Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Hum Mol Genet ; 31(13): 2137-2154, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35075486

ABSTRACT

Retinal diseases exhibit extensive genetic heterogeneity and complex etiology with varying onset and severity. Mutations in over 200 genes can lead to photoreceptor dysfunction and/or cell death in retinal neurodegeneration. To deduce molecular pathways that initiate and/or drive cell death, we adopted a temporal multiomics approach and examined molecular and cellular events in newborn and developing photoreceptors before the onset of degeneration in a widely-used Pde6brd1/rd1 (rd1) mouse, a model of autosomal recessive retinitis pigmentosa caused by PDE6B mutations. Transcriptome profiling of neonatal and developing rods from the rd1 retina revealed early downregulation of genes associated with anabolic pathways and energy metabolism. Quantitative proteomics of rd1 retina showed early changes in calcium signaling and oxidative phosphorylation, with specific partial bypass of complex I electron transfer, which precede the onset of cell death. Concurrently, we detected alterations in central carbon metabolism, including dysregulation of components associated with glycolysis, pentose phosphate and purine biosynthesis. Ex vivo assays of oxygen consumption and transmission electron microscopy validated early and progressive mitochondrial stress and abnormalities in mitochondrial structure and function of rd1 rods. These data uncover mitochondrial overactivation and related metabolic alterations as determinants of early pathology and implicate aberrant calcium signaling as an initiator of higher mitochondrial stress. Our studies thus provide a mechanistic framework with mitochondrial damage and metabolic disruptions as early drivers of photoreceptor cell death in retinal degeneration.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Animals , Cell Death/genetics , Disease Models, Animal , Mice , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/pathology
2.
PLoS Genet ; 16(12): e1009259, 2020 12.
Article in English | MEDLINE | ID: mdl-33362196

ABSTRACT

Rab-GTPases and associated effectors mediate cargo transport through the endomembrane system of eukaryotic cells, regulating key processes such as membrane turnover, signal transduction, protein recycling and degradation. Using developmental transcriptome data, we identified Rabgef1 (encoding the protein RabGEF1 or Rabex-5) as the only gene associated with Rab GTPases that exhibited strong concordance with retinal photoreceptor differentiation. Loss of Rabgef1 in mice (Rabgef1-/-) resulted in defects specifically of photoreceptor morphology and almost complete loss of both rod and cone function as early as eye opening; however, aberrant outer segment formation could only partly account for visual function deficits. RabGEF1 protein in retinal photoreceptors interacts with Rabaptin-5, and RabGEF1 absence leads to reduction of early endosomes consistent with studies in other mammalian cells and tissues. Electron microscopy analyses reveal abnormal accumulation of macromolecular aggregates in autophagosome-like vacuoles and enhanced immunostaining for LC3A/B and p62 in Rabgef1-/- photoreceptors, consistent with compromised autophagy. Transcriptome analysis of the developing Rabgef1-/- retina reveals altered expression of 2469 genes related to multiple pathways including phototransduction, mitochondria, oxidative stress and endocytosis, suggesting an early trajectory of photoreceptor cell death. Our results implicate an essential role of the RabGEF1-modulated endocytic and autophagic pathways in photoreceptor differentiation and homeostasis. We propose that RabGEF1 and associated components are potential candidates for syndromic traits that include a retinopathy phenotype.


Subject(s)
Autophagy , Endocytosis , Guanine Nucleotide Exchange Factors/genetics , Neurogenesis , Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Animals , Female , Guanine Nucleotide Exchange Factors/metabolism , Male , Mice , Mice, Inbred BALB C , Photoreceptor Cells/cytology , Retinal Degeneration/genetics , Transcriptome
3.
Hum Mol Genet ; 27(20): 3612-3626, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30052969

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is caused by deficiency of α-l-iduronidase (IDUA), a lysosomal enzyme involved in the breakdown and recycling of glycosaminoglycans (GAGs). Although enzyme replacement therapy is available, the efficacy of the treatment for neuropathic manifestations is limited. To facilitate drug discovery and model disease pathophysiology, we generated neural stem cells (NSCs) from MPS I patient-derived induced pluripotent stem cells (iPSCs). The NSCs exhibited characteristic disease phenotypes with deficiency of IDUA, accumulation of GAGs and enlargement of lysosomes, in agreement with the severity of clinical subgroups of MPS I. Transcriptome profiling of NSCs revealed 429 genes that demonstrated a more extensive change in expression in the most severe Hurler syndrome subgroup compared to the intermediate Hurler-Scheie or the least severe Scheie syndrome subgroups. Clustering and pathway analysis revealed high concordance of the severity of neurological defects with marked dysregulation of GAG biosynthesis, GAG degradation, lysosomal function and autophagy. Gene ontology (GO) analysis identified a dramatic upregulation of the autophagy pathway, especially in the Hurler syndrome subgroup. We conclude that GAG accumulation in the patient-derived cells disrupts lysosomal homeostasis, affecting multiple related cellular pathways in response to IDUA deficiency. These dysregulated processes likely lead to enhanced autophagy and progressively severe disease states. Our study provides potentially useful targets for clinical biomarker development, disease diagnosis and prognosis, and drug discovery.


Subject(s)
Iduronidase/genetics , Mucopolysaccharidosis I/enzymology , Neural Stem Cells , Phenotype , Cell Line , Child , Child, Preschool , Gene Expression Profiling , Glycosaminoglycans/metabolism , Humans , Iduronidase/metabolism , Induced Pluripotent Stem Cells , Lysosomes , Male , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/metabolism , Mutation
4.
Hum Mutat ; 40(4): 426-443, 2019 04.
Article in English | MEDLINE | ID: mdl-30628748

ABSTRACT

Human RPE65 mutations cause a spectrum of retinal dystrophies that result in blindness. While RPE65 mutations have been almost invariably recessively inherited, a c.1430A>G (p.(D477G)) mutation has been reported to cause autosomal dominant retinitis pigmentosa (adRP). To study the pathogenesis of this human mutation, we have replicated the mutation in a knock-in (KI) mouse model using CRISPR/Cas9-mediated genome editing. Significantly, in contrast to human patients, heterozygous KI mice do not exhibit any phenotypes in visual function tests. When raised in regular vivarium conditions, homozygous KI mice display relatively undisturbed visual functions with minimal retinal structural changes. However, KI/KI mouse retinae are more sensitive to light exposure and exhibit signs of degenerative features when subjected to light stress. We find that instead of merely producing a missense mutant protein, the A>G nucleotide substitution greatly affects appropriate splicing of Rpe65 mRNA by generating an ectopic splice site in comparable context to the canonical one, thereby disrupting RPE65 protein expression. Similar splicing defects were also confirmed for the human RPE65 c.1430G mutant in an in vitro Exontrap assay. Our data demonstrate that a splicing defect is associated with c.1430G pathogenesis, and therefore provide insights in the therapeutic strategy for human patients.


Subject(s)
Alleles , Genetic Predisposition to Disease , Mutation , RNA Splicing , cis-trans-Isomerases/genetics , Animals , Biomarkers , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Genetic Association Studies , Genotype , Humans , Mice , Mice, Transgenic , Phenotype , RNA Splice Sites , Retina/metabolism , Retina/pathology
5.
Mol Vis ; 25: 663-678, 2019.
Article in English | MEDLINE | ID: mdl-31814692

ABSTRACT

PURPOSE: Retinal organoids generated from human pluripotent stem cells exhibit considerable variability during differentiation. Our goals are to assess developmental maturity of the neural retina in vitro and design improved protocols based on objective criteria. METHODS: We performed transcriptome analyses of developing retinal organoids from human embryonic and induced pluripotent stem cell lines and utilized multiple bioinformatic tools for comparative analysis. Immunohistochemistry, immunoblotting and electron microscopy were employed for validation. RESULTS: We show that the developmental variability in organoids was reflected in gene expression profiles and could be evaluated by molecular staging with the human fetal and adult retinal transcriptome data. We also demonstrate that the addition of 9-cis retinal, instead of the widely used all-trans retinoic acid, accelerated rod photoreceptor differentiation in organoid cultures, with higher rhodopsin expression and more mature mitochondrial morphology evident by day 120. CONCLUSION: Our studies provide an objective transcriptome-based modality for determining the differentiation state of retinal organoids and for comparisons across different stem cell lines and platforms, which should facilitate disease modeling and evaluation of therapies in vitro.


Subject(s)
Cell Differentiation , Diterpenes/pharmacology , Human Embryonic Stem Cells/cytology , Organoids/cytology , Retina/cytology , Retinal Rod Photoreceptor Cells/cytology , Retinaldehyde/pharmacology , Transcriptome/genetics , Cell Differentiation/drug effects , Cell Line , Cell Shape/drug effects , Gene Expression Profiling , Human Embryonic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Organoids/drug effects , Organoids/ultrastructure , Retinal Rod Photoreceptor Cells/drug effects , Transcriptome/drug effects
6.
Pharmacol Res ; 115: 1-13, 2017 01.
Article in English | MEDLINE | ID: mdl-27838510

ABSTRACT

Mammalian cells are commonly employed in screening assays to identify active compounds that could potentially affect the progression of different human diseases including retinitis pigmentosa (RP), a class of inherited diseases causing retinal degeneration with compromised vision. Using transcriptome analysis, we compared NIH3T3 cells expressing wildtype (WT) rod opsin with a retinal disease-causing single P23H mutation. Surprisingly, heterologous expression of WT opsin in NIH3T3 cells caused more than a 2-fold change in 783 out of 16,888 protein coding transcripts. The perturbed genes encoded extracellular matrix proteins, growth factors, cytoskeleton proteins, glycoproteins and metalloproteases involved in cell adhesion, morphology and migration. A different set of 347 transcripts was either up- or down-regulated when the P23H mutant opsin was expressed suggesting an altered molecular perturbation compared to WT opsin. Transcriptome perturbations elicited by drug candidates aimed at mitigating the effects of the mutant protein revealed that different drugs targeted distinct molecular pathways that resulted in a similar phenotype selected by a cell-based high-throughput screen. Thus, transcriptome profiling can provide essential information about the therapeutic potential of a candidate drug to restore normal gene expression in pathological conditions.


Subject(s)
Mutation/genetics , Pharmaceutical Preparations/administration & dosage , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/genetics , Rod Opsins/genetics , Transcriptome/genetics , Animals , Cell Line , Disease Models, Animal , Gene Expression Profiling/methods , Mice , NIH 3T3 Cells , Vision, Ocular/drug effects
7.
PLoS Genet ; 8(4): e1002649, 2012.
Article in English | MEDLINE | ID: mdl-22511886

ABSTRACT

A stringent control of homeostasis is critical for functional maintenance and survival of neurons. In the mammalian retina, the basic motif leucine zipper transcription factor NRL determines rod versus cone photoreceptor cell fate and activates the expression of many rod-specific genes. Here, we report an integrated analysis of NRL-centered gene regulatory network by coupling chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-Seq) data from Illumina and ABI platforms with global expression profiling and in vivo knockdown studies. We identified approximately 300 direct NRL target genes. Of these, 22 NRL targets are associated with human retinal dystrophies, whereas 95 mapped to regions of as yet uncloned retinal disease loci. In silico analysis of NRL ChIP-Seq peak sequences revealed an enrichment of distinct sets of transcription factor binding sites. Specifically, we discovered that genes involved in photoreceptor function include binding sites for both NRL and homeodomain protein CRX. Evaluation of 26 ChIP-Seq regions validated their enhancer functions in reporter assays. In vivo knockdown of 16 NRL target genes resulted in death or abnormal morphology of rod photoreceptors, suggesting their importance in maintaining retinal function. We also identified histone demethylase Kdm5b as a novel secondary node in NRL transcriptional hierarchy. Exon array analysis of flow-sorted photoreceptors in which Kdm5b was knocked down by shRNA indicated its role in regulating rod-expressed genes. Our studies identify candidate genes for retinal dystrophies, define cis-regulatory module(s) for photoreceptor-expressed genes and provide a framework for decoding transcriptional regulatory networks that dictate rod homeostasis.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins , Homeostasis , Retina , Retinal Dystrophies , Retinal Rod Photoreceptor Cells/metabolism , Trans-Activators , Animals , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeostasis/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Retina/metabolism , Retina/physiology , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
8.
Am J Hum Genet ; 84(6): 792-800, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19520207

ABSTRACT

Retinitis pigmentosa (RP) refers to a genetically heterogeneous group of progressive neurodegenerative diseases that result in dysfunction and/or death of rod and cone photoreceptors in the retina. So far, 18 genes have been identified for autosomal-dominant (ad) RP. Here, we describe an adRP locus (RP42) at chromosome 7p15 through linkage analysis in a six-generation Scandinavian family and identify a disease-causing mutation, c.449G-->A (p.S150N), in exon 6 of the KLHL7 gene. Mutation screening of KLHL7 in 502 retinopathy probands has revealed three different missense mutations in six independent families. KLHL7 is widely expressed, including expression in rod photoreceptors, and encodes a 75 kDa protein of the BTB-Kelch subfamily within the BTB superfamily. BTB-Kelch proteins have been implicated in ubiquitination through Cullin E3 ligases. Notably, all three putative disease-causing KLHL7 mutations are within a conserved BACK domain; homology modeling suggests that mutant amino acid side chains can potentially fill the cleft between two helices, thereby affecting the ubiquitination complexes. Mutations in an identical region of another BTB-Kelch protein, gigaxonin, have previously been associated with giant axonal neuropathy. Our studies suggest an additional role of the ubiquitin-proteasome protein-degradation pathway in maintaining neuronal health and in disease.


Subject(s)
Autoantigens/genetics , Genes, Dominant , Mutation, Missense/genetics , Polymorphism, Single Nucleotide/genetics , Retinitis Pigmentosa/genetics , Amino Acid Sequence , Autoantigens/metabolism , Chromosomes, Human, Pair 7/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Genetic Linkage , Humans , Immunoblotting , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pedigree , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
9.
Front Cell Neurosci ; 16: 878351, 2022.
Article in English | MEDLINE | ID: mdl-35783089

ABSTRACT

Retinal organoids (ROs) derived from human pluripotent stem cells (hPSCs) recapitulate key features of retinogenesis and provide a promising platform to study retinal development and disease in a human context. Although multiple protocols are currently in use, hPSCs exhibit tremendous variability in differentiation efficiency, with some cell lines consistently yielding few or even no ROs, limiting their utility in research. We report here that early nicotinamide (NAM) treatment significantly improves RO yield across 8 hPSC lines from different donors, including some that would otherwise fail to generate a meaningful number of ROs. NAM treatment promotes neural commitment of hPSCs at the expense of non-neural ectodermal cell fate, which in turn increases eye field progenitor generation. Further analysis suggests that this effect is partially mediated through inhibition of BMP signaling. Our data encourage a broader use of human ROs for disease modeling applications that require the use of multiple patient-specific cell lines.

10.
Stem Cell Reports ; 16(2): 252-263, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33513359

ABSTRACT

Mutations in the photoreceptor transcription factor gene cone-rod homeobox (CRX) lead to distinct retinopathy phenotypes, including early-onset vision impairment in dominant Leber congenital amaurosis (LCA). Using induced pluripotent stem cells (iPSCs) from a patient with CRX-I138fs48 mutation, we established an in vitro model of CRX-LCA in retinal organoids that showed defective photoreceptor maturation by histology and gene profiling, with diminished expression of visual opsins. Adeno-associated virus (AAV)-mediated CRX gene augmentation therapy partially restored photoreceptor phenotype and expression of phototransduction-related genes as determined by single-cell RNA-sequencing. Retinal organoids derived from iPSCs of a second dominant CRX-LCA patient carrying K88N mutation revealed the loss of opsin expression as a common phenotype, which was alleviated by AAV-mediated augmentation of CRX. Our studies provide a proof-of-concept for developing gene therapy of dominant CRX-LCA and other CRX retinopathies.


Subject(s)
Homeodomain Proteins/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Organoids/metabolism , Photoreceptor Cells/metabolism , Retina/metabolism , Trans-Activators/genetics , Adult , Cell Differentiation , Child , Child, Preschool , Dependovirus , Female , Genetic Therapy/methods , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Leber Congenital Amaurosis/pathology , Models, Biological , Mutation , Opsins/metabolism , Organoids/cytology , Phenotype , Retina/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome
11.
Cell Rep ; 31(3): 107525, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320661

ABSTRACT

Aging-associated functional decline is accompanied by alterations in the epigenome. To explore DNA modifications that could influence visual function with age, we perform whole-genome bisulfite sequencing of purified mouse rod photoreceptors at four ages and identify 2,054 differentially methylated regions (DMRs). We detect many DMRs during early stages of aging and in rod regulatory regions, and some of these cluster at chromosomal hotspots, especially on chromosome 10, which includes a longevity interactome. Integration of methylome to age-related transcriptome changes, chromatin signatures, and first-order protein-protein interactions uncover an enrichment of DMRs in altered pathways that are associated with rod function, aging, and energy metabolism. In concordance, we detect reduced basal mitochondrial respiration and increased fatty acid dependency with retinal age in ex vivo assays. Our study reveals age-dependent genomic and chromatin features susceptible to DNA methylation changes in rod photoreceptors and identifies a link between DNA methylation and energy metabolism in aging.


Subject(s)
Aging/genetics , DNA Methylation/genetics , Energy Metabolism/genetics , Retinal Rod Photoreceptor Cells/metabolism , Animals , Genome-Wide Association Study/methods , Humans , Male , Mice
12.
Sci Rep ; 10(1): 5426, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32214115

ABSTRACT

Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a photoreceptor-specific chaperone that stabilizes the effector enzyme of phototransduction, cGMP phosphodiesterase 6 (PDE6). Mutations in the AIPL1 gene cause a severe inherited retinal dystrophy, Leber congenital amaurosis type 4 (LCA4), that manifests as the loss of vision during the first year of life. In this study, we generated three-dimensional (3D) retinal organoids (ROs) from human induced pluripotent stem cells (hiPSCs) derived from an LCA4 patient carrying a Cys89Arg mutation in AIPL1. This study aimed to (i) explore whether the patient hiPSC-derived ROs recapitulate LCA4 disease phenotype, and (ii) generate a clinically relevant resource to investigate the molecular mechanism of disease and safely test novel therapies for LCA4 in vitro. We demonstrate reduced levels of the mutant AIPL1 and PDE6 proteins in patient organoids, corroborating the findings in animal models; however, patient-derived organoids maintained retinal cell cytoarchitecture despite significantly reduced levels of AIPL1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Eye Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Organoids/metabolism , Retina/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/metabolism , Cell Line , Eye Proteins/genetics , Humans , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Mutation/genetics
13.
Nat Genet ; 51(4): 606-610, 2019 04.
Article in English | MEDLINE | ID: mdl-30742112

ABSTRACT

Genome-wide association studies (GWAS) have identified genetic variants at 34 loci contributing to age-related macular degeneration (AMD)1-3. We generated transcriptional profiles of postmortem retinas from 453 controls and cases at distinct stages of AMD and integrated retinal transcriptomes, covering 13,662 protein-coding and 1,462 noncoding genes, with genotypes at more than 9 million common SNPs for expression quantitative trait loci (eQTL) analysis of a tissue not included in Genotype-Tissue Expression (GTEx) and other large datasets4,5. Cis-eQTL analysis identified 10,474 genes under genetic regulation, including 4,541 eQTLs detected only in the retina. Integrated analysis of AMD-GWAS with eQTLs ascertained likely target genes at six reported loci. Using transcriptome-wide association analysis (TWAS), we identified three additional genes, RLBP1, HIC1 and PARP12, after Bonferroni correction. Our studies expand the genetic landscape of AMD and establish the Eye Genotype Expression (EyeGEx) database as a resource for post-GWAS interpretation of multifactorial ocular traits.


Subject(s)
Genetic Predisposition to Disease/genetics , Macular Degeneration/genetics , Quantitative Trait Loci/genetics , Transcriptome/genetics , Case-Control Studies , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Genome-Wide Association Study/methods , Genotype , Humans , Phenotype , Polymorphism, Single Nucleotide/genetics , Retina/physiopathology
14.
Nat Genet ; 51(6): 1067, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31068672

ABSTRACT

In the version of this article initially published, in Supplementary Data 5, the logFC, FC, P value and adjusted P value for advanced AMD versus control (DE 4/1) without age correction did not correspond to the correct gene IDs. The errors have been corrected in the HTML version of the article.

15.
Stem Cell Reports ; 10(1): 300-313, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29233554

ABSTRACT

Pluripotent stem cells can be differentiated into 3D retinal organoids, with major cell types self-patterning into a polarized, laminated architecture. In static cultures, organoid development may be hindered by limitations in diffusion of oxygen and nutrients. Herein, we report a bioprocess using rotating-wall vessel (RWV) bioreactors to culture retinal organoids derived from mouse pluripotent stem cells. Organoids in RWV demonstrate enhanced proliferation, with well-defined morphology and improved differentiation of neurons including ganglion cells and S-cone photoreceptors. Furthermore, RWV organoids at day 25 (D25) reveal similar maturation and transcriptome profile as those at D32 in static culture, closely recapitulating spatiotemporal development of postnatal day 6 mouse retina in vivo. Interestingly, however, retinal organoids do not differentiate further under any in vitro condition tested here, suggesting additional requirements for functional maturation. Our studies demonstrate that bioreactors can accelerate and improve organoid growth and differentiation for modeling retinal disease and evaluation of therapies.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Retina/metabolism , Animals , Mice , Mice, Transgenic , Organoids/cytology , Pluripotent Stem Cells/cytology , Retina/cytology
16.
Invest Ophthalmol Vis Sci ; 58(4): 1941-1953, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28384715

ABSTRACT

Purpose: During retinal degeneration, Müller glia cells respond to photoreceptor loss by undergoing reactive gliosis, with both detrimental and beneficial effects. Increasing our knowledge of the complex molecular response of Müller cells to retinal degeneration is thus essential for the development of new therapeutic strategies. The purpose of this work was to identify new factors involved in Müller cell response to photoreceptor cell death. Methods: Whole transcriptome sequencing was performed from wild-type and degenerating rd10 mouse retinas at P30. The changes in mRNA abundance for several differentially expressed genes were assessed by quantitative RT-PCR (RT-qPCR). Protein expression level and retinal cellular localization were determined by western blot and immunohistochemistry, respectively. Results: Pathway-level analysis from whole transcriptomic data revealed the Hippo/YAP pathway as one of the main signaling pathways altered in response to photoreceptor degeneration in rd10 retinas. We found that downstream effectors of this pathway, YAP and TEAD1, are specifically expressed in Müller cells and that their expression, at both the mRNA and protein levels, is increased in rd10 reactive Müller glia after the onset of photoreceptor degeneration. The expression of Ctgf and Cyr61, two target genes of the transcriptional YAP/TEAD complex, is also upregulated following photoreceptor loss. Conclusions: This work reveals for the first time that YAP and TEAD1, key downstream effectors of the Hippo pathway, are specifically expressed in Müller cells. We also uncovered a deregulation of the expression and activity of Hippo/YAP pathway components in reactive Müller cells under pathologic conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/genetics , Ependymoglial Cells/metabolism , Gene Expression Regulation , Phosphoproteins/genetics , Photoreceptor Cells/metabolism , RNA, Messenger/genetics , Retinal Degeneration/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Blotting, Western , Cell Cycle Proteins , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Ependymoglial Cells/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nuclear Proteins , Phosphoproteins/biosynthesis , Photoreceptor Cells/pathology , Real-Time Polymerase Chain Reaction , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/biosynthesis , YAP-Signaling Proteins
17.
Dev Cell ; 43(6): 763-779.e4, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29233477

ABSTRACT

Clinical and genetic heterogeneity associated with retinal diseases makes stem-cell-based therapies an attractive strategy for personalized medicine. However, we have limited understanding of the timing of key events in the developing human retina, and in particular the factors critical for generating the unique architecture of the fovea and surrounding macula. Here we define three key epochs in the transcriptome dynamics of human retina from fetal day (D) 52 to 136. Coincident histological analyses confirmed the cellular basis of transcriptional changes and highlighted the dramatic acceleration of development in the fovea compared with peripheral retina. Human and mouse retinal transcriptomes show remarkable similarity in developmental stages, although morphogenesis was greatly expanded in humans. Integration of DNA accessibility data allowed us to reconstruct transcriptional networks controlling photoreceptor differentiation. Our studies provide insights into human retinal development and serve as a resource for molecular staging of human stem-cell-derived retinal organoids.


Subject(s)
Neurogenesis/physiology , Retina/cytology , Retina/physiology , Animals , Eye Proteins/genetics , Eye Proteins/physiology , Fovea Centralis/embryology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Macula Lutea/embryology , Mice , Morphogenesis , Neurogenesis/genetics , Neurons/metabolism , Retina/embryology , Retina/growth & development , Sequence Analysis, RNA/methods , Transcriptome
18.
Nucleic Acids Res ; 30(7): e30, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11917036

ABSTRACT

DNA microarrays enable users to obtain information on differences in transcript abundance on a massively parallel scale. Recently, however, data analyses have revealed potential pitfalls related to image acquisition, variability and misclassifications in replicate measurements, cross-hybridization and sensitivity limitations. We have generated a series of analytical tools to address the manufacturing, detection and data analysis components of a microarray experiment. Together, we have used these tools to optimize performance in an expression profiling study. We demonstrate three significant advantages of the Motorola CodeLink platform: sensitivity of one copy per cell, coefficients of variation of 10% in the hybridization signals across slides and across target preparations, and specificity in distinguishing highly homologous sequences. Slides where oligonucleotide probes are spotted in 6-fold redundancy were used to demonstrate the effect of replication on data quality. Lastly, the differential expression ratios obtained with the CodeLink expression platform were validated against those obtained with quantitative reverse transcription-PCR assays for 54 genes.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , RNA/genetics , RNA/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
20.
Sci Signal ; 9(438): ra74, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27460988

ABSTRACT

Photoreceptor cell degeneration leads to visual impairment and blindness in several types of retinal disease. However, the discovery of safe and effective therapeutic strategies conferring photoreceptor cell protection remains challenging. Targeting distinct cellular pathways with low doses of different drugs that produce a functionally synergistic effect could provide a strategy for preventing or treating retinal dystrophies. We took a systems pharmacology approach to identify potential combination therapies using a mouse model of light-induced retinal degeneration. We showed that a combination of U.S. Food and Drug Administration-approved drugs that act on different G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors (GPCRs) exhibited synergistic activity that protected retinas from light-induced degeneration even when each drug was administered at a low dose. In functional assays, the combined effects of these drugs were stimulation of Gi/o signaling by activating the dopamine receptors D2R and D4R, as well as inhibition of Gs and Gq signaling by antagonizing D1R and the α1A-adrenergic receptor ADRA1A, respectively. Moreover, transcriptome analyses demonstrated that such combined GPCR-targeted treatments preserved patterns of retinal gene expression that were more similar to those of the normal retina than did higher-dose monotherapy. Our study thus supports a systems pharmacology approach to identify treatments for retinopathies, an approach that could extend to other complex disorders.


Subject(s)
Adrenergic Agents/pharmacology , Dopamine Agents/pharmacology , Photoreceptor Cells, Vertebrate/metabolism , Receptors, G-Protein-Coupled , Retinal Degeneration , Signal Transduction/drug effects , Animals , Gene Expression Regulation/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Photoreceptor Cells, Vertebrate/pathology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/prevention & control , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL