Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 625
1.
Klin Monbl Augenheilkd ; 241(3): 259-265, 2024 Mar.
Article En | MEDLINE | ID: mdl-38508214

Retinal dystrophies linked to the RPE65 gene are mostly fast-progressing retinal diseases, with childhood onset of night blindness and progressive visual loss up to the middle adult age. Rare phenotypes linked to this gene are known with congenital stationary night blindness or slowly progressing retinitis pigmentosa, as well as an autosomal dominant c.1430A>G (p.Asp477Gly) variant. This review gives an overview of the current knowledge of the clinical phenotypes, as well as experience with the efficacy and safety of the approved gene augmentation therapy voretigene neparvovec.


Night Blindness , Retinal Dystrophies , Retinitis Pigmentosa , Adult , Child , Humans , cis-trans-Isomerases/genetics , Genetic Therapy , Mutation , Night Blindness/therapy , Phenotype , Retinal Dystrophies/diagnosis , Retinal Dystrophies/genetics , Retinal Dystrophies/therapy , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy
2.
Cell Rep Med ; 5(4): 101459, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38518771

Retinitis pigmentosa (RP) is one of the most common forms of hereditary neurodegeneration. It is caused by one or more of at least 3,100 mutations in over 80 genes that are primarily expressed in rod photoreceptors. In RP, the primary rod-death phase is followed by cone death, regardless of the underlying gene mutation that drove the initial rod degeneration. Dampening the oxidation of glycolytic end products in rod mitochondria enhances cone survival in divergent etiological disease models independent of the underlying rod-specific gene mutations. Therapeutic editing of the prolyl hydroxylase domain-containing protein gene (PHD2, also known as Egln1) in rod photoreceptors led to the sustained survival of both diseased rods and cones in both preclinical autosomal-recessive and dominant RP models. Adeno-associated virus-mediated CRISPR-based therapeutic reprogramming of the aerobic glycolysis node may serve as a gene-agnostic treatment for patients with various forms of RP.


Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa , Animals , Humans , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/therapy , Retinal Cone Photoreceptor Cells/metabolism , Disease Models, Animal
3.
EMBO Mol Med ; 16(4): 805-822, 2024 Apr.
Article En | MEDLINE | ID: mdl-38504136

For 15 years, gene therapy has been viewed as a beacon of hope for inherited retinal diseases. Many preclinical investigations have centered around vectors with maximal gene expression capabilities, yet despite efficient gene transfer, minimal physiological improvements have been observed in various ciliopathies. Retinitis pigmentosa-type 28 (RP28) is the consequence of bi-allelic null mutations in the FAM161A, an essential protein for the structure of the photoreceptor connecting cilium (CC). In its absence, cilia become disorganized, leading to outer segment collapses and vision impairment. Within the human retina, FAM161A has two isoforms: the long one with exon 4, and the short one without it. To restore CC in Fam161a-deficient mice shortly after the onset of cilium disorganization, we compared AAV vectors with varying promoter activities, doses, and human isoforms. While all vectors improved cell survival, only the combination of both isoforms using the weak FCBR1-F0.4 promoter enabled precise FAM161A expression in the CC and enhanced retinal function. Our investigation into FAM161A gene replacement for RP28 emphasizes the importance of precise therapeutic gene regulation, appropriate vector dosing, and delivery of both isoforms. This precision is pivotal for secure gene therapy involving structural proteins like FAM161A.


Retinitis Pigmentosa , Animals , Mice , Humans , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retina/metabolism , Exons , Protein Isoforms/genetics , Protein Isoforms/metabolism , Genetic Therapy , Eye Proteins/genetics , Eye Proteins/chemistry , Eye Proteins/metabolism
4.
Neurobiol Dis ; 193: 106436, 2024 Apr.
Article En | MEDLINE | ID: mdl-38341159

Retinitis pigmentosa (RP) is a degenerative disease, caused by genetic mutations that lead to a loss in photoreceptors. For research on RP, rd10 mice, which carry mutations in the phosphodiesterase (PDE) gene, exhibit degenerative patterns comparable to those of patients with RP, making them an ideal model for investigating potential treatments. Although numerous studies have reported the potential of biochemical drugs, gene correction, and stem cell transplantation in decelerating rd10 retinal degeneration, a comprehensive review of these studies has yet to be conducted. Therefore, here, a comparative analysis of rd10 mouse treatment research over the past decade was performed. Our findings suggest that biochemical drugs capable of inhibiting the inflammatory response may be promising therapeutics. Additionally, significant progress has been made in the field of gene therapy; nevertheless, challenges such as strict delivery requirements, bystander editing, and off-target effects still need to be resolved. Nevertheless, secretory function is the only unequivocal protective effect of stem cell transplantation. In summary, this review presents a comprehensive analysis and synthesis of the treatment approaches employing rd10 mice as experimental subjects, describing a clear pathway for future RP treatment research and identifies potential clinical interventions.


Retinal Degeneration , Retinitis Pigmentosa , Mice , Humans , Animals , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retinal Degeneration/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Retina/metabolism
5.
Hum Gene Ther ; 35(5-6): 151-162, 2024 Mar.
Article En | MEDLINE | ID: mdl-38368562

Mutations in the rhodopsin (RHO) gene are the predominant causes of autosomal dominant retinitis pigmentosa (adRP). Given the diverse gain-of-function mutations, therapeutic strategies targeting specific sequences face significant challenges. Here, we provide a universal approach to conquer this problem: we have devised a CRISPR-Cas12i-based, mutation-independent gene knockout and replacement compound therapy carried by a dual AAV2/8 system. In this study, we successfully delayed the progression of retinal degeneration in the classic mouse disease model RhoP23H, and also RhoP347S, a new native mouse mutation model we developed. Our research expands the horizon of potential options for future treatments of RHO-mediated adRP.


Retinal Degeneration , Retinitis Pigmentosa , Mice , Animals , Rhodopsin/genetics , Mice, Knockout , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Mutation , Genes, Dominant
6.
Bull Math Biol ; 86(3): 29, 2024 02 12.
Article En | MEDLINE | ID: mdl-38345678

Both the rod and cone photoreceptors, along with the retinal pigment epithelium have been experimentally and mathematically shown to work interdependently to maintain vision. Further, the theoredoxin-like rod-derived cone viability factor (RdCVF) and its long form (RdCVFL) have proven to increase photoreceptor survival in experimental results. Aerobic glycolysis is the primary source of energy production for photoreceptors and RdCVF accelerates the intake of glucose into the cones. RdCVFL helps mitigate the negative effects of reactive oxidative species and has shown promise in slowing the death of cones in mouse studies. However, this potential treatment and its effects have never been studied in mathematical models. In this work, we examine an optimal control with the treatment of RdCVFL. We mathematically illustrate the potential this treatment might have for treating degenerative retinal diseases such as retinitis pigmentosa, as well as compare this to the results of an updated control model with RdCVF.


Models, Biological , Retinitis Pigmentosa , Animals , Mice , Mathematical Concepts , Retina , Retinal Cone Photoreceptor Cells , Retinitis Pigmentosa/therapy
8.
Int J Mol Sci ; 25(3)2024 Feb 02.
Article En | MEDLINE | ID: mdl-38339118

Variants within the Retinitis Pigmentosa GTPase regulator (RPGR) gene are the predominant cause of X-Linked Retinitis Pigmentosa (XLRP), a common and severe form of inherited retinal disease. XLRP is characterised by the progressive degeneration and loss of photoreceptors, leading to visual loss and, ultimately, bilateral blindness. Unfortunately, there are no effective approved treatments for RPGR-associated XLRP. We sought to investigate the efficacy of RPGRORF15 gene supplementation using a clinically relevant construct in human RPGR-deficient retinal organoids (ROs). Isogenic RPGR knockout (KO)-induced pluripotent stem cells (IPSCs) were generated using established CRISPR/Cas9 gene editing methods targeting RPGR. RPGR-KO and isogenic wild-type IPSCs were differentiated into ROs and utilised to test the adeno associated virus (AAV) RPGR (AAV-RPGR) clinical vector construct. The transduction of RPGR-KO ROs using AAV-RPGR successfully restored RPGR mRNA and protein expression and localisation to the photoreceptor connecting cilium in rod and cone photoreceptors. Vector-derived RPGR demonstrated equivalent levels of glutamylation to WT ROs. In addition, treatment with AAV-RPGR restored rhodopsin localisation within RPGR-KO ROs, reducing mislocalisation to the photoreceptor outer nuclear layer. These data provide mechanistic insights into RPGRORF15 gene supplementation functional potency in human photoreceptor cells and support the previously reported Phase I/II trial positive results using this vector construct in patients with RPGR-associated XLRP, which is currently being tested in a Phase III clinical trial.


Opsins , Retinitis Pigmentosa , Humans , Opsins/genetics , Dependovirus/genetics , Dependovirus/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Reactive Oxygen Species/metabolism , Carrier Proteins/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Genetic Therapy/methods , Mutation
9.
Stem Cell Reports ; 19(2): 254-269, 2024 Feb 13.
Article En | MEDLINE | ID: mdl-38181785

Pluripotent stem cell-based therapy for retinal degenerative diseases is a promising approach to restoring visual function. A clinical study using retinal organoid (RO) sheets was recently conducted in patients with retinitis pigmentosa. However, the graft preparation currently requires advanced skills to identify and excise suitable segments from the transplantable area of the limited number of suitable ROs. This remains a challenge for consistent clinical implementations. Herein, we enabled the enrichment of wild-type (non-reporter) retinal progenitor cells (RPCs) from dissociated ROs using a label-free ghost cytometry (LF-GC)-based sorting system, where a machine-based classifier was trained in advance with another RPC reporter line. The sorted cells reproducibly formed retinal spheroids large enough for transplantation and developed mature photoreceptors in the retinal degeneration rats. This method of enriching early RPCs with no specific surface antigens and without any reporters or chemical labeling is promising for robust preparation of graft tissues during cell-based therapy.


Pluripotent Stem Cells , Retinal Degeneration , Retinitis Pigmentosa , Humans , Animals , Rats , Reactive Oxygen Species , Retina , Pluripotent Stem Cells/transplantation , Retinal Degeneration/therapy , Retinitis Pigmentosa/therapy , Stem Cell Transplantation/methods
10.
Medicina (Kaunas) ; 60(1)2024 Jan 22.
Article En | MEDLINE | ID: mdl-38276069

Retinitis pigmentosa is an inherited disease, in which mutations in different types of genes lead to the death of photoreceptors and the loss of visual function. Although retinitis pigmentosa is the most common type of inherited retinal dystrophy, a clear line of therapy has not yet been defined. In this review, we will focus on the therapeutic aspect and attempt to define the advantages and disadvantages of the protocols of different therapies. The role of some therapies, such as antioxidant agents or gene therapy, has been established for years now. Many clinical trials on different genes and mutations causing RP have been conducted, and the approval of voretigene nepavorec by the FDA has been an important step forward. Nonetheless, even if gene therapy is the most promising type of treatment for these patients, other innovative strategies, such as stem cell transplantation or hyperbaric oxygen therapy, have been shown to be safe and improve visual quality during clinical trials. The treatment of this disease remains a challenge, to which we hope to find a solution as soon as possible.


Hematopoietic Stem Cell Transplantation , Hyperbaric Oxygenation , Retinitis Pigmentosa , Humans , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/genetics , Stem Cell Transplantation , Genetic Therapy
11.
Trials ; 25(1): 89, 2024 Jan 27.
Article En | MEDLINE | ID: mdl-38279157

BACKGROUND: Retinitis pigmentosa (RP) is an inherited disease characterized by a progressive loss of rod photoreceptors of the eye, leading to irreversible blindness. To date, to our knowledge, no clinical prospective studies have been undertaken that could document the effect of interventions that could reverse or reduce the progression of this disease. The application of microcurrent stimulation (ES) of the eye in the treatment of chronic eye diseases such as glaucoma and age-related macular degeneration has been used over several decades and has been reported to have beneficial effects to reduce the progression of these blinding diseases and has been supported by animal studies and smaller clinical studies, but to date, no large randomized clinical trials on the use of microcurrent therapy have been published. More recent clinical reports have also shown beneficial effects of ES on slowing the progression of RP but also lacks data from robust prospective clinical outcome studies. To our knowledge, this is the first prospective randomized study to evaluate the safety and clinical effectiveness of transpalpebral electrical stimulation (TpES) on the progression of RP. METHODS: Randomized prospective study using N-of-1 trial 3 single-blind, crossover comparisons. The intervention period of each comparison is divided into treatment period and control period which are randomized arranged. Twelve participants will be strictly recruited in N-of-1 trial by the researcher in accordance with the inclusion and exclusion criteria. The main outcome of interest examined after each cycle of the 8-week intervention period is the assessment of the visual field (VF). Other variables of interest are best corrected visual acuity (BCVA), retinal function using electroretinogram (ERG), and visual function using NEI VFQ-25 questionnaire. Objective assessments of retinal changes will be undertaken using optical coherence tomography (OCT) and fundus autofluorescence (FAF). DISCUSSION: The trial will evaluate the efficacy and safety of microcurrent stimulation on RP and provide high-quality evidence for clinical application through N-of-1 trial. TRIAL REGISTRATION: Chinese Clinical Trial Registry; ChiCTR2300067357; https://www.chictr.org.cn/showproj.html?proj=174635 . Registered on 5 January 2023.


Retinitis Pigmentosa , Humans , Prospective Studies , Single-Blind Method , Visual Acuity , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/therapy , Blindness , Electric Stimulation/methods , Randomized Controlled Trials as Topic
12.
Article En | MEDLINE | ID: mdl-37037600

The underlying immune state of inherited retinal degenerations (IRDs) and retinitis pigmentosa (RP) has been an emerging area of interest, wherein the consequences have never been greater given the widespread recognition of gene therapy-associated uveitis (GTU) in gene therapy clinical trials. Whereas some evidence suggests that the adaptive immune system may play a role, the majority of studies indicate that the innate immune system is likely the primary driver of neuroinflammation in RP. During retinal degeneration, discrete mechanisms activate resident microglia and promote infiltrating macrophages that can either be protective or detrimental to photoreceptor cell death. This persistent stimulation of innate immunity, overlaid by the introduction of viral antigens as part of gene therapy, has the potential to trigger a complex microglia/macrophage-driven proinflammatory state. A better understanding of the immune pathophysiology in IRD and GTU will be necessary to improve the success of developing novel treatments for IRDs.


Retinal Degeneration , Retinitis Pigmentosa , Uveitis , Humans , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Macrophages , Genetic Therapy , Uveitis/genetics , Uveitis/therapy
13.
Article En | MEDLINE | ID: mdl-37217283

Naturally occurring inherited retinal diseases (IRDs) in cats and dogs provide a rich source of potential models for human IRDs. In many cases, the phenotypes between the species with mutations of the homologous genes are very similar. Both cats and dogs have a high-acuity retinal region, the area centralis, an equivalent to the human macula, with tightly packed photoreceptors and higher cone density. This and the similarity in globe size to that of humans means these large animal models provide information not obtainable from rodent models. The established cat and dog models include those for Leber congenital amaurosis, retinitis pigmentosa (including recessive, dominant, and X-linked forms), achromatopsia, Best disease, congenital stationary night blindness and other synaptic dysfunctions, RDH5-associated retinopathy, and Stargardt disease. Several of these models have proven to be important in the development of translational therapies such as gene-augmentation therapies. Advances have been made in editing the canine genome, which necessitated overcoming challenges presented by the specifics of canine reproduction. Feline genome editing presents fewer challenges. We can anticipate the generation of specific cat and dog IRD models by genome editing in the future.


Cat Diseases , Dog Diseases , Eye Diseases, Hereditary , Retinal Diseases , Retinitis Pigmentosa , Animals , Dogs , Cats , Humans , Cat Diseases/genetics , Dog Diseases/genetics , Eye Diseases, Hereditary/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Proteins , Mutation
14.
Cell Stem Cell ; 30(12): 1585-1596.e6, 2023 12 07.
Article En | MEDLINE | ID: mdl-38065067

Transplantation of induced pluripotent stem cell (iPSC)-derived retinal organoids into retinal disease animal models has yielded promising results, and several clinical trials on iPSC-derived retinal pigment epithelial cell transplantation have confirmed its safety. In this study, we performed allogeneic iPSC-derived retinal organoid sheet transplantation in two subjects with advanced retinitis pigmentosa (jRCTa050200027). The primary endpoint was the survival and safety of the transplanted retinal organoid sheets in the first year post-transplantation. The secondary endpoints were the safety of the transplantation procedure and visual function evaluation. The grafts survived in a stable condition for 2 years, and the retinal thickness increased at the transplant site without serious adverse events in both subjects. Changes in visual function were less progressive than those of the untreated eye during the follow-up. Allogeneic iPSC-derived retinal organoid sheet transplantation is a potential therapeutic approach, and the treatment's safety and efficacy for visual function should be investigated further.


Induced Pluripotent Stem Cells , Retinitis Pigmentosa , Animals , Humans , Retina , Retinitis Pigmentosa/therapy , Vision, Ocular , Organoids
15.
Nat Commun ; 14(1): 8256, 2023 Dec 12.
Article En | MEDLINE | ID: mdl-38086857

Retinitis pigmentosa is an inherited photoreceptor degeneration that begins with rod loss followed by cone loss. This cell loss greatly diminishes vision, with most patients becoming legally blind. Gene therapies are being developed, but it is unknown how retinal function depends on the time of intervention. To uncover this dependence, we utilize a mouse model of retinitis pigmentosa capable of artificial genetic rescue. This model enables a benchmark of best-case gene therapy by removing variables that complicate answering this question. Complete genetic rescue was performed at 25%, 50%, and 70% rod loss (early, mid and late, respectively). Early and mid treatment restore retinal output to near wild-type levels. Late treatment retinas exhibit continued, albeit slowed, loss of sensitivity and signal fidelity among retinal ganglion cells, as well as persistent gliosis. We conclude that gene replacement therapies delivered after 50% rod loss are unlikely to restore visual function to normal. This is critical information for administering gene therapies to rescue vision.


Retinal Degeneration , Retinitis Pigmentosa , Mice , Animals , Humans , Retina , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinal Cone Photoreceptor Cells , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Genetic Therapy , Disease Models, Animal
16.
Zhonghua Yan Ke Za Zhi ; 59(12): 1058-1064, 2023 Dec 11.
Article Zh | MEDLINE | ID: mdl-38061908

Biallelic pathogenic variants in the USH2A gene result in Usher syndrome type Ⅱ and non-syndromic retinitis pigmentosa, both of which entail the progressive loss of photoreceptors leading to blindness. The cDNA of the USH2A gene is extensive, consisting of 15 606 base pairs, rendering it impractical for delivery via adeno-associated virus vectors for gene replacement therapy. Notably, exon 13 has emerged as a focal point for therapeutic intervention, given its predilection for harboring the most pathogenic variants within USH2A. Recent intervention studies targeting USH2A exon 13 through the utilization of antisense oligonucleotides, genome editing, and RNA editing approaches have exhibited promising therapeutic potential. This paper provides a comprehensive overview of the molecular mechanisms, outcome data, and the challenges associated with the application of these interventions in this domain.


Retinitis Pigmentosa , Usher Syndromes , Humans , Usher Syndromes/genetics , Usher Syndromes/therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Exons , Genetic Therapy , Extracellular Matrix Proteins/genetics , Mutation
17.
CRISPR J ; 6(6): 502-513, 2023 Dec.
Article En | MEDLINE | ID: mdl-38108516

Rhodopsin (RHO) mutations such as Pro23His are the leading cause of dominantly inherited retinitis pigmentosa in North America. As with other dominant retinal dystrophies, these mutations lead to production of a toxic protein product, and treatment will require knockdown of the mutant allele. The purpose of this study was to develop a CRISPR-Cas9-mediated transcriptional repression strategy using catalytically inactive Staphylococcus aureus Cas9 (dCas9) fused to the Krüppel-associated box (KRAB) transcriptional repressor domain. Using a reporter construct carrying green fluorescent protein (GFP) cloned downstream of the RHO promoter fragment (nucleotides -1403 to +73), we demonstrate a ∼74-84% reduction in RHO promoter activity in RHOpCRISPRi-treated versus plasmid-only controls. After subretinal transduction of human retinal explants and transgenic Pro23His mutant pigs, significant knockdown of rhodopsin protein was achieved. Suppression of mutant transgene in vivo was associated with a reduction in endoplasmic reticulum (ER) stress and apoptosis markers and preservation of photoreceptor cell layer thickness.


Retinitis Pigmentosa , Rhodopsin , Humans , Animals , Swine , Rhodopsin/genetics , CRISPR-Cas Systems/genetics , Gene Editing , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Alleles
18.
Stem Cell Res Ther ; 14(1): 340, 2023 11 27.
Article En | MEDLINE | ID: mdl-38012786

Inherited retinal diseases (IRDs) can induce severe sight-threatening retinal degeneration and impose a considerable economic burden on patients and society, making efforts to cure blindness imperative. Transgenic animals mimicking human genetic diseases have long been used as a primary research tool to decipher the underlying pathogenesis, but there are still some obvious limitations. As an alternative strategy, patient-derived induced pluripotent stem cells (iPSCs), particularly three-dimensional (3D) organoid technology, are considered a promising platform for modeling different forms of IRDs, including retinitis pigmentosa, Leber congenital amaurosis, X-linked recessive retinoschisis, Batten disease, achromatopsia, and best vitelliform macular dystrophy. Here, this paper focuses on the status of patient-derived iPSCs and organoids in IRDs in recent years concerning disease modeling and therapeutic exploration, along with potential challenges for translating laboratory research to clinical application. Finally, the importance of human iPSCs and organoids in combination with emerging technologies such as multi-omics integration analysis, 3D bioprinting, or microfluidic chip platform are highlighted. Patient-derived retinal organoids may be a preferred choice for more accurately uncovering the mechanisms of human retinal diseases and will contribute to clinical practice.


Induced Pluripotent Stem Cells , Retinal Degeneration , Retinitis Pigmentosa , Animals , Humans , Induced Pluripotent Stem Cells/pathology , Retina/pathology , Retinal Degeneration/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/pathology , Organoids
19.
Arch. Soc. Esp. Oftalmol ; 98(11): 646-655, nov. 2023. tab
Article Es | IBECS | ID: ibc-227204

La retinosis pigmentaria es un grupo de enfermedades de origen hereditario que produce la degeneración de la retina y una disminución de la visión. La Organización Mundial de la Salud reporta alrededor de 1.300 millones de personas afectadas por algún tipo de deficiencia visual a nivel mundial. La prevalencia es de 1 por cada 4.000 habitantes y es la primera causa de ceguera de origen genético, frecuente en los varones con un porcentaje del 60% y el 40% en mujeres. Existe falta de información sobre esta patología en el mundo, principalmente sobre los tratamientos existentes para esta enfermedad, por lo que esta revisión bibliográfica tiene como objetivo actualizar los tratamientos existentes o en estudio e informar de las limitaciones que tiene cada una de estas terapias. Se realizó esta revisión de literatura científica mediante la consulta en bases de datos como PubMed y Web of Sciense; la búsqueda se limitó a artículos de los años 2018 al 2022. Existen varios tipos de terapia en estudios: terapia génica, estimulación eléctrica transcorneal, uso de neuroprotectores, terapia optogénica, trasplantes de células madre y terapia con oligonucleótidos, de los que se hablará en este artículo, tanto de sus beneficios como de las barreras existentes en cada tratamiento experimental. En conclusión, cada una de estas terapias promete un tratamiento viable en el futuro para grupos selectivos de personas con retinosis pigmentaria; sin embargo, algunas terapias han demostrado beneficio al inicio de la enfermedad, perdiendo su eficacia a largo plazo (AU)


Retinitis pigmentosa is a group of inherited diseases that lead to degeneration of the retina and decreased vision. The World Health Organization reports around 1300 million people affected by some type of visual impairment worldwide. The prevalence is 1 in every 4000 inhabitants and it is the first cause of blindness of genetic origin, frequent in men with a percentage of 60% and 40% in women. There is a lack of information on this pathology in the world, mainly on the existing treatments for this disease, so this bibliographic review aims to update the existing or under-study treatments and inform the limitations of each of these therapies. This review of scientific literature was carried out by consulting databases such as PubMed and Web of science, the search will be limited to articles from the years 2018 to 2022. There are several types of therapy in studies: gene therapy, transcorneal electrical stimulation, use of neuroprotectors, optogenic therapy, stem cell transplants and oligonucleotide therapy, which will be discussed in this article, both their benefits and the existing barriers in each experimental treatment. In conclusion, each of these therapies promises a viable treatment in the future for selective groups of people with retinitis pigmentosa, however, some therapies have shown benefit at the beginning of the disease, losing their efficacy in the long term (AU)


Humans , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/genetics , Experimental Development
20.
Exp Biol Med (Maywood) ; 248(20): 1818-1828, 2023 10.
Article En | MEDLINE | ID: mdl-37837380

Rhodopsin (Rho) gene mutation was considered the highest prevalent mutation in autosomal dominant retinitis pigmentosa (ADRP); however, effective therapeutics for ADRP have not been developed. The process of gene editing via the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 system offers the potentiality to provide cures for dominantly inherited disorders. Herein, we generated a CRISPR/SaCas9-mediated gene reduction system to inactivate the Rho mutant, while replacing normal rhodopsin in a rhodopsin mutation mouse model. When Rho-P23H knock-in mice were administered a subretinal injection of the "reduction and replacement" system, the expression of mutant rhodopsin was reduced, and retinal function was improved. Therefore, we concluded that CRISPR/SaCas9-based "reduction and replacement" gene therapy could provide structural and functional benefits for Rho mutant ADRP, as well as new directions for future clinical research on the treatment of such gain-of-function genetic diseases.


Retinal Degeneration , Retinitis Pigmentosa , Mice , Animals , Rhodopsin/genetics , Gene Editing , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retina/metabolism , Mutation , Disease Models, Animal
...