Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36835257

RESUMEN

Age-related macular degeneration (AMD) is the most common cause of blindness in the aged population. However, to date there is no effective treatment for the dry form of the disease, representing 85-90% of cases. AMD is an immensely complex disease which affects, amongst others, both retinal pigment epithelium (RPE) and photoreceptor cells and leads to the progressive loss of central vision. Mitochondrial dysfunction in both RPE and photoreceptor cells is emerging as a key player in the disease. There are indications that during disease progression, the RPE is first impaired and RPE dysfunction in turn leads to subsequent photoreceptor cell degeneration; however, the exact sequence of events has not as yet been fully determined. We recently showed that AAV delivery of an optimised NADH-ubiquinone oxidoreductase (NDI1) gene, a nuclear-encoded complex 1 equivalent from S. cerevisiae, expressed from a general promoter, provided robust benefit in a variety of murine and cellular models of dry AMD; this was the first study employing a gene therapy to directly boost mitochondrial function, providing functional benefit in vivo. However, use of a restricted RPE-specific promoter to drive expression of the gene therapy enables exploration of the optimal target retinal cell type for dry AMD therapies. Furthermore, such restricted transgene expression could reduce potential off-target effects, possibly improving the safety profile of the therapy. Therefore, in the current study, we interrogate whether expression of the gene therapy from the RPE-specific promoter, Vitelliform macular dystrophy 2 (VMD2), might be sufficient to rescue dry AMD models.


Asunto(s)
Terapia Genética , Atrofia Geográfica , Proteínas de Saccharomyces cerevisiae , Anciano , Animales , Humanos , Ratones , Complejo I de Transporte de Electrón/metabolismo , Terapia Genética/métodos , Atrofia Geográfica/genética , Atrofia Geográfica/terapia , Mitocondrias/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
2.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-35163535

RESUMEN

The challenge of developing gene therapies for genetic forms of blindness is heightened by the heterogeneity of these conditions. However, mechanistic commonalities indicate key pathways that may be targeted in a gene-independent approach. Mitochondrial dysfunction and axon degeneration are common features of many neurodegenerative conditions including retinal degenerations. Here we explore the neuroprotective effect afforded by the absence of sterile alpha and Toll/interleukin-1 receptor motif-containing 1 (SARM1), a prodegenerative NADase, in a rotenone-induced mouse model of retinal ganglion cell loss and visual dysfunction. Sarm1 knockout mice retain visual function after rotenone insult, displaying preservation of photopic negative response following rotenone treatment in addition to significantly higher optokinetic response measurements than wild type mice following rotenone. Protection of spatial vision is sustained over time in both sexes and is accompanied by increased RGC survival and additionally preservation of axonal density in optic nerves of Sarm1-/- mice insulted with rotenone. Primary fibroblasts extracted from Sarm1-/- mice demonstrate an increased oxygen consumption rate relative to those from wild type mice, with significantly higher basal, maximal and spare respiratory capacity. Collectively, our data indicate that Sarm1 ablation increases mitochondrial bioenergetics and confers histological and functional protection in vivo in the mouse retina against mitochondrial dysfunction, a hallmark of many neurodegenerative conditions including a variety of ocular disorders.


Asunto(s)
Proteínas del Dominio Armadillo/genética , Proteínas del Citoesqueleto/genética , Fibroblastos/metabolismo , Degeneración Retiniana/prevención & control , Células Ganglionares de la Retina/fisiología , Rotenona/efectos adversos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Fibroblastos/citología , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Consumo de Oxígeno , Cultivo Primario de Células , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/genética
3.
Hum Mol Genet ; 26(R1): R2-R11, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28510639

RESUMEN

While individually classed as rare diseases, hereditary retinal degenerations (IRDs) are the major cause of registered visual handicap in the developed world. Given their hereditary nature, some degree of intergenic heterogeneity was expected, with genes segregating in autosomal dominant, recessive, X-linked recessive, and more rarely in digenic or mitochondrial modes. Today, it is recognized that IRDs, as a group, represent one of the most genetically diverse of hereditary conditions - at least 260 genes having been implicated, with 70 genes identified in the most common IRD, retinitis pigmentosa (RP). However, targeted sequencing studies of exons from known IRD genes have resulted in the identification of candidate mutations in only approximately 60% of IRD cases. Given recent advances in the development of gene-based medicines, characterization of IRD patient cohorts for known IRD genes and elucidation of the molecular pathologies of disease in those remaining unresolved cases has become an endeavor of the highest priority. Here, we provide an outline of progress in this area.


Asunto(s)
Degeneración Retiniana/genética , Secuencia Conservada , Exones , Proteínas del Ojo/genética , Humanos , Mutación , Linaje , Distrofias Retinianas/genética , Retinitis Pigmentosa/genética , Análisis de Secuencia de ADN
4.
Adv Exp Med Biol ; 1185: 275-279, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31884624

RESUMEN

With 329 genes known to be involved in inherited retinal degenerations (IRDs), focus has shifted to generic targets for therapeutics, targets that could provide benefit irrespective of the underlying genetic condition. As one of the most energy-demanding tissues, the retina is acutely sensitive to dysfunction of its energy metabolism. Recent discoveries have shed light on the complex interconnectivity and interdependence of retinal cells on their choice metabolic pathways, highlighting a number of potential targets that could benefit cells in a mutation-independent manner. Some of the latest research on retinal metabolism and mitophagy in photoreceptors and retinal pigment epithelium is discussed, as is how these insights could potentially be used in the design of new therapies.


Asunto(s)
Metabolismo Energético , Células Fotorreceptoras de Vertebrados/fisiología , Retina/fisiología , Degeneración Retiniana , Epitelio Pigmentado de la Retina/fisiología , Humanos , Mitofagia
5.
Trends Genet ; 29(8): 488-97, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23756086

RESUMEN

It has become evident that many human disorders are characterised by mitochondrial dysfunction either at a primary level, due to mutations in genes whose encoded products are involved in oxidative phosphorylation, or at a secondary level, due to the accumulation of mitochondrial DNA (mtDNA) mutations. This has prompted keen interest in the development of cell and animal models and in exploring innovative therapeutic strategies to modulate the mitochondrial deficiencies observed in these diseases. Key advances in these areas are outlined in this review, with a focus on Leber hereditary optic neuropathy (LHON). This exciting field is set to grow exponentially and yield many candidate therapies to treat this class of disease.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/metabolismo , Atrofia Óptica Hereditaria de Leber/genética , Animales , Modelos Animales de Enfermedad , Terapia Genética , Humanos , Mitocondrias/genética , Mutación
6.
Vis Neurosci ; 31(4-5): 289-307, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24949856

RESUMEN

Significant advances have been made over the last decade or two in the elucidation of the molecular pathogenesis of inherited ocular disorders. In particular, remarkable successes have been achieved in exploration of gene-based medicines for these conditions, both in preclinical and in clinical studies. Progress in the development of gene therapies targeted toward correcting the primary genetic defect or focused on modulating secondary effects associated with retinal pathologies are discussed in the review. Likewise, the recent utilization of genes encoding light-sensing molecules to provide new functions to residual retinal cells in the degenerating retina is discussed. While a great deal has been learned over the last two decades, the next decade should result in an increasing number of preclinical studies progressing to human clinical trial, an exciting prospect for patients, those active in research and development and bystanders alike.


Asunto(s)
Terapia Genética , Enfermedades de la Retina/genética , Enfermedades de la Retina/terapia , Animales , Humanos
7.
Pharmaceutics ; 15(2)2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36839646

RESUMEN

AAV gene therapy for ocular disease has become a reality with the market authorisation of LuxturnaTM for RPE65-linked inherited retinal degenerations and many AAV gene therapies currently undergoing phase III clinical trials. Many ocular disorders have a mitochondrial involvement from primary mitochondrial disorders such as Leber hereditary optic neuropathy (LHON), predominantly due to mutations in genes encoding subunits of complex I, to Mendelian and multifactorial ocular conditions such as dominant optic atrophy, glaucoma and age-related macular degeneration. In this study, we have optimised the nuclear yeast gene, NADH-quinone oxidoreductase (NDI1), which encodes a single subunit complex I equivalent, creating a candidate gene therapy to improve mitochondrial function, independent of the genetic mutation driving disease. Optimisation of NDI1 (ophNdi1) substantially increased expression in vivo, protected RGCs and increased visual function, as assessed by optokinetic and photonegative response, in a rotenone-induced murine model. In addition, ophNdi1 increased cellular oxidative phosphorylation and ATP production and protected cells from rotenone insult to a significantly greater extent than wild type NDI1. Significantly, ophNdi1 treatment of complex I deficient patient-derived fibroblasts increased oxygen consumption and ATP production rates, demonstrating the potential of ophNdi1 as a candidate therapy for ocular disorders where mitochondrial deficits comprise an important feature.

8.
Mol Ther ; 19(4): 642-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21224835

RESUMEN

For dominantly inherited disorders development of gene therapies, targeting the primary genetic lesion has been impeded by mutational heterogeneity. An example is rhodopsin-linked autosomal dominant retinitis pigmentosa with over 150 mutations in the rhodopsin gene. Validation of a mutation-independent suppression and replacement gene therapy for this disorder has been undertaken. The therapy provides a means of correcting the genetic defect in a mutation-independent manner thereby circumventing the mutational diversity. Separate adeno-associated virus (AAV) vectors were used to deliver an RNA interference (RNAi)-based rhodopsin suppressor and a codon-modified rhodopsin replacement gene resistant to suppression due to nucleotide alterations at degenerate positions over the RNAi target site. Viruses were subretinally coinjected into P347S mice, a model of dominant rhodopsin-linked retinitis pigmentosa. Benefit in retinal function and structure detected by electroretinography (ERG) and histology, respectively, was observed for at least 5 months. Notably, the photoreceptor cell layer, absent in 5-month-old untreated retinas, contained 3-4 layers of nuclei, whereas photoreceptor ultrastructure, assessed by transmission electron microscopy (TEM) improved significantly. The study provides compelling evidence that codelivered suppression and replacement is beneficial, representing a significant step toward the clinic. Additionally, dual-vector delivery of combined therapeutics represents an exciting approach, which is potentially applicable to other inherited disorders.


Asunto(s)
Terapia Genética/métodos , Retinitis Pigmentosa/terapia , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
9.
Mol Ther Methods Clin Dev ; 25: 236-249, 2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35474956

RESUMEN

Recombinant adeno-associated virus (AAV) vectors are one of the main gene delivery vehicles used in retinal gene therapy approaches; however, there is a need to further improve the efficacy, tropism, and safety of these vectors. In this study, using a CMV-EGFP expression cassette, we characterize the retinal utility of AAV-PHP.eB, a serotype recently developed by in vivo directed evolution, which can cross the blood-brain barrier and target neurons with high efficacy in mice. Systemic and intravitreal delivery of AAV-PHP.eB resulted in the high transduction efficacy of retinal ganglion and horizontal cells, with systemic delivery providing pan-retinal coverage of the mouse retina. Subretinal delivery transduced photoreceptors and retinal pigment epithelium cells robustly. EGFP expression (number of transduced cells and mRNA levels) were similar when the retinas were transduced systemically or intravitreally with AAV-PHP.eB or intravitreally with AAV2/2. Notably, in photoreceptors, EGFP fluorescence intensities and mRNA levels were 50-70 times higher, when subretinal injections with AAV-PHP.eB were compared to AAV2/8. Our results demonstrate the pan-retinal transduction of ganglion cells and extremely efficient transduction of photoreceptor and retinal pigment epithelium cells as the most valuable features of AAV-PHP.eB in the mouse retina.

10.
Exp Eye Res ; 91(4): 500-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20637750

RESUMEN

Retinal degeneration (RD) results from photoreceptor apoptosis. Cell transplantation, one potential therapeutic approach, requires expandable stem cells that can form mature photoreceptors when differentiated. Freshly dissociated primary retinal cells from postnatal day 2-6 (PN2-6) mouse retina can give rise, post-transplantation, to photoreceptors in adult recipients. Unfortunately, incorporation rates are low; moreover, photoreceptor potential is lost if the same PN2-6 cells are cultured prior to transplantation. We investigated the identity of the cells forming photoreceptors post-transplantation, using FACS sorted primary postnatal day (PN) 3-5 Rho-eGFP retinal cells. Higher integration rates were achieved for cells that were expressing Rho-eGFP at PN3-5, indicating that post-mitotic photoreceptor precursors already expressing rhodopsin form the majority of integrating rods. We then investigated improvement of cell culture protocols for retinal progenitor cells (RPCs) derived from PN3-5 retinal cells in vitro. We succeeded in improving RPC survival and growth rates 25-fold, by modifying retinal dissociation, replacing N2 supplement with B27 supplement minus retinoic acid (B27-RA) and coating flasks with fibronectin. However, levels of rhodopsin and similar photoreceptor-specific markers still diminished rapidly during growth in vitro, and did not re-appear after in vitro differentiation. Similarly, transplanted RPCs, whether proliferating or differentiated, did not form photoreceptors in vivo. Cultured RPCs upregulate genes such as Sox2 and nestin, markers of more primitive neural stem cells. Use of these cells for RD treatment will require identification of triggers that favour terminal photoreceptor differentiation and survival in vitro prior to transplantation.


Asunto(s)
Supervivencia Celular , Células Fotorreceptoras de Vertebrados/patología , Retina/patología , Trasplante de Células Madre , Células Madre/patología , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodopsina/genética , Rodopsina/metabolismo
11.
Mol Ther ; 17(4): 593-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19174761

RESUMEN

Mutational heterogeneity represents one of the greatest barriers impeding the progress toward the clinic of gene therapies for many dominantly inherited disorders. A general strategy of gene suppression in conjunction with replacement has been proposed to overcome this mutational heterogeneity. In the current study, various aspects of this strategy are explored for a dominant form of the retinal degeneration, retinitis pigmentosa (RP), caused by mutations in the rhodopsin gene (RHO-adRP). While > 200 mutations have been identified in rhodopsin (RHO), in principle, suppression and replacement may be employed to provide a single mutation-independent therapeutic for this form of the disorder. In the study we demonstrate in a transgenic mouse simulating human RHO-adRP that RNA interference-based suppression, together with gene replacement utilizing the endogenous mouse gene as the replacement, provides significant benefit as evaluated by electroretinography (ERG). Moreover, this is mirrored histologically by preservation of photoreceptors. AAV-based vectors were utilized for in vivo delivery of the therapy to the target cell type, the photoreceptors. The results demonstrate that RNAi-based mutation-independent suppression and replacement can provide benefit for RHO-adRP and promote the therapeutic approach as potentially beneficial for other autosomal dominantly inherited disorders.


Asunto(s)
Dependovirus/genética , Genes Dominantes , Terapia Genética , Retina/fisiopatología , Retinitis Pigmentosa/terapia , Animales , Citometría de Flujo , Ratones , Interferencia de ARN , Retinitis Pigmentosa/fisiopatología
12.
Front Neurosci ; 14: 891, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973439

RESUMEN

With marketing approval of the first ocular gene therapy, and other gene therapies in clinical trial, treatments for inherited retinal degenerations (IRDs) have become a reality. Biallelic mutations in the tubby like protein 1 gene (TULP1) are causative of IRDs in humans; a mouse knock-out model (Tulp1-/-) is characterized by a similar disease phenotype. We developed a Tulp1 supplementation therapy for Tulp1-/- mice. Utilizing subretinal AAV2/5 delivery at postnatal day (p)2-3 and rhodopsin-kinase promoter (GRK1P) we targeted Tulp1 to photoreceptor cells exploring three doses, 2.2E9, 3.7E8, and 1.2E8 vgs. Tulp1 mRNA and TULP1 protein were assessed by RT-qPCR, western blot and immunocytochemistry, and visual function by electroretinography. Our results indicate that TULP1 was expressed in photoreceptors; achieved levels of Tulp1 mRNA and protein were similar to wild type levels at p20. However, the thickness of the outer nuclear layer (ONL) did not improve in treated Tulp1-/- mice. There was a small and transient electroretinography benefit in the treated retinas at 4 weeks of age (not observed by 6 weeks) when using 3.7E8 vg dose. Dark-adapted mixed rod and cone a- and b-wave amplitudes were 24.3 ± 13.5 µV and 52.2 ± 31.7 µV in treated Tulp1-/- mice, which were significantly different (p < 0.001, t-test), from those detected in untreated eyes (7.1 ± 7.0 µV and 9.4 ± 15.1 µV, respectively). Our results indicate that Tulp1 supplementation in photoreceptors may not be sufficient to provide robust benefit in Tulp1-/- mice. As such, further studies are required to fine tune the Tulp1 supplementation therapy, which, in principle, should rescue the Tulp1-/- phenotype.

13.
Sci Rep ; 10(1): 16515, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-33020509

RESUMEN

Retinal ganglion cells (RGCs) are known to be involved in several ocular disorders, including glaucoma and Leber hereditary optic neuropathy (LHON), and hence represent target cells for gene therapies directed towards these diseases. Restricting gene therapeutics to the target cell type in many situations may be preferable compared to ubiquitous transgene expression, stimulating researchers to identify RGC-specific promoters, particularly promoter sequences that may also be appropriate in size to fit readily into recombinant adeno associated viral (AAV) vectors, the vector of choice for many ocular gene therapies. In the current study we analysed EGFP expression driven by various sequences of the putative human NEFH promoter in order to define sequences required for preferential expression in RGCs. EGFP expression profiles from four different potential NEFH promoter constructs were compared in vivo in mice using retinal histology and mRNA expression analysis. Notably, two efficient promoter sequences, one comprising just 199 bp, are presented in the study.


Asunto(s)
Proteínas de Neurofilamentos/genética , Regiones Promotoras Genéticas/genética , Células Ganglionares de la Retina/metabolismo , Animales , Emparejamiento Base , Dependovirus/genética , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Terapia Genética , Vectores Genéticos , Glaucoma/patología , Humanos , Ratones , Ratones de la Cepa 129 , Proteínas de Neurofilamentos/metabolismo , Atrofia Óptica Hereditaria de Leber/patología , Retina/patología , Células Ganglionares de la Retina/fisiología , Transgenes
14.
Front Neurosci ; 14: 571479, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324145

RESUMEN

Optic Atrophy 1 (OPA1) is a mitochondrially targeted GTPase that plays a pivotal role in mitochondrial health, with mutations causing severe mitochondrial dysfunction and typically associated with Dominant Optic Atrophy (DOA), a progressive blinding disease involving retinal ganglion cell loss and optic nerve damage. In the current study, we investigate the use of codon-optimized versions of OPA1 isoform 1 and 7 as potential therapeutic interventions in a range of in vitro and in vivo models of mitochondrial dysfunction. We demonstrate that both isoforms perform equally well in ameliorating mitochondrial dysfunction in OPA1 knockout mouse embryonic fibroblast cells but that OPA1 expression levels require tight regulation for optimal benefit. Of note, we demonstrate for the first time that both OPA1 isoform 1 and 7 can be used independently to protect spatial visual function in a murine model of retinal ganglion cell degeneration caused by mitochondrial dysfunction, as well as providing benefit to mitochondrial bioenergetics in DOA patient derived fibroblast cells. These results highlight the potential value of OPA1-based gene therapy interventions.

15.
Vision Res ; 48(3): 386-91, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17920651

RESUMEN

Mutational heterogeneity in genes causative of dominantly inherited disorders represents a significant barrier for development of therapies directed towards correction of the primary genetic defect. To circumvent the mutational heterogeneity present in rhodopsin- (RHO-) linked autosomal dominant Retinitis Pigmentosa (adRP), a strategy involving suppression and replacement of RHO has been adopted. RNA interference- (RNAi-) mediated suppression of RHO has been explored as has the generation of an RNAi-resistant replacement gene using the degeneracy of the genetic code. Additionally, the functional equivalence of codon-modified replacement genes has been demonstrated in a transgenic animal (RHO-M). Suppression and replacement, while exemplified by adRP, may also be relevant to many other dominantly inherited diseases with the hallmark of mutational heterogeneity.


Asunto(s)
Modelos Animales de Enfermedad , Terapia Genética/métodos , Retinitis Pigmentosa/terapia , Rodopsina/genética , Animales , Células Cultivadas , Electrorretinografía , Expresión Génica , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Rodopsina/biosíntesis
16.
Front Neurosci ; 11: 521, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28983234

RESUMEN

Significant advances in gene therapy have enabled exploration of therapies for inherited retinal disorders, many of which are in preclinical development or clinical evaluation. Gene therapy for retinal conditions has led the way in this growing field. The loss of retinal ganglion cells (RGCs) is a hallmark of a number of retinal disorders. As the field matures innovations that aid in refining therapies and optimizing efficacy are in demand. Gene therapies under development for RGC-related disorders, when delivered with recombinant adeno associated vectors (AAV), have typically been expressed from ubiquitous promoter sequences. Here we describe how a novel promoter from the murine Nefh gene was selected to drive transgene expression in RGCs. The Nefh promoter, in an AAV2/2 vector, was shown to drive preferential EGFP expression in murine RGCs in vivo following intravitreal injection. In contrast, EGFP expression from a CMV promoter was observed not only in RGCs, but throughout the inner nuclear layer and in amacrine cells located within the ganglion cell layer (GCL). Of note, the Nefh promoter sequence is sufficiently compact to be readily accommodated in AAV vectors, where transgene size represents a significant constraint. Moreover, this promoter should in principle provide a more targeted and potentially safer alternative for RGC-directed gene therapies.

18.
Hum Mutat ; 27(3): 260-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16419083

RESUMEN

Extensive mutational heterogeneity presents a significant barrier to the development of therapeutics for RDS-peripherin-linked autosomal-dominant retinitis pigmentosa (RP), for which more than 50 disease-related mutations have been identified to date. Mutation-independent suppression, using RNA interference (RNAi), together with simultaneous expression of a replacement rds gene (r-rds, which has been altered to escape suppression but nevertheless encodes wild-type protein) has been explored in COS-7 cells and mouse retinal explants. The efficacy of small interfering and short hairpin RNAs (si/shRNAs) silencing mouse rds, and the function of r-rds (containing degenerate substitutions in the RNAi target sequence) were analyzed at transcript (RT-PCR) and protein (ELISA) levels in COS-7 cells. "Dual-" and "triple-expression" constructs carrying the shRNA suppressor and the marker EGFP with or without the r-rds cassette were electroporated in vitro into retinal explants from 1-day-old pups. The retinae were dissociated at day 14, and transduced cells were FACS-sorted using the coexpressed EGFP marker and analyzed by RT-PCR. si/shRNAs decreased rds mRNA and protein expression by up to 82%, while r-rds was protected from suppression in COS-7 cells. Similarly, efficient RNAi-mediated suppression of endogenous rds was detected in retinal explants, while concomitant rescue of r-rds was also achieved. These data validate the concept of RNAi-based suppression coupled with replacement technology for the development of therapies targeting RDS-linked autosomal-dominant RP, and suggest that such approaches could potentially be used for other autosomal-dominant diseases with similarly extensive intragenic heterogeneity.


Asunto(s)
Análisis Mutacional de ADN/métodos , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/fisiología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Interferencia de ARN , Retina/metabolismo , Retinitis Pigmentosa/genética , Animales , Células COS , Separación Celular , Chlorocebus aethiops , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Silenciador del Gen , Ratones , Periferinas , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Trends Mol Med ; 11(6): 299-305, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15949772

RESUMEN

Osteogenesis imperfecta (OI) is an incurable genetic brittle-bone disease. Although drug therapy, surgery and physiotherapy represent current treatments for OI, the search is ongoing for effective and innovative new therapies targeting the underlying causes of the disease. In this regard, recent advances in the fields of gene and stem-cell therapies have been considerable. In spite of the many challenges that remain, potential new therapies for OI, which have been tested in cell culture systems, animal models and patients, offer hope for the future development of successful therapies. Recent progress in the field is reviewed here.


Asunto(s)
Osteogénesis Imperfecta/terapia , Trasplante de Células Madre/métodos , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Modelos Genéticos , Mutación , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA