Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Ther ; 31(12): 3441-3456, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37814449

RESUMEN

Adeno-associated virus (AAV) continues to be the gold standard vector for therapeutic gene delivery and has proven especially useful for treating ocular disease. Intravitreal injection (IVtI) is a promising delivery route because it increases accessibility of gene therapies to larger patient populations. However, data from clinical and non-human primate (NHP) studies utilizing currently available capsids indicate that anatomical barriers to AAV and pre-existing neutralizing antibodies can restrict gene expression to levels that are "sub-therapeutic" in a substantial proportion of patients. Here, we performed a combination of directed evolution in NHPs of an AAV2-based capsid library with simultaneous mutations across six surface-exposed variable regions and rational design to identify novel capsid variants with improved retinal transduction following IVtI. Following two rounds of screening in NHP, enriched variants were characterized in intravitreally injected mice and NHPs and shown to have increased transduction relative to AAV2. Lead capsid variant, P2-V1, demonstrated an increased ability to evade neutralizing antibodies in human vitreous samples relative to AAV2 and AAV2.7m8. Taken together, this study further contributed to our understanding of the selective pressures associated with retinal transduction via the vitreous and identified promising novel AAV capsid variants for clinical consideration.


Asunto(s)
Anticuerpos Neutralizantes , Cápside , Humanos , Ratones , Animales , Dependovirus , Inyecciones Intravítreas , Transducción Genética , Primates/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Vectores Genéticos/genética
2.
J Virol ; 95(10)2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33658343

RESUMEN

Adeno-associated viruses (AAVs) have recently emerged as the leading vector for retinal gene therapy. However, AAV vectors which are capable of achieving clinically relevant levels of transgene expression and widespread retinal transduction are still an unmet need. Using rationally designed AAV2-based capsid variants, we investigate the role of capsid hydrophilicity and hydrophobicity as it relates to retinal transduction. We show that hydrophilic, single amino acid (aa) mutations (V387R, W502H, E530K, L583R) in AAV2 negatively impact retinal transduction when heparan sulfate proteoglycan (HSPG) binding remains intact. Conversely, addition of hydrophobic point mutations to an HSPG binding deficient capsid (AAV2ΔHS) lead to increased retinal transduction in both mouse and macaque. Our top performing vector, AAV2(4pMut)ΔHS, achieved robust rod and cone photoreceptor (PR) transduction in macaque, especially in the fovea, and demonstrates the ability to spread laterally beyond the borders of the subretinal injection (SRI) bleb. This study both evaluates biophysical properties of AAV capsids that influence retinal transduction, and assesses the transduction and tropism of a novel capsid variant in a clinically relevant animal model.ImportanceRationally guided engineering of AAV capsids aims to create new generations of vectors with enhanced potential for human gene therapy. By applying rational design principles to AAV2-based capsids, we evaluated the influence of hydrophilic and hydrophobic amino acid (aa) mutations on retinal transduction as it relates to vector administration route. Through this approach we identified a largely deleterious relationship between hydrophilic aa mutations and canonical HSPG binding by AAV2-based capsids. Conversely, the inclusion of hydrophobic aa substitutions on a HSPG binding deficient capsid (AAV2ΔHS), generated a vector capable of robust rod and cone photoreceptor (PR) transduction. This vector AAV2(4pMut)ΔHS also demonstrates a remarkable ability to spread laterally beyond the initial subretinal injection (SRI) bleb, making it an ideal candidate for the treatment of retinal diseases which require a large area of transduction.

3.
Mol Ther ; 25(8): 1866-1880, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28566226

RESUMEN

X-linked retinitis pigmentosa (XLRP) caused by mutations in the RPGR gene is an early onset and severe cause of blindness. Successful proof-of-concept studies in a canine model have recently shown that development of a corrective gene therapy for RPGR-XLRP may now be an attainable goal. In preparation for a future clinical trial, we have here optimized the therapeutic AAV vector construct by showing that GRK1 (rather than IRBP) is a more efficient promoter for targeting gene expression to both rods and cones in non-human primates. Two transgenes were used in RPGR mutant (XLPRA2) dogs under the control of the GRK1 promoter. First was the previously developed stabilized human RPGR (hRPGRstb). Second was a new full-length stabilized and codon-optimized human RPGR (hRPGRco). Long-term (>2 years) studies with an AAV2/5 vector carrying hRPGRstb under control of the GRK1 promoter showed rescue of rods and cones from degeneration and retention of vision. Shorter term (3 months) studies demonstrated comparable preservation of photoreceptors in canine eyes treated with an AAV2/5 vector carrying either transgene under the control of the GRK1 promoter. These results provide the critical molecular components (GRK1 promoter, hRPGRco transgene) to now construct a therapeutic viral vector optimized for RPGR-XLRP patients.


Asunto(s)
Proteínas Portadoras/genética , Proteínas del Ojo/genética , Genes Ligados a X , Terapia Genética , Mutación , Retina/metabolismo , Retinitis Pigmentosa/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Quinasa 1 del Receptor Acoplado a Proteína-G/genética , Expresión Génica , Orden Génico , Genes Reporteros , Vectores Genéticos/genética , Humanos , Fenotipo , Células Fotorreceptoras de Vertebrados/metabolismo , Primates , Regiones Promotoras Genéticas , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/terapia , Transducción Genética , Transgenes , Pruebas de Visión
4.
Mol Ther Methods Clin Dev ; 28: 129-145, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36654798

RESUMEN

Mutations in GUCY2D are associated with severe early-onset retinal dystrophy, Leber congenital amaurosis type 1 (LCA1), a leading cause of blindness in children. Despite a high degree of visual disturbance stemming from photoreceptor dysfunction, patients with LCA1 largely retain normal photoreceptor structure, suggesting that they are good candidates for gene replacement therapy. The purpose of this study was to conduct the preclinical and IND-enabling experiments required to support clinical application of AAV5-hGRK1-GUCY2D in patients harboring biallelic recessive mutations in GUCY2D. Preclinical studies were conducted in mice to evaluate the effect of vector manufacturing platforms and transgene species on the therapeutic response. Dose-ranging studies were conducted in cynomolgus monkeys to establish the minimum dose required for efficient photoreceptor transduction. Good laboratory practice (GLP) studies evaluated systemic biodistribution in rats and toxicology in non-human primates (NHPs). These results expanded our knowledge of dose response for an AAV5-vectored transgene under control of the human rhodopsin kinase (hGRK1) promoter in NHPs with respect to photoreceptor transduction and safety and, in combination with the rat biodistribution and mouse efficacy studies, informed the design of a first-in-human clinical study in patients with LCA1.

5.
Front Neurosci ; 13: 1255, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31824252

RESUMEN

Usher's syndrome is the most common combined blindness-deafness disorder with USH1B, caused by mutations in MYO7A, resulting in the most severe phenotype. The existence of numerous, naturally occurring shaker1 mice harboring variable MYO7A mutations on different genetic backgrounds has complicated the characterization of MYO7A knockout (KO) and heterozygote mice. We generated a novel MYO7A KO mouse (Myo7a - / -) that is easily genotyped, maintained, and confirmed to be null for MYO7A in both the eye and inner ear. Like USH1B patients, Myo7a - / - mice are profoundly deaf, and display near complete loss of inner and outer cochlear hair cells (HCs). No gross structural changes were observed in vestibular HCs. Myo7a - / - mice exhibited modest declines in retinal function but, unlike patients, no loss of retinal structure. We attribute the latter to differential expression of MYO7A in mouse vs. primate retina. Interestingly, heterozygous Myo7a + / - mice had reduced numbers of cochlear HCs and concomitant reductions in auditory function relative to Myo7a +/+ controls. Notably, this is the first report that loss of a single Myo7a allele significantly alters auditory structure and function and suggests that audiological characterization of USH1B carriers is warranted. Maintenance of vestibular HCs in Myo7a - / - mice suggests that gene replacement could be used to correct the vestibular dysfunction in USH1B patients. While Myo7a - / - mice do not exhibit sufficiently robust retinal phenotypes to be used as a therapeutic outcome measure, they can be used to assess expression of vectored MYO7A on a null background and generate valuable pre-clinical data toward the treatment of USH1B.

6.
Hum Gene Ther ; 30(5): 571-589, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30358434

RESUMEN

Mutations in GUCY2D, the gene encoding retinal guanylate cyclase-1 (retGC1), are the leading cause of autosomal dominant cone-rod dystrophy (CORD6). Significant progress toward clinical application of gene replacement therapy for Leber congenital amaurosis (LCA) due to recessive mutations in GUCY2D (LCA1) has been made, but a different approach is needed to treat CORD6 where gain of function mutations cause dysfunction and dystrophy. The CRISPR/Cas9 gene editing system efficiently disrupts genes at desired loci, enabling complete gene knockout or homology directed repair. Here, adeno-associated virus (AAV)-delivered CRISPR/Cas9 was used specifically to edit/disrupt this gene's early coding sequence in mouse and macaque photoreceptors in vivo, thereby knocking out retGC1 expression and demonstrably altering retinal function and structure. Neither preexisting nor induced Cas9-specific T-cell responses resulted in ocular inflammation in macaques, nor did it limit GUCY2D editing. The results show, for the first time, the ability to perform somatic gene editing in primates using AAV-CRISPR/Cas9 and demonstrate the viability this approach for treating inherited retinal diseases in general and CORD6 in particular.


Asunto(s)
Sistemas CRISPR-Cas , Dependovirus/genética , Edición Génica , Guanilato Ciclasa/genética , Receptores de Superficie Celular/genética , Retina/metabolismo , Animales , Secuencia de Bases , Electrorretinografía , Genes Reporteros , Vectores Genéticos/genética , Guanilato Ciclasa/metabolismo , Macaca , Ratones , Ratones Noqueados , Imagen Molecular/métodos , Regiones Promotoras Genéticas , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/genética , Receptores de Superficie Celular/metabolismo , Retina/patología
7.
Hum Gene Ther ; 27(8): 580-97, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27439313

RESUMEN

Adeno-associated virus (AAV) has emerged as the preferred vector for targeting gene expression to the retina. Subretinally injected AAV can efficiently transduce retinal pigment epithelium and photoreceptors in primate retina. Inner and middle primate retina can be transduced by intravitreally delivered AAV, but with low efficiency. This is due to dilution of vector, potential neutralization of capsid because it is not confined to the immune-privileged retinal compartment, and the presence of the inner limiting membrane (ILM), a barrier separating the vitreous from the neural retina. We here describe a novel "subILM" injection method that addresses all three issues. Specifically, vector is placed in a surgically induced, hydrodissected space between the ILM and neural retina. In an initial experiment, we injected viscoelastic (Healon(®)), a substance we confirmed was biocompatible with AAV, to create a subILM bleb and subsequently injected AAV2-GFP into the bleb after irrigation with basic salt solution. For later experiments, we used a Healon-AAV mixture to place single, subILM injections. In all cases, subILM delivery of AAV was well tolerated-no inflammation or gross structural changes were observed by ophthalmological examination or optical coherence tomography. In-life fluorescence imaging revealed profound transgene expression within the area of the subILM injection bleb that persisted for the study duration. Uniform and extensive transduction of retinal ganglion cells (RGCs) was achieved in the areas beneath the subILM bleb. Transduction of Müller glia, ON bipolar cells, and photoreceptors was also observed. Robust central labeling from green fluorescent protein-expressing RGCs confirmed their continued survival, and was observed in the lateral geniculate nucleus, the superior colliculus, and the pretectum. Our results confirm that the ILM is a major barrier to transduction by AAV in primate retina and that, when it is circumvented, the efficiency and depth to which AAV2 promotes transduction of multiple retinal cell classes are greatly enhanced.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/administración & dosificación , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Transgenes/genética , Animales , Proteínas Fluorescentes Verdes/metabolismo , Macaca nemestrina , Masculino , Retina/patología , Células Ganglionares de la Retina/patología , Transducción Genética
8.
Hum Gene Ther ; 27(1): 72-82, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26603570

RESUMEN

Adeno-associated viral (AAV) vectors containing cone-specific promoters have rescued cone photoreceptor function in mouse and dog models of achromatopsia, but cone-specific promoters have not been optimized for use in primates. Using AAV vectors administered by subretinal injection, we evaluated a series of promoters based on the human L-opsin promoter, or a chimeric human cone transducin promoter, for their ability to drive gene expression of green fluorescent protein (GFP) in mice and nonhuman primates. Each of these promoters directed high-level GFP expression in mouse photoreceptors. In primates, subretinal injection of an AAV-GFP vector containing a 1.7-kb L-opsin promoter (PR1.7) achieved strong and specific GFP expression in all cone photoreceptors and was more efficient than a vector containing the 2.1-kb L-opsin promoter that was used in AAV vectors that rescued cone function in mouse and dog models of achromatopsia. A chimeric cone transducin promoter that directed strong GFP expression in mouse and dog cone photoreceptors was unable to drive GFP expression in primate cones. An AAV vector expressing a human CNGB3 gene driven by the PR1.7 promoter rescued cone function in the mouse model of achromatopsia. These results have informed the design of an AAV vector for treatment of patients with achromatopsia.


Asunto(s)
Defectos de la Visión Cromática/genética , Técnicas de Transferencia de Gen , Terapia Genética , Células Fotorreceptoras Retinianas Conos/metabolismo , Enfermedades de la Retina/genética , Animales , Defectos de la Visión Cromática/terapia , Dependovirus/genética , Perros , Expresión Génica , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Células Fotorreceptoras Retinianas Conos/patología , Enfermedades de la Retina/terapia , Opsinas de Bastones/genética
9.
Invest Ophthalmol Vis Sci ; 46(11): 3988-98, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16249472

RESUMEN

PURPOSE: Light-driven protein translocation is responsible for the dramatic redistribution of some proteins in vertebrate rod photoreceptors. In this study, the involvement of microtubules and microfilaments in the light-driven translocation of arrestin and transducin was investigated. METHODS: Pharmacologic reagents were applied to native and transgenic Xenopus tadpoles, to disrupt the microtubules (thiabendazole) and microfilaments (cytochalasin D and latrunculin B) of the rod photoreceptors. Quantitative confocal imaging was used to assess the impact of these treatments on arrestin and transducin translocation. A series of transgenic tadpoles expressing arrestin truncations were also created to identify portions of arrestin that enable arrestin to translocate. RESULTS: Application of cytochalasin D or latrunculin B to disrupt the microfilament organization selectively slowed only transducin movement from the inner to the outer segments. Perturbation of the microtubule cytoskeleton with thiabendazole slowed the translocation of both arrestin and transducin, but only in moving from the outer to the inner segments. Transgenic Xenopus expressing fusions of green fluorescent protein (GFP) with portions of arrestin implicates the C terminus of arrestin as an important portion of the molecule for promoting translocation. This C-terminal region can be used independently to promote translocation of GFP in response to light. CONCLUSIONS: The results show that disruption of the cytoskeletal network in rod photoreceptors has specific effects on the translocation of arrestin and transducin. These effects suggest that the light-driven translocation of visual proteins at least partially relies on an active motor-driven mechanism for complete movement of arrestin and transducin.


Asunto(s)
Citoesqueleto de Actina/fisiología , Arrestina/metabolismo , Luz , Microtúbulos/fisiología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Transducina/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Compuestos Bicíclicos Heterocíclicos con Puentes/toxicidad , Citocalasina D/toxicidad , Proteínas Fluorescentes Verdes/metabolismo , Microscopía Confocal , Microtúbulos/efectos de los fármacos , Transporte de Proteínas/efectos de la radiación , Proteínas Recombinantes de Fusión/metabolismo , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Células Fotorreceptoras Retinianas Bastones/efectos de la radiación , Tiabendazol/toxicidad , Tiazoles/toxicidad , Tiazolidinas , Xenopus laevis
10.
Clin Cancer Res ; 9(10 Pt 2): 3865S-72S, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14506184

RESUMEN

PURPOSE: Immunoglobulins are catabolized in the hepatocytes, primarily by cathepsins. The liver becomes the likely dose-limiting tissue for radiometals, like (90)Y, in radioimmunoconjugates (RICs) used for radioimmunotherapy in combination with bone marrow support. To assess whether in vitro cathepsin-degradable peptide linkers between the chelated radiometal and the antibody decreased hepatic radiation dose, cumulated activity was used as a surrogate for radiation dose. EXPERIMENTAL DESIGN: Four different cathepsin-degradable peptides used to link 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid-chelated (111)In to two different monoclonal antibodies were studied in athymic mouse models of human breast cancer or lymphoma. Measured concentrations of activity during 5 days were used to reflect pharmacokinetic behavior for normal tissues and tumor. With the use of linear regression to fit a monoexponential decay function, cumulated activities in the liver and xenografts were calculated. RESULTS: The pharmacokinetic behavior of the cathepsin-degradable peptide-linked RICs was similar to that for the 2-iminothiolane (2IT) nondegradable linked RICs except for the liver. The liver cumulated activities of peptide-linked RICs were significantly decreased from those of the corresponding 2IT-linked RICs, varying between reductions of 59 and 68%. Cumulated activities of peptide-linked RICs in the xenografts were as great as those of 2IT RICs, so that the therapeutic indices (tumor: liver cumulated activity ratios) were substantially better for cathepsin-degradable peptide-linked RICs. CONCLUSIONS: Cathepsin-degradable peptides used to link chelated radiometals to antibodies reduce liver radiation dose and improve the therapeutic index for radioimmunotherapy given in combination with bone marrow support.


Asunto(s)
Catepsinas/química , Inmunoconjugados/química , Hígado/efectos de la radiación , Animales , Anticuerpos Monoclonales/química , Neoplasias de la Mama/patología , Quelantes/farmacología , Cromatografía Líquida de Alta Presión , Compuestos Heterocíclicos con 1 Anillo/farmacología , Humanos , Radioisótopos de Indio/farmacocinética , Modelos Lineales , Hígado/efectos de los fármacos , Hígado/metabolismo , Linfoma/patología , Ratones , Ratones Desnudos , Modelos Químicos , Trasplante de Neoplasias , Péptidos/química , Radioinmunoterapia/métodos , Factores de Tiempo
11.
Hum Gene Ther ; 26(11): 767-76, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26359319

RESUMEN

The ability to effectively deliver genetic material to vascular endothelial cells remains one of the greatest unmet challenges facing the development of gene therapies to prevent diseases with underlying vascular etiology, such as diabetes, atherosclerosis, and age-related macular degeneration. Herein, we assess the effectiveness of an rAAV2-based capsid mutant vector (Y272F, Y444F, Y500F, Y730F, T491V; termed QuadYF+TV) with strong endothelial cell tropism at transducing the vasculature after systemic administration. Intravenous injection of QuadYF+TV resulted in widespread transduction throughout the vasculature of several major organ systems, as assessed by in vivo bioluminescence imaging and postmortem histology. Robust transduction of lung tissue was observed in QuadYF+TV-injected mice, indicating a role for intravenous gene delivery in the treatment of chronic diseases presenting with pulmonary complications, such as α1-antitrypsin deficiency. The QuadYF+TV vector cross-reacted strongly with AAV2 neutralizing antibodies, however, indicating that a targeted delivery strategy may be required to maximize clinical translatability.


Asunto(s)
Administración Intravenosa , Células Endoteliales/metabolismo , Terapia Genética , Vectores Genéticos/administración & dosificación , Transducción Genética , Enfermedades Vasculares/terapia , Animales , Proteínas de la Cápside/genética , Dependovirus/genética , Células Endoteliales/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación , Enfermedades Vasculares/genética
12.
PLoS One ; 10(6): e0128759, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26052939

RESUMEN

BACKGROUND: Adeno associated virus (AAV) is well known for its ability to deliver transgenes to retina and to mediate improvements in animal models and patients with inherited retinal disease. Although the field is less advanced, there is growing interest in AAV's ability to target cells of the anterior segment. The purpose of our study was to fully articulate a reliable and reproducible method for injecting the anterior chamber (AC) of mice and rats and to investigate the transduction profiles of AAV2- and AAV8-based capsid mutants containing self-complementary (sc) genomes in the anterior segment of the eye. METHODOLOGY/PRINCIPLE FINDINGS: AC injections were performed in C57BL/6 mice and Sprague Dawley rats. The cornea was punctured anterior of the iridocorneal angle. To seal the puncture site and to prevent reflux an air bubble was created in the AC. scAAVs expressing GFP were injected and transduction was evaluated by immunohistochemistry. Both parent serotype and capsid modifications affected expression. scAAV2- based vectors mediated efficient GFP-signal in the corneal endothelium, ciliary non-pigmented epithelium (NPE), iris and chamber angle including trabecular meshwork, with scAAV2(Y444F) and scAAV2(triple) being the most efficient. CONCLUSIONS/SIGNIFICANCE: This is the first study to semi quantitatively evaluate transduction of anterior segment tissues following injection of capsid-mutated AAV vectors. scAAV2- based vectors transduced corneal endothelium, ciliary NPE, iris and trabecular meshwork more effectively than scAAV8-based vectors. Mutagenesis of surface-exposed tyrosine residues greatly enhanced transduction efficiency of scAAV2 in these tissues. The number of Y-F mutations was not directly proportional to transduction efficiency, however, suggesting that proteosomal avoidance alone may not be sufficient. These results are applicable to the development of targeted, gene-based strategies to investigate pathological processes of the anterior segment and may be applied toward the development of gene-based therapies for glaucoma and acquired or inherited corneal anomalies.


Asunto(s)
Cámara Anterior/virología , Cápside/metabolismo , Dependovirus/genética , Mutación/genética , Malla Trabecular/metabolismo , Transducción Genética , Animales , Vectores Genéticos , Proteínas Fluorescentes Verdes , Inyecciones , Ratones Endogámicos C57BL , Ratas Sprague-Dawley
13.
Hum Gene Ther ; 26(9): 575-92, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26247368

RESUMEN

Mutations in GUCY2D are the cause of Leber congenital amaurosis type 1 (LCA1). GUCY2D encodes retinal guanylate cyclase-1 (retGC1), a protein expressed exclusively in outer segments of photoreceptors and essential for timely recovery from photoexcitation. Recent clinical data show that, despite a high degree of visual disturbance stemming from a loss of cone function, LCA1 patients retain normal photoreceptor architecture, except for foveal cone outer segment abnormalities and, in some patients, foveal cone loss. These results point to the cone-rich central retina as a target for GUCY2D replacement. LCA1 gene replacement studies thus far have been conducted in rod-dominant models (mouse) or with vectors and organisms lacking clinical translatability. Here we investigate gene replacement in the Nrl(-/-) Gucy2e(-/-) mouse, an all-cone model deficient in retGC1. We show that AAV-retGC1 treatment fully restores cone function, cone-mediated visual behavior, and guanylate cyclase activity, and preserves cones in treated Nrl(-/-) Gucy2e(-/-) mice over the long-term. A novel finding was that retinal function could be restored to levels above that in Nrl(-/-) controls, contrasting results in other models of retGC1 deficiency. We attribute this to increased cyclase activity in treated Nrl(-/-) Gucy2e(-/-) mice relative to Nrl(-/-) controls. Thus, Nrl(-/-) Gucy2e(-/-) mice possess an expanded dynamic range in ERG response to gene replacement relative to other models. Lastly, we show that a candidate clinical vector, AAV5-GRK1-GUCY2D, when delivered to adult Nrl(-/-) Gucy2e(-/-) mice, restores retinal function that persists for at least 6 months. Our results provide strong support for clinical application of a gene therapy targeted to the cone-rich, central retina of LCA1 patients.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Proteínas del Ojo/genética , Guanilato Ciclasa/genética , Amaurosis Congénita de Leber/terapia , Receptores de Superficie Celular/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Guanilato Ciclasa/metabolismo , Inyecciones Intraoculares , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Opsinas/genética , Opsinas/metabolismo , Receptores de Superficie Celular/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Resultado del Tratamiento , Visión Ocular
14.
Mol Vis ; 10: 857-66, 2004 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-15570221

RESUMEN

PURPOSE: Alternatives to X-ray crystallographic techniques are needed to probe the structure of the hetero-oligomeric lens protein alpha-crystallin. We utilized mass spectrometry for 3 dimensional analysis (MS3D) to study the quaternary structural characteristics of this important lens protein and molecular chaperone. METHODS: We have employed two types of chemical cross-linkers to probe key protein-protein and protein-solvent interactions of alpha-crystallin using MS3D. Native alpha-crystallin was exposed to 3,3'-dithiobis[sulfosuccinimidyl propionate] (DTSSP) and the common fixative, formaldehyde. The reaction products were denatured and enriched in cross-linked and modified species using size exclusion chromatography. Tryptic digests of these fractions were purified using reverse phase HPLC and analyzed by both electrospray and matrix assisted laser desorption mass spectrometry. Comprehensive spectra for each C18 fraction were screened for ions with mass unique to each chemical treatment and candidate sequences matching the experimental data were assigned using MS3D "Links" and "ASAP" software. Selected ions were sequenced by collision induced dissociation. RESULTS: Peptides including residues 164-175 of alphaB-crystallin and residues 1-99 of alphaA-crystallin were modified by formaldehyde and partially hydrolyzed DTSSP. Peptides containing modified lysines 11, 78, and 99 of alphaA-crystallin were sequenced and the modified amino acids identified. In addition, ions corresponding to intramolecular and/or intermolecular cross-links were assigned a sequence based on two criteria. First, the mass values observed were unique to a single cross-linking experiment and were not present in a control where no cross-linker was utilized. Second, two unique ions detected from different cross-linking experiments were correlated in that the structures assigned to the masses were equivalent apart from the structure of the cross-linker. One such correlation was found involving lysine121, within the "highly conserved alpha-crystallin domain" of alphaB-crystallin, cross-linked to either lysine11 or lysine99 of alphaA-crystallin. Another two independent correlations involving lysine72 of alphaB-crystallin were found that indicate cross-linking of two subunits of alphaB-crystallin through this same residue. CONCLUSIONS: Sequences of peptides modified by partially hydrolyzed DTSSP and formaldehyde provide experimental evidence for models of alpha-crystallin quaternary structure that suggest a similar tertiary fold for both alphaA-crystallin and alphaB-crystallin. Analogous to multiple phosphorylations along the N-terminus of alphaB-crystallin, our data indicate that the same region of alphaA-crystallin, up to and including lysine99 is also relatively accessible to modification despite its hydrophobicity. Mass correlation between experiments using different reagents suggests that cross-linking occurred between N-termini of adjacent subunits of alphaB-crystallin in the native complex in support of the amphiphilic, toroidal, or "open micelle" models. In addition, multiple cross-links involving lysine121 of the so called "dimer interface" region within the "highly conserved alpha-crystallin domain" indicate that this region is a site of inter-subunit contacts in the native context.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , Estructura Cuaternaria de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , alfa-Cristalinas/química , Secuencia de Aminoácidos , Animales , Bovinos , Cromatografía Líquida de Alta Presión , Electroforesis en Gel de Poliacrilamida , Formaldehído/farmacología , Cristalino/química , Cristalino/embriología , Datos de Secuencia Molecular , Estructura Molecular , Fragmentos de Péptidos/química , Succinimidas/farmacología , alfa-Cristalinas/efectos de los fármacos , alfa-Cristalinas/aislamiento & purificación
15.
Mol Vis ; 10: 392-8, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15215746

RESUMEN

PURPOSE: The binding of visual arrestin to phosphorylated, activated rhodopsin serves as a model for studying the inactivation process of a large class of G-protein coupled receptor systems. In this study, we combine the use of insertional mutagenesis, fluorescence labeling, and scanning alanine mutagenesis to identify the surface of interaction between arrestin and rhodopsin. METHODS: The ten amino acid myc tag (EQKLISEEDL) was inserted in eleven loop structures that connect betastrands and the tagged arrestins were heterologously expressed in yeast. Binding competition assays were performed with these proteins, using an anti-myc monoclonal antibody. Site specific cysteines were also substituted in selected loop structures in arrestin. These cysteines were labeled with a fluorescent reporter to assess the proximity of the introduced cysteine with rhodopsin in the bound complex. RESULTS: Competitive inhibition of arrestin binding to light activated, phosphorylated rhodopsin with an anti-myc antibody showed that all competitive sites lay along a single surface encompassing the N- and C-terminal domains. Fluorescence labeling of these loop structures and subsequent interaction with rhodopsin indicates close apposition of loops 68-78 and 248-253 to rhodopsin in the receptor bound state. Scanning mutagenesis of loop 248-253 implicates Ser-251 and/or Ser-252 as a potential interaction point with rhodopsin. CONCLUSIONS: Our results clearly suggest a surface of arrestin to which rhodopsin binds upon light activation and phosphorylation. This surface encompasses elements from both the N- and C-terminal domains of arrestin.


Asunto(s)
Arrestina/metabolismo , Rodopsina/metabolismo , Arrestina/química , Unión Competitiva , Mutagénesis Insercional , Fragmentos de Péptidos/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica
17.
Bioresour Technol ; 101(23): 9128-36, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20678927

RESUMEN

Consolidation of bioprocessing steps with lignocellulose is limited by hydrolysate toxicity, the fibrous nature of suspensions, and low activity of cellulase enzymes. Combinations of enzyme dose and treatment conditions improved the flow properties and pumping of acid-pretreated sugarcane bagasse slurries (10% dry weight). Low levels of cellulase enzyme (0.1 and 0.5 FPU/g dry weight acid-pretreated bagasse) were found to reduce viscosities by 77-95% after 6 h, solubilizing 3.5% of the bagasse dry weight. Flow of slurries through small funnels was a useful predictor of success with centrifugal and diaphragm pumps. Equations were derived that describe viscosity and solubilized carbohydrates as a function of time and cellulase dosage. Blending of acid-pretreated bagasse (10% dry weight) with suspensions of acid-pretreated bagasse (10% dry weight) that had been previously digested with cellulase enzymes (low viscosity) did not increase viscosity in a linear fashion. Viscosity of these mixtures remained relatively constant until a threshold level of new fiber was reached, followed by a rapid increase with further additions. Up to 35% fresh acid-pretreated bagasse could be blended with enzyme-digested fiber (5.0 FPU/g dry weight acid-pretreated fiber; 6 h) with only a modest increase in viscosity. The smooth surfaces of enzyme-treated fiber are proposed to hinder the frequency and extent of interactions between fibrils of fresh fiber particles (acid-pretreated) until a threshold concentration is achieved, after which fiber interactions and viscosity increase dramatically. These results were used to model the viscosity in an ideal continuous stirred tank reactor (liquefaction) as a function of residence time and enzyme dosage.


Asunto(s)
Celulasa/metabolismo , Celulosa/metabolismo , Ácidos Fosfóricos/farmacología , Reología/métodos , Saccharum/metabolismo , Reactores Biológicos , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Modelos Biológicos , Polisacáridos/metabolismo , Saccharum/efectos de los fármacos , Solubilidad/efectos de los fármacos , Viscosidad/efectos de los fármacos
18.
Exp Eye Res ; 76(5): 553-63, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12697419

RESUMEN

Subcellular translocation of phototransduction proteins in response to light has previously been detected by immunocytochemistry. This movement is consistent with the hypothesis that migration is part of a basic cellular mechanism regulating photoreceptor sensitivity. In order to monitor the putative migration of arrestin in response to light, we expressed a functional fusion between the signal transduction protein arrestin and green fluorescent protein (GFP) in rod photoreceptors of transgenic Xenopus laevis. In addition to confirming reports that arrestin is translocated, this alternative approach generated unique observations, raising new questions regarding the nature and time scale of migration. Confocal fluorescence microscopy was performed on fixed frozen retinal sections from tadpoles exposed to three different lighting conditions. A consistent pattern of localization emerged in each case. During early light exposure, arrestin-GFP levels diminished in the inner segments (ISs) and simultaneously increased in the outer segments (OSs), initially at the base and eventually at the distal tips as time progressed. Arrestin-GFP reached the distal tips of the photoreceptors by 45-75 min at which time the ratio of arrestin-GFP fluorescence in the OSs compared to the ISs was maximal. When dark-adaptation was initiated after 45 min of light exposure, arrestin-GFP rapidly re-localized to the ISs and axoneme within 30 min. Curiously, prolonged periods of light exposure also resulted in re-localization of arrestin-GFP. Between 150 and 240 min of light adaptation the arrestin-GFP in the ROS gradually declined until the pattern of arrestin-GFP localization was indistinguishable from that of dark-adapted photoreceptors. This distribution pattern was observed over a wide range of lighting intensity (25-2700 lux). Immunocytochemical analysis of arrestin in wild-type Xenopus retinas gave similar results.


Asunto(s)
Adaptación Ocular/fisiología , Arrestina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Animales Modificados Genéticamente , Adaptación a la Oscuridad/fisiología , Proteínas Fluorescentes Verdes , Luz , Proteínas Luminiscentes , Microscopía Confocal , Estimulación Luminosa , Proteínas Recombinantes de Fusión/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA