Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.160
Filtrar
1.
Sci Rep ; 14(1): 10551, 2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719929

RESUMEN

Our purpose was to elucidate the genotype and ophthalmological and audiological phenotype in TUBB4B-associated inherited retinal dystrophy (IRD) and sensorineural hearing loss (SNHL), and to model the effects of all possible amino acid substitutions at the hotspot codons Arg390 and Arg391. Six patients from five families with heterozygous missense variants in TUBB4B were included in this observational study. Ophthalmological testing included best-corrected visual acuity, fundus examination, optical coherence tomography, fundus autofluorescence imaging, and full-field electroretinography (ERG). Audiological examination included pure-tone and speech audiometry in adult patients and auditory brainstem response testing in a child. Genetic testing was performed by disease gene panel analysis based on genome sequencing. The molecular consequences of the substitutions of residues 390 and 391 on TUBB4B and its interaction with α-tubulin were predicted in silico on its three-dimensional structure obtained by homology modelling. Two independent patients had amino acid exchanges at position 391 (p.(Arg391His) or p.(Arg391Cys)) of the TUBB4B protein. Both had a distinct IRD phenotype with peripheral round yellowish lesions with pigmented spots and mild or moderate SNHL, respectively. Yet the phenotype was milder with a sectorial pattern of bone spicules in one patient, likely due to a genetically confirmed mosaicism for p.(Arg391His). Three patients were heterozygous for an amino acid exchange at position 390 (p.(Arg390Gln) or p.(Arg390Trp)) and presented with another distinct retinal phenotype with well demarcated pericentral retinitis pigmentosa. All showed SNHL ranging from mild to severe. One additional patient showed a variant distinct from codon 390 or 391 (p.(Tyr310His)), and presented with congenital profound hearing loss and reduced responses in ERG. Variants at codon positions 390 and 391 were predicted to decrease the structural stability of TUBB4B and its complex with α-tubulin, as well as the complex affinity. In conclusion, the twofold larger reduction in heterodimer affinity exhibited by Arg391 substitutions suggested an association with the more severe retinal phenotype, compared to the substitution at Arg390.


Asunto(s)
Codón , Pérdida Auditiva Sensorineural , Fenotipo , Tubulina (Proteína) , Humanos , Femenino , Tubulina (Proteína)/genética , Tubulina (Proteína)/química , Masculino , Adulto , Pérdida Auditiva Sensorineural/genética , Codón/genética , Persona de Mediana Edad , Mutación Missense , Niño , Linaje , Adolescente , Sustitución de Aminoácidos , Adulto Joven , Retinitis Pigmentosa/genética
2.
Elife ; 122024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727583

RESUMEN

Retinitis pigmentosa (RP) is an inherited retinal disease in which there is a loss of cone-mediated daylight vision. As there are >100 disease genes, our goal is to preserve cone vision in a disease gene-agnostic manner. Previously we showed that overexpressing TXNIP, an α-arrestin protein, prolonged cone vision in RP mouse models, using an AAV to express it only in cones. Here, we expressed different alleles of Txnip in the retinal pigmented epithelium (RPE), a support layer for cones. Our goal was to learn more of TXNIP's structure-function relationships for cone survival, as well as determine the optimal cell type expression pattern for cone survival. The C-terminal half of TXNIP was found to be sufficient to remove GLUT1 from the cell surface, and improved RP cone survival, when expressed in the RPE, but not in cones. Knock-down of HSP90AB1, a TXNIP-interactor which regulates metabolism, improved the survival of cones alone and was additive for cone survival when combined with TXNIP. From these and other results, it is likely that TXNIP interacts with several proteins in the RPE to indirectly support cone survival, with some of these interactions different from those that lead to cone survival when expressed only in cones.


Asunto(s)
Proteínas Portadoras , Modelos Animales de Enfermedad , Células Fotorreceptoras Retinianas Conos , Retinitis Pigmentosa , Animales , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Ratones , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Mutación Missense , Supervivencia Celular , Alelos , Eliminación de Gen , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
3.
Nat Commun ; 15(1): 4316, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773095

RESUMEN

As signalling organelles, cilia regulate their G protein-coupled receptor content by ectocytosis, a process requiring localised actin dynamics to alter membrane shape. Photoreceptor outer segments comprise an expanse of folded membranes (discs) at the tip of highly-specialised connecting cilia, into which photosensitive GPCRs are concentrated. Discs are shed and remade daily. Defects in this process, due to mutations, cause retinitis pigmentosa (RP). Whilst fundamental for vision, the mechanism of photoreceptor disc generation is poorly understood. Here, we show membrane deformation required for disc genesis is driven by dynamic actin changes in a process akin to ectocytosis. We show RPGR, a leading RP gene, regulates actin-binding protein activity central to this process. Actin dynamics, required for disc formation, are perturbed in Rpgr mouse models, leading to aborted membrane shedding as ectosome-like vesicles, photoreceptor death and visual loss. Actin manipulation partially rescues this, suggesting the pathway could be targeted therapeutically. These findings help define how actin-mediated dynamics control outer segment turnover.


Asunto(s)
Actinas , Proteínas del Ojo , Retinitis Pigmentosa , Animales , Actinas/metabolismo , Ratones , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/genética , Cilios/metabolismo , Humanos , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Ratones Noqueados , Ratones Endogámicos C57BL , Membrana Celular/metabolismo
4.
Life Sci Alliance ; 7(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38570189

RESUMEN

Crumbs homolog 1 (CRB1) is one of the key genes linked to retinitis pigmentosa and Leber congenital amaurosis, which are characterized by a high clinical heterogeneity. The Crumbs family member CRB2 has a similar protein structure to CRB1, and in zebrafish, Crb2 has been shown to interact through the extracellular domain. Here, we show that CRB1 and CRB2 co-localize in the human retina and human iPSC-derived retinal organoids. In retina-specific pull-downs, CRB1 was enriched in CRB2 samples, supporting a CRB1-CRB2 interaction. Furthermore, novel interactors of the crumbs complex were identified, representing a retina-derived protein interaction network. Using co-immunoprecipitation, we further demonstrate that human canonical CRB1 interacts with CRB1 and CRB2, but not with CRB3, which lacks an extracellular domain. Next, we explored how missense mutations in the extracellular domain affect CRB1-CRB2 interactions. We observed no or a mild loss of CRB1-CRB2 interaction, when interrogating various CRB1 or CRB2 missense mutants in vitro. Taken together, our results show a stable interaction of human canonical CRB2 and CRB1 in the retina.


Asunto(s)
Amaurosis Congénita de Leber , Retinitis Pigmentosa , Animales , Humanos , Pez Cebra/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Retina/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Portadoras/metabolismo
5.
Mol Vis ; 30: 49-57, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38586605

RESUMEN

RPGR pathogenic variants are the major cause of X-linked retinitis pigmentosa. Here, we report the results from 1,033 clinical DNA tests that included sequencing of RPGR. A total of 184 RPGR variants were identified: 78 pathogenic or likely pathogenic, 14 uncertain, and 92 likely benign or benign. Among the pathogenic and likely pathogenic variants, 23 were novel, and most were frameshift or nonsense mutations (87%) and enriched (67%) in RPGR exon 15 (ORF15). Identical pathogenic variants found in different families were largely on different haplotype backgrounds, indicating relatively frequent, recurrent RPGR mutations. None of the 16 mother/affected son pairs showed de novo mutations; all 16 mothers were heterozygous for the pathogenic variant. These last two observations support the occurrence of most RPGR mutations in the male germline.


Asunto(s)
Proteínas del Ojo , Retinitis Pigmentosa , Humanos , Proteínas del Ojo/genética , Linaje , Mutación , Mutación del Sistema de Lectura , Trastornos de la Visión , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
6.
Neurosci Lett ; 830: 137778, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38621504

RESUMEN

The endoplasmic reticulum (ER) plays an indispensable role in cellular processes, including maintenance of calcium homeostasis, and protein folding, synthesized and processing. Disruptions in these processes leading to ER stress and the accumulation of misfolded proteins can instigate the unfolded protein response (UPR), culminating in either restoration of balanced proteostasis or apoptosis. A key player in this intricate balance is CLCC1, an ER-resident chloride channel, whose essential role extends to retinal development, regulation of ER stress, and UPR. The importance of CLCC1 is further underscored by its interaction with proteins localized to mitochondria-associated endoplasmic reticulum membranes (MAMs), where it participates in UPR induction by MAM proteins. In previous research, we identified a p.(Asp25Glu) pathogenic CLCC1 variant associated with retinitis pigmentosa (RP) (CLCC1 hg38 NC_000001.11; NM_001048210.3, c.75C > A; UniprotKB Q96S66). In attempt to decipher the impact of this variant function, we leveraged liquid chromatography-mass spectrometry (LC-MS) to identify likely CLCC1-interacting proteins. We discovered that the CLCC1 interactome is substantially composed of proteins that localize to ER compartments and that the Asp25Glu variant results in noticeable loss and gain of specific protein interactors. Intriguingly, the analysis suggests that the CLCC1Asp25Glu mutant protein exhibits a propensity for increased interactions with cytoplasmic proteins compared to its wild-type counterpart. To corroborate our LC-MS data, we further scrutinized two novel CLCC1 interactors, Calnexin and SigmaR1, chaperone proteins that localize to the ER and MAMs. Through microscopy, we demonstrate that CLCC1 co-localizes with both proteins, thereby validating our initial findings. Moreover, our results reveal that CLCC1 co-localizes with SigmaR1 not merely at the ER, but also at MAMs. These findings reinforce the notion of CLCC1 interacting with MAM proteins at the ER-mitochondria interface, setting the stage for further exploration into how these interactions impact ER or mitochondria function and lead to retinal degenerative disease when impaired.


Asunto(s)
Retículo Endoplásmico , Receptores sigma , Receptor Sigma-1 , Humanos , Retículo Endoplásmico/metabolismo , Receptores sigma/metabolismo , Receptores sigma/genética , Respuesta de Proteína Desplegada , Células HEK293 , Mitocondrias/metabolismo , Mitocondrias/genética , Membranas Mitocondriales/metabolismo , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
7.
FASEB J ; 38(8): e23606, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38648465

RESUMEN

Rhodopsin mislocalization encompasses various blind conditions. Rhodopsin mislocalization is the primary factor leading to rod photoreceptor dysfunction and degeneration in autosomal dominant retinitis pigmentosa (adRP) caused by class I mutations. In this study, we report a new knock-in mouse model that harbors a class I Q344X mutation in the endogenous rhodopsin gene, which causes rod photoreceptor degeneration in an autosomal dominant pattern. In RhoQ344X/+ mice, mRNA transcripts from the wild-type (Rho) and RhoQ344X mutant rhodopsin alleles are expressed at equal levels. However, the amount of RHOQ344X mutant protein is 2.7 times lower than that of wild-type rhodopsin, a finding consistent with the rapid degradation of the mutant protein. Immunofluorescence microscopy indicates that RHOQ344X is mislocalized to the inner segment and outer nuclear layers of rod photoreceptors in both RhoQ344X/+ and RhoQ344X/Q344X mice, confirming the essential role of the C-terminal VxPx motif in promoting OS delivery of rhodopsin. The mislocalization of RHOQ344X is associated with the concurrent mislocalization of wild-type rhodopsin in RhoQ344X/+ mice. To understand the global changes in proteostasis, we conducted quantitative proteomics analysis and found attenuated expression of rod-specific OS membrane proteins accompanying reduced expression of ciliopathy causative gene products, including constituents of BBSome and axonemal dynein subunit. Those studies unveil a novel negative feedback regulation involving ciliopathy-associated proteins. In this process, a defect in the trafficking signal leads to a reduced quantity of the trafficking apparatus, culminating in a widespread reduction in the transport of ciliary proteins.


Asunto(s)
Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Células Fotorreceptoras Retinianas Bastones , Retinitis Pigmentosa , Rodopsina , Animales , Rodopsina/metabolismo , Rodopsina/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Ratones , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Cilios/metabolismo , Cilios/patología
8.
Nat Commun ; 15(1): 3562, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38670966

RESUMEN

The diagnosis of inherited retinal degeneration (IRD) is challenging owing to its phenotypic and genotypic complexity. Clinical information is important before a genetic diagnosis is made. Metabolomics studies the entire picture of bioproducts, which are determined using genetic codes and biological reactions. We demonstrated that the common diagnoses of IRD, including retinitis pigmentosa (RP), cone-rod dystrophy (CRD), Stargardt disease (STGD), and Bietti's crystalline dystrophy (BCD), could be differentiated based on their metabolite heatmaps. Hundreds of metabolites were identified in the volcano plot compared with that of the control group in every IRD except BCD, considered as potential diagnosing markers. The phenotypes of CRD and STGD overlapped but could be differentiated by their metabolomic features with the assistance of a machine learning model with 100% accuracy. Moreover, EYS-, USH2A-associated, and other RP, sharing considerable similar characteristics in clinical findings, could also be diagnosed using the machine learning model with 85.7% accuracy. Further study would be needed to validate the results in an external dataset. By incorporating mass spectrometry and machine learning, a metabolomics-based diagnostic workflow for the clinical and molecular diagnoses of IRD was proposed in our study.


Asunto(s)
Aprendizaje Automático , Metabolómica , Degeneración Retiniana , Retinitis Pigmentosa , Enfermedad de Stargardt , Humanos , Metabolómica/métodos , Diagnóstico Diferencial , Degeneración Retiniana/diagnóstico , Degeneración Retiniana/sangre , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Masculino , Femenino , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/sangre , Retinitis Pigmentosa/metabolismo , Enfermedad de Stargardt/genética , Adulto , Persona de Mediana Edad , Adolescente , Adulto Joven , Biomarcadores/sangre , Metaboloma , Niño , Distrofias de Conos y Bastones/diagnóstico , Distrofias de Conos y Bastones/genética , Distrofias de Conos y Bastones/sangre , Distrofias de Conos y Bastones/metabolismo , Espectrometría de Masas , Degeneración Macular/sangre , Degeneración Macular/diagnóstico , Degeneración Macular/genética
9.
Genes (Basel) ; 15(4)2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38674450

RESUMEN

Retinitis pigmentosa is a group of genetically determined retinal dystrophies characterized by primary photoreceptor apoptosis and can occur in isolated or syndromic conditions. This study reviewed the clinical data of 15 patients with syndromic retinitis pigmentosa from a Rare Disease Reference Center in Brazil and the results of their next-generation sequencing tests. Five males and ten females participated, with the mean ages for ocular disease onset, fundoscopic diagnosis, and molecular evaluation being 9, 19, and 29 years, respectively. Bardet-Biedl syndrome (n = 5) and Usher syndrome (n = 3) were the most frequent diagnoses, followed by other rare conditions. Among the patients, fourteen completed molecular studies, with three negative results and eleven revealing findings in known genes, including novel variants in MKKS (c.432_435del, p.Phe144Leufs*14), USH2A (c.(7301+1_7302-1)_(9369+1_9370-1)del), and CEP250 (c.5383dup, p.Glu1795Glyfs*13, and c.5050del, p.Asp1684Thrfs*9). Except for Kearn-Sayre, all presented an autosomal recessive inheritance pattern with 64% homozygosity results. The long gap between symptom onset and diagnosis highlights the diagnostic challenges faced by the patients. This study reaffirms the clinical heterogeneity of syndromic retinitis pigmentosa and underscores the pivotal role of molecular analysis in advancing our understanding of these diseases.


Asunto(s)
Retinitis Pigmentosa , Humanos , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/diagnóstico , Masculino , Femenino , Adulto , Adolescente , Niño , Adulto Joven , Síndromes de Usher/genética , Síndromes de Usher/patología , Síndromes de Usher/diagnóstico , Brasil/epidemiología , Persona de Mediana Edad , Secuenciación de Nucleótidos de Alto Rendimiento , Síndrome de Bardet-Biedl/genética , Mutación
10.
Elife ; 122024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38661530

RESUMEN

Retinitis pigmentosa (RP), a heterogenous group of inherited retinal disorder, causes slow progressive vision loss with no effective treatments available. Mutations in the rhodopsin gene (RHO) account for ~25% cases of autosomal dominant RP (adRP). In this study, we describe the disease characteristics of the first-ever reported mono-allelic copy number variation (CNV) in RHO as a novel cause of adRP. We (a) show advanced retinal degeneration in a male patient (68 years of age) harboring four transcriptionally active intact copies of rhodopsin, (b) recapitulated the clinical phenotypes using retinal organoids, and (c) assessed the utilization of a small molecule, Photoregulin3 (PR3), as a clinically viable strategy to target and modify disease progression in RP patients associated with RHO-CNV. Patient retinal organoids showed photoreceptors dysgenesis, with rod photoreceptors displaying stunted outer segments with occasional elongated cilia-like projections (microscopy); increased RHO mRNA expression (quantitative real-time PCR [qRT-PCR] and bulk RNA sequencing); and elevated levels and mislocalization of rhodopsin protein (RHO) within the cell body of rod photoreceptors (western blotting and immunohistochemistry) over the extended (300 days) culture time period when compared against control organoids. Lastly, we utilized PR3 to target NR2E3, an upstream regulator of RHO, to alter RHO expression and observed a partial rescue of RHO protein localization from the cell body to the inner/outer segments of rod photoreceptors in patient organoids. These results provide a proof-of-principle for personalized medicine and suggest that RHO expression requires precise control. Taken together, this study supports the clinical data indicating that RHO-CNV associated adRPdevelops as a result of protein overexpression, thereby overloading the photoreceptor post-translational modification machinery.


Asunto(s)
Variaciones en el Número de Copia de ADN , Retinitis Pigmentosa , Rodopsina , Anciano , Humanos , Masculino , Organoides/metabolismo , Organoides/efectos de los fármacos , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Rodopsina/genética , Rodopsina/metabolismo
11.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(4): 456-460, 2024 Apr 10.
Artículo en Chino | MEDLINE | ID: mdl-38565512

RESUMEN

OBJECTIVE: To explore the genetic basis for a patient with autosomal dominant retinitis pigmentosa (RP). METHODS: A male patient with RP treated at Gansu Provincial Maternal and Child Health Care Hospital in September 2019 was selected as the study subject. Clinical data was collected. Peripheral blood samples of the patient and his parents were subjected to whole exome sequencing (WES). Candidate variant was validated by Sanger sequencing and bioinformatic analysis. RESULTS: The patient, a 29-year-old male, developed night blindness, amblyopia, visual field defects and optic disc abnormalities since childhood. Gene sequencing revealed that he has harbored a heterozygous c.942G>C (p.Lys314Asn) variant of the IMPDH1 gene, which was inherited from his mother, whilst his father was of the wild type. Based on the guidelines from the American College of Medical Genetics and Genomics, the c.942G>C variant was predicted as likely pathogenic (PM1+PM2_Supporting+PP3+PP1). CONCLUSION: The c.942G>C (p.Lys314Asn) variant in the IMPDH1 gene probably underlay the RP in this patient.


Asunto(s)
Retinitis Pigmentosa , Adulto , Femenino , Humanos , Masculino , Biología Computacional , Genómica , Heterocigoto , IMP Deshidrogenasa , Madres , Mutación , Retinitis Pigmentosa/genética
12.
Gene ; 912: 148367, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38485037

RESUMEN

Retinitis pigmentosa 1-like 1 (RP1L1) is a component of photoreceptor cilia. Pathogenic variants in RP1L1 cause photoreceptor diseases, suggesting that RP1L1 plays an important role in photoreceptor biology, although its exact function is unknown. To date, RP1L1 variants have been associated with occult macular dystrophy (cone degeneration) and retinitis pigmentosa (rod degeneration). Here, we summarize the reported RP1L1-associated photoreceptor pathogenic mutations. The association between RP1L1 and other diseases (mainly several tumors) is also summarized and RP1L1 is included in a wider range of diseases. Finally, it is necessary to further explore the influence mechanism of RP1L1 gene on the health of photoreceptors and how it participates in the occurrence and development of tumors.


Asunto(s)
Degeneración Macular , Neoplasias , Retinitis Pigmentosa , Humanos , Proteínas del Ojo/genética , Degeneración Macular/genética , Neoplasias/genética , Retinitis Pigmentosa/genética
13.
EMBO Mol Med ; 16(4): 805-822, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38504136

RESUMEN

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.


Asunto(s)
Retinitis Pigmentosa , Animales , Ratones , Humanos , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Retinitis Pigmentosa/metabolismo , Retina/metabolismo , Exones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Terapia Genética , Proteínas del Ojo/genética , Proteínas del Ojo/química , Proteínas del Ojo/metabolismo
14.
Cell Rep Med ; 5(4): 101459, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38518771

RESUMEN

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.


Asunto(s)
Células Fotorreceptoras Retinianas Bastones , Retinitis Pigmentosa , Animales , Humanos , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/terapia , Células Fotorreceptoras Retinianas Conos/metabolismo , Modelos Animales de Enfermedad
15.
Klin Monbl Augenheilkd ; 241(3): 259-265, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38508214

RESUMEN

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.


Asunto(s)
Ceguera Nocturna , Distrofias Retinianas , Retinitis Pigmentosa , Adulto , Niño , Humanos , cis-trans-Isomerasas/genética , Terapia Genética , Mutación , Ceguera Nocturna/terapia , Fenotipo , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/genética , Distrofias Retinianas/terapia , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia
17.
Proc Natl Acad Sci U S A ; 121(11): e2316118121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38442152

RESUMEN

Retinitis pigmentosa (RP) is a common form of retinal dystrophy that can be caused by mutations in any one of dozens of rod photoreceptor genes. The genetic heterogeneity of RP represents a significant challenge for the development of effective therapies. Here, we present evidence for a potential gene-independent therapeutic strategy based on targeting Nr2e3, a transcription factor required for the normal differentiation of rod photoreceptors. Nr2e3 knockout results in hybrid rod photoreceptors that express the full complement of rod genes, but also a subset of cone genes. We show that germline deletion of Nr2e3 potently protects rods in three mechanistically diverse mouse models of retinal degeneration caused by bright-light exposure (light damage), structural deficiency (rhodopsin-deficient Rho-/- mice), or abnormal phototransduction (phosphodiesterase-deficient rd10 mice). Nr2e3 knockout confers strong neuroprotective effects on rods without adverse effects on their gene expression, structure, or function. Furthermore, in all three degeneration models, prolongation of rod survival by Nr2e3 knockout leads to lasting preservation of cone morphology and function. These findings raise the possibility that upregulation of one or more cone genes in Nr2e3-deficient rods may be responsible for the neuroprotective effects we observe.


Asunto(s)
Fármacos Neuroprotectores , Distrofias Retinianas , Retinitis Pigmentosa , Animales , Ratones , Células Fotorreceptoras Retinianas Conos , Retinitis Pigmentosa/genética , Modelos Animales de Enfermedad , Células Germinativas , Receptores Nucleares Huérfanos
18.
Int J Mol Sci ; 25(5)2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38474159

RESUMEN

PRPH2, one of the most frequently inherited retinal dystrophy (IRD)-causing genes, implies a high phenotypic variability. This study aims to analyze the PRPH2 mutational spectrum in one of the largest cohorts worldwide, and to describe novel pathogenic variants and genotype-phenotype correlations. A study of 220 patients from 103 families recruited from a database of 5000 families. A molecular diagnosis was performed using classical molecular approaches and next-generation sequencing. Common haplotypes were ascertained by analyzing single-nucleotide polymorphisms. We identified 56 variants, including 11 novel variants. Most of them were missense variants (64%) and were located in the D2-loop protein domain (77%). The most frequently occurring variants were p.Gly167Ser, p.Gly208Asp and p.Pro221_Cys222del. Haplotype analysis revealed a shared region in families carrying p.Leu41Pro or p.Pro221_Cys222del. Patients with retinitis pigmentosa presented an earlier disease onset. We describe the largest cohort of IRD families associated with PRPH2 from a single center. Most variants were located in the D2-loop domain, highlighting its importance in interacting with other proteins. Our work suggests a likely founder effect for the variants p.Leu41Pro and p.Pro221_Cys222del in our Spanish cohort. Phenotypes with a primary rod alteration presented more severe affectation. Finally, the high phenotypic variability in PRPH2 hinders the possibility of drawing genotype-phenotype correlations.


Asunto(s)
Distrofias Retinianas , Retinitis Pigmentosa , Humanos , Análisis Mutacional de ADN , Mutación , Mutación Missense , Fenotipo , Distrofias Retinianas/genética , Retinitis Pigmentosa/genética
19.
Exp Eye Res ; 241: 109856, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38479725

RESUMEN

Rhodopsin-mediated autosomal dominant retinitis pigmentosa (RHO-adRP) causes progressive vision loss and is potentially incurable, accounting for 25% of adRP cases. Studies on RHO-adRP mechanism were at large based on the biochemical and cellular properties, especially class-3. Nonetheless, the absence of an appropriate model for class-3 RHO-adRP has impeded comprehensive exploration. Here, induced pluripotent stem cells (iPSCs) were generated from a healthy control and two sibling RP patients with the same point mutation, c.403C>T (p.R135W). The first three-dimensional (3D) retinal organoid model of a class-3 RHO point mutation from patient-derived iPSCs was generated. Significant defects were observed in rod photoreceptors in terms of localization, morphology, transcriptional profiling and single cell resolution, to better understand the human disease resulting from RHO mutations from a developmental perspective. This first human model of class-3 RHO-adRP provides a representation of patient's retina in vitro and displays features of RHO-adRP retinal organoids relevant for therapeutic development.


Asunto(s)
Retina , Retinitis Pigmentosa , Humanos , Retinitis Pigmentosa/genética , Mutación , Rodopsina/genética , Organoides
20.
Cells ; 13(6)2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38534367

RESUMEN

We report a novel RPGR missense variant co-segregated with a familial X-linked retinitis pigmentosa (XLRP) case. The brothers were hemizygous for this variant, but only the proband presented with primary ciliary dyskinesia (PCD). Thus, we aimed to elucidate the role of the RPGR variant and other modifier genes in the phenotypic variability observed in the family and its impact on motile cilia. The pathogenicity of the variant on the RPGR protein was evaluated by in vitro studies transiently transfecting the mutated RPGR gene, and immunofluorescence analysis on nasal brushing samples. Whole-exome sequencing was conducted to identify potential modifier variants. In vitro studies showed that the mutated RPGR protein could not localise to the cilium and impaired cilium formation. Accordingly, RPGR was abnormally distributed in the siblings' nasal brushing samples. In addition, a missense variant in CEP290 was identified. The concurrent RPGR variant influenced ciliary mislocalisation of the protein. We provide a comprehensive characterisation of motile cilia in this XLRP family, with only the proband presenting PCD symptoms. The variant's pathogenicity was confirmed, although it alone does not explain the respiratory symptoms. Finally, the CEP290 gene may be a potential modifier for respiratory symptoms in patients with RPGR mutations.


Asunto(s)
Trastornos de la Motilidad Ciliar , Retinitis Pigmentosa , Humanos , Masculino , Trastornos de la Motilidad Ciliar/genética , Proteínas del Ojo/metabolismo , Genes Modificadores , Mutación , Retinitis Pigmentosa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...