Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 31(7): 2266-2285, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-36934299

RESUMEN

The human T cell leukemia virus type 1 (HTLV-1) is a pathogenic retrovirus that persists as a provirus in the genome of infected cells and can lead to adult T cell leukemia (ATL). Worldwide, more than 10 million people are infected and approximately 5% of these individuals will develop ATL, a highly aggressive cancer that is currently incurable. In the last years, genome editing tools have emerged as promising antiviral agents. In this proof-of-concept study, we use substrate-linked directed evolution (SLiDE) to engineer Cre-derived site-specific recombinases to excise the HTLV-1 proviral genome from infected cells. We identified a conserved loxP-like sequence (loxHTLV) present in the long terminal repeats of the majority of virus isolates. After 181 cycles of SLiDE, we isolated a designer-recombinase (designated RecHTLV), which efficiently recombines the loxHTLV sequence in bacteria and human cells with high specificity. Expression of RecHTLV in human Jurkat T cells resulted in antiviral activity when challenged with an HTLV-1 infection. Moreover, expression of RecHTLV in chronically infected SP cells led to the excision of HTLV-1 proviral DNA. Our data suggest that recombinase-mediated excision of the HTLV-1 provirus represents a promising approach to reduce proviral load in HTLV-1-infected individuals, potentially preventing the development of HTLV-1-associated diseases.


Asunto(s)
Virus Linfotrópico T Tipo 1 Humano , Paraparesia Espástica Tropical , Adulto , Humanos , Virus Linfotrópico T Tipo 1 Humano/genética , Paraparesia Espástica Tropical/tratamiento farmacológico , Paraparesia Espástica Tropical/genética , Provirus/genética , Antivirales
2.
Nat Commun ; 13(1): 422, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35058465

RESUMEN

Despite advances in nuclease-based genome editing technologies, correcting human disease-causing genomic inversions remains a challenge. Here, we describe the potential use of a recombinase-based system to correct the 140 kb inversion of the F8 gene frequently found in patients diagnosed with severe Hemophilia A. Employing substrate-linked directed molecular evolution, we develop a coupled heterodimeric recombinase system (RecF8) achieving 30% inversion of the target sequence in human tissue culture cells. Transient RecF8 treatment of endothelial cells, differentiated from patient-derived induced pluripotent stem cells (iPSCs) of a hemophilic donor, results in 12% correction of the inversion and restores Factor VIII mRNA expression. In this work, we present designer-recombinases as an efficient and specific means towards treatment of monogenic diseases caused by large gene inversions.


Asunto(s)
Inversión Cromosómica/genética , Factor VIII/genética , Recombinasas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Diferenciación Celular , Células Clonales , Evolución Molecular Dirigida , Células Endoteliales/citología , Células Endoteliales/metabolismo , Exones/genética , Células HEK293 , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Secuencias Invertidas Repetidas/genética , Recombinación Genética/genética , Especificidad por Sustrato , Secuenciación Completa del Genoma
3.
Nucleic Acids Res ; 45(16): 9726-9740, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28934476

RESUMEN

As part of the HIV infection cycle, viral DNA inserts into the genome of host cells such that the integrated DNA encoding the viral proteins is flanked by long terminal repeat (LTR) regions from the retrovirus. In an effort to develop novel genome editing techniques that safely excise HIV provirus from cells, Tre, an engineered version of Cre recombinase, was designed to target a 34-bp sequence within the HIV-1 LTR (loxLTR). The sequence targeted by Tre lacks the symmetry present in loxP, the natural DNA substrate for Cre. We report here the crystal structure of a catalytically inactive (Y324F) mutant of this engineered Tre recombinase in complex with the loxLTR DNA substrate. We also report that 17 of the 19 amino acid changes relative to Cre contribute to the altered specificity, even though many of these residues do not contact the DNA directly. We hypothesize that some mutations increase the flexibility of the Cre tetramer and that this, along with flexibility in the DNA, enable the engineered enzyme and DNA substrate to adopt complementary conformations.


Asunto(s)
Duplicado del Terminal Largo de VIH , VIH-1/genética , Integrasas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , ADN Viral/química , ADN Viral/metabolismo , Duplicado del Terminal Largo de VIH/genética , Integrasas/química , Integrasas/genética , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Conformación Proteica , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/genética
4.
Sci Rep ; 6: 30130, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27444945

RESUMEN

Precise genome engineering is instrumental for biomedical research and holds great promise for future therapeutic applications. Site-specific recombinases (SSRs) are valuable tools for genome engineering due to their exceptional ability to mediate precise excision, integration and inversion of genomic DNA in living systems. The ever-increasing complexity of genome manipulations and the desire to understand the DNA-binding specificity of these enzymes are driving efforts to identify novel SSR systems with unique properties. Here, we describe two novel tyrosine site-specific recombination systems designated Nigri/nox and Panto/pox. Nigri originates from Vibrio nigripulchritudo (plasmid VIBNI_pA) and recombines its target site nox with high efficiency and high target-site selectivity, without recombining target sites of the well established SSRs Cre, Dre, Vika and VCre. Panto, derived from Pantoea sp. aB, is less specific and in addition to its native target site, pox also recombines the target site for Dre recombinase, called rox. This relaxed specificity allowed the identification of residues that are involved in target site selectivity, thereby advancing our understanding of how SSRs recognize their respective DNA targets.


Asunto(s)
ADN Nucleotidiltransferasas/genética , Genoma/genética , Recombinación Genética/genética , Animales , ADN/genética , Ingeniería Genética/métodos , Humanos , Plásmidos/genética , Tirosina/genética
5.
Nat Biotechnol ; 34(4): 401-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26900663

RESUMEN

Current combination antiretroviral therapies (cART) efficiently suppress HIV-1 reproduction in humans, but the virus persists as integrated proviral reservoirs in small numbers of cells. To generate an antiviral agent capable of eradicating the provirus from infected cells, we employed 145 cycles of substrate-linked directed evolution to evolve a recombinase (Brec1) that site-specifically recognizes a 34-bp sequence present in the long terminal repeats (LTRs) of the majority of the clinically relevant HIV-1 strains and subtypes. Brec1 efficiently, precisely and safely removes the integrated provirus from infected cells and is efficacious on clinical HIV-1 isolates in vitro and in vivo, including in mice humanized with patient-derived cells. Our data suggest that Brec1 has potential for clinical application as a curative HIV-1 therapy.


Asunto(s)
Antivirales/farmacología , Evolución Molecular Dirigida/métodos , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Provirus/efectos de los fármacos , Recombinasas/farmacología , Integración Viral/efectos de los fármacos , Animales , Antivirales/metabolismo , Secuencia de Bases , Células Cultivadas , VIH-1/genética , Humanos , Ratones , Datos de Secuencia Molecular , Provirus/genética , Recombinasas/metabolismo , Integración Viral/genética
6.
J Int AIDS Soc ; 17(4 Suppl 3): 19706, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25397454

RESUMEN

Current drugs against HIV can suppress the progression to AIDS but cannot clear the patient from the virus. Because of potential side effects of these drugs and the possible development of drug resistance, finding a cure for HIV infection remains a high priority of HIV/AIDS research. We recently generated a recombinase (termed Tre) tailored to efficiently eradicate the provirus from the host genome of HIV-1 infected cells by specifically targeting a sequence that is present in the long terminal repeats (LTRs) of the viral DNA [1]. In vivo analyses in HIV-infected humanized mice demonstrated highly significant antiviral effects of Tre recombinase [2]. However, the fact that Tre recognizes a particular HIV-1 subtype A strain may limit its broad therapeutic application. To advance our Tre-based strategy towards a universally efficient cure, we have engineered a new, universal recombinase (uTre) applicable to the majority of HIV-1 infections by the various virus strains and subtypes. We employed the search tool SeLOX [3] in order to find a well-conserved HIV-1 proviral sequence that could serve as target site for a universal Tre from sequences compiled in the Los Alamos HIV Sequence Database. We selected a candidate (termed loxLTRu) with a mean conservation rate of 94% throughout the major HIV-1 subtype groups A, B and C. We applied loxLTRu as substrate in our established substrate-linked protein evolution (SLiPE) process [4] and evolved the uTre recombinase in 142 evolution cycles. Highly specific enzymatic activity on loxLTRu is demonstrated for uTre in both Escherichia coli and human cells. Naturally occurring viral variants with single mutations within the loxLTRu sequence are also shown to be efficiently targeted by uTre, further increasing the range of applicability of the recombinase. Potential off-target sites in the human genome are not recombined by uTre. Furthermore, uTre expression in primary human T cells shows no obvious Tre-related cytopathic or genotoxic effects. Finally, uTre expressing mice show no undesired phenotypes during their normal lifespan. We have developed a broad-range HIV-1 LTR specific recombinase that has the potential to be effective against the vast majority of HIV-1 strains and to cure HIV-1 infected cells from the infection. These results strongly encouraged us in our confidence that a Tre recombinase-mediated HIV eradication strategy may become a valuable component of a future therapy for HIV-infected patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...