Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 44(8): e76, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-26762978

RESUMO

The CRISPR/Cas9 system facilitates precise DNA modifications by generating RNA-guided blunt-ended double-strand breaks. We demonstrate that guide RNA pairs generate deletions that are repaired with a high level of precision by non-homologous end-joining in mammalian cells. We present a method called knock-in blunt ligation for exploiting these breaks to insert exogenous PCR-generated sequences in a homology-independent manner without loss of additional nucleotides. This method is useful for making precise additions to the genome such as insertions of marker gene cassettes or functional elements, without the need for homology arms. We successfully utilized this method in human and mouse cells to insert fluorescent protein cassettes into various loci, with efficiencies up to 36% in HEK293 cells without selection. We also created versions of Cas9 fused to the FKBP12-L106P destabilization domain in an effort to improve Cas9 performance. Our in vivo blunt-end cloning method and destabilization-domain-fused Cas9 variant increase the repertoire of precision genome engineering approaches.


Assuntos
Proteínas Associadas a CRISPR/genética , Sistemas CRISPR-Cas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Reparo do DNA por Junção de Extremidades/genética , Engenharia Genética/métodos , Animais , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Genoma/genética , Células HEK293 , Humanos , Camundongos , Edição de RNA/genética , RNA Guia de Cinetoplastídeos/genética
2.
Mol Ther ; 24(4): 685-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26916285

RESUMO

Limb girdle muscular dystrophies types 2B (LGMD2B) and 2D (LGMD2D) are degenerative muscle diseases caused by mutations in the dysferlin and alpha-sarcoglycan genes, respectively. Using patient-derived induced pluripotent stem cells (iPSC), we corrected the dysferlin nonsense mutation c.5713C>T; p.R1905X and the most common alpha-sarcoglycan mutation, missense c.229C>T; p.R77C, by single-stranded oligonucleotide-mediated gene editing, using the CRISPR/Cas9 gene-editing system to enhance the frequency of homology-directed repair. We demonstrated seamless, allele-specific correction at efficiencies of 0.7-1.5%. As an alternative, we also carried out precise gene addition strategies for correction of the LGMD2B iPSC by integration of wild-type dysferlin cDNA into the H11 safe harbor locus on chromosome 22, using dual integrase cassette exchange (DICE) or TALEN-assisted homologous recombination for insertion precise (THRIP). These methods employed TALENs and homologous recombination, and DICE also utilized site-specific recombinases. With DICE and THRIP, we obtained targeting efficiencies after selection of ~20%. We purified iPSC corrected by all methods and verified rescue of appropriate levels of dysferlin and alpha-sarcoglycan protein expression and correct localization, as shown by immunoblot and immunocytochemistry. In summary, we demonstrate for the first time precise correction of LGMD iPSC and validation of expression, opening the possibility of cell therapy utilizing these corrected iPSC.


Assuntos
Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas de Membrana/genética , Proteínas Musculares/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Mutação , Alelos , Sistemas CRISPR-Cas , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Disferlina , Feminino , Terapia Genética , Recombinação Homóloga , Humanos , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/patologia , Sarcoglicanas/genética
3.
Nucleic Acids Res ; 42(5): e34, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24304893

RESUMO

To reveal the full potential of human pluripotent stem cells, new methods for rapid, site-specific genomic engineering are needed. Here, we describe a system for precise genetic modification of human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We identified a novel human locus, H11, located in a safe, intergenic, transcriptionally active region of chromosome 22, as the recipient site, to provide robust, ubiquitous expression of inserted genes. Recipient cell lines were established by site-specific placement of a 'landing pad' cassette carrying attP sites for phiC31 and Bxb1 integrases at the H11 locus by spontaneous or TALEN-assisted homologous recombination. Dual integrase cassette exchange (DICE) mediated by phiC31 and Bxb1 integrases was used to insert genes of interest flanked by phiC31 and Bxb1 attB sites at the H11 locus, replacing the landing pad. This system provided complete control over content, direction and copy number of inserted genes, with a specificity of 100%. A series of genes, including mCherry and various combinations of the neural transcription factors LMX1a, FOXA2 and OTX2, were inserted in recipient cell lines derived from H9 ESC, as well as iPSC lines derived from a Parkinson's disease patient and a normal sibling control. The DICE system offers rapid, efficient and precise gene insertion in ESC and iPSC and is particularly well suited for repeated modifications of the same locus.


Assuntos
Células-Tronco Embrionárias/metabolismo , Genoma Humano , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutagênese Insercional/métodos , Animais , Linhagem Celular , Células Cultivadas , Cromossomos Humanos Par 11 , Expressão Gênica , Loci Gênicos , Genômica/métodos , Recombinação Homóloga , Humanos , Integrases/metabolismo , Camundongos , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Mol Ther ; 26(10): 2327, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30245159
6.
Circulation ; 126(11 Suppl 1): S20-8, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22965984

RESUMO

BACKGROUND: Human cardiac progenitor cells (hCPCs) are a promising cell source for regenerative repair after myocardial infarction. Exploitation of their full therapeutic potential may require stable genetic modification of the cells ex vivo. Safe genetic engineering of stem cells, using facile methods for site-specific integration of transgenes into known genomic contexts, would significantly enhance the overall safety and efficacy of cellular therapy in a variety of clinical contexts. METHODS AND RESULTS: We used the phiC31 site-specific recombinase to achieve targeted integration of a triple fusion reporter gene into a known chromosomal context in hCPCs and human endothelial cells. Stable expression of the reporter gene from its unique chromosomal integration site resulted in no discernible genomic instability or adverse changes in cell phenotype. Namely, phiC31-modified hCPCs were unchanged in their differentiation propensity, cellular proliferative rate, and global gene expression profile when compared with unaltered control hCPCs. Expression of the triple fusion reporter gene enabled multimodal assessment of cell fate in vitro and in vivo using fluorescence microscopy, bioluminescence imaging, and positron emission tomography. Intramyocardial transplantation of genetically modified hCPCs resulted in significant improvement in myocardial function 2 weeks after cell delivery, as assessed by echocardiography (P=0.002) and MRI (P=0.001). We also demonstrated the feasibility and therapeutic efficacy of genetically modifying differentiated human endothelial cells, which enhanced hind limb perfusion (P<0.05 at day 7 and 14 after transplantation) on laser Doppler imaging. CONCLUSIONS: The phiC31 integrase genomic modification system is a safe, efficient tool to enable site-specific integration of reporter transgenes in progenitor and differentiated cell types.


Assuntos
Células-Tronco Fetais/transplante , Terapia Genética/métodos , Membro Posterior/irrigação sanguínea , Isquemia/cirurgia , Mutagênese Insercional/métodos , Infarto do Miocárdio/cirurgia , Animais , Diferenciação Celular , Divisão Celular , Cromossomos Humanos Par 19/genética , Células Endoteliais/citologia , Feminino , Coração Fetal/citologia , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Humanos , Integrases , Peptídeos e Proteínas de Sinalização Intracelular , Isquemia/fisiopatologia , Luciferases de Vaga-Lume/genética , Proteínas Luminescentes/genética , Imageamento por Ressonância Magnética , Camundongos , Camundongos SCID , Proteínas/genética , Distribuição Aleatória , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Timidina Quinase/genética , Transgenes , Proteínas de Transporte Vesicular , Proteínas Virais/genética , Integração Viral , Proteína Vermelha Fluorescente
7.
Stem Cells ; 29(11): 1696-704, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21898697

RESUMO

Induced pluripotent stem cells (iPSCs) have revolutionized the stem cell field. iPSCs are most often produced by using retroviruses. However, the resulting cells may be ill-suited for clinical applications. Many alternative strategies to make iPSCs have been developed, but the nonintegrating strategies tend to be inefficient, while the integrating strategies involve random integration. Here, we report a facile strategy to create murine iPSCs that uses plasmid DNA and single transfection with sequence-specific recombinases. PhiC31 integrase was used to insert the reprogramming cassette into the genome, producing iPSCs. Cre recombinase was then used for excision of the reprogramming genes. The iPSCs were demonstrated to be pluripotent by in vitro and in vivo criteria, both before and after excision of the reprogramming cassette. This strategy is comparable with retroviral approaches in efficiency, but is nonhazardous for the user, simple to perform, and results in nonrandom integration of a reprogramming cassette that can be readily deleted. We demonstrated the efficiency of this reprogramming and excision strategy in two accessible cell types, fibroblasts and adipose stem cells. This simple strategy produces pluripotent stem cells that have the potential to be used in a clinical setting.


Assuntos
DNA Nucleotidiltransferases/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Plasmídeos/genética , Tecido Adiposo/citologia , Animais , Southern Blotting , Células Cultivadas , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , DNA Nucleotidiltransferases/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Reação em Cadeia da Polimerase
8.
Nat Med ; 8(10): 1166-70, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12244305

RESUMO

Current gene-transfer technologies display limitations in achieving effective gene delivery. Among these limitations are difficulties in stably integrating large corrective sequences into the genomes of long-lived progenitor-cell populations. Current larger-capacity viral vectors suffer from biosafety concerns, whereas plasmid-based approaches have poor efficiency of stable gene transfer. These barriers hinder genetic correction of many severe inherited human diseases, such as the blistering skin disorder recessive dystrophic epidermolysis bullosa (RDEB), caused by mutations in the large COL7A1 gene. To circumvent these barriers, we used the phi C31 bacteriophage integrase, which stably integrates large DNA sequences containing a specific 285-base-pair attB sequence into genomic 'pseudo-attP sites'. phi C31 integrase-based gene transfer stably integrated the COL7A1 cDNA into genomes of primary epidermal progenitor cells from four unrelated RDEB patients. Skin regenerated using these cells displayed stable correction of hallmark RDEB disease features, including Type VII collagen protein expression, anchoring fibril formation and dermal-epidermal cohesion. These findings establish a practical approach to nonviral genetic correction of severe human genetic disorders requiring stable genomic integration of large DNA sequences.


Assuntos
Epidermólise Bolhosa Distrófica/genética , Técnicas de Transferência de Genes , Terapia Genética , Animais , Bacteriófagos/genética , Bacteriófagos/metabolismo , Membrana Basal/ultraestrutura , Células Cultivadas , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Epidermólise Bolhosa Distrófica/patologia , Epidermólise Bolhosa Distrófica/terapia , Vetores Genéticos , Humanos , Imuno-Histoquímica , Integrases/genética , Integrases/metabolismo , Queratinócitos/fisiologia , Camundongos , Camundongos SCID , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Pele/anatomia & histologia , Pele/metabolismo , Pele/patologia , Transplante de Pele , Células-Tronco/citologia , Células-Tronco/fisiologia
9.
Mol Ther ; 17(1): 112-20, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19002165

RESUMO

phiC31 integrase is a sequence-specific phage recombinase that can recombine two short DNA sequences called attB and attP. The enzyme can also promote genomic integration of plasmids carrying attB into native mammalian sequences having partial identity to attP. To increase the efficiency of integration, we mutated the phiC31 integrase gene and screened the mutants in human cells in an assay for higher recombination frequency between attB and attP. We report in this article the isolation of a mutant, P2 that has twice the chromosomal integration frequency of wild-type phiC31 integrase, at both a preintegrated chromosomal attP site and at endogenous pseudo attP sequences in cultured human cells. In mouse liver, P2-mediated integration provided therapeutic long-term levels of human factor IX that were double those generated by wild-type phiC31 integrase. We also describe an additional mutant, P3 that combines the mutations of P2 with further changes and possesses an elevated specificity for integration at a chromosomally placed attP site in human cells. Forty-four percent of colonies carrying integration events mediated by P3 have integration at the placed attP site. These mutant integrases are useful for gene therapy and genome modification, and they demonstrate the feasibility of engineering phiC31 integrase toward more desirable properties.


Assuntos
Integrases/genética , Integrases/metabolismo , Recombinação Genética/genética , Sítios de Ligação Microbiológicos/genética , Linhagem Celular , Humanos , Mutação , Especificidade por Substrato/genética
10.
J Mol Biol ; 432(13): 3950-3955, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32339531

RESUMO

The phage-derived phiC31 integrase is a useful tool for mediating sequence-specific genomic integration in mammalian cells, recombining donor plasmids bearing the attB recognition site with introduced genomic attP sites or endogeneous pseudo-attP sites having partial identity to attP. In most prior studies, phiC31 integrase has been introduced as plasmid DNA or mRNA. The current report examines whether phiC31 integrase functions efficiently in mammalian cells when co-nucleofected as a purified protein, along with attB-containing donor plasmids or PCR fragments. We describe preparation of phiC31 integrase protein and evidence that it can mediate genomic integration in human 293 cells, including PCR evidence for integration at an endogenous pseudo-attP site. This work demonstrates for the first time the ability of 605- and 613-amino-acid versions of phiC31 integrase protein to mediate efficient, site-specific integration into the genome of human cells when co-nucleofected with full-sizedattB-containing donor plasmids or linear 2.5-kb PCR fragments. This protein-mediated approach may be especially useful for integration of exogenous sequences into valuable therapeutic target cells, such as hematopoietic stem cells or T cells, that are sensitive to introduced DNA.


Assuntos
Sítios de Ligação Microbiológicos/genética , Integrases/genética , Siphoviridae/genética , Integração Viral/genética , Linhagem Celular , Genoma Humano/genética , Genômica/métodos , Humanos , Plasmídeos/genética , Siphoviridae/enzimologia
11.
Mol Ther Methods Clin Dev ; 15: 294-304, 2019 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-31890729

RESUMO

We delivered plasmid DNA encoding therapeutic genes to the muscles of mouse models of limb girdle muscular dystrophy (LGMD) 2A, 2B, and 2D, deficient in calpain3, dysferlin, and alpha-sarcoglycan, respectively. We also delivered the human follistatin gene, which has the potential to increase therapeutic benefit. After intramuscular injection of DNA, electroporation was applied to enhance delivery to muscle fibers. When plasmids encoding the human calpain3 or dysferlin cDNA sequences were injected into quadriceps muscles of LGMD2A and LGMD2B mouse models, respectively, in 3-month studies, robust levels of calpain3 and dysferlin proteins were detected. We observed a statistically significant decrease in Evans blue dye penetration in LGMD2B mouse muscles after delivery of the dysferlin gene, consistent with repair of the muscle membrane defect in these mice. The therapeutic value of delivery of the genes for alpha-sarcoglycan and follistatin was documented by significant drops in Evans blue dye penetration in gastrocnemius muscles of LGMD2D mice. These results indicated for the first time that a combined gene therapy involving both alpha-sarcoglycan and follistatin would be valuable for LGMD2D patients. We suggest that this non-viral gene delivery method should be explored for its translational potential in patients.

12.
Curr Opin Mol Ther ; 10(5): 464-70, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18830922

RESUMO

Gene therapy for hemophilia A has fallen short of success despite several clinical trials conducted over the past decade. Challenges to its success include vector immunogenicity, insufficient transgene expression levels of Factor VIII, and inhibitor antibody formation. Gene therapy has been dominated by the use of viral vectors, as well as the immunogenic and oncogenic concerns that accompany these strategies. Because of the complexity of viral vectors, the development of nonviral DNA delivery methods may provide an efficient and safe alternative for the treatment of hemophilia A. New types of nonviral strategies, such as DNA integrating vectors, and the success of several nonviral animal studies, suggest that nonviral gene therapy has curative potential and justifies its clinical development.


Assuntos
Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos , Hemofilia A/terapia , Transgenes , Ensaios Clínicos como Assunto , Fator VIII/biossíntese , Fator VIII/genética , Terapia Genética/tendências , Hemofilia A/genética , Humanos
13.
J Neurosci Methods ; 173(2): 299-305, 2008 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-18606184

RESUMO

Stem cells can potentially be utilized in combined gene/cell therapies for neural diseases. We examined the ability of the non-viral phiC31 integrase system to promote stable transgene expression in mouse neural progenitor cells (mNPCs). phiC31 integrase catalyzes the sequence-specific integration of attB-containing plasmids into pseudo attP sites in mammalian genomes, to produce long-term transgene expression. We achieved gene transfer by co-nucleofection of a plasmid carrying the luciferase marker gene and an attB site and a plasmid expressing integrase in mNPCs that had been generated in a neurosphere preparation. Luciferase expression was quantified in live cells for 8 weeks, revealing persistence of gene expression. Sequence-specific integration at a preferred pseudo attP site in the mouse genome was detected by using PCR. Furthermore, sustained transgene expression was demonstrated in genetically modified NPCs that were cultured in conditions that promoted either growth or differentiation into neurons and astrocytes. Our results demonstrate that the phiC31 integrase system produces stable transgene expression in adult mNPCs and their progeny and may be useful in strategies for combating neurodegenerative disorders.


Assuntos
Terapia Genética/métodos , Integrases/genética , Neurônios/metabolismo , Transplante de Células-Tronco/métodos , Células-Tronco/metabolismo , Transfecção/métodos , Transgenes/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Eletroporação/métodos , Regulação da Expressão Gênica/genética , Genes Reporter/genética , Vetores Genéticos/genética , Sobrevivência de Enxerto/genética , Luciferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , Esferoides Celulares , Tempo , Fatores de Tempo
14.
Methods Mol Biol ; 435: 165-73, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370075

RESUMO

phiC31 integrase is a site-specific recombinase from a bacteriophage that has become a useful tool in mammalian cells. The enzyme normally performs precise, unidirectional recombination between two attachment or att sites called attB and attP. We have shown that an attP site preintegrated into a mammalian chromosome can serve as a target for integration of an introduced plasmid carrying an attB site. Recombination leads to precise integration of the plasmid into the chromosome at the attP site. This reaction is useful for placing introduced genes into the same chromosomal environment, in order to minimize position effects associated with random integration. Because phiC31 integrase can also mediate integration at endogenous sequences that resemble attP, called pseudo attP sites, a selection system is used that yields integration only at the desired preintegrated attP site. This chapter provides a protocol that features a simple antibiotic selection to isolate cell lines in which the introduced plasmid has integrated at the desired attP site. A polymerase chain reaction assay is also presented to verify correct chromosomal placement of the introduced plasmid. This integration system based on phiC31 integrase supplies a simple method to obtain repeated integration at the same chromosomal site in mammalian cells.


Assuntos
Integrases/genética , Sítios de Ligação Microbiológicos , Bacteriófagos/enzimologia , Bacteriófagos/genética , Linhagem Celular , Técnicas Genéticas , Humanos , Integrases/metabolismo , Plasmídeos/genética , Reação em Cadeia da Polimerase
15.
J Mol Biol ; 357(1): 28-48, 2006 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-16414067

RESUMO

The site-specific integrase from bacteriophage phiC31 functions in mammalian cells and is being applied for genetic engineering, including gene therapy. The phiC31 integrase catalyzes precise, unidirectional recombination between its 30-40-bp attP and attB recognition sites. In mammalian cells, the enzyme also mediates integration of plasmids bearing attB into native sequences that have partial sequence identity with attP, termed pseudo attP sites. Here, we analyzed the features of phiC31-mediated integration into pseudo attP sites in the human genome. Sequence analysis of 196 independent integration events derived from three cell lines revealed approximately 101 integration sites: 56% of the events were recurrent integrations distributed among 19 pseudo attP sequences. Bioinformatics analysis revealed a approximately 30-bp palindromic consensus sequence motif shared by all of the repeat occurrences and most of the single occurrence sites, verifying that phiC31-mediated integration into pseudo attP sites is significantly guided by DNA sequence recognition. The most favored unique sequence in these cell lines occurred at chromosome 19q13.31 and accounted for 7.5% of integration events. Other frequent integration sites were in three specific sequences in subfamilies of ERVL and L1 repetitive sequences, accounting for an additional 17.9% of integration events. Integrations could occur in either orientation at a pseudo attP site, were often accompanied by small deletions, and typically occurred in a single copy per cell. A number of aberrant events were also described, including large deletions and chromosome rearrangements. phiC31 integrase-mediated integration only slightly favored genes and did not favor promoter regions. Gene density and expression studies suggested chromatin context effects. An analysis of the safety of integration sites in terms of proximity to cancer genes suggested minimal cancer risk. We conclude that integration systems derived from phiC31 integrase have great potential utility.


Assuntos
Sítios de Ligação Microbiológicos , Bacteriófagos/enzimologia , Genoma Humano , Integrases/metabolismo , Animais , Bacteriófagos/genética , Sequência de Bases , Linhagem Celular , Cromossomos Humanos , Biologia Computacional , Humanos , Hibridização in Situ Fluorescente , Integrases/genética , Dados de Sequência Molecular , Recombinação Genética , Homologia de Sequência do Ácido Nucleico
16.
Nat Biotechnol ; 20(11): 1124-8, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12379870

RESUMO

We used the integrase from phage phiC31 to integrate the human Factor IX (hFIX) gene permanently into specific sites in the mouse genome. A plasmid containing attB and an expression cassette for hFIX was delivered to the livers of mice by using high-pressure tail vein injection. When an integrase expression plasmid was co-injected, hFIX serum levels increased more than tenfold to approximately 4 microg/ml, similar to normal FIX levels, and remained stable throughout the more than eight months of the experiment. hFIX levels persisted after partial hepatectomy, suggesting genomic integration of the vector. Site-specific integration was proven by characterizing and quantifying genomic integration in the liver at the DNA level. Integration was documented at two pseudo-attP sites, native sequences with partial identity to attP, with one site highly predominant. This study demonstrates in vivo gene transfer in an animal by site-specific genomic integration.


Assuntos
Fator IX/biossíntese , Fator IX/genética , Fígado/metabolismo , Mutagênese Sítio-Dirigida , Animais , Regulação da Expressão Gênica , Terapia Genética/métodos , Genoma , Injeções Intravenosas , Integrases/genética , Integrases/metabolismo , Fígado/efeitos dos fármacos , Fígado/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/administração & dosagem , Plasmídeos/genética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Análise de Sequência de DNA
17.
Bio Protoc ; 7(5): e2163, 2017 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-34458476

RESUMO

The incorporation of the CRISPR/Cas9 bacterial immune system into the genetic engineering toolbox has led to the development of several new methods for genome manipulation ( Auer et al., 2014 ; Byrne et al., 2015 ). We took advantage of the ability of Cas9 to generate blunt-ended double-strand breaks ( Jinek et al., 2012 ) to introduce exogenous DNA in a highly precise manner through the exploitation of non-homologous end-joining DNA repair machinery ( Geisinger et al., 2016 ). This protocol has been successfully applied to traditional immortalized cell lines and human induced pluripotent stem cells. Here we present a generalized protocol for knock-in blunt ligation, using HEK293 cells as an example.

18.
Methods Mol Biol ; 1642: 69-85, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28815494

RESUMO

When constructing transgenic cell lines via plasmid DNA integration, precise targeting to a desired genomic location is advantageous. It is also often advantageous to remove the bacterial backbone, since bacterial elements can lead to the epigenetic silencing of neighboring DNA. The least cumbersome method to remove the plasmid backbone is recombinase-mediated cassette exchange (RMCE). RMCE is accomplished by arranging recombinase sites in the genome and in a donor plasmid such that a recombinase can both integrate the donor plasmid and excise its bacterial backbone. When a single recombinase is used for RMCE, recombination between undesired pairings of the sites can lead to a significant number of unwanted cell lines. To reduce the frequency with which these side products occur, several variants of RMCE that increase desired outcomes have been developed. Nevertheless, an important feature lacking from these improved RMCE methods is that none have fully utilized the recombinases that have been demonstrated to be the most robust and stringent at performing genomic integration in plants and animals, i.e., the phiC31 and Bxb1 phage integrases. To address this need, we have developed an RMCE protocol using these two serine integrases that we call dual integrase cassette exchange (DICE). Our DICE system provides a means to achieve high-precision DNA integration at a desired location and is especially well suited for repeated recombination into the same locus. In this chapter, we provide our most current protocols for using DICE in feeder-free human-induced pluripotent stem cells .


Assuntos
Marcação de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Integrases/genética , Recombinação Genética , Proteínas Virais/genética , Bacteriófagos/genética , Bacteriófagos/metabolismo , Linhagem Celular , Células Clonais , Genes Reporter , Loci Gênicos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Integrases/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase/métodos , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Transfecção/métodos , Proteínas Virais/metabolismo , Proteína Vermelha Fluorescente
19.
Mol Ther Methods Clin Dev ; 7: 123-131, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29159199

RESUMO

Mutations in the gene for dysferlin cause a degenerative disorder of skeletal muscle known as limb girdle muscular dystrophy 2B. To achieve gene delivery of plasmids encoding dysferlin to hind limb muscles of dysferlin knockout mice, we used a vascular injection method that perfused naked plasmid DNA into all major muscle groups of the hind limb. We monitored delivery by luciferase live imaging and western blot, confirming strong dysferlin expression that persisted over the 3-month time course of the experiment. Co-delivery of the follistatin gene, which may promote muscle growth, was monitored by ELISA. Immunohistochemistry documented the presence of dysferlin in muscle fibers in treated limbs, and PCR confirmed the presence of plasmid DNA. Because dysferlin is involved in repair of the sarcolemmal membrane, dysferlin loss leads to fragile sarcolemmal membranes that can be detected by permeability to Evan's blue dye. We showed that after gene therapy with a plasmid encoding both dysferlin and follistatin, statistically significant reduction in Evan's blue dye permeability was present in hamstring muscles. These results suggest that vascular delivery of plasmids carrying these therapeutic genes may lead to simple and effective approaches for improving the clinical condition of limb girdle muscular dystrophy 2B.

20.
Hum Gene Ther ; 17(8): 871-6, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16942446

RESUMO

Peripheral vascular disease (PVD), characterized by insufficient blood supply to extremities, can be a devastating illness. Although many gene therapy strategies for PVD using vascular endothelial growth factor (VEGF) have resulted in increased blood vessel formation, the vessels are often impermanent and regress after therapy, probably because of the short-lived VEGF expression mediated by gene therapy vectors (14 days or less). phiC31 integrase is a recombinase originally isolated from a bacteriophage of Streptomyces. This integrase performs efficient chromosomal integration of plasmid DNA into mammalian genomes that results in long-term transgene expression. In this study, gene transfer was achieved by intramuscular injection of VEGF and integrase plasmid DNAs into the tibialis anterior muscle in the mouse hindlimb, followed by electroporation of the muscle with needle electrodes. We observed VEGF levels significantly above background 40 days after injection in animals that received phiC31 integrase and the VEGF plasmid. Site-specific integration of plasmid DNA in the chromosomes of muscle tissue was verified by polymerase chain reaction at a common integration site. These results suggest the possible utility of the phiC31 integrase system to treat ischemic disease.


Assuntos
Bacteriófagos , Membro Posterior/irrigação sanguínea , Integrases , Isquemia/terapia , Plasmídeos , Fator A de Crescimento do Endotélio Vascular , Proteínas Virais , Animais , Bacteriófagos/enzimologia , Bacteriófagos/genética , Eletroporação/métodos , Feminino , Terapia Genética/métodos , Integrases/genética , Isquemia/genética , Camundongos , Recombinação Genética/genética , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA