zif-VP64-Ep1 containing transcription cassettes CMV- Zif-VP64 and CMV-eGFP-S/MAR transfected a)K562 cells; b)murine ß-YAC bone marrow cells (BMC); c)human haematopoietic progenitor CD34+ cells, with transfection efficiencies of 46.3 ± 5.2%, 23.0 ± 2.1% and 24.2 ± 2.4% respectively. K562 transfections generated stable cell lines running for 28 weeks with and without selection, with increased levels of γ-globin mRNA by 3.3 ± 0.13, of γ-globin protein by 6.75 ± 3.25 and HbF protein by 2 ± 0.2 fold, while the vector remained episomal and non integrated. In murine ß-YAC BMCs the vector mediated the activation of the silent human γ-globin gene and in CD34+ cells, increased γ-globin mRNA, albeit only transiently. A second vector Zif-VP64-Ep2, with both transcription cassettes carrying promoter SFFV instead of CMV and the addition of ß-globin Replicator, transferred into CD34+ cells, produced CD34+ eGFP+ cells, that generated colonies in colony forming cell cultures. Importantly, these were 100% fluorescent, with 2.11 ± 0.13 fold increased γ-globin mRNA, compared to non-transfected cells. We consider these episomal vectors valid, safer alternatives to viral vectors.


Assuntos
Vetores Genéticos , Células-Tronco Hematopoéticas/metabolismo , Regiões de Interação com a Matriz , Plasmídeos , Regiões Promotoras Genéticas , Globinas beta/biossíntese , Células-Tronco Hematopoéticas/citologia , Humanos , Células K562 , Globinas beta/genética
4.
Bioorg Med Chem ; 25(21): 5952-5961, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28988625

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Biotina/imunologia , Haptenos/imunologia , Cetonas/imunologia , Administração Oral , Animais , Anticorpos Monoclonais/farmacocinética , Reações Antígeno-Anticorpo/efeitos dos fármacos , Biotina/administração & dosagem , Haptenos/administração & dosagem , Cetonas/administração & dosagem , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular
5.
Bioconjug Chem ; 27(10): 2271-2275, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27666414

RESUMO

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.


Assuntos
Imunoconjugados/química , Proteínas Recombinantes de Fusão/química , Albumina Sérica/química , Anticorpos/química , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Imunoconjugados/sangue , Imunoconjugados/metabolismo , Lisina/química , Domínios Proteicos , Engenharia de Proteínas/métodos , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/metabolismo , Rodaminas/química , Trastuzumab/química
6.
PLoS One ; 11(3): e0150037, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26933881

RESUMO

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.


Assuntos
Fatores Ativadores da Transcrição/genética , Infecções por HIV/genética , HIV-1/genética , Latência Viral/genética , Replicação Viral/genética , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/genética , Células HEK293 , Infecções por HIV/tratamento farmacológico , Repetição Terminal Longa de HIV/genética , HIV-1/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Latência Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Vorinostat
7.
Nat Protoc ; 10(11): 1842-59, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26492140

RESUMO

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.


Assuntos
Marcação de Genes/métodos , Biologia Molecular/métodos , Recombinases/metabolismo , Células Cultivadas , Humanos , Transporte Proteico , Células-Tronco
8.
PLoS One ; 10(9): e0139123, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26414179

RESUMO

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.


Assuntos
Genoma Humano , Recombinases/metabolismo , Sequência de Bases , Domínio Catalítico , Loci Gênicos , Humanos , Dados de Sequência Molecular , Mutação/genética , Receptores CCR5/genética , Recombinases/química , Recombinação Genética , Serina/química , Serina/metabolismo , Especificidade por Substrato
9.
Bioorg Med Chem Lett ; 25(8): 1684-1687, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25791455

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Frutose-Bifosfato Aldolase/química , Haptenos/química , Fragmentos Fab das Imunoglobulinas/química , beta-Lactamas/química , Anticorpos Monoclonais/imunologia , Reações Antígeno-Anticorpo , Biotina/química , Biotina/metabolismo , Frutose-Bifosfato Aldolase/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Ligação Proteica , Estabilidade Proteica , Estreptavidina/química , Estreptavidina/metabolismo
10.
Mol Ther Nucleic Acids ; 4: e232, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25756962

RESUMO

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.

11.
Chem Commun (Camb) ; 51(15): 3196-9, 2015 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-25604598

RESUMO

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.


Assuntos
Compostos Heterocíclicos/química , Cetonas/química , Espécies Reativas de Oxigênio/química , Tiofenos/química , Doxorrubicina/química , Luz , Azul de Metileno/química , Fármacos Fotossensibilizantes/química , Pró-Fármacos , Rosa Bengala/química
12.
Methods Enzymol ; 546: 79-91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25398336

RESUMO

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.


Assuntos
DNA Nucleotidiltransferases/química , DNA Nucleotidiltransferases/metabolismo , Engenharia de Proteínas/métodos , Sequência de Aminoácidos , Animais , DNA Nucleotidiltransferases/genética , Marcação de Genes , Genoma Humano , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Dedos de Zinco
13.
Angew Chem Int Ed Engl ; 53(44): 11783-6, 2014 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-25196737

RESUMO

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.


Assuntos
Lisina/química , Albumina Sérica/síntese química , Humanos
14.
Bioconjug Chem ; 25(8): 1402-7, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25099687

RESUMO

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.


Assuntos
Imunoconjugados/química , Sulfonas/química , Anticorpos Monoclonais Humanizados/química , Sítios de Ligação , Linhagem Celular , Humanos , Imunoconjugados/sangue , Fragmentos Fc das Imunoglobulinas/química , Modelos Moleculares , Oxidiazóis/química , Conformação Proteica , Estabilidade Proteica , Anticorpos de Cadeia Única/química , Especificidade por Substrato , Trastuzumab
15.
Acc Chem Res ; 47(8): 2309-18, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-24877793

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Genoma Humano , DNA/química , DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Desoxirribonucleases/química , Desoxirribonucleases/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/química , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Humanos , Estrutura Terciária de Proteína , Recombinases/química , Recombinases/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Dedos de Zinco
16.
ACS Chem Biol ; 9(8): 1662-7, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24936957

RESUMO

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.


Assuntos
Cisteína/química , Histidina/química , Proteínas/administração & dosagem , Dedos de Zinco , Sequência de Aminoácidos , Linhagem Celular , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas/química
17.
J Am Chem Soc ; 136(13): 5047-56, 2014 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-24611715

RESUMO

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.


Assuntos
Engenharia de Proteínas/métodos , Recombinases/química , Recombinases/genética , Recombinação Genética , Dedos de Zinco , Sequência de Aminoácidos , Domínio Catalítico , DNA/genética , Evolução Molecular Direcionada/métodos , Fator IX/genética , Genoma Humano , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Recombinases/metabolismo , alfa-Galactosidase/genética
18.
ACS Med Chem Lett ; 5(2): 133-137, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24563723

RESUMO

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.

19.
Nucleic Acids Res ; 42(7): 4755-66, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24452803

RESUMO

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.


Assuntos
Recombinases/química , Recombinases/genética , Dedos de Zinco , Domínio Catalítico , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Evolução Molecular Direcionada , Genoma Humano , Humanos , Mutagênese , Recombinases/metabolismo , Recombinação Genética , Especificidade por Substrato
20.
PLoS One ; 9(1): e85755, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465685

RESUMO

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.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Endonucleases/administração & dosagem , Marcação de Genes/métodos , Engenharia Genética/métodos , Proliferação de Células , Peptídeos Penetradores de Células/genética , Endonucleases/genética , Células HEK293 , Células HeLa , Humanos , Mutagênese Sítio-Dirigida , Receptores CCR5/genética , Receptores CCR5/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...