Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 39(8): e102468, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32154600

RESUMEN

Vertebrate vision relies on the daily phagocytosis and lysosomal degradation of photoreceptor outer segments (POS) within the retinal pigment epithelium (RPE). However, how these events are controlled by light is largely unknown. Here, we show that the light-responsive miR-211 controls lysosomal biogenesis at the beginning of light-dark transitions in the RPE by targeting Ezrin, a cytoskeleton-associated protein essential for the regulation of calcium homeostasis. miR-211-mediated down-regulation of Ezrin leads to Ca2+ influx resulting in the activation of calcineurin, which in turn activates TFEB, the master regulator of lysosomal biogenesis. Light-mediated induction of lysosomal biogenesis and function is impaired in the RPE from miR-211-/- mice that show severely compromised vision. Pharmacological restoration of lysosomal biogenesis through Ezrin inhibition rescued the miR-211-/- phenotype, pointing to a new therapeutic target to counteract retinal degeneration associated with lysosomal dysfunction.


Asunto(s)
Calcio/metabolismo , Proteínas del Citoesqueleto/metabolismo , Regulación de la Expresión Génica , Lisosomas/metabolismo , MicroARNs/metabolismo , Animales , Autofagia , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/genética , Regulación hacia Abajo , Luz , Lisosomas/ultraestructura , Ratones , Ratones Noqueados , MicroARNs/genética , Fagocitosis , Fagosomas/metabolismo , Fagosomas/ultraestructura , Epitelio Pigmentado de la Retina/metabolismo
2.
Am J Hum Genet ; 108(2): 295-308, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33508235

RESUMEN

Retinitis pigmentosa (RP) is a group of progressive retinal degenerations of mostly monogenic inheritance, which cause blindness in about 1:3,500 individuals worldwide. Heterozygous variants in the rhodopsin (RHO) gene are the most common cause of autosomal dominant RP (adRP). Among these, missense variants at C-terminal proline 347, such as p.Pro347Ser, cause severe adRP recurrently in European affected individuals. Here, for the first time, we use CRISPR/Cas9 to selectively target the p.Pro347Ser variant while preserving the wild-type RHO allele in vitro and in a mouse model of adRP. Detailed in vitro, genomic, and biochemical characterization of the rhodopsin C-terminal editing demonstrates a safe downregulation of p.Pro347Ser expression leading to partial recovery of photoreceptor function in a transgenic mouse model treated with adeno-associated viral vectors. This study supports the safety and efficacy of CRISPR/Cas9-mediated allele-specific editing and paves the way for a permanent and precise correction of heterozygous variants in dominantly inherited retinal diseases.


Asunto(s)
Edición Génica , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Rodopsina/genética , Alelos , Animales , Sistemas CRISPR-Cas , Línea Celular , Dependovirus/genética , Modelos Animales de Enfermedad , Electrorretinografía , Terapia Genética , Humanos , Mutación INDEL , Ratones , Ratones Transgénicos , Mutación Missense , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Retina/fisiopatología , Rodopsina/metabolismo
3.
Hum Mol Genet ; 28(R1): R108-R118, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31238338

RESUMEN

Retinal gene therapy has advanced considerably in the past three decades. Initial efforts have been devoted to comprehensively explore and optimize the transduction abilities of gene delivery vectors, define the appropriate intraocular administration routes and obtain evidence of efficacy in animal models of inherited retinal diseases (IRDs). Successful translation in clinical trials of the initial promising proof-of-concept studies led to the important milestone of the first approved product for retinal gene therapy in both US and Europe. The unprecedented clinical development observed during the last decade in the field is however highlighting new challenges that will need to be overcome to bring gene therapy to fruition to a larger patient population within and beyond the realm of IRDs.


Asunto(s)
Terapia Genética , Retina/metabolismo , Enfermedades de la Retina/genética , Enfermedades de la Retina/terapia , Animales , Ensayos Clínicos como Asunto , Edición Génica , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Predisposición Genética a la Enfermedad , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Transducción Genética , Transgenes , Investigación Biomédica Traslacional
4.
Eur Heart J ; 41(26): 2487-2497, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-31289820

RESUMEN

AIMS: Here, we aimed to determine the therapeutic effect of longevity-associated variant (LAV)-BPIFB4 gene therapy on atherosclerosis. METHODS AND RESULTS: ApoE knockout mice (ApoE-/-) fed a high-fat diet were randomly allocated to receive LAV-BPIFB4, wild-type (WT)-BPIFB4, or empty vector via adeno-associated viral vector injection. The primary endpoints of the study were to assess (i) vascular reactivity and (ii) atherosclerotic disease severity, by Echo-Doppler imaging, histology and ultrastructural analysis. Moreover, we assessed the capacity of the LAV-BPIFB4 protein to shift monocyte-derived macrophages of atherosclerotic mice and patients towards an anti-inflammatory phenotype. LAV-BPIFB4 gene therapy rescued endothelial function of mesenteric and femoral arteries from ApoE-/- mice; this effect was blunted by AMD3100, a CXC chemokine receptor type 4 (CXCR4) inhibitor. LAV-BPIFB4-treated mice showed a CXCR4-mediated shift in the balance between Ly6Chigh/Ly6Clow monocytes and M2/M1 macrophages, along with decreased T cell proliferation and elevated circulating levels of interleukins IL-23 and IL-27. In vitro conditioning with LAV-BPIFB4 protein of macrophages from atherosclerotic patients resulted in a CXCR4-dependent M2 polarization phenotype. Furthermore, LAV-BPIFB4 treatment of arteries explanted from atherosclerotic patients increased the release of atheroprotective IL-33, while inhibiting the release of pro-inflammatory IL-1ß, inducing endothelial nitric oxide synthase phosphorylation and restoring endothelial function. Finally, significantly lower plasma BPIFB4 was detected in patients with pathological carotid stenosis (>25%) and intima media thickness >2 mm. CONCLUSION: Transfer of the LAV of BPIFB4 reduces the atherogenic process and skews macrophages towards an M2-resolving phenotype through modulation of CXCR4, thus opening up novel therapeutic possibilities in cardiovascular disease.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Anciano , Animales , Apolipoproteínas E , Aterosclerosis/genética , Grosor Intima-Media Carotídeo , Femenino , Humanos , Inflamación , Péptidos y Proteínas de Señalización Intercelular , Longevidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Persona de Mediana Edad , Fosfoproteínas , Receptores CXCR4
5.
Circ Res ; 121(5): 525-536, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28620067

RESUMEN

RATIONALE: Mutations in the cardiac Ryanodine Receptor gene (RYR2) cause dominant catecholaminergic polymorphic ventricular tachycardia (CPVT), a leading cause of sudden death in apparently healthy individuals exposed to emotions or physical exercise. OBJECTIVE: We investigated the efficacy of allele-specific silencing by RNA interference to prevent CPVT phenotypic manifestations in our dominant CPVT mice model carriers of the heterozygous mutation R4496C in RYR2. METHODS AND RESULTS: We developed an in vitro mRNA and protein-based assays to screen multiple siRNAs for their ability to selectively silence mutant RYR2-R4496C mRNA over the corresponding wild-type allele. For the most performant of these siRNAs (siRYR2-U10), we evaluated the efficacy of an adeno-associated serotype 9 viral vector (AAV9) expressing miRYR2-U10 in correcting RyR2 (Ryanodine Receptor type 2 protein) function after in vivo delivery by intraperitoneal injection in neonatal and adult RyR2R4496C/+ (mice heterozygous for the R4496C mutation in the RyR2) heterozygous CPVT mice. Transcriptional analysis showed that after treatment with miRYR2-U10, the ratio between wild-type and mutant RYR2 mRNA was doubled (from 1:1 to 2:1) confirming the ability of miRYR2-U10 to selectively inhibit RYR2-R4496C mRNA, whereas protein quantification showed that total RyR2 was reduced by 15% in the heart of treated mice. Furthermore, AAV9-miRYR2-U10 effectively (1) reduced isoproterenol-induced delayed afterdepolarizations and triggered activity in infected cells, (2) reduced adrenergically mediated ventricular tachycardia in treated mice, (3) reverted ultrastructural abnormalities of junctional sarcoplasmic reticulum and transverse tubules, and (4) attenuated mitochondrial abnormalities. CONCLUSIONS: The study demonstrates that allele-specific silencing with miRYR2-U10 prevents life-threatening arrhythmias in CPVT mice, suggesting that the reduction of mutant RyR2 may be a novel therapeutic approach for CPVT.


Asunto(s)
Alelos , Arritmias Cardíacas/genética , Heterocigoto , Mutación/genética , ARN Mensajero/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Animales , Animales Recién Nacidos , Arritmias Cardíacas/patología , Arritmias Cardíacas/prevención & control , Células Cultivadas , Silenciador del Gen/fisiología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , ARN Mensajero/ultraestructura , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Canal Liberador de Calcio Receptor de Rianodina/ultraestructura
6.
Mol Ther ; 26(2): 524-541, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29292161

RESUMEN

Retinal gene transfer with adeno-associated viral (AAV) vectors holds great promise for the treatment of inherited retinal degenerations (IRDs). One limit of AAV is its transfer capacity of about 5 kb, which can be expanded to about 9 kb, using dual AAV vectors. This strategy would still not suffice for treatment of IRDs such as Usher syndrome type 1D or Alström syndrome type I (ALMS) due to mutations in CDH23 or ALMS1, respectively. To overcome this limitation, we generated triple AAV vectors, with a maximal transfer capacity of about 14 kb. Transcriptomic analysis following triple AAV transduction showed the expected full-length products along a number of aberrant transcripts. However, only the full-length transcripts are efficiently translated in vivo. We additionally showed that approximately 4% of mouse photoreceptors are transduced by triple AAV vectors and showed correct localization of recombinant ALMS1. The low-photoreceptor transduction levels might justify the modest and transient improvement we observe in the retina of a mouse model of ALMS. However, the levels of transduction mediated by triple AAV vectors in pig retina reached 40% of those observed with single vectors, and this bodes well for further improving the efficiency of triple AAV vectors in the retina.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Recombinación Genética , Retina/metabolismo , Transducción Genética , Animales , Cadherinas/genética , Cadherinas/metabolismo , Expresión Génica , Regulación Viral de la Expresión Génica , Técnicas de Transferencia de Gen , Genes Reporteros , Terapia Genética , Vectores Genéticos/administración & dosificación , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Porcinos , Transcripción Genética , Transgenes
7.
Retina ; 39(7): 1399-1409, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29642238

RESUMEN

PURPOSE: To investigate the natural history of Stargardt disease over a multiyear follow-up. METHODS: We reviewed medical records of Stargardt disease patients, with clinical diagnosis of Stargardt disease at a single institution, which was also supported by molecular diagnosis. All patients underwent best-corrected visual acuity, fundus photography, optical coherence tomography, and full-field electroretinography. RESULTS: The study cohort consisted of 157 Stargardt disease patients aged 30.4 ± 1.1 years. Longitudinal analysis (mean follow-up: 3 years) showed a significant worsening of best-corrected visual acuity at an average rate of 1.5 Early Treatment Diabetic Retinopathy Study letters/year (P < 0.001), an enlargement of retinal pigment epithelium lesion area by optical coherence tomography at an average linear rate of 0.10 mm/year (P < 0.001), and a thinning of central macular thickness at a mean rate of -1.42 µm/year (P < 0.001). Survival analysis showed that patients with 2 alleles harboring likely-null variants, on average, reached most severe disease stage, i.e., legal blindness, alteration in both dark-adapted and light-adapted electroretinographic responses, and retinal pigment epithelium lesion area larger than 2.5 mm significantly earlier than patients with at least one allele harboring a missense variant. CONCLUSION: The current longitudinal study showed a significant genotype-phenotype correlation characterization, because patients harboring 2 likely-null alleles reach a severe disease stage about 10 years earlier than patients with at least one missense allele.


Asunto(s)
Angiografía con Fluoresceína/métodos , Predicción , Oftalmoscopía/métodos , Epitelio Pigmentado de la Retina/patología , Enfermedad de Stargardt/diagnóstico , Tomografía de Coherencia Óptica/métodos , Agudeza Visual , Transportadoras de Casetes de Unión a ATP/genética , Adulto , Progresión de la Enfermedad , Electrorretinografía , Femenino , Estudios de Seguimiento , Fondo de Ojo , Estudios de Asociación Genética , Humanos , Incidencia , Italia/epidemiología , Masculino , Mutación , Estudios Retrospectivos , Enfermedad de Stargardt/epidemiología , Enfermedad de Stargardt/genética
8.
Hum Mol Genet ; 24(7): 1843-55, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452428

RESUMEN

Selenoprotein N (SEPN1) is a broadly expressed resident protein of the endoplasmic reticulum (ER) whose loss-of-function inexplicably leads to human muscle disease. We found that SEPN1 levels parallel those of endoplamic reticulum oxidoreductin 1 (ERO1), an ER protein thiol oxidase, and that SEPN1's redox activity defends the ER from ERO1-generated peroxides. Moreover, we have defined the redox-regulated interactome of SEPN1 and identified the ER calcium import SERCA2 pump as a redox-partner of SEPN1. SEPN1 enhances SERCA2 activity by reducing luminal cysteines that are hyperoxidized by ERO1-generated peroxides. Cells lacking SEPN1 are hypersensitive to ERO1 overexpression and conspicuously defective in ER calcium re-uptake. After being muscle-transduced with an adeno-associated virus driving ERO1α, SEPN1 knockout mice unmasks a myopathy that resembles the dense core disease due to human mutations in SEPN1, whereas the combined attenuation of ERO1α and SEPN1 enhances cell fitness. These observations reveal the involvement of SEPN1 in ER redox and calcium homeostasis and that an ERO1 inhibitor, restoring redox-dependent calcium homeostasis, may ameliorate the myopathy of SEPN1 deficiency.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Selenoproteínas/metabolismo , Animales , Cisteína/metabolismo , Retículo Endoplásmico/genética , Humanos , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Músculo Esquelético/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Oxidación-Reducción , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Peróxidos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Selenoproteínas/genética
9.
Hum Mol Genet ; 24(23): 6811-25, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26420842

RESUMEN

Stargardt disease (STGD1) due to mutations in the large ABCA4 gene is the most common inherited macular degeneration in humans. We have shown that dual adeno-associated viral (AAV) vectors effectively transfer ABCA4 to the retina of Abca4-/- mice. However, they express both lower levels of transgene compared with a single AAV and truncated proteins. To increase productive dual AAV concatemerization, which would overcome these limitations, we have explored the use of either various regions of homology or heterologous inverted terminal repeats (ITR). In addition, we tested the ability of various degradation signals to decrease the expression of truncated proteins. We found the highest levels of transgene expression using regions of homology based on either alkaline phosphatase or the F1 phage (AK). The use of heterologous ITR does not decrease the levels of truncated proteins relative to full-length ABCA4 and impairs AAV vector production. Conversely, the inclusion of the CL1 degradation signal results in the selective degradation of truncated proteins from the 5'-half without affecting full-length protein production. Therefore, we developed dual AAV hybrid ABCA4 vectors including homologous ITR2, the photoreceptor-specific G protein-coupled receptor kinase 1 promoter, the AK region of homology and the CL1 degradation signal. We show that upon subretinal administration these vectors are both safe in pigs and effective in Abca4-/- mice. Our data support the use of improved dual AAV vectors for gene therapy of STGD1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos , Degeneración Macular/congénito , Administración Oftálmica , Animales , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Células HEK293 , Humanos , Degeneración Macular/genética , Degeneración Macular/terapia , Ratones , Retina/metabolismo , Enfermedad de Stargardt , Porcinos , Secuencias Repetidas Terminales , Transgenes
10.
Lancet ; 388(10045): 661-72, 2016 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-27375040

RESUMEN

BACKGROUND: Safety and efficacy have been shown in a phase 1 dose-escalation study involving a unilateral subretinal injection of a recombinant adeno-associated virus (AAV) vector containing the RPE65 gene (AAV2-hRPE65v2) in individuals with inherited retinal dystrophy caused by RPE65 mutations. This finding, along with the bilateral nature of the disease and intended use in treatment, prompted us to determine the safety of administration of AAV2-hRPE65v2 to the contralateral eye in patients enrolled in the phase 1 study. METHODS: In this follow-on phase 1 trial, one dose of AAV2-hRPE65v2 (1.5 × 10(11) vector genomes) in a total volume of 300 µL was subretinally injected into the contralateral, previously uninjected, eyes of 11 children and adults (aged 11-46 years at second administration) with inherited retinal dystrophy caused by RPE65 mutations, 1.71-4.58 years after the initial subretinal injection. We assessed safety, immune response, retinal and visual function, functional vision, and activation of the visual cortex from baseline until 3 year follow-up, with observations ongoing. This study is registered with ClinicalTrials.gov, number NCT01208389. FINDINGS: No adverse events related to the AAV were reported, and those related to the procedure were mostly mild (dellen formation in three patients and cataracts in two). One patient developed bacterial endophthalmitis and was excluded from analyses. We noted improvements in efficacy outcomes in most patients without significant immunogenicity. Compared with baseline, pooled analysis of ten participants showed improvements in mean mobility and full-field light sensitivity in the injected eye by day 30 that persisted to year 3 (mobility p=0.0003, white light full-field sensitivity p<0.0001), but no significant change was seen in the previously injected eyes over the same time period (mobility p=0.7398, white light full-field sensitivity p=0.6709). Changes in visual acuity from baseline to year 3 were not significant in pooled analysis in the second eyes or the previously injected eyes (p>0.49 for all time-points compared with baseline). INTERPRETATION: To our knowledge, AAV2-hRPE65v2 is the first successful gene therapy administered to the contralateral eye. The results highlight the use of several outcome measures and help to delineate the variables that contribute to maximal benefit from gene augmentation therapy in this disease. FUNDING: Center for Cellular and Molecular Therapeutics at The Children's Hospital of Philadelphia, Spark Therapeutics, US National Institutes of Health, Foundation Fighting Blindness, Institute for Translational Medicine and Therapeutics, Research to Prevent Blindness, Center for Advanced Retinal and Ocular Therapeutics, Mackall Foundation Trust, F M Kirby Foundation, and The Research Foundation-Flanders.


Asunto(s)
Ceguera/genética , Ceguera/terapia , Dependovirus , Terapia Genética/métodos , Mutación , Lóbulo Occipital/fisiopatología , Visión Ocular , cis-trans-Isomerasas/genética , Administración Oftálmica , Adolescente , Adulto , Edad de Inicio , Ceguera/patología , Ceguera/fisiopatología , Niño , Medicina Basada en la Evidencia , Femenino , Estudios de Seguimiento , Terapia Genética/efectos adversos , Vectores Genéticos , Humanos , Inyecciones Intraoculares , Modelos Lineales , Masculino , Persona de Mediana Edad , Seguridad del Paciente , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retratamiento
11.
Circ Res ; 117(4): 333-45, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26034043

RESUMEN

RATIONALE: Long living individuals show delay of aging, which is characterized by the progressive loss of cardiovascular homeostasis, along with reduced endothelial nitric oxide synthase activity, endothelial dysfunction, and impairment of tissue repair after ischemic injury. OBJECTIVE: Exploit genetic analysis of long living individuals to reveal master molecular regulators of physiological aging and new targets for treatment of cardiovascular disease. METHODS AND RESULTS: We show that the polymorphic variant rs2070325 (Ile229Val) in bactericidal/permeability-increasing fold-containing-family-B-member-4 (BPIFB4) associates with exceptional longevity, under a recessive genetic model, in 3 independent populations. Moreover, the expression of BPIFB4 is instrumental to maintenance of cellular and vascular homeostasis through regulation of protein synthesis. BPIFB4 phosphorylation/activation by protein-kinase-R-like endoplasmic reticulum kinase induces its complexing with 14-3-3 and heat shock protein 90, which is facilitated by the longevity-associated variant. In isolated vessels, BPIFB4 is upregulated by mechanical stress, and its knock-down inhibits endothelium-dependent vasorelaxation. In hypertensive rats and old mice, gene transfer of longevity-associated variant-BPIFB4 restores endothelial nitric oxide synthase signaling, rescues endothelial dysfunction, and reduces blood pressure levels. Furthermore, BPIFB4 is implicated in vascular repair. BPIFB4 is abundantly expressed in circulating CD34(+) cells of long living individuals, and its knock-down in endothelial progenitor cells precludes their capacity to migrate toward the chemoattractant SDF-1. In a murine model of peripheral ischemia, systemic gene therapy with longevity-associated variant-BPIFB4 promotes the recruitment of hematopoietic stem cells, reparative vascularization, and reperfusion of the ischemic muscle. CONCLUSIONS: Longevity-associated variant-BPIFB4 may represent a novel therapeutic tool to fight endothelial dysfunction and promote vascular reparative processes.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Longevidad/genética , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas 14-3-3/metabolismo , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Presión Sanguínea , Movimiento Celular , Modelos Animales de Enfermedad , Europa (Continente) , Femenino , Estudios de Asociación Genética , Terapia Genética , Genotipo , Células HEK293 , Proteínas HSP90 de Choque Térmico/metabolismo , Miembro Posterior , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipertensión/terapia , Péptidos y Proteínas de Señalización Intercelular , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/terapia , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenotipo , Fosforilación , Interferencia de ARN , Ratas Endogámicas SHR , Transducción de Señal , Estrés Mecánico , Transfección , Estados Unidos , Vasodilatación , eIF-2 Quinasa/metabolismo
12.
Mol Ther ; 24(12): 2054-2063, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27658524

RESUMEN

Enzyme replacement therapy (ERT) is the standard of care for several lysosomal storage diseases (LSDs). ERT, however, requires multiple and costly administrations and has limited efficacy. We recently showed that a single high dose administration of adeno-associated viral vector serotype 8 (AAV2/8) is at least as effective as weekly ERT in a mouse model of mucopolysaccharidosis type VI (MPS VI). However, systemic administration of high doses of AAV might result in both cell-mediated immune responses and insertional mutagenesis. Here we evaluated whether the combination of low doses of AAV2/8 with a less frequent (monthly) than canonical (weekly) ERT schedule may be as effective as the single treatments at high doses or frequent regimen. A greater reduction of both urinary glycosaminoglycans, considered a sensitive biomarker of therapeutic efficacy, and storage in the myocardium and heart valves was observed in mice receiving the combined than the single therapies. Importantly, these levels of correction were similar to those we obtained in a previous study following either high doses of AAV2/8 or weekly ERT. Our data show that low-dose gene therapy can be used as a means to rarify ERT administration, thus reducing both the risks and costs associated with either therapies.


Asunto(s)
Terapia Combinada/métodos , Terapia de Reemplazo Enzimático/métodos , Terapia Genética/métodos , Enfermedades por Almacenamiento Lisosomal/terapia , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Glicosaminoglicanos/orina , Humanos , Enfermedades por Almacenamiento Lisosomal/orina , Ratones , Resultado del Tratamiento
13.
Mol Ther ; 24(2): 276-286, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26639405

RESUMEN

Cerebrospinal fluid administration of recombinant adeno-associated viral (rAAV) vectors has been demonstrated to be effective in delivering therapeutic genes to the central nervous system (CNS) in different disease animal models. However, a quantitative and qualitative analysis of transduction patterns of the most promising rAAV serotypes for brain targeting in large animal models is missing. Here, we characterize distribution, transduction efficiency, and cellular targeting of rAAV serotypes 1, 2, 5, 7, 9, rh.10, rh.39, and rh.43 delivered into the cisterna magna of wild-type pigs. rAAV9 showed the highest transduction efficiency and the widest distribution capability among the vectors tested. Moreover, rAAV9 robustly transduced both glia and neurons, including the motor neurons of the spinal cord. Relevant cell transduction specificity of the glia was observed after rAAV1 and rAAV7 delivery. rAAV7 also displayed a specific tropism to Purkinje cells. Evaluation of biochemical and hematological markers suggested that all rAAV serotypes tested were well tolerated. This study provides a comprehensive CNS transduction map in a useful preclinical large animal model enabling the selection of potentially clinically transferable rAAV serotypes based on disease specificity. Therefore, our data are instrumental for the clinical evaluation of these rAAV vectors in human neurodegenerative diseases.


Asunto(s)
Sistema Nervioso Central/metabolismo , Dependovirus/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/líquido cefalorraquídeo , Proteínas Fluorescentes Verdes/metabolismo , Animales , Dependovirus/inmunología , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Especificidad de Órganos , Serogrupo , Porcinos , Transducción Genética , Transgenes
14.
Retina ; 37(8): 1581-1590, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27828912

RESUMEN

PURPOSE: To evaluate differences in the visual phenotype and natural history of Usher syndrome caused by mutations in MYO7A or USH2A, the most commonly affected genes of Usher syndrome Type I (USH1) and Type II (USH2), respectively. METHODS: Eighty-eight patients with a clinical diagnosis of USH1 (26 patients) or USH2 (62 patients) were retrospectively evaluated. Of these, 48 patients had 2 disease-causing mutations in MYO7A (10 USH1 patients), USH2A (33 USH2 patients), and other USH (5 patients) genes. Clinical investigation included best-corrected visual acuity, Goldmann visual field, fundus photography, electroretinography, and audiologic and vestibular assessments. Longitudinal analysis was performed over a median follow-up time of 3.5 years. RESULTS: Patients carrying mutations in MYO7A had a younger age of onset of hearing and visual impairments than those carrying mutations in USH2A, leading to an earlier diagnosis of the disease in the former patients. Longitudinal analysis showed that visual acuity and visual field decreased more rapidly in subjects carrying MYO7A mutations than in those carrying USH2A mutations (mean annual exponential rates of decline of 3.92 vs. 3.44% and of 8.52 vs. 4.97%, respectively), and the former patients reached legal blindness on average 15 years earlier than the latter. CONCLUSION: The current study confirmed a more severe progression of the retinal disease in USH1 patients rather than in USH2 patients. Furthermore, most visual symptoms (i.e., night blindness, visual acuity worsening) occurred at an earlier age in USH1 patients carrying mutations in MYO7A.


Asunto(s)
ADN/genética , Proteínas de la Matriz Extracelular/genética , Mutación , Miosinas/genética , Síndromes de Usher/genética , Agudeza Visual , Campos Visuales , Adolescente , Adulto , Análisis Mutacional de ADN , Progresión de la Enfermedad , Electrorretinografía , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Miosina VIIa , Miosinas/metabolismo , Fenotipo , Retina/diagnóstico por imagen , Retina/fisiopatología , Estudios Retrospectivos , Factores de Tiempo , Tomografía de Coherencia Óptica , Síndromes de Usher/diagnóstico , Síndromes de Usher/fisiopatología , Adulto Joven
16.
Mol Ther ; 23(5): 885-895, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25669433

RESUMEN

Although in the last decades the molecular underpinnings of the cell cycle have been unraveled, the acquired knowledge has been rarely translated into practical applications. Here, we investigate the feasibility and safety of triggering proliferation in vivo by temporary suppression of the cyclin-dependent kinase inhibitor, p21. Adeno-associated virus (AAV)-mediated, acute knockdown of p21 in intact skeletal muscles elicited proliferation of multiple, otherwise quiescent cell types, notably including satellite cells. Compared with controls, p21-suppressed muscles exhibited a striking two- to threefold expansion in cellularity and increased fiber numbers by 10 days post-transduction, with no detectable inflammation. These changes partially persisted for at least 60 days, indicating that the muscles had undergone lasting modifications. Furthermore, morphological hyperplasia was accompanied by 20% increases in maximum strength and resistance to fatigue. To assess the safety of transiently suppressing p21, cells subjected to p21 knockdown in vitro were analyzed for γ-H2AX accumulation, DNA fragmentation, cytogenetic abnormalities, ploidy, and mutations. Moreover, the differentiation competence of p21-suppressed myoblasts was investigated. These assays confirmed that transient suppression of p21 causes no genetic damage and does not impair differentiation. Our results establish the basis for further exploring the manipulation of the cell cycle as a strategy in regenerative medicine.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular , Aberraciones Cromosómicas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Dependovirus/clasificación , Dependovirus/genética , Fibroblastos , Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Ratones , Contracción Muscular/genética , Mutación , Interferencia de ARN , ARN Interferente Pequeño/genética , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Serogrupo , Transducción Genética
17.
Adv Exp Med Biol ; 854: 533-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427456

RESUMEN

Gene therapy clinical trials with gene augmentation therapy for Leber Congenital Amaurosis have shown partial reversal of retinal dysfunction. Most studies described the effect of treatment in a single eye and limited evidence is reported in literature about patients treated in both eyes. In this chapter, we present the findings of a young patient treated in both eyes. Efficacy of the treatment was assessed with Best Corrected Visual Acuity, Goldman Visual Field testing, Esterman computerized binocular visual field and Microperimetric testing. Post-treatment results showed improvement of visual function in both eyes, in particular, a strong amelioration was observed after the first injection, by using conventional monocular tests. Moreover, the treatment in the second eye resulted in a further improvement of binocular visual functionality, as easily detected by computerized binocular visual field. In conclusion, our data suggest that gene therapy can inhibit retinal degeneration and can be safe and effective in restoring visual functionality in young subjects treated in both eyes. Finally, new outcome measurements, in particular binocular computerized visual field parameters, can therefore be useful to quantify overall visual gain in patients undergoing gene therapy in both eyes.


Asunto(s)
Ojo/metabolismo , Terapia Genética/métodos , Amaurosis Congénita de Leber/terapia , cis-trans-Isomerasas/metabolismo , Niño , Dependovirus/genética , Estudios de Seguimiento , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Inyecciones , Italia , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/fisiopatología , Resultado del Tratamiento , Visión Binocular/fisiología , Visión Ocular/fisiología , Agudeza Visual/fisiología , Campos Visuales/fisiología , cis-trans-Isomerasas/genética
18.
Circulation ; 129(25): 2673-81, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24888331

RESUMEN

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia is an inherited arrhythmogenic disorder characterized by sudden cardiac death in children. Drug therapy is still insufficient to provide full protection against cardiac arrest, and the use of implantable defibrillators in the pediatric population is limited by side effects. There is therefore a need to explore the curative potential of gene therapy for this disease. We investigated the efficacy and durability of viral gene transfer of the calsequestrin 2 (CASQ2) wild-type gene in a catecholaminergic polymorphic ventricular tachycardia knock-in mouse model carrying the CASQ2(R33Q/R33Q) (R33Q) mutation. METHODS AND RESULTS: We engineered an adeno-associated viral vector serotype 9 (AAV9) containing cDNA of CASQ2 wild-type (AAV9-CASQ2) plus the green fluorescent protein (GFP) gene to infect newborn R33Q mice studied by in vivo and in vitro protocols at 6, 9, and 12 months to investigate the ability of the infection to prevent the disease and adult R33Q mice studied after 2 months to assess whether the AAV9-CASQ2 delivery could revert the catecholaminergic polymorphic ventricular tachycardia phenotype. In both protocols, we observed the restoration of physiological expression and interaction of CASQ2, junctin, and triadin; the rescue of electrophysiological and ultrastructural abnormalities in calcium release units present in R33Q mice; and the lack of life-threatening arrhythmias. CONCLUSIONS: Our data demonstrate that viral gene transfer of wild-type CASQ2 into the heart of R33Q mice prevents and reverts severe manifestations of catecholaminergic polymorphic ventricular tachycardia and that this curative effect lasts for 1 year after a single injection of the vector, thus posing the rationale for the design of a clinical trial.


Asunto(s)
Envejecimiento , Calsecuestrina/genética , Dependovirus/genética , Taquicardia Ventricular/terapia , Animales , Proteínas de Unión al Calcio/metabolismo , Calsecuestrina/metabolismo , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Oxigenasas de Función Mixta/metabolismo , Proteínas Musculares/metabolismo , Mutación/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología , Resultado del Tratamiento
19.
Mol Ther ; 22(1): 10-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24247928

RESUMEN

Mutations in human MPV17 cause a hepatocerebral form of mitochondrial DNA depletion syndrome (MDS) hallmarked by early-onset liver failure, leading to premature death. Liver transplantation and frequent feeding using slow-release carbohydrates are the only available therapies, although surviving patients eventually develop slowly progressive peripheral and central neuropathy. The physiological role of Mpv17, including its functional link to mitochondrial DNA (mtDNA) maintenance, is still unclear. We show here that Mpv17 is part of a high molecular weight complex of unknown composition, which is essential for mtDNA maintenance in critical tissues, i.e. liver, of a Mpv17 knockout mouse model. On a standard diet, Mpv17-/- mouse shows hardly any symptom of liver dysfunction, but a ketogenic diet (KD) leads these animals to liver cirrhosis and failure. However, when expression of human MPV17 is carried out by adeno-associated virus (AAV)-mediated gene replacement, the Mpv17 knockout mice are able to reconstitute the Mpv17-containing supramolecular complex, restore liver mtDNA copy number and oxidative phosphorylation (OXPHOS) proficiency, and prevent liver failure induced by the KD. These results open new therapeutic perspectives for the treatment of MPV17-related liver-specific MDS.


Asunto(s)
ADN Mitocondrial , Dependovirus/genética , Dieta Cetogénica/efectos adversos , Vectores Genéticos/genética , Fallo Hepático/etiología , Fallo Hepático/terapia , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Genotipo , Humanos , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Cirrosis Hepática/terapia , Fallo Hepático/patología , Fallo Hepático/prevención & control , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Peso Molecular , Fenotipo , Multimerización de Proteína
20.
Mol Ther ; 22(5): 901-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24448160

RESUMEN

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is an autosomal recessive disorder caused by mutations in TYMP, enconding thymidine phosphorylase (TP). TP deficiency results in systemic accumulation of thymidine and deoxyuridine, which interferes with mitochondrial DNA (mtDNA) replication and leads to mitochondrial dysfunction. To date, the only treatment available for MNGIE patients is allogeneic hematopoietic stem cell transplantation, which is associated with high morbidity and mortality. Here, we report that AAV2/8-mediated transfer of the human TYMP coding sequence (hcTYMP) under the control of a liver-specific promoter prevents the biochemical imbalances in a murine model of MNGIE. hcTYMP expression was restricted to liver, and a dose as low as 2 × 10(11) genome copies/kg led to a permanent reduction in systemic nucleoside levels to normal values in about 50% of treated mice. Higher doses resulted in reductions to normal or slightly below normal levels in virtually all mice treated. The nucleoside reduction achieved by this treatment prevented deoxycytidine triphosphate (dCTP) depletion, which is the limiting factor affecting mtDNA replication in this disease. These results demonstrate that the use of AAV to direct TYMP expression in liver is feasible as a potentially safe gene therapy strategy for MNGIE.


Asunto(s)
Terapia Genética , Seudoobstrucción Intestinal/genética , Seudoobstrucción Intestinal/terapia , Encefalomiopatías Mitocondriales/genética , Encefalomiopatías Mitocondriales/terapia , Timidina Fosforilasa/genética , Animales , ADN Mitocondrial/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Homeostasis/genética , Humanos , Seudoobstrucción Intestinal/patología , Hígado/metabolismo , Ratones , Encefalomiopatías Mitocondriales/patología , Distrofia Muscular Oculofaríngea , Mutación , Oftalmoplejía/congénito , Timidina/metabolismo , Timidina Fosforilasa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA