Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Biotechnol ; 37(8): 945-952, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31359006

RESUMEN

Engineered nucleases have gained broad appeal for their ability to mediate highly efficient genome editing. However the specificity of these reagents remains a concern, especially for therapeutic applications, given the potential mutagenic consequences of off-target cleavage. Here we have developed an approach for improving the specificity of zinc finger nucleases (ZFNs) that engineers the FokI catalytic domain with the aim of slowing cleavage, which should selectively reduce activity at low-affinity off-target sites. For three ZFN pairs, we engineered single-residue substitutions in the FokI domain that preserved full on-target activity but showed a reduction in off-target indels of up to 3,000-fold. By combining this approach with substitutions that reduced the affinity of zinc fingers, we developed ZFNs specific for the TRAC locus that mediated 98% knockout in T cells with no detectable off-target activity at an assay background of ~0.01%. We anticipate that this approach, and the FokI variants we report, will enable routine generation of nucleases for gene editing with no detectable off-target activity.


Asunto(s)
División del ADN , Edición Génica/métodos , Linfocitos T , Secuencia de Bases , ADN/genética , ADN/metabolismo , Citometría de Flujo , Células Madre Hematopoyéticas , Humanos , Células K562 , Dominios Proteicos , ARN Mensajero
2.
Mol Ther ; 27(4): 866-877, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30902585

RESUMEN

It has previously been shown that engineered zinc finger nucleases (ZFNs) can be packaged into adeno-associated viruses (AAVs) and delivered intravenously into mice, non-human primates, and most recently, humans to induce highly efficient therapeutic genome editing in the liver. Lipid nanoparticles (LNPs) are synthetic delivery vehicles that enable repeat administration and are not limited by the presence of preexisting neutralizing antibodies in patients. Here, we show that mRNA encoding ZFNs formulated into LNP can enable >90% knockout of gene expression in mice by targeting the TTR or PCSK9 gene, at mRNA doses 10-fold lower than has ever been reported. Additionally, co-delivering mRNA-LNP containing ZFNs targeted to intron 1 of the ALB locus with AAV packaged with a promoterless human IDS or FIX therapeutic transgene can result in high levels of targeted integration and subsequent therapeutically relevant levels of protein expression in mice. Finally, we show repeat administration of ZFN mRNA-LNP after a single AAV donor dose results in significantly increased levels of genome editing and transgene expression compared to a single dose. These results demonstrate LNP-mediated ZFN mRNA delivery can drive highly efficient levels of in vivo genome editing and can potentially offer a new treatment modality for a variety of diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Edición Génica/métodos , Nanopartículas/administración & dosificación , ARN Mensajero/administración & dosificación , Nucleasas con Dedos de Zinc/administración & dosificación , Animales , Células Cultivadas , Dependovirus/genética , Femenino , Técnicas de Inactivación de Genes , Vectores Genéticos , Hepatocitos/metabolismo , Intrones/genética , Lípidos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Prealbúmina/genética , Proproteína Convertasa 9/genética , ARN Mensajero/genética , Transgenes/genética , Nucleasas con Dedos de Zinc/farmacología
3.
Stem Cells Transl Med ; 7(6): 477-486, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29589874

RESUMEN

As a powerful regulator of cellular homeostasis and metabolism, adenosine is involved in diverse neurological processes including pain, cognition, and memory. Altered adenosine homeostasis has also been associated with several diseases such as depression, schizophrenia, or epilepsy. Based on its protective properties, adenosine has been considered as a potential therapeutic agent for various brain disorders. Since systemic application of adenosine is hampered by serious side effects such as vasodilatation and cardiac suppression, recent studies aim at improving local delivery by depots, pumps, or cell-based applications. Here, we report on the characterization of adenosine-releasing human embryonic stem cell-derived neuroepithelial stem cells (long-term self-renewing neuroepithelial stem [lt-NES] cells) generated by zinc finger nuclease (ZFN)-mediated knockout of the adenosine kinase (ADK) gene. ADK-deficient lt-NES cells and their differentiated neuronal and astroglial progeny exhibit substantially elevated release of adenosine compared to control cells. Importantly, extensive adenosine release could be triggered by excitation of differentiated neuronal cultures, suggesting a potential activity-dependent regulation of adenosine supply. Thus, ZFN-modified neural stem cells might serve as a useful vehicle for the activity-dependent local therapeutic delivery of adenosine into the central nervous system. Stem Cells Translational Medicine 2018;7:477-486.


Asunto(s)
Adenosina/metabolismo , Edición Génica/métodos , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Adenosina/análisis , Adenosina Quinasa/deficiencia , Adenosina Quinasa/genética , Animales , Línea Celular , Cromatografía Líquida de Alta Presión , Células Madre Embrionarias Humanas/citología , Humanos , Cariotipificación , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Neuronas/citología , Polimorfismo de Nucleótido Simple , Nucleasas con Dedos de Zinc/genética
4.
Stem Cell Reports ; 7(2): 139-48, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27396937

RESUMEN

Wiskott-Aldrich syndrome (WAS) is an X-linked primary immunodeficiency disease caused by mutations in the gene encoding the WAS protein (WASp). Here, induced pluripotent stem cells (iPSCs) were derived from a WAS patient (WAS-iPSC) and the endogenous chromosomal WAS locus was targeted with a wtWAS-2A-eGFP transgene using zinc finger nucleases (ZFNs) to generate corrected WAS-iPSC (cWAS-iPSC). WASp and GFP were first expressed in the earliest CD34(+)CD43(+)CD45(-) hematopoietic precursor cells and later in all hematopoietic lineages examined. Whereas differentiation to non-lymphoid lineages was readily obtained from WAS-iPSCs, in vitro T lymphopoiesis from WAS-iPSC was deficient with few CD4(+)CD8(+) double-positive and mature CD3(+) T cells obtained. T cell differentiation was restored for cWAS-iPSCs. Similarly, defects in natural killer cell differentiation and function were restored on targeted correction of the WAS locus. These results demonstrate that the defects exhibited by WAS-iPSC-derived lymphoid cells were fully corrected and suggests the potential therapeutic use of gene-corrected WAS-iPSCs.


Asunto(s)
Terapia Genética , Células Madre Pluripotentes Inducidas/patología , Linfopoyesis , Síndrome de Wiskott-Aldrich/patología , Síndrome de Wiskott-Aldrich/terapia , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Células Asesinas Naturales/metabolismo , Linfocitos T/inmunología , Proteína del Síndrome de Wiskott-Aldrich/genética
5.
Nat Biotechnol ; 34(4): 424-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26950749

RESUMEN

Gene therapy with genetically modified human CD34(+) hematopoietic stem and progenitor cells (HSPCs) may be safer using targeted integration (TI) of transgenes into a genomic 'safe harbor' site rather than random viral integration. We demonstrate that temporally optimized delivery of zinc finger nuclease mRNA via electroporation and adeno-associated virus (AAV) 6 delivery of donor constructs in human HSPCs approaches clinically relevant levels of TI into the AAVS1 safe harbor locus. Up to 58% Venus(+) HSPCs with 6-16% human cell marking were observed following engraftment into mice. In HSPCs from patients with X-linked chronic granulomatous disease (X-CGD), caused by mutations in the gp91phox subunit of the NADPH oxidase, TI of a gp91phox transgene into AAVS1 resulted in ∼15% gp91phox expression and increased NADPH oxidase activity in ex vivo-derived neutrophils. In mice transplanted with corrected HSPCs, 4-11% of human cells in the bone marrow expressed gp91phox. This method for TI into AAVS1 may be broadly applicable to correction of other monogenic diseases.


Asunto(s)
Antígenos CD34/química , Terapia Genética/métodos , Enfermedad Granulomatosa Crónica/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Animales , Células Cultivadas , Humanos , Ratones , Ratones Transgénicos
6.
Sci Rep ; 6: 21645, 2016 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-26898342

RESUMEN

We describe a fundamentally novel feat of animal genetic engineering: the precise and efficient substitution of an agronomic haplotype into a domesticated species. Zinc finger nuclease in-embryo editing of the RELA locus generated live born domestic pigs with the warthog RELA orthologue, associated with resilience to African Swine Fever. The ability to efficiently achieve interspecies allele introgression in one generation opens unprecedented opportunities for agriculture and basic research.


Asunto(s)
Resistencia a la Enfermedad/genética , Edición Génica/métodos , Ingeniería Genética , Ligasas/genética , Fiebre Porcina Africana/genética , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/patogenicidad , Alelos , Animales , Genoma , Haplotipos , Porcinos
7.
Blood ; 126(15): 1777-84, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26297739

RESUMEN

Site-specific genome editing provides a promising approach for achieving long-term, stable therapeutic gene expression. Genome editing has been successfully applied in a variety of preclinical models, generally focused on targeting the diseased locus itself; however, limited targeting efficiency or insufficient expression from the endogenous promoter may impede the translation of these approaches, particularly if the desired editing event does not confer a selective growth advantage. Here we report a general strategy for liver-directed protein replacement therapies that addresses these issues: zinc finger nuclease (ZFN) -mediated site-specific integration of therapeutic transgenes within the albumin gene. By using adeno-associated viral (AAV) vector delivery in vivo, we achieved long-term expression of human factors VIII and IX (hFVIII and hFIX) in mouse models of hemophilia A and B at therapeutic levels. By using the same targeting reagents in wild-type mice, lysosomal enzymes were expressed that are deficient in Fabry and Gaucher diseases and in Hurler and Hunter syndromes. The establishment of a universal nuclease-based platform for secreted protein production would represent a critical advance in the development of safe, permanent, and functional cures for diverse genetic and nongenetic diseases.


Asunto(s)
Albúminas/genética , Terapia de Reemplazo Enzimático , Terapia Genética , Genoma , Hígado/metabolismo , Transgenes/fisiología , Albúminas/metabolismo , Animales , Dependovirus/genética , Endonucleasas , Enfermedad de Fabry/genética , Enfermedad de Fabry/terapia , Factor IX/genética , Factor VIII/genética , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/terapia , Vectores Genéticos/administración & dosificación , Hemofilia A/genética , Hemofilia A/terapia , Hemofilia B/genética , Hemofilia B/terapia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lisosomas/enzimología , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/terapia , Mucopolisacaridosis II/genética , Mucopolisacaridosis II/terapia , Regiones Promotoras Genéticas/genética , Edición de ARN , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dedos de Zinc
8.
Blood ; 125(17): 2597-604, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25733580

RESUMEN

Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the ß-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the ß-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.


Asunto(s)
Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Terapia Genética , Células Madre Hematopoyéticas/metabolismo , Mutación , Globinas beta/genética , Anemia de Células Falciformes/patología , Animales , Antígenos CD34/análisis , Secuencia de Bases , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Endodesoxirribonucleasas/metabolismo , Sangre Fetal/trasplante , Sitios Genéticos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Datos de Secuencia Molecular , Dedos de Zinc
9.
Stem Cell Reports ; 4(4): 569-77, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25772471

RESUMEN

Recently developed reprogramming and genome editing technologies make possible the derivation of corrected patient-specific pluripotent stem cell sources-potentially useful for the development of new therapeutic approaches. Starting with skin fibroblasts from patients diagnosed with cystic fibrosis, we derived and characterized induced pluripotent stem cell (iPSC) lines. We then utilized zinc-finger nucleases (ZFNs), designed to target the endogenous CFTR gene, to mediate correction of the inherited genetic mutation in these patient-derived lines via homology-directed repair (HDR). We observed an exquisitely sensitive, homology-dependent preference for targeting one CFTR allele versus the other. The corrected cystic fibrosis iPSCs, when induced to differentiate in vitro, expressed the corrected CFTR gene; importantly, CFTR correction resulted in restored expression of the mature CFTR glycoprotein and restoration of CFTR chloride channel function in iPSC-derived epithelial cells.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Marcación de Gen , Células Madre Pluripotentes Inducidas/metabolismo , Alelos , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Endonucleasas/genética , Endonucleasas/metabolismo , Expresión Génica , Marcación de Gen/métodos , Vectores Genéticos/genética , Genotipo , Recombinación Homóloga , Humanos , Células Madre Pluripotentes Inducidas/citología , Mutación , Reparación del ADN por Recombinación , Análisis de Secuencia de ADN , Dedos de Zinc/genética
10.
Nat Med ; 18(5): 807-815, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22466705

RESUMEN

The transfer of high-avidity T cell receptor (TCR) genes isolated from rare tumor-specific lymphocytes into polyclonal T cells is an attractive cancer immunotherapy strategy. However, TCR gene transfer results in competition for surface expression and inappropriate pairing between the exogenous and endogenous TCR chains, resulting in suboptimal activity and potentially harmful unpredicted antigen specificities of the resultant TCRs. We designed zinc-finger nucleases (ZFNs) that promoted the disruption of endogenous TCR ß- and α-chain genes. Lymphocytes treated with ZFNs lacked surface expression of CD3-TCR and expanded with the addition of interleukin-7 (IL-7) and IL-15. After lentiviral transfer of a TCR specific for the Wilms tumor 1 (WT1) antigen, these TCR-edited cells expressed the new TCR at high levels, were easily expanded to near purity and were superior at specific antigen recognition compared to donor-matched, unedited TCR-transferred cells. In contrast to unedited TCR-transferred cells, the TCR-edited lymphocytes did not mediate off-target reactivity while maintaining their anti-tumor activity in vivo, thus showing that complete editing of T cell specificity generates tumor-specific lymphocytes with improved biosafety profiles.


Asunto(s)
Desoxirribonucleasas de Localización Especificada Tipo II/farmacología , Lentivirus/genética , Leucemia/inmunología , Receptores de Antígenos de Linfocitos T/genética , Especificidad del Receptor de Antígeno de Linfocitos T , Dedos de Zinc , Secuencia de Bases , Técnicas de Transferencia de Gen , Humanos , Células Jurkat , Datos de Secuencia Molecular , Linfocitos T/inmunología , Proteínas WT1/genética
11.
Nature ; 478(7369): 391-4, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21993621

RESUMEN

Human induced pluripotent stem cells (iPSCs) represent a unique opportunity for regenerative medicine because they offer the prospect of generating unlimited quantities of cells for autologous transplantation, with potential application in treatments for a broad range of disorders. However, the use of human iPSCs in the context of genetically inherited human disease will require the correction of disease-causing mutations in a manner that is fully compatible with clinical applications. The methods currently available, such as homologous recombination, lack the necessary efficiency and also leave residual sequences in the targeted genome. Therefore, the development of new approaches to edit the mammalian genome is a prerequisite to delivering the clinical promise of human iPSCs. Here we show that a combination of zinc finger nucleases (ZFNs) and piggyBac technology in human iPSCs can achieve biallelic correction of a point mutation (Glu342Lys) in the α(1)-antitrypsin (A1AT, also known as SERPINA1) gene that is responsible for α(1)-antitrypsin deficiency. Genetic correction of human iPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene-targeting technology that is currently available and crucially prevents contamination of the host genome with residual non-human sequences. Our results provide the first proof of principle, to our knowledge, for the potential of combining human iPSCs with genetic correction to generate clinically relevant cells for autologous cell-based therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Reparación del Gen Blanco , Deficiencia de alfa 1-Antitripsina/genética , alfa 1-Antitripsina/genética , Animales , Línea Celular , Elementos Transponibles de ADN/genética , Hepatocitos/metabolismo , Hepatocitos/trasplante , Humanos , Hígado/citología , Ratones , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Albúmina Sérica Humana , Factores de Tiempo , alfa 1-Antitripsina/metabolismo
12.
Nature ; 475(7355): 217-21, 2011 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-21706032

RESUMEN

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


Asunto(s)
Reparación del ADN/genética , Modelos Animales de Enfermedad , Marcación de Gen/métodos , Terapia Genética/métodos , Genoma/genética , Hemofilia B/genética , Hemostasis , Animales , Secuencia de Bases , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Endonucleasas/química , Endonucleasas/genética , Endonucleasas/metabolismo , Exones/genética , Factor IX/análisis , Factor IX/genética , Vectores Genéticos/genética , Células HEK293 , Hemofilia B/fisiopatología , Humanos , Intrones/genética , Hígado/metabolismo , Regeneración Hepática , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Fenotipo , Homología de Secuencia , Dedos de Zinc
13.
Nat Biotechnol ; 29(2): 143-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21179091

RESUMEN

Nucleases that cleave unique genomic sequences in living cells can be used for targeted gene editing and mutagenesis. Here we develop a strategy for generating such reagents based on transcription activator-like effector (TALE) proteins from Xanthomonas. We identify TALE truncation variants that efficiently cleave DNA when linked to the catalytic domain of FokI and use these nucleases to generate discrete edits or small deletions within endogenous human NTF3 and CCR5 genes at efficiencies of up to 25%. We further show that designed TALEs can regulate endogenous mammalian genes. These studies demonstrate the effective application of designed TALE transcription factors and nucleases for the targeted regulation and modification of endogenous genes.


Asunto(s)
Técnicas Químicas Combinatorias/métodos , Ingeniería Genética , Mutagénesis Sitio-Dirigida/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Sitios de Unión , ADN/genética , ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Genoma , Humanos , Células K562 , Datos de Secuencia Molecular , Receptores CCR5/genética , Factor A de Crecimiento Endotelial Vascular/genética , Xanthomonas
14.
J Neurosci ; 30(49): 16469-74, 2010 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21147986

RESUMEN

Loss of dopaminergic neurons is primarily responsible for the onset and progression of Parkinson's disease (PD); thus, neuroprotective and/or neuroregenerative strategies remain critical to the treatment of this increasingly prevalent disease. Here we explore a novel approach to neurotrophic factor-based therapy by engineering zinc finger protein transcription factors (ZFP TFs) that activate the expression of the endogenous glial cell line-derived neurotrophic factor (GDNF) gene. We show that GDNF activation can be achieved with exquisite genome-wide specificity. Furthermore, in a rat model of PD, striatal delivery of an adeno-associated viral vector serotype 2 encoding the GDNF activator resulted in improvements in forelimb akinesia, sensorimotor neglect, and amphetamine-induced rotations caused by 6-hydroxydopamine (6-OHDA) lesion. Our results suggest that an engineered ZFP TF can drive sufficient GDNF expression in the brain to provide functional neuroprotection against 6-OHDA; therefore, targeted activation of the endogenous gene may provide a method for delivering appropriate levels of GDNF to PD patients.


Asunto(s)
Terapia Genética/métodos , Factores Neurotróficos Derivados de la Línea Celular Glial/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/terapia , Ingeniería de Proteínas/métodos , Anfetamina/administración & dosificación , Animales , Línea Celular , Modelos Animales de Enfermedad , Dopaminérgicos/administración & dosificación , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/fisiología , Factores Neurotróficos Derivados de la Línea Celular Glial/biosíntesis , Factores Neurotróficos Derivados de la Línea Celular Glial/genética , Proteínas Fluorescentes Verdes/genética , Haplorrinos , Humanos , Lentivirus/fisiología , Ratones , Análisis por Micromatrices/métodos , Actividad Motora/efectos de los fármacos , Oxidopamina/toxicidad , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/etiología , ARN Mensajero/metabolismo , Ratas , Factores de Tiempo , Transfección , Tirosina 3-Monooxigenasa/metabolismo , Dedos de Zinc/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA