Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Gene Ther ; 21(6): 618-28, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24784447

RESUMEN

Effective gene therapy strategies for the treatment of kidney disorders remain elusive. We report an optimized kidney-targeted gene delivery strategy using recombinant adeno-associated virus (rAAV) administered via retrograde renal vein injection in mice. Renal vein injection of rAAV consistently resulted in superior kidney transduction compared with tail vein injection using as little as half the tail vein dose. We compared rAAV5, 6, 8 and 9, containing either green fluorescent protein (GFP) or luciferase reporter genes driven by the Cytomegalovirus promoter. We demonstrated that although rAAV6 and 8 injected via renal vein transduced the kidney, transgene expression was mainly restricted to the medulla. Transgene expression was systematically low after rAAV5 injection, attributed to T-cell immune response, which could be overcome by transient immunosuppression. However, rAAV9 was the only serotype that permitted high-transduction efficiency of both the cortex and medulla. Moreover, both the glomeruli and tubules were targeted, with a higher efficiency within the glomeruli. To improve the specificity of kidney-targeted gene delivery with rAAV9, we used the parathyroid hormone receptor 'kidney-specific' promoter. We obtained a more efficient transgene expression within the kidney, and a significant reduction in other tissues. Our work represents the first comprehensive and clinically relevant study for kidney gene delivery.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/administración & dosificación , Venas Renales/efectos de los fármacos , Animales , Terapia Genética/métodos , Inyecciones , Riñón/efectos de los fármacos , Riñón/fisiología , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Receptor de Hormona Paratiroídea Tipo 1/genética , Cola (estructura animal) , Transducción Genética , Transgenes
2.
Methods Mol Biol ; 1937: 227-234, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30706400

RESUMEN

Genetic nephropathies represent a challenging class of disorders to be treated by gene therapy. This is primarily due to the filtering properties of the kidney itself, which does not allow the vehicle carrying the transgene of interest to remain long enough in the organ to penetrate efficiently into the nephrotic cells. Also, the kidney has a complex anatomical structure composed of different cell types compartmentalized within isolated anatomic structures that limit their access. Here, we describe a simple surgical procedure to deliver recombinant adeno-associated virus (rAAV) to the whole kidney based on the hydraulic force of the retrograde renal vein injection. In its clinical form, this procedure would correspond to a renal venography where a catheter is threaded retrograde from the femoral vein under fluoroscopic guidance.


Asunto(s)
Vectores Genéticos/administración & dosificación , Riñón/cirugía , Venas Renales/virología , Animales , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Inyecciones Intravenosas , Riñón/inervación , Ratones , Modelos Animales , Transducción Genética , Transgenes , Procedimientos Quirúrgicos Urológicos
3.
Sci Rep ; 8(1): 8214, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29844458

RESUMEN

Genome editing is the introduction of directed modifications in the genome, a process boosted to therapeutic levels by designer nucleases. Building on the experience of ex vivo gene therapy for severe combined immunodeficiencies, it is likely that genome editing of haematopoietic stem/progenitor cells (HSPC) for correction of inherited blood diseases will be an early clinical application. We show molecular evidence of gene correction in a mouse model of primary immunodeficiency. In vitro experiments in DNA-dependent protein kinase catalytic subunit severe combined immunodeficiency (Prkdc scid) fibroblasts using designed zinc finger nucleases (ZFN) and a repair template demonstrated molecular and functional correction of the defect. Following transplantation of ex vivo gene-edited Prkdc scid HSPC, some of the recipient animals carried the expected genomic signature of ZFN-driven gene correction. In some primary and secondary transplant recipients we detected double-positive CD4/CD8 T-cells in thymus and single-positive T-cells in blood, but no other evidence of immune reconstitution. However, the leakiness of this model is a confounding factor for the interpretation of the possible T-cell reconstitution. Our results provide support for the feasibility of rescuing inherited blood disease by ex vivo genome editing followed by transplantation, and highlight some of the challenges.


Asunto(s)
Edición Génica , Inmunodeficiencia Combinada Grave/genética , Animales , Proteína Quinasa Activada por ADN/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID , Proteínas Nucleares/genética
4.
Endocrinology ; 157(4): 1363-71, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26812160

RESUMEN

Hypothyroidism is the most frequent and earliest endocrine complication in cystinosis, a multisystemic lysosomal storage disease caused by defective transmembrane cystine transporter, cystinosin (CTNS gene). We recently demonstrated in Ctns(-/-) mice that altered thyroglobulin biosynthesis associated with endoplasmic reticulum stress, combined with defective lysosomal processing, caused hypothyroidism. In Ctns(-/-) kidney, hematopoietic stem cell (HSC) transplantation provides long-term functional and structural protection. Tissue repair involves transfer of cystinosin-bearing lysosomes from HSCs differentiated as F4/80 macrophages into deficient kidney tubular cells, via tunneling nanotubes that cross basement laminae. Here we evaluated the benefit of HSC transplantation for cystinotic thyroid and investigated the underlying mechanisms. HSC engraftment in Ctns(-/-) thyroid drastically decreased cystine accumulation, normalized the TSH level, and corrected the structure of a large fraction of thyrocytes. In the thyroid microenvironment, HSCs differentiated into a distinct, mixed macrophage/dendritic cell lineage expressing CD45 and major histocompatibility complex II but low CD11b and F4/80. Grafted HSCs closely apposed to follicles and produced tunneling nanotube-like extensions that crossed follicular basement laminae. HSCs themselves further squeezed into follicles, allowing extensive contact with thyrocytes, but did not transdifferentiate into Nkx2.1-expressing cells. Our observations revealed significant differences of basement lamina porosity between the thyroid and kidney and/or intrinsic macrophage invasive properties once in the thyroid microenvironment. The contrast between extensive thyrocyte protection and low HSC abundance at steady state suggests multiple sequential encounters and/or remanent impact. This is the first report demonstrating the potential of HSC transplantation to correct thyroid disease and supports a major multisystemic benefit of stem cell therapy for cystinosis.


Asunto(s)
Cistinosis/terapia , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas/métodos , Glándula Tiroides/fisiopatología , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animales , Diferenciación Celular , Cistina/metabolismo , Cistinosis/genética , Cistinosis/fisiopatología , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Glándula Tiroides/metabolismo , Tirotropina/metabolismo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda