Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.633
Filtrar
1.
Transl Vis Sci Technol ; 13(5): 1, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38691083

RESUMEN

Purpose: This feasibility study investigated the practicability of collecting and analyzing tear proteins from preterm infants at risk of retinopathy of prematurity (ROP). We sought to identify any tear proteins which might be implicated in the pathophysiology of ROP as well as prognostic markers. Methods: Schirmer's test was used to obtain tear samples from premature babies, scheduled for ROP screening, after parental informed consent. Mass spectrometry was used for proteomic analysis. Results: Samples were collected from 12 infants, which were all adequate for protein analysis. Gestational age ranged from 25 + 6 to 31 + 1 weeks. Postnatal age at sampling ranged from 19 to 66 days. One infant developed self-limiting ROP. Seven hundred one proteins were identified; 261 proteins identified in the majority of tear samples, including several common tear proteins, were used for analyses. Increased risk of ROP as determined by the postnatal growth ROP (G-ROP) criteria was associated with an increase in lactate dehydrogenase B chain in tears. Older infants demonstrated increased concentration of immunoglobulin complexes within their tear samples and two sets of twins in the cohort showed exceptionally similar proteomes, supporting validity of the analysis. Conclusions: Tear sampling by Schirmer test strips and subsequent proteomic analysis by mass spectrometry in preterm infants is feasible. A larger study is required to investigate the potential use of tear proteomics in identification of ROP. Translational Relevance: Tear sampling and subsequent mass spectrometry in preterm infants is feasible. Investigation of the premature tear proteome may increase our understanding of retinal development and provide noninvasive biomarkers for identification of treatment-warranted ROP.


Asunto(s)
Biomarcadores , Proteínas del Ojo , Estudios de Factibilidad , Edad Gestacional , Recien Nacido Prematuro , Proteómica , Retinopatía de la Prematuridad , Lágrimas , Humanos , Retinopatía de la Prematuridad/diagnóstico , Retinopatía de la Prematuridad/metabolismo , Proteómica/métodos , Recién Nacido , Femenino , Lágrimas/química , Lágrimas/metabolismo , Masculino , Biomarcadores/metabolismo , Biomarcadores/análisis , Proteínas del Ojo/metabolismo , Proteínas del Ojo/análisis , Lactante , Espectrometría de Masas/métodos
2.
Proc Natl Acad Sci U S A ; 121(20): e2321711121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38713624

RESUMEN

During development, neural stem cells in the cerebral cortex, also known as radial glial cells (RGCs), generate excitatory neurons, followed by production of cortical macroglia and inhibitory neurons that migrate to the olfactory bulb (OB). Understanding the mechanisms for this lineage switch is fundamental for unraveling how proper numbers of diverse neuronal and glial cell types are controlled. We and others recently showed that Sonic Hedgehog (Shh) signaling promotes the cortical RGC lineage switch to generate cortical oligodendrocytes and OB interneurons. During this process, cortical RGCs generate intermediate progenitor cells that express critical gliogenesis genes Ascl1, Egfr, and Olig2. The increased Ascl1 expression and appearance of Egfr+ and Olig2+ cortical progenitors are concurrent with the switch from excitatory neurogenesis to gliogenesis and OB interneuron neurogenesis in the cortex. While Shh signaling promotes Olig2 expression in the developing spinal cord, the exact mechanism for this transcriptional regulation is not known. Furthermore, the transcriptional regulation of Olig2 and Egfr has not been explored. Here, we show that in cortical progenitor cells, multiple regulatory programs, including Pax6 and Gli3, prevent precocious expression of Olig2, a gene essential for production of cortical oligodendrocytes and astrocytes. We identify multiple enhancers that control Olig2 expression in cortical progenitors and show that the mechanisms for regulating Olig2 expression are conserved between the mouse and human. Our study reveals evolutionarily conserved regulatory logic controlling the lineage switch of cortical neural stem cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Corteza Cerebral , Receptores ErbB , Proteínas Hedgehog , Proteínas del Tejido Nervioso , Células-Madre Neurales , Neurogénesis , Factor de Transcripción 2 de los Oligodendrocitos , Factor de Transcripción PAX6 , Animales , Neurogénesis/fisiología , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Receptores ErbB/metabolismo , Receptores ErbB/genética , Ratones , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Factor de Transcripción PAX6/metabolismo , Factor de Transcripción PAX6/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Proteína Gli3 con Dedos de Zinc/metabolismo , Proteína Gli3 con Dedos de Zinc/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factores de Transcripción Paired Box/metabolismo , Factores de Transcripción Paired Box/genética , Neuroglía/metabolismo , Neuroglía/citología , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/citología , Linaje de la Célula , Humanos
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.
CRISPR J ; 7(2): 100-110, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38579141

RESUMEN

Inherited retinal diseases (IRDs) are a heterogeneous group of blinding genetic disorders caused by pathogenic variants in genes expressed in the retina. In this study, we sought to develop a method for rapid evaluation of IRD gene variant pathogenicity by inducing expression of retinal genes in patient-derived fibroblasts using CRISPR-activation (CRISPRa). We demonstrate CRISPRa of CRB1 expression in fibroblasts derived from patients with retinitis pigmentosa, enabling investigation of pathogenic mechanisms associated with specific variants. We show the CRB1 c.4005 + 1G>A variant caused exon 11 skipping in CRISPR-activated fibroblasts and retinal organoids (ROs) derived from the same RP12 patient. The c.652 + 5G>C variant was shown to enhance exon 2 skipping in CRISPR-activated fibroblasts and differentially affected CRB1 isoform expression in fibroblasts and ROs. Our study demonstrates an accessible platform for transcript screening of IRD gene variants in patient-derived fibroblasts, which can potentially be applied for rapid pathogenicity assessments of any gene variant.


Asunto(s)
Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Especies Reactivas de Oxígeno/metabolismo , Virulencia , Edición Génica , Expresión Génica , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
6.
Exp Eye Res ; 243: 109887, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38609044

RESUMEN

The pathophysiology of Primary Open Angle Glaucoma (POAG) remains poorly understood. Through proteomic analysis of aqueous humour (AH) from POAG patients, we aim to identify changes in protein composition of these samples compared to control samples. High resolution mass spectrometry-based TMT6plex quantitative proteomics analysis is performed on AH samples collected from POAG patients, and compared against a control group of patients with cataracts. Data are available via ProteomeXchange with identifier PXD033153. 1589 proteins were quantified from the aqueous samples using Proteome Discoverer version 2.2 software. Among these proteins, 210 were identified as unique master proteins. The proteins which were up or down-regulated by ±3 fold-change were considered significant. Human neuroblastoma full-length cDNA clone CS0DD006YL02 was significantly upregulated in patients with severe POAG on >2 medications, while actin, cytoplasmic 1, V2-7 protein (fragment), immunoglobulin-like polypeptide 1 and phosphatidylethanolamine-binding protein 4 were only present in these patients with severe POAG on >2 medications. Beta-crystallin B1 and B2, Gamma-crystallin C, D and S were significantly downregulated in the severe POAG ≤2 glaucoma medications group. Beta-crystallin B2, Gamma-crystallin D and GCT-A9 light chain variable region (fragment) were significantly downregulated in the non-severe POAG group. Actin, cytoplasmic 1 was significantly upregulated in subjects with severe POAG who required more than 2 glaucoma medications. Crystallins (Beta-crystallin B1 and B2, Gamma-crystallin C, D and S) were significantly downregulated in subjects with severe POAG who required less than 2 glaucoma medications.


Asunto(s)
Humor Acuoso , Proteínas del Ojo , Glaucoma de Ángulo Abierto , Proteómica , Humanos , Glaucoma de Ángulo Abierto/metabolismo , Humor Acuoso/metabolismo , Femenino , Masculino , Proteínas del Ojo/metabolismo , Anciano , Persona de Mediana Edad , Proteómica/métodos , Presión Intraocular/fisiología , Pueblo Asiatico
7.
Exp Eye Res ; 243: 109903, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38642601

RESUMEN

Pseudoexfoliation syndrome (PEX) is characterized by the deposition of fibrous pseudoexfoliation material (PEXM) in the eye, and secondary glaucoma associated with this syndrome has a faster and more severe clinical course. The incidence of PEX and pseudoexfoliative glaucoma (PEXG) exhibits ethnic clustering; however, few proteomic studies related to PEX and PEXG have been conducted in Asian populations. Therefore, we aimed to conduct proteomic analysis on the aqueous humor (AH) obtained from Uyghur patients with cataracts, those with PEX and cataracts, and those with PEXG and cataracts to better understand the molecular mechanisms of the disease and identify its potential biomarkers. To this end, AH was collected from patients with cataracts (n = 10, control group), PEX with cataracts (n = 10, PEX group), and PEXG with cataracts (n = 10, PEXG group) during phacoemulsification. Label-free quantitative proteomic techniques combined with bioinformatics were used to identify and analyze differentially expressed proteins (DEPs) in the AH of PEX and PEXG groups. Then, independent AH samples (n = 12, each group) were collected to validate DEPs by enzyme-linked immunosorbent assay (ELISA). The PEX group exhibited 25 DEPs, while the PEXG group showed 44 DEPs, both compared to the control group. Subsequently, we found three newly identified proteins in both PEX and PEXG groups, wherein FRAS1-related extracellular matrix protein 2 (FREM2) and osteoclast-associated receptor (OSCAR) exhibited downregulation, whereas coagulation Factor IX (F9) displayed upregulation. Bioinformatics analysis suggested that extracellular matrix interactions, abnormal blood-derived proteins, and lysosomes were mainly involved in the process of PEX and PEXG, and the PPI network further revealed F9 may serve as a potential biomarker for both PEX and PEXG. In conclusion, this study provides new information for understanding the proteomics of AH in PEX and PEXG.


Asunto(s)
Humor Acuoso , Síndrome de Exfoliación , Proteínas del Ojo , Proteómica , Humanos , Síndrome de Exfoliación/metabolismo , Humor Acuoso/metabolismo , Proteómica/métodos , Masculino , Femenino , Anciano , Proteínas del Ojo/metabolismo , China/epidemiología , Glaucoma de Ángulo Abierto/metabolismo , Persona de Mediana Edad , Biomarcadores/metabolismo , Ensayo de Inmunoadsorción Enzimática , Catarata/metabolismo , Presión Intraocular/fisiología
8.
Int J Biol Macromol ; 267(Pt 1): 131274, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38569991

RESUMEN

The vitreous is a vital component of the eye, occupying a substantial portion of its volume and maintaining its structure. This study delves into the presence and significance of intrinsically disordered proteins (IDPs) within the vitreous, utilizing a dataset of 1240 vitreous proteins previously discovered in the vitreous proteome by Murthy et al.in five healthy subjects. The results indicate that 26.9 % of vitreous proteins are highly disordered, 68.8 % possess moderate disorder, and only 4.3 % are highly ordered. A complex interaction network among these proteins suggests their biological importance, and approximately 25 % may undergo liquid-liquid phase separation (LLPS). These findings offer new perspectives on the vitreous' molecular composition and behavior, potentially impacting our understanding of eye-related diseases, physiological changes such as vitreous syneresis. Further research is needed to translate these insights into clinical applications, although the intrinsic protein disorder and its association with LLPS appears to play a role in vitreous proteome function.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Proteoma , Cuerpo Vítreo , Humanos , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteoma/metabolismo , Cuerpo Vítreo/metabolismo , Proteínas del Ojo/metabolismo
9.
JCI Insight ; 9(8)2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38646933

RESUMEN

Inherited retinal dystrophies (IRDs) are progressive diseases leading to vision loss. Mutation in the eyes shut homolog (EYS) gene is one of the most frequent causes of IRD. However, the mechanism of photoreceptor cell degeneration by mutant EYS has not been fully elucidated. Here, we generated retinal organoids from induced pluripotent stem cells (iPSCs) derived from patients with EYS-associated retinal dystrophy (EYS-RD). In photoreceptor cells of RD organoids, both EYS and G protein-coupled receptor kinase 7 (GRK7), one of the proteins handling phototoxicity, were not in the outer segment, where they are physiologically present. Furthermore, photoreceptor cells in RD organoids were vulnerable to light stimuli, and especially to blue light. Mislocalization of GRK7, which was also observed in eys-knockout zebrafish, was reversed by delivering control EYS into photoreceptor cells of RD organoids. These findings suggest that avoiding phototoxicity would be a potential therapeutic approach for EYS-RD.


Asunto(s)
Células Madre Pluripotentes Inducidas , Organoides , Distrofias Retinianas , Pez Cebra , Animales , Humanos , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Luz/efectos adversos , Mutación , Organoides/metabolismo , Retina/metabolismo , Retina/patología , Distrofias Retinianas/terapia , Distrofias Retinianas/genética , Distrofias Retinianas/metabolismo
10.
Int J Mol Sci ; 25(6)2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38542364

RESUMEN

Retinitis pigmentosa 11 is an untreatable, dominantly inherited retinal disease caused by heterozygous mutations in pre-mRNA processing factor 31 PRPF31. The expression level of PRPF31 is linked to incomplete penetrance in affected families; mutation carriers with higher PRPF31 expression can remain asymptomatic. The current study explores an antisense oligonucleotide exon skipping strategy to treat RP11 caused by truncating mutations within PRPF31 exon 12 since it does not appear to encode any domains essential for PRPF31 protein function. Cells derived from a patient carrying a PRPF31 1205C>A nonsense mutation were investigated; PRPF31 transcripts encoded by the 1205C>A allele were undetectable due to nonsense-mediated mRNA decay, resulting in a 46% reduction in PRPF31 mRNA, relative to healthy donor cells. Antisense oligonucleotide-induced skipping of exon 12 rescued the open reading frame with consequent 1.7-fold PRPF31 mRNA upregulation in the RP11 patient fibroblasts. The level of PRPF31 upregulation met the predicted therapeutic threshold of expression inferred in a non-penetrant carrier family member harbouring the same mutation. This study demonstrated increased PRPF31 expression and retention of the nuclear translocation capability for the induced PRPF31 isoform. Future studies should evaluate the function of the induced PRPF31 protein on pre-mRNA splicing in retinal cells to validate the therapeutic approach for amenable RP11-causing mutations.


Asunto(s)
Oligonucleótidos Antisentido , Precursores del ARN , Retinitis Pigmentosa , Humanos , Precursores del ARN/genética , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/uso terapéutico , Sistemas de Lectura Abierta , Mutación , Codón sin Sentido , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Linaje
11.
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
12.
Exp Eye Res ; 242: 109872, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38514024

RESUMEN

X-linked retinoschisis (XLRS) is an early onset degenerative retinal disease characterized by cystic lesions in the middle layers of the retina. These structural changes are accompanied by a loss of visual acuity and decreased contrast sensitivity. XLRS is caused by mutations in the gene Rs1 which encodes the secreted protein Retinoschisin 1. Young Rs1-mutant mouse models develop key hallmarks of XLRS including intraretinal schisis and abnormal electroretinograms. The electroretinogram (ERG) comprises activity of multiple cellular generators, and it is not known how and when each of these is impacted in Rs1 mutant mice. Here we use an ex vivo ERG system and pharmacological blockade to determine how ERG components generated by photoreceptors, ON-bipolar, and Müller glial cells are impacted in Rs1 mutants and to determine the time course of these changes. We report that ERG abnormalities begin near eye-opening and that all ERG components are involved.


Asunto(s)
Moléculas de Adhesión Celular , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas del Ojo , Retinosquisis , Animales , Retinosquisis/genética , Retinosquisis/fisiopatología , Ratones , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Ratones Endogámicos C57BL , Mutación , Células Ependimogliales/patología , Células Ependimogliales/metabolismo , Masculino , Células Bipolares de la Retina/patología , Células Bipolares de la Retina/metabolismo
13.
Exp Eye Res ; 242: 109861, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38522635

RESUMEN

Amyloid-beta (Aß), a family of aggregation-prone and neurotoxic peptides, has been implicated in the pathophysiology of age-related macular degeneration (AMD). We have previously shown that oligomeric and fibrillar species of Aß42 exerted retinal toxicity in rats, but while the consequences of exposure to amyloid were related to intracellular effects, the mechanism of Aß42 internalization in the retina is not well characterized. In the brain, the 67 kDa laminin receptor (67LR) participates in Aß-related neuronal cell death. A short peptide derived from pigment epithelium-derived factor (PEDF), formerly designated PEDF-335, was found to mitigate experimental models of ischemic retinopathy via targeting of 67LR. In the present study, we hypothesized that 67LR mediates the uptake of pathogenic Aß42 assemblies in the retina, and that targeting of this receptor by PEDF-335 may limit the internalization of Aß, thereby ameliorating its retinotoxicity. To test this assumption ARPE-19 cells in culture were incubated with PEDF-335 before treatment with fibrillar or oligomeric structures of Aß42. Immunostaining confirmed that PEDF-335 treatment substantially prevented amyloid internalization into ARPE-19 cells and maintained their viability in the presence of toxic oligomeric and fibrillar Aß42 entities in vitro. FRET competition assay was performed and confirmed the binding of PEDF-335 to 67LR in RPE-like cells. Wild-type rats were treated with intravitreal PEDF-335 in the experimental eye 2 days prior to administration of retinotoxic Aß42 oligomers or fibrils to both eyes. Retinal function was assessed by electroretinography through 6 weeks post injection. The ERG responses in rats treated with oligomeric or fibrillar Aß42 assemblies were near-normal in eyes previously treated with intravitreal PEDF-335, whereas those measured in the control eyes treated with injection of the Aß42 assemblies alone showed pathologic attenuation of the retinal function through 6 weeks. The retinal presence of 67LR was determined ex vivo by immunostaining and western blotting. Retinal staining demonstrated the constitutional expression of 67LR mainly in the retinal nuclear layers. In the presence of Aß42, the levels of 67LR were increased, although its retinal distribution remained largely unaltered. In contrast, no apparent differences in the retinal expression level of 67LR were noted following exposure to PEDF-335 alone, and its pattern of localization in the retina remained similarly concentrated primarily in the inner and outer nuclear layers. In summary, we found that PEDF-335 confers protection against Aß42-mediated retinal toxicity, with significant effects noted in cells as well as in vivo in rats. The effects of PEDF-335 in the retina are potentially mediated via binding to 67LR and by at least partial inhibition of Aß42 internalization. These results suggest that PEDF-335 may merit further consideration in the development of targeted inhibition of amyloid-related toxicity in the retina. More broadly, our observations provide evidence on the importance of extracellular versus intracellular Aß42 in the retina and suggest concepts on the molecular mechanism of Aß retinal pathogenicity.


Asunto(s)
Péptidos beta-Amiloides , Electrorretinografía , Proteínas del Ojo , Factores de Crecimiento Nervioso , Serpinas , Animales , Serpinas/metabolismo , Proteínas del Ojo/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Ratas , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Fragmentos de Péptidos/toxicidad , Modelos Animales de Enfermedad , Receptores de Laminina/metabolismo , Masculino , Retina/efectos de los fármacos , Retina/metabolismo , Humanos , Inyecciones Intravítreas , Western Blotting , Enfermedades de la Retina/prevención & control , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/inducido químicamente , Células Cultivadas
14.
Invest Ophthalmol Vis Sci ; 65(3): 31, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38517429

RESUMEN

Purpose: This study aimed to investigate the impact of 21 NDP mutations located at the dimer interface, focusing on their potential effects on protein assembly, secretion efficiency, and activation of the Norrin/ß-catenin signaling pathway. Methods: The expression level, secretion efficiency, and protein assembly of mutations were analyzed using Western blot. The Norrin/ß-catenin signaling pathway activation ability after overexpression of mutants or supernatant incubation of mutant proteins was tested in HEK293STF cells. The mutant norrin and wild-type (WT) FZD4 were overexpressed in HeLa cells to observe their co-localization. Immunofluorescence staining was conducted in HeLa cells to analyze the subcellular localization of Norrin and the Retention Using Selective Hook (RUSH) assay was used to dynamically observe the secretion process of WT and mutant Norrin. Results: Four mutants (A63S, E66K, H68P, and L103Q) exhibited no significant differences from WT in all evaluations. The other 17 mutants presented abnormalities, including inadequate protein assembly, reduced secretion, inability to bind to FZD4 on the cell membrane, and decreased capacity to activate Norrin/ß-catenin signaling pathway. The RUSH assay revealed the delay in endoplasmic reticulum (ER) exit and impairment of Golgi transport. Conclusions: Mutations at the Norrin dimer interface may lead to abnormal protein assembly, inability to bind to FZD4, and decreased secretion, thus contributing to compromised Norrin/ß-catenin signaling. Our results shed light on the pathogenic mechanisms behind a significant proportion of NDP gene mutations in familial exudative vitreoretinopathy (FEVR) or Norrie disease.


Asunto(s)
Proteínas del Ojo , Receptores Frizzled , Enfermedades de la Retina , Humanos , beta Catenina/genética , beta Catenina/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Receptores Frizzled/genética , Células HeLa , Mutación , Enfermedades de la Retina/genética , Proteínas del Tejido Nervioso/genética
15.
Sci Rep ; 14(1): 6958, 2024 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-38521856

RESUMEN

Mutations in myocilin (MYOC) are the leading known genetic cause of primary open-angle glaucoma, responsible for about 4% of all cases. Mutations in MYOC cause a gain-of-function phenotype in which mutant myocilin accumulates in the endoplasmic reticulum (ER) leading to ER stress and trabecular meshwork (TM) cell death. Therefore, knocking out myocilin at the genome level is an ideal strategy to permanently cure the disease. We have previously utilized CRISPR/Cas9 genome editing successfully to target MYOC using adenovirus 5 (Ad5). However, Ad5 is not a suitable vector for clinical use. Here, we sought to determine the efficacy of adeno-associated viruses (AAVs) and lentiviruses (LVs) to target the TM. First, we examined the TM tropism of single-stranded (ss) and self-complimentary (sc) AAV serotypes as well as LV expressing GFP via intravitreal (IVT) and intracameral (IC) injections. We observed that LV_GFP expression was more specific to the TM injected via the IVT route. IC injections of Trp-mutant scAAV2 showed a prominent expression of GFP in the TM. However, robust GFP expression was also observed in the ciliary body and retina. We next constructed lentiviral particles expressing Cas9 and guide RNA (gRNA) targeting MYOC (crMYOC) and transduction of TM cells stably expressing mutant myocilin with LV_crMYOC significantly reduced myocilin accumulation and its associated chronic ER stress. A single IVT injection of LV_crMYOC in Tg-MYOCY437H mice decreased myocilin accumulation in TM and reduced elevated IOP significantly. Together, our data indicates, LV_crMYOC targets MYOC gene editing in TM and rescues a mouse model of myocilin-associated glaucoma.


Asunto(s)
Proteínas del Citoesqueleto , Glaucoma de Ángulo Abierto , Glicoproteínas , Animales , Ratones , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/terapia , Glaucoma de Ángulo Abierto/metabolismo , Presión Intraocular/genética , Lentivirus/genética , Malla Trabecular/metabolismo
16.
Cell Rep ; 43(4): 114010, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38536817

RESUMEN

Although the small GTPase RAB37 acts as an organizer of autophagosome biogenesis, the upstream regulatory mechanism of autophagy via guanosine diphosphate (GDP)-guanosine triphosphate (GTP) exchange in maintaining retinal function has not been determined. We found that retinitis pigmentosa GTPase regulator (RPGR) is a guanine nucleotide exchange factor that activates RAB37 by accelerating GDP-to-GTP exchange. RPGR directly interacts with RAB37 via the RPGR-RCC1-like domain to promote autophagy through stimulating exchange. Rpgr knockout (KO) in mice leads to photoreceptor degeneration owing to autophagy impairment in the retina. Notably, the retinopathy phenotypes of Rpgr KO retinas are rescued by the adeno-associated virus-mediated transfer of pre-trans-splicing molecules, which produce normal Rpgr mRNAs via trans-splicing in the Rpgr KO retinas. This rescue upregulates autophagy through the re-expression of RPGR in KO retinas to accelerate GDP-to-GTP exchange; thus, retinal homeostasis reverts to normal. Taken together, these findings provide an important missing link for coordinating RAB37 GDP-GTP exchange via the RPGR and retinal homeostasis by autophagy regulation.


Asunto(s)
Autofagia , Proteínas Portadoras , Proteínas del Ojo , Factores de Intercambio de Guanina Nucleótido , Ratones Noqueados , Retina , Proteínas de Unión al GTP rab , Animales , Retina/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab/genética , Ratones , Humanos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/genética , Células HEK293 , Ratones Endogámicos C57BL , Guanosina Trifosfato/metabolismo , Guanosina Difosfato/metabolismo , Unión Proteica
17.
Cont Lens Anterior Eye ; 47(3): 102134, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38472014

RESUMEN

PURPOSE: To explore the biomechanical proteins different between low myopic corneas and moderate to high myopic corneas. METHODS: A total of 27 myopic corneas were used for the Tandem Mass Tag (TMT) proteomics analysis. Differentially expressed proteins (DEPs) were clustered with fold changes > 1.20 or < 0.83 and p < 0.05. Proteins and Proteins Interactions (PPIs) were conducted to find hub proteins; Uniprot database was to screen proteins with biomechanical functions, and Parallel Reaction Monitoring (PRM) was performed to verify the TMT results. Pearson analysis was used to reveal the correlations between myopic degrees and biomechanical proteins. The Immunofluorescence (IF) staining was used to observe the protein distributions. RESULTS: In total, 34 DEPs were observed between moderate myopic corneas and low myopic corneas; 103 DEPs were observed between high myopic corneas and low myopic corneas, 20 proteins overlapped. The PPIs analysis showed keratin 2, keratins 10 and PRSS1 were hub proteins. The Uniprot function analysis suggested keratin 2 and keratin 10 exhibited biomechanical functions. The PRM demonstrated keratin 2 and keratin 10 levels were significantly lower in moderate and high myopic corneas, which was consistent with the TMT proteomics results. IF staining also demonstrated keratin 2 and keratin 10 were less distributed in moderate and high myopic corneas than in low myopic corneas. CONCLUSIONS: The levels of biomechanical proteins keratin 2 and keratin 10 are significantly lower in moderate and high myopic corneas than in low myopic corneas.


Asunto(s)
Córnea , Proteínas del Ojo , Miopía , Proteómica , Humanos , Femenino , Miopía/metabolismo , Miopía/fisiopatología , Córnea/metabolismo , Masculino , Adulto , Fenómenos Biomecánicos , Proteínas del Ojo/metabolismo , Adulto Joven , Persona de Mediana Edad
18.
Int J Mol Sci ; 25(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38474133

RESUMEN

The human photoreceptor function is dependent on a highly specialised cilium. Perturbation of cilial function can often lead to death of the photoreceptor and loss of vision. Retinal ciliopathies are a genetically diverse range of inherited retinal disorders affecting aspects of the photoreceptor cilium. Despite advances in the understanding of retinal ciliopathies utilising animal disease models, they can often lack the ability to accurately mimic the observed patient phenotype, possibly due to structural and functional deviations from the human retina. Human-induced pluripotent stem cells (hiPSCs) can be utilised to generate an alternative disease model, the 3D retinal organoid, which contains all major retinal cell types including photoreceptors complete with cilial structures. These retinal organoids facilitate the study of disease mechanisms and potential therapies in a human-derived system. Three-dimensional retinal organoids are still a developing technology, and despite impressive progress, several limitations remain. This review will discuss the state of hiPSC-derived retinal organoid technology for accurately modelling prominent retinal ciliopathies related to genes, including RPGR, CEP290, MYO7A, and USH2A. Additionally, we will discuss the development of novel gene therapy approaches targeting retinal ciliopathies, including the delivery of large genes and gene-editing techniques.


Asunto(s)
Ciliopatías , Células Madre Pluripotentes Inducidas , Degeneración Retiniana , Animales , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Retina/metabolismo , Degeneración Retiniana/metabolismo , Terapia Genética , Organoides/metabolismo , Ciliopatías/metabolismo , Proteínas del Ojo/metabolismo
19.
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
20.
Biosci Rep ; 44(3)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38451099

RESUMEN

In addition to aquaporin (AQP) classes AQP1, AQP4 and AQP9 known to be expressed in mammalian brain, our recent transcriptomic analyses identified AQP0 and AQP11 in human cortex and hippocampus at levels correlated with age and Alzheimer's disease (AD) status; however, protein localization remained unknown. Roles of AQP0 and AQP11 in transporting hydrogen peroxide (H2O2) in lens and kidney prompted our hypothesis that up-regulation in brain might similarly be protective. Established cell lines for astroglia (1321N1) and neurons (SHSY5Y, differentiated with retinoic acid) were used to monitor changes in transcript levels for human AQPs (AQP0 to AQP12) in response to inflammation (simulated with 10-100 ng/ml lipopolysaccharide [LPS], 24 h), and hypoxia (5 min N2, followed by 0 to 24 h normoxia). AQP transcripts up-regulated in both 1321N1 and SHSY5Y included AQP0, AQP1 and AQP11. Immunocytochemistry in 1321N1 cells confirmed protein expression for AQP0 and AQP11 in plasma membrane and endoplasmic reticulum; AQP11 increased 10-fold after LPS and AQP0 increased 0.3-fold. In SHSY5Y cells, AQP0 expression increased 0.2-fold after 24 h LPS; AQP11 showed no appreciable change. Proposed peroxiporin roles were tested using melondialdehyde (MDA) assays to quantify lipid peroxidation levels after brief H2O2. Boosting peroxiporin expression by LPS pretreatment lowered subsequent H2O2-induced MDA responses (∼50%) compared with controls; conversely small interfering RNA knockdown of AQP0 in 1321N1 increased lipid peroxidation (∼17%) after H2O2, with a similar trend for AQP11 siRNA. Interventions that increase native brain peroxiporin activity are promising as new approaches to mitigate damage caused by aging and neurodegeneration.


Asunto(s)
Acuaporinas , Astrocitos , Proteínas del Ojo , Neuronas , Neuroprotección , Estrés Oxidativo , Humanos , Acuaporinas/genética , Acuaporinas/metabolismo , Astrocitos/metabolismo , Línea Celular , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/toxicidad , Lipopolisacáridos/farmacología , Neuronas/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA