Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 17218, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36241656

RESUMEN

To delineate the genetic bases of primary congenital glaucoma (PCG), we ascertained a large cohort consisting of 48 consanguineous families. Of these, we previously reported 26 families with mutations in CYP1B1 and six families with LTBP2, whereas the genetic bases responsible for PCG in 16 families remained elusive. We employed next-generation whole exome sequencing to delineate the genetic basis of PCG in four of these 16 familial cases. Exclusion of linkage to reported PCG loci was established followed by next-generation whole exome sequencing, which was performed on 10 affected individuals manifesting cardinal systems of PCG belonging to four unresolved families along with four control samples consisting of genomic DNAs of individuals harboring mutations in CYP1B1 and LTBP2. The analyses of sequencing datasets failed to identify potential causal alleles in the 10 exomes whereas c.1169G > A (p. Arg390His) in CYP1B1 and c.3427delC (p.Gln1143Argfs*35) in LTBP2 were identified in the control samples. Taken together, next-generation whole exome sequencing failed to delineate the genetic basis of PCG in familial cases excluded from mutations in CYP1B1 and LTBP2. These data strengthen the notion that compound heterozygous coding variants or non-coding variants might contribute to PCG.


Asunto(s)
Exoma , Glaucoma , Consanguinidad , Exoma/genética , Glaucoma/congénito , Glaucoma/genética , Humanos , Proteínas de Unión a TGF-beta Latente/genética , Mutación , Secuenciación del Exoma
2.
Hum Genome Var ; 9(1): 31, 2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36075891

RESUMEN

Here we report a consanguineous Pakistani family with multiple affected individuals with autosomal recessive congenital cataract (arCC). Exclusion analysis established linkage to chromosome 22q, and Sanger sequencing coupled with PCR-based chromosome walking identified a large homozygous genomic deletion. Our data suggest that this deletion leads to CRYBB2-CRYBB2P1 fusion, consisting of exons 1-5 of CRYBB2 and exon 6 of CRYBB2P1, the latter of which harbors the c.463 C > T (p.Gln155*) mutation, and is responsible for arCC.

3.
Autophagy ; 18(9): 2198-2215, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35343376

RESUMEN

FYCO1 (FYVE and coiled-coil domain containing 1) is an adaptor protein, expressed ubiquitously and required for microtubule-dependent, plus-end-directed transport of macroautophagic/autophagic vesicles. We have previously shown that loss-of-function mutations in FYCO1 cause cataracts with no other ocular and/or extra-ocular phenotype. Here, we show fyco1 homozygous knockout (fyco1-/-) mice recapitulate the cataract phenotype consistent with a critical role of FYCO1 and autophagy in lens morphogenesis. Transcriptome coupled with proteome and metabolome profiling identified many autophagy-associated genes, proteins, and lipids respectively perturbed in fyco1-/- mice lenses. Flow cytometry of FYCO1 (c.2206C>T) knock-in (KI) human lens epithelial cells revealed a decrease in autophagic flux and autophagic vesicles resulting from the loss of FYCO1. Transmission electron microscopy showed cellular organelles accumulated in FYCO1 (c.2206C>T) KI lens-like organoid structures and in fyco1-/- mice lenses. In summary, our data confirm the loss of FYCO1 function results in a diminished autophagic flux, impaired organelle removal, and cataractogenesis.Abbreviations: CC: congenital cataracts; DE: differentially expressed; ER: endoplasmic reticulum; FYCO1: FYVE and coiled-coil domain containing 1; hESC: human embryonic stem cell; KI: knock-in; OFZ: organelle-free zone; qRT-PCR: quantitative real-time PCR; PE: phosphatidylethanolamine; RNA-Seq: RNA sequencing; SD: standard deviation; sgRNA: single guide RNA; shRNA: shorthairpin RNA; TEM: transmission electron microscopy; WT: wild type.


Asunto(s)
Catarata , Cristalino , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Autofagia , Catarata/genética , Catarata/metabolismo , Diferenciación Celular , Retículo Endoplásmico/metabolismo , Humanos , Cristalino/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Factores de Transcripción/metabolismo
4.
PLoS Genet ; 17(10): e1009848, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34662339

RESUMEN

Patients with inherited retinal dystrophies (IRDs) were recruited from two understudied populations: Mexico and Pakistan as well as a third well-studied population of European Americans to define the genetic architecture of IRD by performing whole-genome sequencing (WGS). Whole-genome analysis was performed on 409 individuals from 108 unrelated pedigrees with IRDs. All patients underwent an ophthalmic evaluation to establish the retinal phenotype. Although the 108 pedigrees in this study had previously been examined for mutations in known IRD genes using a wide range of methodologies including targeted gene(s) or mutation(s) screening, linkage analysis and exome sequencing, the gene mutations responsible for IRD in these 108 pedigrees were not determined. WGS was performed on these pedigrees using Illumina X10 at a minimum of 30X depth. The sequence reads were mapped against hg19 followed by variant calling using GATK. The genome variants were annotated using SnpEff, PolyPhen2, and CADD score; the structural variants (SVs) were called using GenomeSTRiP and LUMPY. We identified potential causative sequence alterations in 61 pedigrees (57%), including 39 novel and 54 reported variants in IRD genes. For 57 of these pedigrees the observed genotype was consistent with the initial clinical diagnosis, the remaining 4 had the clinical diagnosis reclassified based on our findings. In seven pedigrees (12%) we observed atypical causal variants, i.e. unexpected genotype(s), including 4 pedigrees with causal variants in more than one IRD gene within all affected family members, one pedigree with intrafamilial genetic heterogeneity (different affected family members carrying causal variants in different IRD genes), one pedigree carrying a dominant causative variant present in pseudo-recessive form due to consanguinity and one pedigree with a de-novo variant in the affected family member. Combined atypical and large structural variants contributed to about 20% of cases. Among the novel mutations, 75% were detected in Mexican and 50% found in European American pedigrees and have not been reported in any other population while only 20% were detected in Pakistani pedigrees and were not previously reported. The remaining novel IRD causative variants were listed in gnomAD but were found to be very rare and population specific. Mutations in known IRD associated genes contributed to pathology in 63% Mexican, 60% Pakistani and 45% European American pedigrees analyzed. Overall, contribution of known IRD gene variants to disease pathology in these three populations was similar to that observed in other populations worldwide. This study revealed a spectrum of mutations contributing to IRD in three populations, identified a large proportion of novel potentially causative variants that are specific to the corresponding population or not reported in gnomAD and shed light on the genetic architecture of IRD in these diverse global populations.


Asunto(s)
Etnicidad/genética , Degeneración Retiniana/genética , Consanguinidad , Análisis Mutacional de ADN/métodos , Exoma/genética , Proteínas del Ojo/genética , Femenino , Estudios de Asociación Genética/métodos , Ligamiento Genético/genética , Genotipo , Humanos , Masculino , México , Mutación/genética , Pakistán , Linaje , Retina/patología , Secuenciación del Exoma/métodos , Secuenciación Completa del Genoma/métodos
5.
Sci Rep ; 11(1): 18801, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34552108

RESUMEN

Here, we report a multi OMIC (transcriptome, proteome, and metabolome) approach to investigate molecular changes in lens fiber cells (FC) of mice exposed to cigarette smoke (CS). Pregnant mice were placed in a whole-body smoke chamber and a few days later pups were born, which were exposed to CS for 5 hours/day, 5 days/week for a total of 3½ months. We examined the mice exposed to CS for CS-related cataractogenesis after completion of the CS exposure but no cataracts were observed. Lenses of CS-exposed and age-matched, untreated control mice were extracted and lens FC were subjected to multi OMIC profiling. We identified 348 genes, 130 proteins, and 14 metabolites exhibiting significant (p < 0.05) differential levels in lens FC of mice exposed to CS, corresponding to 3.6%, 4.3%, and 5.0% of the total genes, protein, and metabolites, respectively identified in this study. Our multi OMIC approach confirmed that only a small fraction of the transcriptome, the proteome, and the metabolome was perturbed in the lens FC of mice exposed to CS, which suggests that exposure of CS had a minimal effect on the mouse lens. It is worth noting that while our results confirm that CS exposure does not have a substantial impact on the molecular landscape of the mouse lens FC, we cannot rule out that CS exposure for longer durations and/or in combination with other morbidities or environmental factors would have a more robust effect and/or result in cataractogenesis.


Asunto(s)
Catarata/etiología , Cristalino/efectos de los fármacos , Contaminación por Humo de Tabaco/efectos adversos , Animales , Femenino , Perfilación de la Expresión Génica , Exposición por Inhalación/efectos adversos , Cristalino/metabolismo , Masculino , Metabolómica , Ratones , Ratones Endogámicos C57BL , Embarazo , Proteómica
6.
Stem Cell Reports ; 16(9): 2320-2335, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34358452

RESUMEN

Here, we evaluate the efficacy of cryopreserved human embryonic stem cell (hESC)-derived corneal endothelial cells (CECs) to form a functional monolayer of corneal endothelium (CE) in rabbits and monkeys. We injected cryopreserved hESC-derived CECs into the anterior chamber of rabbits and monkeys either immediately after mechanical scraping of the central CE or a few days later when corneal edema developed. All preclinical models developed deturgesced and clear corneas following the injection of cryopreserved hESC-derived CECs and remained comparable to the corneas of the untreated eye. Confocal scanning microscopy confirmed an intact structure of hexagonal/polygonal cells and immunohistochemical analysis illustrated a monolayer expressing barrier and pump function proteins in the regenerated CE. The necropsy examination confirmed no remarkable change in multiple tissues assessed for teratoma formation. In conclusion, our data demonstrate the efficacy of cryopreserved hESC-derived CECs to form a functional CE on the denuded Descemet's membrane.


Asunto(s)
Trasplante de Células , Trasplante de Córnea , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Corneal/citología , Células Madre Pluripotentes/citología , Animales , Biomarcadores , Diferenciación Celular/genética , Trasplante de Células/métodos , Células Cultivadas , Trasplante de Córnea/métodos , Criopreservación , Técnica del Anticuerpo Fluorescente , Haplorrinos , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Inmunofenotipificación , Células Madre Pluripotentes/metabolismo , Regeneración
7.
Invest Ophthalmol Vis Sci ; 62(3): 3, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33651877

RESUMEN

Purpose: To investigate changes at a molecular level in the mouse corneal endothelium (CE) exposed to chronic cigarette smoke (CS). Methods: Pregnant mice (gestation days 18-20) were placed in a whole-body exposure smoking chamber, and a few days later pups were born. After 3.5 months of CS exposure, a ConfoScan4 scanning microscope was used to examine the corneal endothelial cells (CECs) of CS-exposed and control (Ct) mice. The CE was peeled under a microscope and maintained as four biological replicates (two male and two female) for CS-exposed and Ct mice; each replicate consisted of 16 CEs. The proteome of the CE was investigated through mass spectrometry. Results: The CE images of CS-exposed and Ct mice revealed a difference in the shape of CECs accompanied by a nearly 10% decrease in CEC density (P < 0.00003) following CS exposure. Proteome profiling identified a total of 524 proteins exhibiting statistically significant changes in CE from CS-exposed mice. Importantly, proteins associated with Descemet's membrane (DM), including COL4α1, COL4α2, COL4α3, COL4α4, COL4α5, COL4α6, COL8α1, COL8α2, and FN1, among others, exhibited diminished protein levels in the CE of CS-exposed mice. Conclusions: Our data confirm that exposure to CS results in reduced CEC density accompanied by diminished levels of multiple collagen and extracellular matrix proteins associated with DM.


Asunto(s)
Fumar Cigarrillos/efectos adversos , Pérdida de Celulas Endoteliales de la Córnea/etiología , Lámina Limitante Posterior/metabolismo , Proteínas del Ojo/metabolismo , Proteoma/metabolismo , Animales , Cámaras de Exposición Atmosférica , Pérdida de Celulas Endoteliales de la Córnea/metabolismo , Pérdida de Celulas Endoteliales de la Córnea/patología , Femenino , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Embarazo , Preñez
8.
Hum Genet ; 140(4): 649-666, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33389129

RESUMEN

Peroxisomes, single-membrane intracellular organelles, play an important role in various metabolic pathways. The translocation of proteins from the cytosol to peroxisomes depends on peroxisome import receptor proteins and defects in peroxisome transport result in a wide spectrum of peroxisomal disorders. Here, we report a large consanguineous family with autosomal recessive congenital cataracts and developmental defects. Genome-wide linkage analysis localized the critical interval to chromosome 12p with a maximum two-point LOD score of 4.2 (θ = 0). Next-generation exome sequencing identified a novel homozygous missense variant (c.653 T > C; p.F218S) in peroxisomal biogenesis factor 5 (PEX5), a peroxisome import receptor protein. This missense mutation was confirmed by bidirectional Sanger sequencing. It segregated with the disease phenotype in the family and was absent in ethnically matched control chromosomes. The lens-specific knockout mice of Pex5 recapitulated the cataractous phenotype. In vitro import assays revealed a normal capacity of the mutant PEX5 to enter the peroxisomal Docking/Translocation Module (DTM) in the presence of peroxisome targeting signal 1 (PTS1) cargo protein, be monoubiquitinated and exported back into the cytosol. Importantly, the mutant PEX5 protein was unable to form a stable trimeric complex with peroxisomal biogenesis factor 7 (PEX7) and a peroxisome targeting signal 2 (PTS2) cargo protein and, therefore, failed to promote the import of PTS2 cargo proteins into peroxisomes. In conclusion, we report a novel missense mutation in PEX5 responsible for the defective import of PTS2 cargo proteins into peroxisomes resulting in congenital cataracts and developmental defects.


Asunto(s)
Catarata/genética , Mutación Missense , Señales de Direccionamiento al Peroxisoma , Receptor de la Señal 1 de Direccionamiento al Peroxisoma/genética , Peroxisomas/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico Activo , Catarata/congénito , Catarata/metabolismo , Cromosomas Humanos Par 12 , Consanguinidad , Femenino , Ligamiento Genético , Humanos , Cristalino/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptor de la Señal 1 de Direccionamiento al Peroxisoma/metabolismo , Proteína Sequestosoma-1/metabolismo , Secuenciación del Exoma
9.
Exp Eye Res ; 202: 108343, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33159909

RESUMEN

Metabolomics is a study of the entire repertoire of metabolites in a cell at a particular time point. Here, we investigate the mouse lens at multiple embryonic and postnatal time points to establish the metabolome profile during early lens development. The lenses were isolated at six time points including embryonic day 15 (E15) and E18 and postnatal day 0 (P0), P3, P6, and P9. A total of four biological replicates of each time point, each consisting of 25 mg of lens tissue were preserved. Sample preparation was performed by protein precipitation followed by centrifugation to remove proteins and recover metabolites. The resulting extract was subjected to reverse phase/ultra-performance liquid chromatography-tandem mass spectrometry. Metabolome profiling identified a total of 353 metabolites in mouse lens, marked with an abundance of collagen, antioxidant, glycosaminoglycans, lipid, amino acid, and energy-related metabolites. A comparative metabolome analysis identified >200 metabolites exhibiting increased levels (p < 0.05) at latter time points relative to E15. Principal component analysis revealed distinct metabolomic signatures running from E15 to P9 while random forest analysis categorized lipid-, amino acid-, and nucleotide-related metabolites contributing significantly to the separation of the time points. To the best of our knowledge, this is the first report investigating the mouse lens metabolome at multiple embryonic and postnatal time points.


Asunto(s)
Proteínas del Ojo/metabolismo , Cristalino/metabolismo , Metaboloma/fisiología , Animales , Animales Recién Nacidos , Cromatografía Liquida , Ratones , Ratones Endogámicos C57BL , Modelos Animales
10.
Sci Data ; 7(1): 350, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33051442

RESUMEN

Here we report whole genome sequencing of four individuals (H3, H4, H5, and H6) from a family of Pakistani descent. Whole genome sequencing yielded 1084.92, 894.73, 1068.62, and 1005.77 million mapped reads corresponding to 162.73, 134.21, 160.29, and 150.86 Gb sequence data and 52.49x, 43.29x, 51.70x, and 48.66x average coverage for H3, H4, H5, and H6, respectively. We identified 3,529,659, 3,478,495, 3,407,895, and 3,426,862 variants in the genomes of H3, H4, H5, and H6, respectively, including 1,668,024 variants common in the four genomes. Further, we identified 42,422, 39,824, 28,599, and 35,206 novel variants in the genomes of H3, H4, H5, and H6, respectively. A major fraction of the variants identified in the four genomes reside within the intergenic regions of the genome. Single nucleotide polymorphism (SNP) genotype based comparative analysis with ethnic populations of 1000 Genomes database linked the ancestry of all four genomes with the South Asian populations, which was further supported by mitochondria based haplogroup analysis. In conclusion, we report whole genome sequencing of four individuals of Pakistani descent.


Asunto(s)
Genoma Humano , Secuenciación Completa del Genoma , Humanos , Pakistán , Polimorfismo de Nucleótido Simple
11.
Stem Cell Res ; 46: 101813, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32474394

RESUMEN

Here, we report proteome profiling of peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cell (iPSC)-derived, lens-like organoids termed lentoid bodies at two differentiation time points. A small aliquot of the blood sample was ascertained to collect PBMCs that were reprogrammed to iPSCs. The PBMC-originated, iPSCs were differentiated to lentoid bodies employing the "fried egg" method. Quantitative real-time PCR (qRT-PCR) analysis revealed increased expression levels of lens-associated markers in lentoid bodies while transmission electron microscopy identified closely packed lens epithelial- and differentiating fiber-like cells in lentoid bodies. Total cellular protein was extracted from lentoid bodies at differentiation day 25 and mass spectrometry identified a total of 9,473 proteins. The low counts of crystallin proteins at differentiation day 25 prompted us to re-examine the proteome at differentiation day 35 as we reasoned that 10 additional days of differentiation will increase the crystallin count. However, we did not detect any substantial increase in crystallin protein counts at differentiation day 35. In conclusion, we report generation and proteome profiles of PBMC-originated, iPSC-derived lentoid bodies at multiple differentiation time points.


Asunto(s)
Cristalinas , Células Madre Pluripotentes Inducidas , Cristalino , Diferenciación Celular , Leucocitos Mononucleares , Proteoma
12.
Mol Vis ; 26: 334-344, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32355443

RESUMEN

Purpose: This study was designed to identify the pathogenic variants in three consanguineous families with congenital cataracts segregating as a recessive trait. Methods: Consanguineous families with multiple individuals manifesting congenital cataracts were ascertained. All participating members underwent an ophthalmic examination. A small aliquot of the blood sample was collected from all participating individuals, and genomic DNAs were extracted. Homozygosity-based linkage analysis was performed using short tandem repeat (STR) markers. The haplotypes were constructed with alleles of the STR markers, and the two-point logarithm of odds (LOD) scores were calculated. The candidate gene was sequenced bidirectionally to identify the disease-causing mutations. Results: Linkage analysis localized the disease interval to chromosome 3p in three families. Subsequently, bidirectional Sanger sequencing identified two novel mutations-a single base deletion resulting in a frameshift (c.3196delC; p.His1066IlefsTer10) mutation and a single base substitution resulting in a nonsense (c.4270C>T; p.Arg1424Ter) mutation-and a known missense (c.4127T>C, p.Leu1376Pro) mutation in FYCO1. All three mutations showed complete segregation with the disease phenotype and were absent in 96 ethnically matched control individuals. Conclusions: We report two novel mutations and a previously reported mutation in FYCO1 in three large consanguineous families. Taken together, mutations in FYCO1 contribute nearly 15% to the total genetic load of autosomal recessive congenital cataracts in this cohort.


Asunto(s)
Catarata/genética , Proteínas Asociadas a Microtúbulos/genética , Adulto , Alelos , Catarata/sangre , Catarata/congénito , Catarata/patología , Niño , Preescolar , Cromosomas Humanos Par 3/genética , Codón sin Sentido , Consanguinidad , Familia , Femenino , Mutación del Sistema de Lectura , Genes Recesivos , Ligamiento Genético , Predisposición Genética a la Enfermedad , Haplotipos , Homocigoto , Humanos , Lactante , Masculino , Repeticiones de Microsatélite , Proteínas Asociadas a Microtúbulos/sangre , Mutación Missense , Pakistán , Linaje , Filogenia
13.
Mol Vis ; 26: 14-25, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32165823

RESUMEN

Purpose: Primary congenital glaucoma (PCG) is a genetically heterogeneous disorder caused by developmental defects in the anterior chamber and trabecular meshwork. This disease is an important cause of childhood blindness. In this study, we aim to identify the genetic determinants of PCG in three consanguineous families of Pakistani descent. Methods: Affected members of all three families underwent detailed ophthalmological examination including slit-lamp biomicroscopy. Blood samples were collected from affected and healthy members of all three families, and genomic DNA was extracted. Linkage analysis was performed for the known or reported loci of PCG to localize the disease interval, and logarithm of odds (LOD) scores were calculated. All protein-coding exons of the candidate gene, latent transforming growth factor-beta binding protein 2 (LTBP2), were bidirectionally sequenced to identify the disease-causing mutation. Results: Short tandem repeat (STR) marker-based linkage analysis localized the critical interval to chromosome 14q with a maximum two-point LOD score of 2.86 (PKGL076), 2.8 (PKGL015), and 2.92 (PKGL042). Bidirectional Sanger sequencing of LTBP2 revealed three novel pathogenic variants, i.e., c.3028G>A (p.Asp1010Asn), c.3427delC (p.Gln1143Argfs*35), and c.5270G>A (p.Cys1757Tyr) in PKGL076, PKGL015, and PKGL042, respectively. All three mutations segregated with the disease phenotype in their respective families and were absent in 200 ethnically matched normal chromosomes. Conclusions: We identified three novel mutations, p.D1010N, p.Q1143Rfs*35, and p.C1757Y, in LTBP2 responsible for PCG.


Asunto(s)
Cromosomas Humanos Par 14/genética , Glaucoma/genética , Proteínas de Unión a TGF-beta Latente/genética , Adolescente , Alelos , Niño , Preescolar , Análisis Mutacional de ADN , Evolución Molecular , Exones , Femenino , Ligamiento Genético , Glaucoma/congénito , Glaucoma/fisiopatología , Humanos , Proteínas de Unión a TGF-beta Latente/sangre , Masculino , Mutación , Pakistán , Linaje , Análisis de Secuencia de ADN
14.
PLoS One ; 14(12): e0225010, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31815953

RESUMEN

PURPOSE: To investigate the genetic basis of autosomal recessive congenital cataracts (arCC) in a large consanguineous Pakistani family. METHODS: All participating members of family, PKCC074 underwent an ophthalmic examination. Slit-lamp photographs were ascertained for affected individuals that have not been operated for the removal of the cataractous lens. A small aliquot of the blood sample was collected from all participating individuals and genomic DNAs were extracted. A genome-wide scan was performed with polymorphic short tandem repeat (STR) markers and the logarithm of odds (LOD) scores were calculated. All coding exons and exon-intron boundaries of HSF4 were sequenced and expression of Hsf4 in mouse ocular lens was investigated. The C-terminal FLAG-tagged wild-type and mutant HSF4b constructs were prepared to examine the nuclear localization pattern of the mutant protein. RESULTS: The ophthalmological examinations suggested that nuclear cataracts are present in affected individuals. Genome-wide linkage analyses localized the critical interval to a 10.95 cM (14.17 Mb) interval on chromosome 16q with a maximum two-point LOD score of 4.51 at θ = 0. Sanger sequencing identified a novel missense mutation: c.433G>C (p.Ala145Pro) that segregated with the disease phenotype in the family and was not present in ethnically matched controls. Real-time PCR analysis identified the expression of HSF4 in mouse lens as early as embryonic day 15 with a steady level of expression thereafter. The immunofluorescence tracking confirmed that both wild-type and mutant HSF4 (p.Ala145Pro) proteins localized to the nucleus. CONCLUSION: Here, we report a novel missense mutation in HSF4 associated with arCC in a familial case of Pakistani descent.


Asunto(s)
Catarata/genética , Ligamiento Genético , Factores de Transcripción del Choque Térmico/genética , Cristalino/metabolismo , Repeticiones de Microsatélite , Mutación Missense , Animales , Catarata/congénito , Catarata/metabolismo , Consanguinidad , Femenino , Genes Recesivos , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Escala de Lod , Masculino , Ratones , Pakistán , Linaje
15.
Sci Rep ; 9(1): 18552, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31811247

RESUMEN

The ocular lens serves as an excellent system to investigate the intricate details of development and differentiation. Generation of lentoid bodies or lens-like structures using pluripotent stem cells is important for understanding the processes critical for lens morphogenesis and the mechanism of cataractogenesis. We previously reported the generation of peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cells (iPSCs). Here, we report generation of lentoid bodies from human embryonic stem cells (hESCs) and (PBMC)-originated, iPSCs employing the "fried egg" method with brief modifications. The ultrastructure analysis of hESC- and iPSC-derived lentoid bodies identified closely packed lens epithelial- and differentiating fiber-like cells. In addition, we performed RNA sequencing (RNA-Seq) based transcriptome profiling of hESC- and iPSC-derived lentoid bodies at differentiation day 25. Next-generation RNA sequencing (RNA-Seq) of hESC- and iPSC-derived lentoid bodies detected expression (≥0.659 RPKM) of 13,975 and 14,003 genes, respectively. Comparative transcriptome analysis of hESC- and iPSC-derived lentoid bodies revealed 13,563 (>96%) genes common in both datasets. Among the genes common in both transcriptome datasets, 12,856 (~95%) exhibited a quantitatively similar expression profile. Next, we compared the mouse lens epithelial and fiber cell transcriptomes with hESC- and iPSC-derived lentoid bodies transcriptomes and identified > 96% overlap with lentoid body transcriptomes. In conclusion, we report first-time comparative transcriptome analysis of hESC- and iPSC-derived lentoid bodies at differentiation day 25.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Cristalino/crecimiento & desarrollo , Transcriptoma/fisiología , Anciano , Línea Celular , Reprogramación Celular/fisiología , Células Madre Embrionarias Humanas/fisiología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Cristalino/citología , Leucocitos Mononucleares/fisiología , Masculino , Cultivo Primario de Células , RNA-Seq
16.
Exp Eye Res ; 176: 252-257, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30196069

RESUMEN

The corneal endothelium (CE), a monolayer of hexagonal cells constitutes the innermost layer of the cornea that is critical in maintaining clarity by mediating hydration through barrier and pump functions. Corneal endothelial cells (CECs) have limited proliferative potential and therefore generation of CECs has been undertaken by many groups. We previously reported generation of CECs from peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cells (iPSCs). In here, we extend our analysis through next-generation seqeuncing based transcriptome profiling of H9 human embryonic stem cell (hESC)- and human PBMC-originated, iPSC-derived CECs. The differentiating CECs on day 20 (D20) exhibited a tightly packed hexagonal/polygonal shape expressing zona occludens-1 (ZO-1) and N-cadherin at the cell boundaries. Next-generation RNA sequencing of hESC- and iPSC-derived CECs detected expression (≥0.659 RPKM) of 13,546 and 13,536 genes, respectively. Comparative transcriptome analysis of hESC- and iPSC-derived CECs revealed 13,208 (>96%) genes common in both transcriptomes. Among the 13,208 genes common in these transcriptomes, 12,580 (>95%) exhibited a quantitatively similar expression. To the best of our knowledge, this is the first report presenting comparative transcriptome analysis of hESC- and iPSC-derived CECs.


Asunto(s)
Endotelio Corneal/citología , Perfilación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Células Madre Pluripotentes Inducidas/citología , Anciano , Biomarcadores/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Endotelio Corneal/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Microscopía de Contraste de Fase , Transcriptoma , Proteína de la Zonula Occludens-1/metabolismo
17.
Sci Data ; 5: 180174, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30204152

RESUMEN

Here we report next-generation based whole genome sequencing of two individuals (H1 and H2) from a family of Pakistani descent. The genomic DNA was used to prepare paired-end libraries for whole-genome sequencing. Deep sequencing yielded 706.49 and 778.12 million mapped reads corresponding to 70.64 and 77.81 Gb sequence data and 23× and 25× average coverage for H1 and H2, respectively. Notably, a total of 448,544 and 470,683 novel variants, not present in the single nucleotide polymorphism database (dbSNP), were identified in H1 and H2, respectively. Comparative analysis identified 2,415,852 variants common in both genomes including 240,181 variants absent in the dbSNP. Principal component analysis linked the ancestry of both genomes with South Asian populations. In conclusion, we report whole genome sequences of two individuals from a family of Pakistani descent.


Asunto(s)
Pueblo Asiatico/genética , Genoma Humano , Secuenciación Completa del Genoma , Familia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Pakistán , Polimorfismo de Nucleótido Simple
18.
Sci Rep ; 8(1): 11162, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30042402

RESUMEN

We previously investigated the transcriptome and proteome profiles of the murine ocular lens at six developmental time points including two embryonic (E15 and E18) and four postnatal time points (P0, P3, P6, and P9). Here, we extend our analyses to identify novel transcripts and peptides in developing  mouse lens. We identified a total of 9,707 novel transcripts and 325 novel fusion genes in developing mouse lens. Additionally, we identified 13,281 novel alternative splicing (AS) events in mouse lens including 6,990 exon skipping (ES), 2,447 alternative 3' splice site (A3SS), 1,900 alternative 5' splice site (A5SS), 1,771 mutually exclusive exons (MXE), and 173 intron retention (IR). Finally, we integrated our OMIC (Transcriptome and Proteome) datasets identifying 20 novel peptides in mouse lens. All 20 peptides were validated through matching MS/MS spectra of synthetic peptides. To the best of our knowledge, this is the first report integrating OMIC datasets to identify novel peptides in developing murine lens.


Asunto(s)
Empalme Alternativo/genética , Cristalino/embriología , Cristalino/crecimiento & desarrollo , Organogénesis/genética , Péptidos/genética , Proteoma/genética , Transcriptoma/genética , Algoritmos , Animales , Cromatografía Liquida , Bases de Datos Genéticas , Exones/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Intrones/genética , Ratones , Embarazo , Sitios de Empalme de ARN/genética , Análisis de Secuencia de ARN , Espectrometría de Masas en Tándem
19.
Invest Ophthalmol Vis Sci ; 59(6): 2437-2444, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29847650

RESUMEN

Purpose: Corneal endothelial cells (CECs) are critical in maintaining clarity of the cornea. This study was initiated to develop peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cell (iPSC)-derived CECs. Methods: We isolated PBMCs and programmed the mononuclear cells to generate iPSCs, which were differentiated to CECs through the neural crest cells (NCCs). The morphology of differentiating iPSCs was examined at regular intervals by phase contrast microscopy. In parallel, the expression of pluripotent and corneal endothelium (CE)-associated markers was investigated by quantitative real-time PCR (qRT-PCR). The molecular architecture of the iPSC-derived CECs and human corneal endothelium (hCE) was examined by mass spectrometry-based proteome sequencing. Results: The PBMC-originated, iPSC-derived CECs were tightly adherent, exhibiting a hexagonal-like shape, one of the cardinal characteristics of CECs. The CE-associated markers expressed at significantly higher levels in iPSC-derived CECs at days 13, 20, and 30 compared with their respective levels in iPSCs. It is of importance that only residual expression levels of pluripotency markers were detected in iPSC-derived CECs. Cryopreservation of iPSC-derived CECs did not affect the tight adherence of CECs and their hexagonal-like shape while expressing high levels of CE-associated markers. Mass spectrometry-based proteome sequencing identified 10,575 proteins in the iPSC-derived CEC proteome. In parallel, we completed proteome profiling of the hCE identifying 6345 proteins. Of these, 5763 proteins were identified in the iPSC-derived CECs, suggesting that 90.82% of the hCE proteome overlaps with the iPSC-derived CEC proteome. Conclusions: We have successfully developed a personalized approach to generate CECs that closely mimic the molecular architecture of the hCE. To the best of our knowledge, this is the first report describing the development of PBMC-originated, iPSC-derived CECs.


Asunto(s)
Endotelio Corneal/citología , Perfilación de la Expresión Génica/métodos , Marcadores Genéticos/genética , Células Madre Pluripotentes Inducidas/citología , Leucocitos Mononucleares/citología , Proteoma/genética , Anciano , Diferenciación Celular/fisiología , Células Cultivadas , Criopreservación , Células Madre Embrionarias/citología , Endotelio Corneal/metabolismo , Citometría de Flujo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Espectrometría de Masas , Microscopía de Contraste de Fase , Persona de Mediana Edad , Cresta Neural/citología , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Invest Ophthalmol Vis Sci ; 59(1): 100-107, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29332127

RESUMEN

Purpose: We previously completed a comprehensive profile of the mouse lens transcriptome. Here, we investigate the proteome of the mouse lens through mass spectrometry-based protein sequencing at the same embryonic and postnatal time points. Methods: We extracted mouse lenses at embryonic day 15 (E15) and 18 (E18) and postnatal day 0 (P0), 3 (P3), 6 (P6), and 9 (P9). The lenses from each time point were preserved in three distinct pools to serve as biological replicates for each developmental stage. The total cellular protein was extracted from the lens, digested with trypsin, and labeled with isobaric tandem mass tags (TMT) for three independent TMT experiments. Results: A total of 5404 proteins were identified in the mouse ocular lens in at least one TMT set, 4244 in two, and 3155 were present in all three TMT sets. The majority of the proteins exhibited steady expression at all six developmental time points; nevertheless, we identified 39 proteins that exhibited an 8-fold differential (higher or lower) expression during the developmental time course compared to their respective levels at E15. The lens proteome is composed of diverse proteins that have distinct biological properties and functional characteristics, including proteins associated with cataractogenesis and autophagy. Conclusions: We have established a comprehensive profile of the developing murine lens proteome. This repository will be helpful in identifying critical components of lens development and processes essential for the maintenance of its transparency.


Asunto(s)
Proteínas del Ojo/genética , Perfilación de la Expresión Génica/métodos , Cristalino/metabolismo , Espectrometría de Masas/métodos , Proteoma/genética , ARN Mensajero/genética , Animales , Animales Recién Nacidos , Proteínas del Ojo/metabolismo , Ratones , Modelos Animales , Proteoma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...