Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Exp Eye Res ; 179: 106-114, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30445045

RESUMO

The Cacna1f gene encodes the α1F subunit of an L-type voltage-gated calcium channel, Cav1.4. In photoreceptor synaptic terminals, Cav1.4 channels mediate glutamate release and postsynaptic responses associated with visual signal transmission. We have discovered a new Cacna1f mutation in nob9 mice, which display more severe phenotypes than do nob2 mice. To characterize the nob9 phenotype at different ages, we examined the murine fundus, applied retinal optical coherence tomography, measured flash electroretinograms (ERGs) in vivo, and analyzed the retinal histology in vitro. After identifying the X-linked recessive inheritance trait, we sequenced Cacna1f as the candidate gene. Mutations in this gene were detected by polymerase chain reaction (PCR) and confirmed by restriction fragment length polymorphism. Morphologically, an early-onset of retinal disorder was detected, and the degeneration of the outer plexiform layers progressed rapidly. Moreover, the mutant mice showed drastically reduced scotopic ERGs with increasing age. In 14-month-old nob9 retinas, immunostaining of cone opsins demonstrated a reduction in the number of short-wavelength opsins (S-opsins) to 54% of wild-type levels, and almost no middle-wavelength opsins (M-opsins) were observed. No cone ERGs could be detected from residual cones, in which S-opsins abnormally migrated to inner segments of the photoreceptors. The mutations of the Cacna1f gene in nob9 mice involved both a single nucleotide G to A transition and a 10-nucleotide insertion, the latter resulting in a frame-shift mutation in exon 14.


Assuntos
Canais de Cálcio/genética , Modelos Animais de Doenças , Mutação da Fase de Leitura , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/genética , Animais , Canais de Cálcio Tipo L , Eletrorretinografia , Éxons , Técnicas de Genotipagem , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Visão Noturna/fisiologia , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Retina/fisiopatologia , Degeneração Retiniana/metabolismo , Degeneração Retiniana/fisiopatologia , Opsinas de Bastonetes/metabolismo , Tomografia de Coerência Óptica
2.
Mol Vis ; 24: 17-28, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29386880

RESUMO

Purpose: Blue cone monochromacy (BCM) is an X-linked congenital vision disorder characterized by complete loss or severely reduced L- and M-cone function. Patients with BCM display poor visual acuity, severely impaired color discrimination, myopia, nystagmus, and minimally detectable cone-mediated electroretinogram. Recent studies of patients with BCM with adaptive optics scanning laser ophthalmoscopy (AOSLO) showed that they have a disrupted cone mosaic with reduced numbers of cones in the fovea that is normally dominated by L- and M-cones. The remaining cones in the fovea have significantly shortened outer segments but retain sufficient structural integrity to serve as potential gene therapy targets. In this study, we tested whether exogenously expressed human L- and M-opsins can rescue M-cone function in an M-opsin knockout (Opn1mw-/- ) mouse model for BCM. Methods: Adeno-associated virus type 5 (AAV5) vectors expressing OPN1LW, OPN1MW, or C-terminal tagged OPN1LW-Myc, or OPN1MW-HA driven by a cone-specific promoter were injected subretinally into one eye of Opn1mw-/- mice, while the contralateral eye served as the uninjected control. Expression of cone pigments was determined with western blotting and their cellular localization identified with immunohistochemistry. M-cone function was analyzed with electroretinogram (ERG). Antibodies against cone phototransduction proteins were used to study cone outer segment (OS) morphology in untreated and treated Opn1mw-/- eyes. Results: We showed that cones in the dorsal retina of the Opn1mw-/- mouse do not form outer segments, resembling cones that lack outer segments in the human BCM fovea. We further showed that AAV5-mediated expression of either human M- or L-opsin individually or combined promotes regrowth of cone outer segments and rescues M-cone function in the treated Opn1mw-/- dorsal retina. Conclusions: Exogenously expressed human opsins can regenerate cone outer segments and rescue M-cone function in Opn1mw-/- mice, thus providing a proof-of-concept gene therapy in an animal model of BCM.


Assuntos
Defeitos da Visão Cromática/terapia , Fóvea Central/metabolismo , Terapia Genética/métodos , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Opsinas de Bastonetes/genética , Animais , Defeitos da Visão Cromática/genética , Defeitos da Visão Cromática/metabolismo , Defeitos da Visão Cromática/patologia , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Fóvea Central/patologia , Expressão Gênica , Teste de Complementação Genética , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Oftalmoscopia , Regiões Promotoras Genéticas , Segmento Externo das Células Fotorreceptoras da Retina/patologia , Opsinas de Bastonetes/metabolismo , Transgenes
3.
Hum Mol Genet ; 24(13): 3699-707, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25855802

RESUMO

The CNGA3(-/-)/Nrl(-/-) mouse is a cone-dominant model with Cnga3 channel deficiency, which partially mimics the all cone foveal structure of human achromatopsia 2 with CNGA3 mutations. Although subretinal (SR) AAV vector administration can transfect retinal cells efficiently, the injection-induced retinal detachment can cause retinal damage, particularly when SR vector bleb includes the fovea. We therefore explored whether cone function-structure could be rescued in CNGA3(-/-)/Nrl(-/-) mice by intravitreal (IVit) delivery of tyrosine to phenylalanine (Y-F) capsid mutant AAV8. We find that AAV-mediated CNGA3 expression can restore cone function and rescue structure following IVit delivery of AAV8 (Y447, 733F) vector. Rescue was assessed by restoration of the cone-mediated electroretinogram (ERG), optomotor responses, and cone opsin immunohistochemistry. Demonstration of gene therapy in a cone-dominant mouse model by IVit delivery provides a potential alternative vector delivery mode for safely transducing foveal cones in achromatopsia patients and in other human retinal diseases affecting foveal function.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Defeitos da Visão Cromática/genética , Defeitos da Visão Cromática/terapia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Proteínas do Olho/genética , Terapia Genética , Células Fotorreceptoras Retinianas Cones/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Defeitos da Visão Cromática/metabolismo , Defeitos da Visão Cromática/fisiopatologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Proteínas do Olho/metabolismo , Feminino , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
J Neurosci ; 33(29): 11745-53, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23864662

RESUMO

Phosphodiesterase-6 (PDE6) is the key effector enzyme of the vertebrate phototransduction pathway in rods and cones. Rod PDE6 catalytic core is composed of two distinct subunits, PDE6α and PDE6ß, whereas two identical PDE6α' subunits form the cone PDE6 catalytic core. It is not known whether this difference in PDE6 catalytic subunit identity contributes to the functional differences between rods and cones. To address this question, we expressed cone PDE6α' in the photoreceptor cells of the retinal degeneration 10 (rd10) mouse that carries a mutation in rod PDEß subunit. We show that adeno-associated virus-mediated subretinal delivery of PDE6α' rescues rod electroretinogram responses and preserves retinal structure, indicating that cone PDE6α' can couple effectively to the rod phototransduction pathway. We also show that restoration of light sensitivity in rd10 rods is attributable to assembly of PDE6α' with rod PDE6γ. Single-cell recordings revealed that, surprisingly, rods expressing cone PDE6α' are twofold more sensitive to light than wild-type rods, most likely because of the slower shutoff of their light responses. Unlike in wild-type rods, the response kinetics in PDE6α'-treated rd10 rods accelerated with increasing flash intensity, indicating a possible direct feedback modulation of cone PDE6α' activity. Together, these results demonstrate that cone PDE6α' can functionally substitute for rod PDEαß in vivo, conferring treated rods with distinct physiological properties.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Domínio Catalítico , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Técnicas de Transferência de Genes , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Células Fotorreceptoras Retinianas Cones/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo
5.
Nat Med ; 13(6): 685-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17515894

RESUMO

Loss of cone function in the central retina is a pivotal event in the development of severe vision impairment for many prevalent blinding diseases. Complete achromatopsia is a genetic defect resulting in cone vision loss in 1 in 30,000 individuals. Using adeno-associated virus (AAV) gene therapy, we show that it is possible to target cones and rescue both the cone-mediated electroretinogram response and visual acuity in the Gnat2 ( cpfl3 ) mouse model of achromatopsia.


Assuntos
Defeitos da Visão Cromática/terapia , Modelos Animais de Doenças , Terapia Genética , Células Fotorreceptoras Retinianas Cones/fisiologia , Animais , Defeitos da Visão Cromática/genética , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Camundongos , Camundongos Transgênicos
6.
Biochem Biophys Rep ; 37: 101646, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38333050

RESUMO

Adeno-associated virus (AAV) vectors have been widely used in therapy to treat hereditary retinal diseases. But its transduction efficiency by intravitreal injection still needs to be improved. In this study, we investigated the transduction efficiency of AAV-DJ (K137R)-GFP in different retinal cells of normal mice, as well as the therapy effection of AAV-DJ (K137R)-Rs1 on retinal function and structure in Rs1-KO mice. The intravitreal injection of AAV-DJ (K137R)-GFP demonstrated that this vector transduced cells in all layers of the retina, including the inner nuclear layer and photoreceptor layer. The intravitreal injection of AAV-DJ (K137R)-Rs1 found that 3 months post-injection of this vector improved retinal function and structure in Rs1-KO mice. Our conclusion is that AAV-DJ (K137R) vector can efficiently and safely penetrate the inner limiting membrane and transduce different layers of retinal cells in the long term, as well as being able to continuously and efficiently express target therapeutic proteins, making it a candidate therapeutic vector for X-linked retinoschisis (XLRS).

7.
Mol Vis ; 19: 2579-89, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24367157

RESUMO

Mouse models are useful tools for developing potential therapies for human inherited retinal diseases, such as retinitis pigmentosa (RP), since more strains are being identified with the same mutant genes and phenotypes as humans with corresponding retinal degenerative diseases. Mutations in the beta subunit of the human rod phosphodiesterase (PDE6B) gene are a common cause of autosomal recessive RP (arRP). This article focuses on two well-established naturally occurring mouse models of arRP caused by spontaneous mutations in Pde6b, their discovery, phenotype, mechanism of degeneration, strengths and limitations, and therapeutic approaches to restore vision and delay disease progression. Viral vector, especially adeno-associated viral vector (AAV) -mediated gene replacement therapy, pharmacological treatment, cell-based therapy and other approaches that extend the therapeutic window of treatment, is a potentially promising strategy for improving photoreceptor function and significantly slowing the process of retinal degeneration.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Mutação , Retina/enzimologia , Retinose Pigmentar/genética , Retinose Pigmentar/terapia , Adenoviridae/genética , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos , Humanos , Camundongos , Retina/patologia , Retinose Pigmentar/enzimologia , Retinose Pigmentar/patologia
8.
Zhonghua Yan Ke Za Zhi ; 49(6): 521-5, 2013 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-24119965

RESUMO

OBJECTIVE: To investigate the distribution and biological roles of voltage-dependent calcium channel (VDCC) α1F subunit in murine retina. METHODS: Experimental study.α1F(-/-) (homozygous mutant) mice (n = 35) and α1F(+/+) (wild type) mice (n = 35) were used in this study. Immunohistochemistry was performed to determine the expression of VDCC α1F subunit in the mouse retina. Retinae in α1F(-/-) mice and age-matched control mice at 3, 6, 9, 14-day and 3-month after birth were paraffin embedded, sectioned and HE stained, and full-field electroretinogram (ERG) were also recorded at these time points.Statistics were based on independent samples t-test. RESULTS: (1) α1F subunit was absent in α1F(-/-) mice retina. But in α1F(+/+) mice retina, α1F subunit was expressed most strongly in the outer plexiform layer (OPL), less in the inner plexiform layer (IPL) and ganglion cell layer (GCL). (2) OPL thickness in the subunit deficient mice gradually reduced after birth and lost at adult age. (3) In dark-adapted ERGs,standard response showed that the b-wave amplitude of α1F(-/-) mice [(163.8 ± 26.7) µV] significantly decreased compared with that of α1F(+/+) mice [(408.4 ± 54.5) µV] (t = -9.017, P = 0.000), whereas the a-wave amplitude of α1F(-/-) group [(208.2 ± 27.3) µV] was similar to that of control group [(196.0 ± 24.2) µV] (t = 0.748, P = 0.476). CONCLUSION: This study demonstrates that the lack of VDCC α1F subunit affect the structure and function in the OPL of the murine retina.


Assuntos
Canais de Cálcio Tipo L/genética , Retina/patologia , Retina/fisiopatologia , Animais , Eletrorretinografia , Homozigoto , Camundongos , Camundongos Knockout , Mutação
9.
Front Med (Lausanne) ; 10: 1291795, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38264046

RESUMO

Leber congenital amaurosis (LCA) is the most common genetic cause of congenital visual impairment in infants and children. Patients with LCA who harbor RPE65 mutations exhibit a deficiency in photoreceptor rhodopsin, leading to severe night blindness and visual impairment following birth. Since either gene replacement therapy or anti-apoptosis therapy alone cannot maintain both functional and morphological normality for a long time in the animal model, we propose a robust treatment strategy, that is, gene replacement therapy combined with anti-apoptotic therapy to protect photoreceptors from further degeneration while compensating for lost RPE65 function. Here, rd12 mice were injected subretinally at postnatal day 14 with four vector administrations, respectively. At 6 months after treatment, it was discovered that injection of three vectors, AAV8 (Y733F)-CBA-hRPE65, AAV8(Y733F)-CBA-hRPE65-BCL-2-L10 and mixture of half-dose AAV8(Y733F)-CBA-hRPE65 and half-dose AAV8 (Y733F)-CBA-BCL-2-L10, could partially restore the visual function of rd12 mice. Meanwhile, these treated eyes also exhibited a thicker outer nuclear layer (ONL) structure. However, despite the fact that the eyes of rd12 mice injected with the AAV8 (Y733F)-CBA-BCL-2-L10 vector displayed a slightly thicker ONL structure compared to untreated eyes, the visual function of the treated eyes did not recover. Continuing the observation period to 12 months after treatment, we found that compared to rd12 mice at 6-month post-treatment, rd12 mice injected with AAV8 (Y733F)-CBA-hRPE65 or mixture of half-dose AAV8(Y733F)-CBA-hRPE65 and half-dose AAV8 (Y733F)-CBA-BCL-2-L10 exhibited varying degrees of decline in both visual function and ONL thickness. However, in the case of rd12 mice injected with the AAV8(Y733F)-CBA-hRPE65-BCL-2-L10 vector, the ONL thickness remains consistent at both 6 and 12 months after treatment. These mice continued to maintain a relatively strong visual function and showed restoration in the levels of RPE65 and Rhodopsin protein expression. Our findings illustrate that early postnatal treatment with AAV vectors containing both the hRPE65 gene and the Bcl-2L10 anti-apoptotic gene provide enhanced and sustained retinal protection.

10.
Front Med (Lausanne) ; 10: 1220075, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37636578

RESUMO

X-linked retinoschisis is more common in male children and rare in females. Clinically, male patients mainly present with early onset visual impairment or vision loss, and retinal retinoschisis due to division of the inner retina. We report a long-term observation of a female patient with familial foveal retinoschisis (FFR) caused by CRB1 gene with complex heterozygotic mutation. The initial symptoms of the female patient reported in this study were very similar to some early manifestations of X-linked retinoschisis (XLRS) caused by RS1 mutations involving macular fovea. However, as time going on, the splitting height at retinal fovea of FFR gradually decreased, and the splitting extent at retinal fovea of FFR gradually decreased.

11.
Mol Ther ; 19(2): 234-42, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21139570

RESUMO

The retinal degeneration 10 (rd10) mouse is a well-characterized model of autosomal recessive retinitis pigmentosa (RP), which carries a spontaneous mutation in the ß subunit of rod cGMP-phosphodiesterase (PDEß). Rd10 mouse exhibits photoreceptor dysfunction and rapid rod photoreceptor degeneration followed by cone degeneration and remodeling of the inner retina. Here, we evaluate whether gene replacement using the fast-acting tyrosine-capsid mutant AAV8 (Y733F) can provide long-term therapy in this model. AAV8 (Y733F)-smCBA-PDEß was subretinally delivered to postnatal day 14 (P14) rd10 mice in one eye only. Six months after injection, spectral domain optical coherence tomography (SD-OCT), electroretinogram (ERG), optomotor behavior tests, and immunohistochemistry showed that AAV8 (Y733F)-mediated PDEß expression restored retinal function and visual behavior and preserved retinal structure in treated rd10 eyes for at least 6 months. This is the first demonstration of long-term phenotypic rescue by gene therapy in an animal model of PDEß-RP. It is also the first example of tyrosine-capsid mutant AAV8 (Y733F)-mediated correction of a retinal phenotype. These results lay the groundwork for the development of PDEß-RP gene therapy trial and suggest that tyrosine-capsid mutant AAV vectors may be effective for treating other rapidly degenerating models of retinal degeneration.


Assuntos
Capsídeo/metabolismo , Dependovirus/genética , Vetores Genéticos/genética , Retinose Pigmentar/terapia , Animais , Western Blotting , Modelos Animais de Doenças , Eletrorretinografia , Terapia Genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Retinose Pigmentar/genética , Retinose Pigmentar/patologia
12.
Mol Ther ; 19(2): 293-301, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21045809

RESUMO

Vectors based on adeno-associated virus serotype 2 (AAV2) have been used extensively in many gene-delivery applications, including several successful clinical trials for one type of Leber congenital amaurosis in the retina. Many studies have focused on improving AAV2 transduction efficiency and cellular specificity by genetically engineering its capsid. We have previously shown that vectors-containing single-point mutations of capsid surface tyrosines in serotypes AAV2, AAV8, and AAV9 displayed significantly increased transduction efficiency in the retina compared with their wild-type counterparts. In the present study, we evaluated the transduction characteristics of AAV2 vectors containing combinations of multiple tyrosine to phenylalanine mutations in seven highly conserved surface-exposed capsid tyrosine residues following subretinal or intravitreal delivery in adult mice. The multiply mutated vectors exhibited different in vivo transduction properties, with some having a unique ability of transgene expression in all retinal layers. Such novel vectors may be useful in developing valuable new therapeutic strategies for the treatment of many genetic diseases.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Retina/metabolismo , Tirosina/genética , Animais , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Mutação , Mutação Puntual/genética , Retina/patologia
13.
Proc Natl Acad Sci U S A ; 106(42): 17681-6, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-19815523

RESUMO

Rod and cone photoreceptors use similar but distinct sets of phototransduction proteins to achieve different functional properties, suitable for their role as dim and bright light receptors, respectively. For example, rod and cone visual pigments couple to distinct variants of the heterotrimeric G protein transducin. However, the role of the structural differences between rod and cone transducin alpha subunits (Talpha) in determining the functional differences between rods and cones is unknown. To address this question, we studied the translocation and signaling properties of rod Talpha expressed in cones and cone Talpha expressed in rods in three mouse strains: rod Talpha knockout, cone Talpha GNAT2(cpfl3) mutant, and rod and cone Talpha double mutant rd17 mouse. Surprisingly, although the rod/cone Talpha are only 79% identical, exogenously expressed rod or cone Talpha localized and translocated identically to endogenous Talpha in each photoreceptor type. Moreover, exogenously expressed rod or cone Talpha rescued electroretinogram responses (ERGs) in mice lacking functional cone or rod Talpha, respectively. Ex vivo transretinal ERG and single-cell recordings from rd17 retinas treated with rod or cone Talpha showed comparable rod sensitivity and response kinetics. These results demonstrate that cone Talpha forms a functional heterotrimeric G protein complex in rods and that rod and cone Talpha couple equally well to the rod phototransduction cascade. Thus, rod and cone transducin alpha-subunits are functionally interchangeable and their signaling properties do not contribute to the intrinsic light sensitivity differences between rods and cones. Additionally, the technology used here could be adapted for any such homologue swap desired.


Assuntos
Proteínas do Olho/química , Proteínas do Olho/fisiologia , Proteínas Heterotriméricas de Ligação ao GTP/química , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Animais , Eletrorretinografia , Potenciais Evocados Visuais , Proteínas do Olho/genética , Proteínas Heterotriméricas de Ligação ao GTP/deficiência , Proteínas Heterotriméricas de Ligação ao GTP/genética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Estimulação Luminosa , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
Zhonghua Yan Ke Za Zhi ; 48(8): 755-8, 2012 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-23141518

RESUMO

Achromatopsia is an early onset retinal dystrophy that causes severe visual impairment. To date, four genes have been found to be implicated in achromatopsia-associated mutations: guanine nucleotide-binding protein (GNAT2), cyclic nucleotide-gated channel alpha-3 (CNGA3), cyclic nucleotide-gated channel beta-3 (CNGB3) and phosphodiesterase 6C (PDE6C). Even with early onset, the slow progress and the good responses to gene therapy in animal models render achromatopsia a very attractive candidate for human gene therapy after the successful of the Phase I clinical trials of Leber's congenital amaurosis. With the development of molecular genetics and the therapeutic gene replacement technology, the adeno-associated viral (AAV) vector-mediated gene therapy for achromatopsia in the preclinical animal experiments achieved encouraging progress in the past years. This article briefly reviews the recent research achievements of achromatopsia with gene therapy.


Assuntos
Defeitos da Visão Cromática/terapia , Terapia Genética , Humanos
15.
Eur J Ophthalmol ; : 11206721221136324, 2022 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-36325687

RESUMO

PURPOSE: Bardet-Biedl syndrome (BBS) is a rare autosomal-recessive inherited disorder characterized by multisystem anomalies. The objective of this study was to detect and analyse pathogenic variants in four Chinese families with BBS. METHODS: Comprehensive clinical examinations were performed to investigate and evaluate the phenotypes of the affected individuals from four families. Genomic DNA was extracted from peripheral blood. Next-generation sequencing (NGS) was performed for four families, and the presence of pathogenic variants was confirmed via Sanger sequencing. RESULTS: There were two males and three females with a mean age of 16.00 years. All probands displayed the primary clinical features of BBS. Mutation screening demonstrated four novel mutations: c.613C>T; p.Q205* in the BBS5 gene, c.1391C>G; p.S464* in the BBS10 gene, and c.155delC; p.S52* and c.1584T>G; p.Y528* in the BBS12 gene. Two previously reported mutations were also identified, including c.534 + 1G>T in the BBS2 gene and c.539G>A; p.G180E in the BBS10 gene. The bioinformatic analysis revealed that all the detected mutations in BBS genes were disease causing. CONCLUSIONS: This study identified four novel BBS gene mutations in these Chinese families and further expanded the genotypic spectrum of BBS, thus contributing to the literature and understanding of this multisystem disease.

16.
Proc Natl Acad Sci U S A ; 105(39): 15112-7, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18809924

RESUMO

The RPE65 gene encodes the isomerase of the retinoid cycle, the enzymatic pathway that underlies mammalian vision. Mutations in RPE65 disrupt the retinoid cycle and cause a congenital human blindness known as Leber congenital amaurosis (LCA). We used adeno-associated virus-2-based RPE65 gene replacement therapy to treat three young adults with RPE65-LCA and measured their vision before and up to 90 days after the intervention. All three patients showed a statistically significant increase in visual sensitivity at 30 days after treatment localized to retinal areas that had received the vector. There were no changes in the effect between 30 and 90 days. Both cone- and rod-photoreceptor-based vision could be demonstrated in treated areas. For cones, there were increases of up to 1.7 log units (i.e., 50 fold); and for rods, there were gains of up to 4.8 log units (i.e., 63,000 fold). To assess what fraction of full vision potential was restored by gene therapy, we related the degree of light sensitivity to the level of remaining photoreceptors within the treatment area. We found that the intervention could overcome nearly all of the loss of light sensitivity resulting from the biochemical blockade. However, this reconstituted retinoid cycle was not completely normal. Resensitization kinetics of the newly treated rods were remarkably slow and required 8 h or more for the attainment of full sensitivity, compared with <1 h in normal eyes. Cone-sensitivity recovery time was rapid. These results demonstrate dramatic, albeit imperfect, recovery of rod- and cone-photoreceptor-based vision after RPE65 gene therapy.


Assuntos
Cegueira/terapia , Proteínas de Transporte/genética , Proteínas do Olho/genética , Terapia Genética , Isomerases/genética , Células Fotorreceptoras Retinianas Bastonetes/fisiopatologia , Retinoides/metabolismo , Cegueira/patologia , Cegueira/fisiopatologia , Dependovirus/genética , Humanos , Células Fotorreceptoras Retinianas Cones/enzimologia , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Cones/fisiopatologia , Células Fotorreceptoras Retinianas Bastonetes/enzimologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Visão Ocular/fisiologia , cis-trans-Isomerases
17.
Zhonghua Yan Ke Za Zhi ; 47(1): 83-7, 2011 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-21418933

RESUMO

Leber congenital amaurosis (LCA) is an early onset retinal dystrophy that causes severe visual impairment. With the development of molecular genetics and the therapeutic gene replacement technology, the adeno-associated viral (AAV) vector-mediated gene therapy for LCA achieved encouraging progress in the past decade. The success of the Phase I clinical trials of human RPE65 gene therapy for LCA II patients makes it a pioneer in the field of retinal gene therapy and brings light to the cure of other hereditary retinopathy. This article briefly reviews the recent developments in the preclinical animal experiments and Phase I clinical trials for LCA.


Assuntos
Terapia Genética , Amaurose Congênita de Leber/terapia , Ensaios Clínicos Fase I como Assunto , Humanos
18.
Exp Eye Res ; 90(5): 546-54, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20138034

RESUMO

To clarify whether transduction efficiency and cell type specificity of self-complementary (sc) AAV5 vectors are similar to those of standard, single-stranded AAV5 vectors in normal retina, one micro liter of scAAV5-smCBA-GFP vector (1 x 10(12) genome-containing particles/ml) and AAV5-smCBA-GFP vector (1 x 10(12) genome-containing particles/ml) were subretinally or intravitreally (in both cases through the cornea) injected into the right and left eyes of adult C57BL/6J mice, respectively. On post-injection day (PID) 1, 2, 5, 7, 10, 14, 21, 28 and 35, eyes were enucleated; retinal pigment epithelium (RPE) wholemounts, neuroretinal wholemounts and eyecup sections were prepared to evaluate green fluorescent protein (GFP) expression by fluorescent microscopy. GFP expression following trans-cornea subretinal injection of scAAV5-smCBA-GFP vector was first detected in RPE wholemounts around PID 1 and in neuroretinal wholemounts between PID 2 and 5; GFP expression peaked and stabilized between PID 10-14 in RPE wholemounts and between P14 and P21 in neuroretinal wholemounts with strong, homogeneous green fluorescence covering the entire wholemounts. The frozen sections supported the following findings from the wholemounts: GFP expression appeared first in RPE around PID 1-2 and soon spread to photoreceptors (PR) cells; by PID 7, moderate GFP expression was found mainly in PR and RPE layers; between PID 14 and 21, strong and homogenous GFP expression was observed in RPE and PR cells. GFP expression following subretinal injection of AAV5-smCBA-GFP was first detected in RPE wholemounts around PID 5-7 and in neuroretinal wholemounts around PID 7-10; ssAAV5-mediated GFP expression peaked at PID 21 in RPE wholemounts and around PID 28 in neuroretinal wholemounts; sections from AAV5 treated eyes also supported findings obtained from wholemounts: GFP expression was first detected in RPE and then spread to the PR cells. Peak GFP expression in RPE mediated by scAAV5 was similar to that mediated by AAV5. However, peak GFP expression mediated by scAAV5 in PR cells was stronger than that mediated by AAV5. No GFP fluorescence was detected in any retinal cells (RPE wholemounts, neuroretinal wholemounts and retinal sections) after trans-cornea intravitreal delivery of either scAAV5-GFP or AAV5-GFP. Neither scAAV5 nor AAV5 can transduce retinal cells following trans-cornea intravitreal injection. The scAAV5 vector used in this study directs an earlier onset of transgene expression than the matched AAV5 vector, and has stronger transgene expression in PR cells following subretinal injection. Our data confirm the previous reports that scAAV vectors have an earlier onset than the standard, single strand AAV vectors (Natkunarajah et al., 2008; Yokoi et al., 2007). scAAV5 vectors may be more useful than standard, single-stranded AAV vector when addressing certain RPE and/or PR cell-related models of retinal dystrophy, particularly for mouse models of human retinitis pigmentosa that require rapid and robust transgene expression to prevent early degeneration in PR cells.


Assuntos
Dependovirus/genética , Regulação da Expressão Gênica/fisiologia , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Injeções , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Retina/efeitos dos fármacos , Transgenes , Corpo Vítreo
19.
Mol Ther ; 17(3): 463-71, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19066593

RESUMO

Vectors derived from adeno-associated viruses (AAVs) have become important gene delivery tools for the treatment of many inherited ocular diseases in well-characterized animal models. Previous studies have determined that the viral capsid plays an essential role in the cellular tropism and efficiency of transgene expression. Recently, it was shown that phosphorylation of surface-exposed tyrosine residues from AAV2 capsid targets the viral particles for ubiquitination and proteasome- mediated degradation, and mutations of these tyrosine residues lead to highly efficient vector transduction in vitro and in vivo. Because the tyrosine residues are highly conserved in other AAV serotypes, in this study we evaluated the intraocular transduction characteristics of vectors containing point mutations in surface- exposed capsid tyrosine residues in AAV serotypes 2, 8, and 9. Several of these novel AAV mutants were found to display a strong and widespread transgene expression in many retinal cells after subretinal or intravitreal delivery compared with their wild-type counterparts. For the first time, we show efficient transduction of the ganglion cell layer by AAV serotype 8 or 9 mutant vectors, thus providing additional tools besides AAV2 for targeting these cells. These enhanced AAV vectors have a great potential for future therapeutic applications for retinal degenerations and ocular neovascular diseases.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Retina/metabolismo , Transgenes/genética , Animais , Dependovirus/classificação , Cistos Glanglionares , Vetores Genéticos/administração & dosagem , Injeções , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Tirosina/genética , Tirosina/metabolismo
20.
Adv Exp Med Biol ; 664: 639-46, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20238068

RESUMO

Achromatopsia is an autosomal recessive retinal disease involving loss of cone function that afflicts approximately 1 in 30,000 individuals. Patients with achromatopsia usually have visual acuities lower than 20/200 because of the central vision loss, photophobia, complete color blindness and reduced cone-mediated electroretinographic (ERG) amplitudes. Mutations in three genes have been found to be the primary causes of achromatopsia, including CNGB3 (beta subunit of the cone cyclic nucleotide-gated cation channel), CNGA3 (alpha subunit of the cone cyclic nucleotide-gated cation channel), and GNAT2 (cone specific alpha subunit of transducin). Naturally occurring mouse models with mutations in Cnga3 (cpfl5 mice) and Gnat2 (cpfl3 mice) were discovered at The Jackson Laboratory. A natural occurring canine model with CNGB3 mutations has also been found. These animal models have many of the central phenotypic features of the corresponding human diseases. Using adeno-associated virus (AAV)-mediated gene therapy, we and others show that cone function can be restored in all three models. These data suggest that human achromatopsia may be a good candidate for corrective gene therapy.


Assuntos
Defeitos da Visão Cromática/genética , Defeitos da Visão Cromática/terapia , Terapia Genética , Animais , Defeitos da Visão Cromática/fisiopatologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/uso terapêutico , Cães , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/uso terapêutico , Humanos , Camundongos , Mutação/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa