Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Front Med (Lausanne) ; 11: 1418566, 2024.
Article in English | MEDLINE | ID: mdl-39247635

ABSTRACT

Objective: To investigate the pathogenesis of Primary Angle-Closure Glaucoma (PACG) and its relationship with the anatomical structure of the anterior segment by obtaining biometric parameters using the IOL-Master 700. Methods: A retrospective case-control study was conducted. Clinical data from 39 PACG patients and 40 normal controls treated at the Aier Eye Hospital affiliated with Wuhan University from January to December 2022 were collected. Anterior chamber depth (AC), white-to-white (WTW), lens thickness (LT), central corneal thickness (CCT), axial length (AL), corneal curvature (K1), corneal curvature (K2), and lens position (LP) were measured using the IOL-Master 700 to analyze the characteristics and differences in the anterior segment structure of both groups. Statistical methods included independent sample t-tests and logistic regression analysis. Results: Significant differences were found in the anterior segment biometric parameters between PACG patients and normal controls (p < 0.05). Anterior chamber depth, white-to-white, lens thickness, central corneal thickness, axial length, and K2 were all related to the occurrence of PACG (p < 0.05). The occurrence of PACG was negatively correlated with ACD, CCT, and AL (OR = 0.12-0.64, p < 0.05), and positively correlated with LT. Conclusion: Compared to the normal control group, PACG patients in the Hubei region have a smaller anterior segment space, narrower angles, thicker lens, thinner cornea, shorter axial length, flatter cornea, and more anteriorly positioned lens.

2.
Neurosci Bull ; 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285154

ABSTRACT

The looming stimulus-evoked flight response to approaching predators is a defensive behavior in most animals. However, how looming stimuli are detected in the retina and transmitted to the brain remains unclear. Here, we report that a group of GABAergic retinal ganglion cells (RGCs) projecting to the superior colliculus (SC) transmit looming signals from the retina to the brain, mediating the looming-evoked flight behavior by releasing GABA. GAD2-Cre and vGAT-Cre transgenic mice were used in combination with Cre-activated anterograde or retrograde tracer viruses to map the inputs to specific GABAergic RGC circuits. Optogenetic technology was used to assess the function of SC-projecting GABAergic RGCs (scpgRGCs) in the SC. FDIO-DTA (Flp-dependent Double-Floxed Inverted Open reading frame-Diphtheria toxin) combined with the FLP (Florfenicol, Lincomycin & Prednisolone) approach was used to ablate or silence scpgRGCs. In the mouse retina, GABAergic RGCs project to different brain areas, including the SC. ScpgRGCs are monosynaptically connected to parvalbumin-positive SC neurons known to be required for the looming-evoked flight response. Optogenetic activation of scpgRGCs triggers GABA-mediated inhibition in SC neurons. Ablation or silencing of scpgRGCs compromises looming-evoked flight responses without affecting image-forming functions. Our study reveals that scpgRGCs control the looming-evoked flight response by regulating SC neurons via GABA, providing novel insight into the regulation of innate defensive behaviors.

3.
Heliyon ; 9(12): e23002, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38144322

ABSTRACT

Background: Neurodegenerative retinal diseases such as retinitis pigmentosa are serious disorders that may cause irreversible visual impairment. Ferroptosis is a novel type of programmed cell death, and the involvement of ferroptosis in retinal degeneration is still unclear. This study aimed to investigate the related ferroptosis genes in a mice model of retinal degeneration induced by light damage. Methods: A public dataset of GSE10528 deriving from the Gene Expression Omnibus database was analyzed to identify the differentially expressed genes (DEGs). Gene set enrichment analysis between light damage and control group was conducted. The differentially expressed ferroptosis-related genes (DE-FRGs) were subsequently identified by intersecting the DEGs with a ferroptosis genes dataset retrieved from the FerrDb database. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were further performed using the DE-FRGs. A protein-protein interaction (PPI) network was constructed to identify hub ferroptosis-related genes (HFRGs). The microRNAs (miRNAs)-HFRGs, transcription factors (TFs)-HFRGs networks as well as target drugs potentially interacting with HFRGs were analyzed utilizing bioinformatics algorithms. Results: A total of 932 DEGs were identified between the light damage and control group. Among these, 25 genes were associated with ferroptosis. GO and KEGG analyses revealed that these DE-FRGs were mainly enriched in apoptotic signaling pathway, response to oxidative stress and autophagy, ferroptosis, necroptosis and cytosolic DNA-sensing pathway. Through PPI network analysis, six hub ferroptosis-related genes (Jun, Stat3, Hmox1, Atf3, Hspa5 and Ripk1) were ultimately identified. All of them were upregulated in light damage retinas, as verified by the GSE146176 dataset. Bioinformatics analyses predicated that 116 miRNAs, 23 TFs and several potential therapeutic compounds might interact with the identified HFRGs. Conclusion: Our study may provide novel potential biomarkers, therapeutic targets and new insights into the ferroptosis landscape in retinal neurodegenerative diseases.

4.
Exp Eye Res ; 210: 108728, 2021 09.
Article in English | MEDLINE | ID: mdl-34390734

ABSTRACT

PURPOSE: Activation of bone morphogenetic protein (BMP) 4 signaling promotes the survival of retinal ganglion cell (RGC) after acute injury. Chordin-like 1 (CHRDL1) is an endogenous BMP antagonist. In this study, we researched whether CHRDL1 was involved in BMP4 signaling and regulation of RGC degeneration in a mouse model of glaucoma. METHODS: Magnetic microbeads were intracameral injected to induce experimental glaucoma in a mouse model. A recombinant adeno-associated virus (rAAV) system was designed for overexpression of BMP4 or CHRDL1 in mouse retina. Immunohistochemistry and hematoxylin-eosin (HE) stains were performed to identify changes in retinal morphology. Electroretinogram (ERG) recordings were used to assess changes in visual function. RESULTS: The mRNA expression levels of Bmp4 and its downstream BMPRIa, small mothers against decapentaplegic 1 (Smad1), were significantly upregulated in retinas with glaucoma. RGC survival was significantly enhanced in the beads + AAV-BMP4 group and significantly reduced in the beads + AAV-CHRDL1 group, compared with the beads + AAV-EGFP group. Similar results were observed in retinal explant culture in vitro. Consistent with these findings, the photopic negative response (PhNR)responses in ERG, which indicate RGC function, were restored in mice overexpressing BMP4, whereas a-wave and b-wave responses were not. Activation of CHRLD1 inhibited Smad1/5/8 phosphorylation and exacerbated RGC damage. The expression of Glial fibrillary acidic protein (GFAP) was decreased significantly in beads + AAV-BMP4 group. CONCLUSIONS: BMP4 promoted RGC survival and visual function in an experimental glaucoma model. Activation of CHRDL1 exaggerated RGC degeneration by inhibiting the BMP4/Smad1/5/8 pathway. The mechanism of BMP4/Smad1/5/8 pathway may be related to the inhibition of glial cell activation. Our studies suggested that BMP4 and CHRLD1 might serve as therapeutic targets in glaucoma.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Disease Models, Animal , Eye Proteins/metabolism , Gene Expression Regulation/physiology , Glaucoma/metabolism , Nerve Tissue Proteins/metabolism , Retinal Ganglion Cells/physiology , Animals , Bone Morphogenetic Protein 4/antagonists & inhibitors , Cell Survival , Dependovirus/genetics , Electroretinography , Genetic Vectors , Glaucoma/physiopathology , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Intraocular Pressure/physiology , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retina/physiopathology
5.
Invest Ophthalmol Vis Sci ; 62(7): 1, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34061953

ABSTRACT

Purpose: The neuronal ELAV-like proteins (nElavls; Elavl2, Elavl3, Elavl4) have been known to regulate neuronal differentiation, maintenance, and axonogenesis in the brain. However, the specific role of nElavls in retina remains unclear. Here, we attempted to identify the expression pattern of Elavl2 during retinogenesis and aimed to decipher the function of Elavl2 in the retina. Methods: We have used the Cre-loxP system to conditionally inactivate Elavl2 in order to examine its role in developing retina. Eyes were collected for histology, immunohistochemistry, and TUNEL analysis to identify the structure of retina, and examined by RNA sequencing to analyze the function and pathway enrichment of differentially expressed genes in transgenic mice. Moreover, the mechanism by which Elavl2 regulates the differentiation of amacrine cells (ACs) was explored by RNA immunoprecipitation assays. Finally, eyes were functionally assessed by whole-cell patch-clamp, electroretinography (ERG) and optomotor response. Results: Elavl2 was expressed in retinal progenitor cells and retinal ganglion cells (RGCs), ACs, and horizontal cells. Retina-specific ablation of Elavl2 led to the loss of ACs and the transcription factors involved in ACs differentiation were also downregulated. In addition, the spontaneous activities of RGCs were obviously increased in Elavl2-deficient mice. Meanwhile, the loss of ACs that induced by Elavl2 deficiency lead to a decrease in ERG responses and visual acuity. Conclusions: Elavl2 is an intrinsic factor that involved in the differentiation of ACs subtype during retinogenesis, and essential for maintaining the normal retinal function.


Subject(s)
Amacrine Cells/physiology , ELAV-Like Protein 2/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Retina , Animals , Cell Differentiation , Electroretinography/methods , Gene Expression Regulation, Developmental , Interneurons/physiology , Mice , Mice, Transgenic , Patch-Clamp Techniques/methods , Retina/embryology , Retina/growth & development , Retina/physiology , Retinal Ganglion Cells/physiology , Transcriptome
6.
Microvasc Res ; 136: 104146, 2021 07.
Article in English | MEDLINE | ID: mdl-33610563

ABSTRACT

The goal of this study was to evaluate the effects of CM082, a novel vascular endothelial growth factor (VEGF) receptor-2 tyrosine kinase inhibitor, on human umbilical vein endothelial cells (HUVECs), and oxygen-induced retinopathy (OIR) mice. HUVECs were stimulated with rHuVEGF165 and then treated with CM082 to assess the antiangiogenic effects of CM082; subsequently, proliferation, wound-healing migration, Transwell invasion, tube formation assays, and Western blotting were performed in vitro. Retinal neovascularization tufts, avascular area, and TUNEL assays were estimated for OIR mice after intraperitoneal injection with CM082. CM082 significantly inhibited proliferation, migration, invasion, and tube formation induced by stimulation of HUVECs with rHuVEGF165; this inhibitory effect was mediated by blocking VEGFR2 activation. CM082 significantly inhibited retinal neovascularization and avascular area and did not increase apoptosis in the retina of OIR mice. The findings demonstrated that CM082 exhibits highly antiangiogenic effects in HUVECs and OIR mice. Thus, it may serve as an alternative treatment for neovascular eye disease in the future.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Indoles/pharmacology , Neovascularization, Physiologic/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Pyrrolidines/pharmacology , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/drug therapy , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hyperoxia/complications , Mice, Inbred C57BL , Retinal Neovascularization/enzymology , Retinal Neovascularization/etiology , Retinal Neovascularization/pathology , Retinopathy of Prematurity/enzymology , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/pathology , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32364220

ABSTRACT

Choroideremia is a complex form of blindness-causing retinal degeneration. The aim of the present study was to investigate the pathogenic variant and molecular etiology associated with choroideremia in a Chinese family. All available family members underwent detailed ophthalmological examinations. Whole exome sequencing, bioinformatics analysis, Sanger sequencing, and co-segregation analysis of family members were used to validate sequencing data and confirm the presence of the disease-causing gene variant. The proband was diagnosed with choroideremia on the basis of clinical manifestations. Whole exome sequencing showed that the proband had a hemizygous variant in the CHM gene, c.22delG p. (Glu8Serfs*4), which was confirmed by Sanger sequencing and found to co-segregate with choroideremia. The variant was classified as likely pathogenic and has not previously been described. These results expand the spectrum of variants in the CHM gene, thus potentially enriching the understanding of the molecular basis of choroideremia. Moreover, they may provide insight for future choroideremia diagnosis and gene therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Choroideremia/genetics , Exome Sequencing , Genetic Variation , Vision, Ocular/genetics , Adult , Asian People/genetics , Child , China , Choroideremia/diagnosis , Choroideremia/ethnology , Choroideremia/physiopathology , Female , Genetic Predisposition to Disease , Heredity , Humans , Male , Middle Aged , Pedigree , Phenotype
8.
Front Cell Dev Biol ; 8: 128, 2020.
Article in English | MEDLINE | ID: mdl-32211407

ABSTRACT

Although an increasing number of disease genes have been identified, the exact cellular mechanisms of retinitis pigmentosa (RP) remain largely unclear. Retinal organoids (ROs) derived from the induced pluripotent stem cells (iPSCs) of patients provide a potential but unvalidated platform for deciphering disease mechanisms and an advantageous tool for preclinical testing of new treatments. Notably, early-onset RP has been extensively recapitulated by patient-iPSC-derived ROs. However, it remains a challenge to model late-onset disease in a dish due to its chronicity, complexity, and instability. Here, we generated ROs from late-onset RP proband-derived iPSCs harboring a PDE6B mutation. Transcriptome analysis revealed a remarkably distinct gene expression profile in the patient ROs at differentiation day (D) 230. Changes in the expression genes regulating cGMP hydrolysis prompted the elevation of cGMP levels, which was verified by a cGMP enzyme-linked immunosorbent assay (ELISA) in patient ROs. Furthermore, significantly higher cGMP levels in patient ROs than in control ROs at D193 and D230 might lead to impaired formation of synaptic connections and the connecting cilium in photoreceptor cells. In this study, we established the first late-onset RP model with a consistent phenotype using an in vitro cell culture system and provided new insights into the PDE6B-related mechanism of RP.

9.
Biochem Genet ; 57(4): 540-554, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30652268

ABSTRACT

CNGA1 encodes a membrane protein on rod photoreceptor related to phototransduction. The present study was to identify a novel mutation of CNGA1 associated with autosomal recessive retinitis pigmentosa by using next generation sequencing of a Chinese family. Next generation sequencing and Sanger sequencing has identified a compound heterozygous mutation in CNGA1 gene, c0.472 del C (reported) and c0.829G>A (novel mutation, same as c0.622G>A according to NM_000087.3) of the proband. SIFT and Polyphen-2 predicted the CNGA1 G622A site to be possibly deleterious. Evolutionary conservation analysis of amino acid residues showed this aspartic acid is highly conserved between species, and protein structure prediction by I-TASSER server indicated that the D208N mutation induced a large disappear of interactions between S2 and S4. Flag-tagged CNGA1 and mutant G622A cDNA were generated and inserted into pCIG-eGFP vectors. Transfection of human embryonic kidney 293T cells was performed with lipofectamine. Interestingly, western blot and immunofluorescence results indicated that the expression of mutant CNGA1 (D208N) decreased significantly, especially on the membrane of transfected HEK293T cells. The novel variant c0.622G>A (p. D208N) in this study enriched the CNGA1 mutation spectrum. Besides, this mutant was predicted "possibly damaging" due to bioinformatics analysis and validated by laboratorial experiments. Our study suggests that this mutation lead to the CNGA1 protein reduction from the cell membrane.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Mutation, Missense , Retinitis Pigmentosa/genetics , Adult , Amino Acid Substitution , Cell Membrane/metabolism , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/deficiency , Female , Genes, Recessive , HEK293 Cells , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Male , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Pedigree , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinitis Pigmentosa/metabolism , Sequence Analysis, DNA , Sequence Deletion , Transfection
11.
Stem Cell Reports ; 10(4): 1267-1281, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29526738

ABSTRACT

Retinitis pigmentosa (RP) is an irreversible, inherited retinopathy in which early-onset nyctalopia is observed. Despite the genetic heterogeneity of RP, RPGR mutations are the most common causes of this disease. Here, we generated induced pluripotent stem cells (iPSCs) from three RP patients with different frameshift mutations in the RPGR gene, which were then differentiated into retinal pigment epithelium (RPE) cells and well-structured retinal organoids possessing electrophysiological properties. We observed significant defects in photoreceptor in terms of morphology, localization, transcriptional profiling, and electrophysiological activity. Furthermore, shorted cilium was found in patient iPSCs, RPE cells, and three-dimensional retinal organoids. CRISPR-Cas9-mediated correction of RPGR mutation rescued photoreceptor structure and electrophysiological property, reversed the observed ciliopathy, and restored gene expression to a level in accordance with that in the control using transcriptome-based analysis. This study recapitulated the pathogenesis of RPGR using patient-specific organoids and achieved targeted gene therapy of RPGR mutations in a dish as proof-of-concept evidence.


Subject(s)
Ciliopathies/therapy , Genetic Therapy , Induced Pluripotent Stem Cells/pathology , Organoids/pathology , Photoreceptor Cells/pathology , Retina/pathology , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/therapy , Cell Differentiation , Ciliopathies/pathology , Ciliopathies/physiopathology , Eye Proteins/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Photoreceptor Cells/metabolism , Potassium Channels/metabolism , Retinitis Pigmentosa/physiopathology
12.
Invest Ophthalmol Vis Sci ; 58(2): 801-811, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28152141

ABSTRACT

Purpose: Accumulating evidence has demonstrated that excessive immunoreaction plays a prominent role in the pathogenesis of dry AMD. Toll-like receptor 3 (TLR3) can be activated by double-stranded (ds)RNA in retinal pigment epithelia and trigger an innate immunity-mediated inflammatory response. However, its role in photoreceptor cells, the effectors of AMD geographic atrophy, remains unclear. Methods: The expression of TLR3 was examined in mouse retina and in a murine photoreceptor cell line (661W). Retinal structure, function, and cell death in the polyinosine-polycytidylic acid (poly I:C)-treated retina were investigated by optical coherence tomography, electroretinography (ERG), and immunostaining. Cytokine and chemokine expression as well as cell death were measured in poly I:C-exposed 661W cells and explant retinas. By comparing the RNA sequencing (seq) data of 661W cells and murine retina, we comprehensively investigated the contribution of photoreceptor in poly I:C-induced retinal immune response. Results: Toll-like receptor 3 was highly expressed in the inner segment of the photoreceptor and in 661W cells. We found poly I:C induced significant retinal structural damages and impairment of ERG responses. Focal ERG demonstrated that injected and parainjected zones were functionally damaged by poly I:C. In addition, poly I:C acted on cultured photoreceptor cells directly and evoked an inflammatory response that exhibited similarities with the immune response in mouse retina. Moreover, TLR3 activation initiated cell death in murine photoreceptor cells in vivo and in vitro. Additionally, poly I:C initiated immune response in explant retinas. Conclusions: We deciphered the TLR3-mediated inflammatory response in photoreceptor cells. Our findings suggested TLR3-mediated inflammatory response in photoreceptor cells may play an important role in dry AMD, offering new insights of potential treatments targeting photoreceptor immunity.


Subject(s)
Cell Death/physiology , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Toll-Like Receptor 3/metabolism , Analysis of Variance , Animals , Cell Death/drug effects , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Electroretinography , Interferon Inducers/pharmacology , Mice , Mice, Inbred C57BL , Neuroglia/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/immunology , Poly I-C/pharmacology , Retina/drug effects , Retina/physiopathology , Sequence Analysis, RNA , Tomography, Optical Coherence
13.
Sci Rep ; 7: 43062, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28216641

ABSTRACT

Precursor messenger RNA (Pre-mRNA) splicing is an essential biological process in eukaryotic cells. Genetic mutations in many spliceosome genes confer human eye diseases. Mutations in the pre-mRNA splicing factor, RP9 (also known as PAP1), predispose autosomal dominant retinitis pigmentosa (adRP) with an early onset and severe vision loss. However, underlying molecular mechanisms of the RP9 mutation causing photoreceptor degeneration remains fully unknown. Here, we utilize the CRISPR/Cas9 system to generate both the Rp9 gene knockout (KO) and point mutation knock in (KI) (Rp9, c.A386T, P.H129L) which is analogous to the reported one in the retinitis pigmentosa patients (RP9, c.A410T, P.H137L) in 661 W retinal photoreceptor cells in vitro. We found that proliferation and migration were significantly decreased in the mutated cells. Gene expression profiling by RNA-Seq demonstrated that RP associated genes, Fscn2 and Bbs2, were down-regulated in the mutated cells. Furthermore, pre-mRNA splicing of the Fscn2 gene was markedly affected. Our findings reveal a functional relationship between the ubiquitously expressing RP9 and the disease-specific gene, thereafter provide a new insight of disease mechanism in RP9-related retinitis pigmentosa.


Subject(s)
Cell Proliferation , Gene Expression Profiling , Point Mutation , RNA Splicing Factors/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Cell Movement , Gene Expression Regulation , Gene Knockout Techniques , Humans , Mice , Microfilament Proteins/metabolism , Proteins , RNA Splicing Factors/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology , Sequence Analysis, RNA
14.
BMJ Open ; 6(4): e010649, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27036142

ABSTRACT

OBJECTIVES: Infantile nystagmus (IN) is a genetically heterogeneous condition characterised by involuntary rhythmic oscillations of the eyes accompanied by different degrees of vision impairment. Two genes have been identified as mainly causing IN: FRMD7 and GPR143. The aim of our study was to identify the genetic basis of both sporadic IN and X-linked IN. DESIGN: Prospective analysis. PATIENTS: Twenty Chinese patients, including 15 sporadic IN cases and 5 from X-linked IN families, were recruited and underwent molecular genetic analysis. We first performed PCR-based DNA sequencing of the entire coding region and the splice junctions of the FRMD7 and GPR143 genes in participants. Mutational analysis and co-segregation confirmation were then performed. SETTING: All clinical examinations and genetic experiments were performed in the Eye Hospital of Wenzhou Medical University. RESULTS: Two mutations in the FRMD7 gene, including one novel nonsense mutation (c.1090C>T, p.Q364X) and one reported missense mutation (c.781C>G, p.R261G), were identified in two of the five (40%) X-linked IN families. However, none of putative mutations were identified in FRMD7 or GPR143 in any of the sporadic cases. CONCLUSIONS: The results suggest that mutations in FRMD7 appeared to be the major genetic cause of X-linked IN, but not of sporadic IN. Our findings provide further insights into FRMD7 mutations, which could be helpful for future genetic diagnosis and genetic counselling of Chinese patients with nystagmus.


Subject(s)
Asian People/genetics , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Eye Proteins/genetics , Genetic Diseases, X-Linked/genetics , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mutation, Missense , Nystagmus, Congenital/genetics , Nystagmus, Pathologic/genetics , China/epidemiology , DNA Mutational Analysis/methods , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Predisposition to Disease , Genetic Testing , Humans , Infant , Male , Nystagmus, Congenital/diagnosis , Nystagmus, Pathologic/diagnosis , Prospective Studies
15.
Mol Med Rep ; 12(4): 5983-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26260533

ABSTRACT

Retinitis pigmentosa (RP) is an inherited retinopathy, which affects the photoreceptors in the retina. Lysophosphatidylcholine acyltransferase (LPCAT) is a critical phospholipid biosynthesis enzyme, which promotes the conversion of lysophosphatidylcholine into phosphatidylcholine in the remodeling pathway of PC biosynthesis. A previous study reported a homozygous insertion in the LPCAT1 gene in mice exhibiting retinal degeneration (rd11). However, whether genetic mutations in LPCAT1 predispose individuals to RP remains to be elucidated. Therefore, the aim of the present study was to investigate whether LPCAT1 mutations exist in patients with RP. A total of 50 unrelated patients diagnosed with either a sporadic or recessive inheritance pattern of RP were recruited in the present study. All of the patients were comprehensively screened for genes associated with the predisposition of RP, and no pathogenic mutations were identified. Reverse transcription-polymerase chain reaction and Sanger sequencing were performed to investigate the coding regions and exon­intron boundaries of the LPCAT1 gene in the recruited patients. In total, three genetic variations in the coding regions, which lead to amino acid changes, were identified. Although two of these mutations were predicted to be pathogenic, co­segregation analysis in the pedigrees excluded these as disease­causing mutations. In addition, the LPCAT1 gene was screen in a panel of RP patients who exhibited no identifiable mutations in any of the known RP­associated genes. No disease­causing mutations in the LPCAT1 gene were identified, indicating that LPCAT1 either does not confer a genetic predisposition to RP, or that the incidence of mutations in LPCAT1 is particularly rare in patients with RP.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/genetics , Retinitis Pigmentosa/genetics , Adolescent , Adult , Aged , Animals , Asian People/genetics , Child , Child, Preschool , DNA Mutational Analysis , Exons , Female , Genetic Testing , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mutation , Pedigree , Retina/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL