Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 32(5): 1445-1460, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38504520

RESUMO

Age-related macular degeneration (AMD) is the most common cause of untreatable blindness in the developed world. Recently, CDHR1 has been identified as the cause of a subset of AMD that has the appearance of the "dry" form, or geographic atrophy. Biallelic variants in CDHR1-a specialized protocadherin highly expressed in cone and rod photoreceptors-result in blindness from shortened photoreceptor outer segments and progressive photoreceptor cell death. Here we demonstrate long-term morphological, ultrastructural, functional, and behavioral rescue following CDHR1 gene therapy in a relevant murine model, sustained to 23-months after injection. This represents the first demonstration of rescue of a monogenic cadherinopathy in vivo. Moreover, the durability of CDHR1 gene therapy seems to be near complete-with morphological findings of the rescued retina not obviously different from wildtype throughout the lifespan of the mouse model. A follow-on clinical trial in patients with CDHR1-associated retinal degeneration is warranted. Hypomorphic CDHR1 variants may mimic advanced dry AMD. Accurate clinical classification is now critical, as their pathogenesis and treatment are distinct.


Assuntos
Proteínas Relacionadas a Caderinas , Caderinas , Modelos Animais de Doenças , Terapia Genética , Proteínas do Tecido Nervoso , Células Fotorreceptoras Retinianas Cones , Degeneração Retiniana , Células Fotorreceptoras Retinianas Bastonetes , Animais , Camundongos , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Caderinas/genética , Caderinas/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Degeneração Retiniana/etiologia , Humanos , Terapia Genética/métodos , Degeneração Macular/terapia , Degeneração Macular/genética , Degeneração Macular/patologia , Degeneração Macular/etiologia , Degeneração Macular/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(49): e2208707119, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36445968

RESUMO

Pathogenic variants in the Retinitis pigmentosa GTPase regulator (RPGR) gene lead to a clinically severe form of X-linked retinal dystrophy. However, it remains unclear why some variants cause a predominant rod, while others result in a cone-dominated phenotype. Post-translational glutamylation of the photoreceptor-specific RPGRORF15 isoform by the TTLL5 enzyme is essential for its optimal function in photoreceptors, and loss of TTLL5 leads to retinal dystrophy with a cone phenotype. Here we show that RPGR retinal disease, studied in a single cohort of 116 male patients, leads to a clear progressive shift from rod- to cone-dominating phenotype as the RPGRORF15 variant location approaches the distal part of the Open Reading Frame 15 (ORF15) region. The rod photoreceptor involvement on the contrary diminishes along the RGPR sequence, and the variants associated with the cone only phenotype are located predominantly in the very distal part, including the C-terminal basic domain. Moreover, these distal truncating RPGRORF15 variants disrupt the interaction with TTLL5 and lead to a significant impairment of RPGR glutamylation. Thus, consistent with the phenotype of TTLL5 pathogenic variants, our study shows that RPGRORF15 variants, which disrupt its basic domain and the interaction with TTLL5, also impair RPGR glutamylation and lead to the cone phenotype. This has implications for ongoing gene therapy clinical trials where the application of RPGR with impaired glutamylation may be less effective in treating RGPR dystrophies and may even convert a rod-cone dystrophy into a cone dystrophy phenotype.


Assuntos
Distrofias de Cones e Bastonetes , Distrofias Retinianas , Humanos , Masculino , Distrofias de Cones e Bastonetes/genética , Distrofias de Cones e Bastonetes/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Fases de Leitura Aberta/genética , Fases de Leitura Aberta/fisiologia , Fenótipo , Células Fotorreceptoras Retinianas Cones/metabolismo , Distrofias Retinianas/genética , Distrofias Retinianas/metabolismo , Ácido Glutâmico/metabolismo
3.
Int J Mol Sci ; 25(3)2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38338978

RESUMO

Age-related macular degeneration (AMD) is the leading cause of irreversible vision loss among the elderly in the developed world. Whilst AMD is a multifactorial disease, the involvement of the complement system in its pathology is well documented, with single-nucleotide polymorphisms (SNPs) in different complement genes representing an increased risk factor. With several complement inhibitors explored in clinical trials showing limited success, patients with AMD are still without a reliable treatment option. This indicates that there is still a gap of knowledge in the functional implications and manipulation of the complement system in AMD, hindering the progress towards translational treatments. Since the discovery of the CRISPR/Cas system and its development into a powerful genome engineering tool, the field of molecular biology has been revolutionised. Genetic variants in the complement system have long been associated with an increased risk of AMD, and a variety of haplotypes have been identified to be predisposing/protective, with variation in complement genes believed to be the trigger for dysregulation of the cascade leading to inflammation. AMD-haplotypes (SNPs) alter specific aspects of the activation and regulation of the complement cascade, providing valuable insights into the pathogenic mechanisms of AMD with important diagnostic and therapeutic implications. The effect of targeting these AMD-related SNPs on the regulation of the complement cascade has been poorly explored, and the CRISPR/Cas system provides an ideal tool with which to explore this avenue. Current research concentrates on the association events of specific AMD-related SNPs in complement genes without looking into the effect of targeting these SNPs and therefore influencing the complement system in AMD pathogenesis. This review will explore the current understanding of manipulating the complement system in AMD pathogenesis utilising the genomic manipulation powers of the CRISPR/Cas systems. A number of AMD-related SNPs in different complement factor genes will be explored, with a particular emphasis on factor H (CFH), factor B (CFB), and complement C3 (C3).


Assuntos
Fator B do Complemento , Degeneração Macular , Humanos , Idoso , Haplótipos , Degeneração Macular/genética , Degeneração Macular/terapia , Degeneração Macular/patologia , Ativação do Complemento/genética , Fatores de Risco , Polimorfismo de Nucleotídeo Único
4.
Int J Mol Sci ; 24(8)2023 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-37108761

RESUMO

This review considers research into the treatment of Usher syndrome, a deaf-blindness syndrome inherited in an autosomal recessive manner. Usher syndrome mutations are markedly heterogeneous, involving many different genes, and research grants are limited due to minimal patient populations. Furthermore, gene augmentation therapies are impossible in all but three Usher syndromes as the cDNA sequence exceeds the 4.7 kb AAV packaging limit. It is, therefore, vital to focus research efforts on alternative tools with the broadest applicability. The CRISPR field took off in recent years following the discovery of the DNA editing activity of Cas9 in 2012. New generations of CRISPR tools have succeeded the original CRISPR/Cas9 model to enable more sophisticated genomic amendments such as epigenetic modification and precise sequence alterations. This review will evaluate the most popular CRISPR tools to date: CRISPR/Cas9, base editing, and prime editing. It will consider these tools in terms of applicability (in relation to the ten most prevalent USH2A mutations), safety, efficiency, and in vivo delivery potential with the intention of guiding future research investment.


Assuntos
Edição de Genes , Síndromes de Usher , Humanos , Sistemas CRISPR-Cas/genética , Síndromes de Usher/genética , Síndromes de Usher/terapia , Terapia Genética , Epigênese Genética
5.
Int J Mol Sci ; 23(19)2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36232969

RESUMO

Inherited retinal degenerations are a leading cause of blindness in the UK. Significant advances have been made to tackle this issue in recent years, with a pioneering FDA approved gene therapy treatment (Luxturna®), which targets a loss of function mutation in the RPE65 gene. However, there remain notable shortcomings to this form of gene replacement therapy. In particular, the lack of viability for gene sequences exceeding the 4.7 kb adeno-associated virus (AAV) packaging limit or for toxic gain of function mutations. The USH2A gene at ~15.7 kb for instance is too large for AAV delivery: a safe and effective vehicle capable of transducing photoreceptor cells for gene replacement therapy. Usher Syndrome is a clinically and genetically heterogenous deaf-blindness syndrome with autosomal recessive inheritance. The USH2A gene encodes the protein usherin, which localises to the photoreceptor cilium and cochlear hair cells. Mutations in the USH2A gene cause Usher Syndrome type II (USH2), which is the most common subtype of Usher Syndrome and the focus of this review. To date, researchers have been unable to create an efficient, safe editing tool that is small enough to fit inside a single AAV vector for delivery into human cells. This article reviews the potential of CRISPR technology, derived from bacterial defence mechanisms, to overcome these challenges; delivering tools to precisely edit and correct small insertions, deletions and base transitions in USH2A without the need to deliver the full-length gene. Such an ultra-compact therapy could make strides in combating a significant cause of blindness in young people.


Assuntos
Degeneração Retiniana , Síndromes de Usher , Adolescente , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Proteínas da Matriz Extracelular/metabolismo , Humanos , Mutação , Degeneração Retiniana/genética , Síndromes de Usher/genética , Síndromes de Usher/terapia
6.
Int J Mol Sci ; 23(19)2022 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-36232975

RESUMO

Non-viral gene therapy has the potential to overcome several shortcomings in viral vector-based therapeutics. Methods of in vivo plasmid delivery have developed over recent years to increase the efficiency of non-viral gene transfer, yet further improvements still need to be made to improve their translational capacity. Gene therapy advances for inherited retinal disease have been particularly prominent over the recent decade but overcoming physical and physiological barriers present in the eye remains a key obstacle in the field of non-viral ocular drug delivery. Minicircles are circular double-stranded DNA vectors that contain expression cassettes devoid of bacterial DNA, thereby limiting the risks of innate immune responses induced by such elements. To date, they have not been extensively used in pre-clinical studies yet remain a viable vector option for the treatment of inherited retinal disease. Here, we explore the potential of minicircle DNA delivery to the neural retina as a gene therapy approach. We consider the advantages of minicircles as gene therapy vectors as well as review the challenges involved in optimising their delivery to the neural retina.


Assuntos
Terapia Genética , Doenças Retinianas , DNA/genética , DNA Bacteriano , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Humanos , Retina
7.
Gene Ther ; 28(5): 265-276, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33750925

RESUMO

Dry age-related macular degeneration (AMD) is characterised by loss of central vision and currently has no approved medical treatment. Dysregulation of the complement system is thought to play an important role in disease pathology and supplementation of Complement Factor I (CFI), a key regulator of the complement system, has the potential to provide a treatment option for AMD. In this study, we demonstrate the generation of AAV constructs carrying the human CFI sequence and expression of CFI in cell lines and in the retina of C57BL/6 J mice. Four codon optimised constructs were compared to the most common human CFI sequence. All constructs expressed CFI protein; however, most codon optimised sequences resulted in significantly reduced CFI secretion compared to the non-optimised CFI sequence. In vivo expression analysis showed that CFI was predominantly expressed in the RPE and photoreceptors. Secreted protein in vitreous humour was demonstrated to be functionally active. The findings presented here have led to the formulation of an AAV-vectored gene therapy product currently being tested in a first-in-human clinical trial in subjects with geographic atrophy secondary to dry AMD (NCT03846193).


Assuntos
Fator I do Complemento , Degeneração Macular , Animais , Fator I do Complemento/genética , Terapia Genética , Humanos , Degeneração Macular/genética , Degeneração Macular/terapia , Camundongos , Camundongos Endogâmicos C57BL , Retina
8.
Mol Vis ; 27: 233-242, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34012226

RESUMO

Purpose: The classic Kozak consensus is a critical genetic element included in gene therapy transgenes to encourage the translation of the therapeutic coding sequence. Despite optimizations of other transgene elements, the Kozak consensus has not yet been considered for potential tissue-specific sequence refinement. We screened the -9 to -1 region relative to the AUG start codon of retina-specific genes to identify whether a Kozak consensus that is different from the classic sequence may be more appropriate for inclusion in gene therapy transgenes that treat inherited retinal disease. Methods: Sequences for 135 genes known to cause nonsyndromic inherited retinal disease were extracted from the NCBI database, and the -9 to -1 nucleotides were compared. This panel was then refined to 75 genes with specific retinal functions, for which the -9 to -1 nucleotides were placed in front of a GFP transcript sequence and RNAfold predictions performed. These were compared with a GFP sequence with the classic Kozak consensus (GCCGCCACC), and sequences from retinal genes with minimum free energy (MFE) predictions greater than the reference sequence were selected to generate an optimized Kozak consensus sequence. The original Kozak consensus and the refined retina Kozak consensus were placed upstream of the Renilla luciferase coding sequence, which were used to transfect retinoblastoma cell lines Y-79 and WERI-RB-1 and HEK 293T/17 cells. Results: The nucleotide frequencies of the original panel of genes were determined to be comparable to the classic Kozak consensus. RNAfold analysis of a GFP transcript with the classic Kozak sequence in the 5' untranslated region (UTR) generated an MFE prediction of -503.3 kcal/mol. RNAfold analysis was then performed with a GFP transcript containing each -9 to -1 Kozak sequence of 75 retinal genes. Thirty-eight of the 75 genes provided a greater MFE value than -503.3 kcal/mol and exhibited an absence of stable secondary structures before the AUG codon. The -9 to -1 nucleotide frequencies of these genes identified a Kozak consensus of ACCGAGACC, differing from the classic Kozak consensus at positions -9, -5, and -4. Applying this sequence to the GFP transcript increased the MFE prediction to -500.1 kcal/mol. The newly identified retina Kozak sequence was also applied to Renilla luciferase plus the REP1 and RPGR transcripts used in current clinical trials. In all examples, the predicted transcript MFE score increased when compared with the current transcript sequences containing classic Kozak consensus sequences. In vitro transfections identified a 7%-9% increase in Renilla activity when incorporating the optimized Kozak sequence. Conclusions: The Kozak consensus is a critical element of eukaryotic genes; therefore, it is a required feature of gene therapy transgenes. To date, the classic sequence of GCCRCC (-6 to -1) has typically been incorporated in gene therapy transgenes, but the analysis described here suggests that, for vectors targeting the retina, using a Kozak consensus derived from retinal genes can provide increased expression of the target product.


Assuntos
Regiões 5' não Traduzidas/genética , Códon de Iniciação/genética , Terapia Genética , RNA Mensageiro/genética , Doenças Retinianas/genética , Sequência Consenso , Bases de Dados Factuais , Vetores Genéticos , Humanos , Doenças Retinianas/terapia , Transfecção , Transgenes/genética
9.
Exp Eye Res ; 208: 108603, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33964272

RESUMO

PURPOSE: To validate the Cdhr1-/- mouse as a model for human CDHR1-associated retinal degeneration, which may present as cone-rod dystrophy or geographic atrophy. METHODS: Deep phenotyping of Cdhr1-/-(n = 56) and C57BL6J wildtype control mice (n = 45) was undertaken using in vivo multimodal retinal imaging and dark- and light-adapted electroretinography (ERG) over 15 months to evaluate rod- and cone-photoreceptor responses and retinal morphology. RESULTS: Cdhr1-/- retinas exhibited outer retinal thinning on optical coherence tomography (OCT) at 1-month versus C57BL6J (mean 14.6% reduction; P < 0.0001), with progressive degeneration to 15 months. The OCT layer representing photoreceptor outer segments was more significantly shortened in Cdhr1-/- eyes at 1 month (mean 33.7% reduction; P < 0.0001), remained stable to 3 months and was not identifiable at later timepoints. Outer retinal thinning was more pronounced at inferior versus superior retinal locations in Cdhr1-/- eyes (P < 0.002 at 3-9 months). Dark-adapted ERG identified severe functional deficits in Cdhr1-/- mice at 1 month of age versus C57BL6J (mean 62% reduction) that continued to decline to 15 months (P < 0.0001). Light-adapted flicker identified severe deficits in cone function at 1 month (mean 70% reduction), with improved function to 3 months followed by progressive decline (P < 0.0001). CONCLUSIONS: The Cdhr1-/- mouse exhibits structural and functional evidence of progressive outer retinal degeneration at a slow rate. Early functional deficits affecting both rod and cone photoreceptors in the context of relatively mild structural changes reflect the human phenotype. This study validates the use of the Cdhr1-/- mouse for the pre-clinical evaluation of therapeutics for human CDHR1-associated retinal degeneration.


Assuntos
Caderinas/genética , DNA/genética , Mutação , Proteínas do Tecido Nervoso/genética , Degeneração Retiniana/genética , Animais , Proteínas Relacionadas a Caderinas , Caderinas/biossíntese , Análise Mutacional de DNA , Modelos Animais de Doenças , Eletrorretinografia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Fenótipo , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/diagnóstico , Degeneração Retiniana/metabolismo , Tomografia de Coerência Óptica/métodos
10.
Mol Ther ; 28(3): 830-844, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32027843

RESUMO

The authors describe retinal reconstruction and restoration of visual function in heritably blind mice missing the rhodopsin gene using a novel method of ex vivo gene therapy and cell transplantation. Photoreceptor precursors with the same chromosomal genetic mutation were treated ex vivo using minicircle DNA, a non-viral technique that does not present the packaging limitations of adeno-associated virus (AAV) vectors. Following transplantation, genetically modified cells reconstructed a functional retina and supported vision in blind mice harboring the same founder gene mutation. Gene delivery by minicircles showed comparable long-term efficiency to AAV in delivering the missing gene, representing the first non-viral system for robust treatment of photoreceptors. This important proof-of-concept finding provides an innovative convergence of cell and gene therapies for the treatment of hereditary neurodegenerative disease and may be applied in future studies toward ex vivo correction of patient-specific cells to provide an autologous source of tissue to replace lost photoreceptors in inherited retinal blindness. This is the first report using minicircles in photoreceptor progenitors and the first to transplant corrected photoreceptor precursors to restore vision in blind animals.


Assuntos
DNA/administração & dosagem , Terapia Genética , Células-Tronco Neurais/metabolismo , Células Fotorreceptoras/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Transplante de Células-Tronco , Animais , Diferenciação Celular , Células Cultivadas , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Ordem dos Genes , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Camundongos , Camundongos Knockout , Plasmídeos/genética , Rodopsina/genética , Transplante de Células-Tronco/métodos , Transdução Genética , Transgenes
11.
Proc Natl Acad Sci U S A ; 114(42): 11211-11216, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-28973921

RESUMO

Optogenetic strategies to restore vision in patients who are blind from end-stage retinal degenerations aim to render remaining retinal cells light sensitive once photoreceptors are lost. Here, we assessed long-term functional outcomes following subretinal delivery of the human melanopsin gene (OPN4) in the rd1 mouse model of retinal degeneration using an adeno-associated viral vector. Ectopic expression of OPN4 using a ubiquitous promoter resulted in cellular depolarization and ganglion cell action potential firing. Restoration of the pupil light reflex, behavioral light avoidance, and the ability to perform a task requiring basic image recognition were restored up to 13 mo following injection. These data suggest that melanopsin gene therapy via a subretinal route may be a viable and stable therapeutic option for the treatment of end-stage retinal degeneration in humans.


Assuntos
Terapia Genética/métodos , Degeneração Retiniana/terapia , Opsinas de Bastonetes/genética , Animais , Dependovirus , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C3H , Visão Ocular
12.
Int J Mol Sci ; 21(17)2020 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-32872311

RESUMO

Many genetic diseases and undesirable traits are due to base-pair alterations in genomic DNA. Base-editing, the newest evolution of clustered regularly interspaced short palindromic repeats (CRISPR)-Cas-based technologies, can directly install point-mutations in cellular DNA without inducing a double-strand DNA break (DSB). Two classes of DNA base-editors have been described thus far, cytosine base-editors (CBEs) and adenine base-editors (ABEs). Recently, prime-editing (PE) has further expanded the CRISPR-base-edit toolkit to all twelve possible transition and transversion mutations, as well as small insertion or deletion mutations. Safe and efficient delivery of editing systems to target cells is one of the most paramount and challenging components for the therapeutic success of BEs. Due to its broad tropism, well-studied serotypes, and reduced immunogenicity, adeno-associated vector (AAV) has emerged as the leading platform for viral delivery of genome editing agents, including DNA-base-editors. In this review, we describe the development of various base-editors, assess their technical advantages and limitations, and discuss their therapeutic potential to treat debilitating human diseases.


Assuntos
Dependovirus/genética , Edição de Genes/métodos , RNA Guia de Cinetoplastídeos/genética , Sistemas CRISPR-Cas , Predisposição Genética para Doença , Humanos , Mutação Puntual , Tropismo
13.
Int J Mol Sci ; 21(7)2020 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-32230903

RESUMO

The treatment of dominantly inherited retinal diseases requires silencing of the pathogenic allele. RNA interference to suppress gene expression suffers from wide-spread off-target effects, while CRISPR-mediated gene disruption creates permanent changes in the genome. CRISPR interference uses a catalytically inactive 'dead' Cas9 directed by a guide RNA to block transcription of chosen genes without disrupting the DNA. It is highly specific and potentially reversible, increasing its safety profile as a therapy. Pre-clinical studies have demonstrated the versatility of CRISPR interference for gene silencing both in vivo and in ex vivo modification of iPSCs for transplantation. Applying CRISPR interference techniques for the treatment of autosomal dominant inherited retinal diseases is promising but there are few in vivo studies to date. This review details how CRISPR interference might be used to treat retinal diseases and addresses potential challenges for clinical translation.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Doenças Retinianas/genética , Doenças Retinianas/metabolismo , Alelos , Animais , Sistemas CRISPR-Cas , Expressão Gênica , Técnicas de Silenciamento de Genes , Inativação Gênica , Terapia Genética , Humanos , Células-Tronco Pluripotentes Induzidas , Interferência de RNA , RNA Guia de Cinetoplastídeos/metabolismo , Doenças Retinianas/terapia , Transcrição Gênica
14.
Int J Mol Sci ; 21(3)2020 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-31991730

RESUMO

RNA editing aims to treat genetic disease through altering gene expression at the transcript level. Pairing site-directed RNA-targeting mechanisms with engineered deaminase enzymes allows for the programmable correction of G>A and T>C mutations in RNA. This offers a promising therapeutic approach for a range of genetic diseases. For inherited retinal degenerations caused by point mutations in large genes not amenable to single-adeno-associated viral (AAV) gene therapy such as USH2A and ABCA4, correcting RNA offers an alternative to gene replacement. Genome editing of RNA rather than DNA may offer an improved safety profile, due to the transient and potentially reversible nature of edits made to RNA. This review considers the current site-directing RNA editing systems, and the potential to translate these to the clinic for the treatment of inherited retinal degeneration.


Assuntos
Edição de Genes , Terapia Genética , Edição de RNA , Retina/metabolismo , Transgenes , Adenosina Desaminase/metabolismo , Animais , Sistemas CRISPR-Cas , Imunofluorescência , Marcação de Genes , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Humanos , Proteínas de Ligação a RNA/metabolismo , Retina/patologia , Doenças Retinianas/genética , Doenças Retinianas/terapia
15.
Int J Mol Sci ; 21(14)2020 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-32674481

RESUMO

Effective treatment of retinal diseases with adeno-associated virus (AAV)-mediated gene therapy is highly dependent on the proportion of successfully transduced cells. However, due to inflammatory reactions at high vector doses, adjunctive treatment may be necessary to enhance the therapeutic outcome. Hydroxychloroquine and chloroquine are anti-malarial drugs that have been successfully used in the treatment of autoimmune diseases. Evidence suggests that at high concentrations, hydroxychloroquine and chloroquine can impact viral infection and replication by increasing endosomal and lysosomal pH. This effect has led to investigations into the potential benefits of these drugs in the treatment of viral infections, including human immunodeficiency virus and severe acute respiratory syndrome coronavirus-2. However, at lower concentrations, hydroxychloroquine and chloroquine appear to exert immunomodulatory effects by inhibiting nucleic acid sensors, including toll-like receptor 9 and cyclic GMP-AMP synthase. This dose-dependent effect on their mechanism of action supports observations of increased viral infections associated with lower drug doses. In this review, we explore the immunomodulatory activity of hydroxychloroquine and chloroquine, their impact on viral infections, and their potential to improve the efficacy and safety of retinal gene therapy by reducing AAV-induced immune responses. The safety and practicalities of delivering hydroxychloroquine into the retina will also be discussed.


Assuntos
Cloroquina/uso terapêutico , Terapia Genética , Hidroxicloroquina/uso terapêutico , Doenças Retinianas/terapia , Viroses/tratamento farmacológico , Animais , Betacoronavirus/efeitos dos fármacos , Cloroquina/farmacologia , Dependovirus/genética , Humanos , Hidroxicloroquina/farmacologia , Imunomodulação/efeitos dos fármacos , Doenças Retinianas/patologia , SARS-CoV-2
16.
Int J Mol Sci ; 21(17)2020 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-32842706

RESUMO

Retinitis pigmentosa (RP) is a generic term for a group of genetic diseases characterized by loss of rod and cone photoreceptor cells. Although the genetic causes of RP frequently only affect the rod photoreceptor cells, cone photoreceptors become stressed in the absence of rods and undergo a secondary degeneration. Changes in the gene expression profile of cone photoreceptor cells are likely to occur prior to observable physiological changes. To this end, we sought to achieve greater understanding of the changes in cone photoreceptor cells early in the degeneration process of the Rho-/- mouse model. To account for gene expression changes attributed to loss of cone photoreceptor cells, we normalized PCR in the remaining number of cones to a cone cell reporter (OPN1-GFP). Gene expression profiles of key components involved in the cone phototransduction cascade were correlated with tests of retinal cone function prior to cell loss. A significant downregulation of the photoreceptor transcription factor Crx was observed, which preceded a significant downregulation in cone opsin transcripts that coincided with declining cone function. Our data add to the growing understanding of molecular changes that occur prior to cone dysfunction in a model of rod-cone dystrophy. It is of interest that gene supplementation of CRX by adeno-associated viral vector delivery prior to cone cell loss did not prevent cone photoreceptor degeneration in this mouse model.


Assuntos
Distrofias de Cones e Bastonetes/genética , Distrofias de Cones e Bastonetes/fisiopatologia , Animais , Distrofias de Cones e Bastonetes/terapia , Modelos Animais de Doenças , Eletrorretinografia , Regulação da Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Proteínas de Fluorescência Verde/genética , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/farmacologia , Humanos , Camundongos Transgênicos , Oftalmoscopia , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Rodopsina/genética , Opsinas de Bastonetes/genética , Tomografia de Coerência Óptica , Transativadores/genética , Transativadores/farmacologia , Visão Ocular/genética
17.
Mol Ther ; 25(8): 1854-1865, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28549772

RESUMO

X-linked retinitis pigmentosa (XLRP) is generally a severe form of retinitis pigmentosa, a neurodegenerative, blinding disorder of the retina. 70% of XLRP cases are due to mutations in the retina-specific isoform of the gene encoding retinitis pigmentosa GTPase regulator (RPGRORF15). Despite successful RPGRORF15 gene replacement with adeno-associated viral (AAV) vectors being established in a number of animal models of XLRP, progression to human trials has not yet been possible. The inherent sequence instability in the purine-rich region of RPGRORF15 (which contains highly repetitive nucleotide sequences) leads to unpredictable recombination errors during viral vector cloning. While deleted RPGR may show some efficacy in animal models, which have milder disease, the therapeutic effect of a mutated RPGR variant in patients with XLRP cannot be predicted. Here, we describe an optimized gene replacement therapy for human XLRP disease using an AAV8 vector that reliably and consistently produces the full-length correct RPGR protein. The glutamylation pattern in the RPGR protein derived from the codon-optimized sequence is indistinguishable from the wild-type variant, implying that codon optimization does not significantly alter post-translational modification. The codon-optimized sequence has superior stability and expression levels in vitro. Significantly, when delivered by AAV8 vector and driven by the rhodopsin kinase promoter, the codon-optimized RPGR rescues the disease phenotype in two relevant animal models (Rpgr-/y and C57BL/6JRd9/Boc) and shows good safety in C57BL6/J wild-type mice. This work provides the basis for clinical trial development to treat patients with XLRP caused by RPGR mutations.


Assuntos
Proteínas de Transporte/genética , Códon , Dependovirus/genética , Proteínas do Olho/genética , Genes Ligados ao Cromossomo X , Terapia Genética , Vetores Genéticos/genética , Retinose Pigmentar/genética , Animais , Modelos Animais de Doenças , Expressão Gênica , Camundongos , Mutação , Fenótipo , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Estabilidade de RNA , Retinose Pigmentar/diagnóstico , Retinose Pigmentar/terapia , Transdução Genética , Transgenes
18.
Yale J Biol Med ; 90(4): 611-623, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29259525

RESUMO

The use of adeno-associated viral (AAV) vectors for gene therapy treatments of inherited disorders has accelerated over the past decade with multiple clinical trials ongoing in varying tissue types and new ones initiating every year. These vectors are exhibiting low-immunogenicity across the clinical trials in addition to showing evidence of efficacy, making it clear they are the current standard vector for any potential gene therapy treatment. However, AAV vectors do have a limitation in their packaging capacity, being capable of holding no more than ~5kb of DNA and in a therapeutic transgene scenario, this length of DNA would need to include genetic control elements in addition to the gene coding sequence (CDS) of interest. Given that numerous diseases are caused by mutations in genes with a CDS exceeding 3.5kb, this makes packaging into a single AAV capsid not possible for larger genes. Due to this problem, yet with the desire to use AAV vectors, research groups have adapted the standard AAV gene therapy approach to enable delivery of such large genes to target cells using dual AAV vector systems. Here we review the AAV dual vector strategies currently employed and highlight the virtues and drawbacks of each method plus the likelihood of success with such approaches.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Transgenes , Animais , DNA de Cadeia Simples , Humanos , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Sequências Repetidas Terminais
19.
Mol Diagn Ther ; 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38955952

RESUMO

Gene therapies have emerged as promising treatments in clinical development for various retinal disorders, offering hope to patients with inherited degenerative eye conditions. Several gene therapies have already shown remarkable success in clinical trials, with significant improvements observed in visual acuity and the preservation of retinal function. A multitude of gene therapies have now been delivered safely in human clinical trials for a wide range of inherited retinal disorders but there are some gaps in the reported trial data. Some of the most exciting treatment options are not under peer review and information is only available in press release form. Whilst many trials appear to have delivered good outcomes of safety, others have failed to meet primary endpoints and therefore not proceeded to phase III. Despite this, such trials have enabled researchers to learn how best to assess and monitor patient outcomes, which will guide future trials to greater success. In this review, we consider recent and ongoing clinical trials for a variety of potential retinal gene therapy treatments and discuss the positive and negative issues related to these trials. We discuss the treatment potential following clinical trials as well as the potential risks of some treatments under investigation. As these therapies continue to advance through rigorous testing and regulatory approval processes, they hold the potential to revolutionise the landscape of retinal disorder treatments, providing renewed vision and enhancing the quality of life for countless individuals worldwide.

20.
Cells ; 12(3)2023 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-36766782

RESUMO

Inherited retinal diseases (IRD) are a clinically and genetically heterogenous group of diseases and a leading cause of blindness in the working-age population. Even though gene augmentation therapies have shown promising results, they are only feasible to treat a small number of autosomal recessive IRDs, because the size of the gene is limited by the vector used. DNA editing however could potentially correct errors regardless of the overall size of the gene and might also be used to correct dominant mutations. Prime editing is a novel CRISPR/Cas9 based gene editing tool that enables precise correction of point mutations, insertions, and deletions without causing double strand DNA breaks. Due to its versatility and precision this technology may be a potential treatment option for virtually all genetic causes of IRD. Since its initial description, the prime editing technology has been further improved, resulting in higher efficacy and a larger target scope. Additionally, progress has been achieved concerning the size-related delivery issue of the prime editor components. This review aims to give an overview of these recent advancements and discusses prime editing as a potential treatment for IRDs.


Assuntos
Sistemas CRISPR-Cas , Doenças Retinianas , Humanos , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Doenças Retinianas/genética , Doenças Retinianas/terapia , Terapia Genética/métodos , Mutação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA