Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Oncol ; 18(4): 918-938, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37975412

ABSTRACT

MYCN proto-oncogene, bHLH transcription factor (MYCN) amplification is associated with aggressive retinoblastoma (RB) and neuroblastoma (NB) cancer recurrence that is resistant to chemotherapies. Therefore, there is an urgent need to identify new therapeutic tools. This study aimed to evaluate the potential repurposing of ceftriaxone for the treatment of MYCN-amplified RB and NB, based on the clinical observations that the drug was serendipitously found to decrease the volume of the MYCN-driven RB subtype. Using patient-derived tumor organoids and tumor cell lines, we demonstrated that ceftriaxone is a potent and selective growth inhibitor targeting MYCN-driven RB and NB cells. Profiling of drug-induced transcriptomic changes, cell-cycle progression, and apoptotic death indicated cell-cycle arrest and death of drug-treated MYCN-amplified tumor cells. Drug target identification, using an affinity-based proteomic and molecular docking approach, and functional studies of the target proteins revealed that ceftriaxone targeted DEAD-box helicase 3 X-linked (DDX3X), thereby inhibiting translation in MYCN-amplified tumors but not in MYCN-nonamplified cells. The data suggest the feasibility of repurposing ceftriaxone as an anticancer drug and provide insights into the mechanism of drug action, highlighting DDX3X as a potential target for treating MYCN-driven tumors.


Subject(s)
Neuroblastoma , Retinal Neoplasms , Retinoblastoma , Humans , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Ceftriaxone , Retinoblastoma/drug therapy , Retinoblastoma/genetics , Molecular Docking Simulation , Proteomics , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/metabolism , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Neoplastic
2.
Sci Rep ; 13(1): 10290, 2023 06 25.
Article in English | MEDLINE | ID: mdl-37357228

ABSTRACT

Olfactomedin4 (Olfm4) is expressed in normal mouse prostate. However, Olfm4+ cells in the murine prostate have not been well characterized. In this study, we generated an Olfm4eGFP reporter mouse line with C57BL/6 mice and investigated the distribution of Olfm4/eGFP-expressing cells during postnatal development from P1, P7, P14, P20, P42, P56 to adult male mouse prostate and urethral tube. We observed Olfm4/eGFP expression in urogenital and prostatic epithelial cells during early postnatal development, which persisted into adulthood in urethral-tube and anterior-prostate (AP) epithelium. We found Olfm4+ cells are E-cadherin+/CD44+/Foxa1+ and some of subpopulation are Ck8+/Ck5+/Sca-1-/Ck4-/Syn- in the adult mouse AP epithelium. Functional studies of single-cell preparations of Olfm4/eGFP-expressing cells isolated from adult Olfm4eGFP mouse prostate demonstrated that Olfm4+ cells can grow and form colonies, spheres, or organoids in culture. Bioinformatic analysis of Olfm4+ cells using single-cell RNA sequencing meta data in adult mouse urethra (GSE145865) identified upregulation of genes related to cell and tissue migration and development, as well as upregulation of xenobiotic metabolism signaling pathways. In conclusion, Olfm4eGFP mouse is a novel model to further study Olfm4's biological functions and Olfm4+ cells may contribute importantly to cellular processes supporting development and homeostasis of the epithelium in murine prostate and urethral tube.


Subject(s)
Glycoproteins , Prostate , Mice , Male , Animals , Prostate/metabolism , Mice, Inbred C57BL , Epithelium/metabolism , Glycoproteins/metabolism , Granulocyte Colony-Stimulating Factor/metabolism
3.
J Exp Clin Cancer Res ; 42(1): 39, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36726110

ABSTRACT

BACKGROUND: Recurrence of retinoblastoma (RB) following chemoreduction is common and is often managed with local (intra-arterial/intravitreal) chemotherapy. However, some tumors are resistant to even local administration of maximum feasible drug dosages, or effective tumor control and globe preservation may be achieved at the cost of vision loss due to drug-induced retinal toxicity. The aim of this study was to identify drugs with improved antitumor activity and more favorable retinal toxicity profiles via screening of potentially repurposable FDA-approved drugs in patient-derived tumor organoids. METHODS: Genomic profiling of five RB organoids and the corresponding parental tissues was performed. RB organoids were screened with 133 FDA-approved drugs, and candidate drugs were selected based on cytotoxicity and potency. RNA sequencing was conducted to generate a drug signature from RB organoids, and the effects of drugs on cell cycle progression and proliferative tumor cone restriction were examined. Drug toxicity was assessed with human embryonic stem cell-derived normal retinal organoids. The efficacy/toxicity profiles of candidate drugs were compared with those of drugs in clinical use. RESULTS: RB organoids maintained the genomic features of the parental tumors. Sunitinib was identified as highly cytotoxic against both classical RB1-deficient and novel MYCN-amplified RB organoids and inhibited proliferation while inducing differentiation in RB. Sunitinib was a more effective suppressor of proliferative tumor cones in RB organoids and had lower toxicity in normal retinal organoids than either melphalan or topotecan. CONCLUSION: The efficacy and retinal toxicity profiles of sunitinib suggest that it could potentially be repurposed for local chemotherapy of RB.


Subject(s)
Antineoplastic Agents , Retinal Neoplasms , Retinoblastoma , Humans , Retinoblastoma/drug therapy , Retinoblastoma/genetics , Retinoblastoma/pathology , Sunitinib/pharmacology , Sunitinib/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Retinal Neoplasms/drug therapy , Retinal Neoplasms/genetics , Retinal Neoplasms/pathology , Organoids/metabolism
4.
Sci Rep ; 10(1): 21924, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318499

ABSTRACT

Olfactomedin 4 (OLFM4) is expressed in normal prostate epithelial cells and immortalized normal human prostate epithelial cells (RWPE1), but the identity of OLFM4-expressing cells within these populations and OLFM4's physiological functions in these cells have not been elucidated. Using single-cell RNA sequencing analysis, we found here that OLFM4 was expressed in multiple stem/progenitor-like cell populations in both the normal prostate epithelium and RWPE1 cells and was frequently co-expressed with KRT13 and LY6D in RWPE1 cells. Functionally, OLFM4-knockout RWPE1 cells exhibited enhanced proliferation of the stem/progenitor-like cell population, shifts stem/progenitor-like cell division to favor symmetric division and differentiated into higher levels PSA expression cells in organoid assays compared with OLFM4-wild RWPE1 cells. Bulk-cell RNA sequencing analysis pinpointed that cMYC expression were enhanced in the OLFM4-knockout RWPE1 cells compared with OLFM4-wild cells. Molecular and signaling pathway studies revealed an increase in the WNT/APC/MYC signaling pathway gene signature, as well as that of MYC target genes that regulate multiple biological processes, in OLFM4-knockout RWPE1 cells. These findings indicated that OLFM4 is co-expressed with multiple stem/progenitor cell marker genes in prostate epithelial cells and acts as a novel mediator in prostate stem/progenitor cell proliferation and differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/biosynthesis , Prostate/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Stem Cells/metabolism , Cell Line, Transformed , Granulocyte Colony-Stimulating Factor/genetics , Humans , Male , Prostate/cytology , Proto-Oncogene Proteins c-myc/genetics , Stem Cells/cytology
5.
Invest Ophthalmol Vis Sci ; 61(14): 8, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33270844

ABSTRACT

Purpose: MYCNOS (MYCN opposite strand) is co-amplified with MYCN in pediatric cancers, including retinoblastoma. MYCNOS encodes several RNA variants whose functions have not been elucidated in retinoblastoma. Thus, we attempted to identify MYCNOS variants in retinoblastoma and aimed to decipher the role of MYCNOS variant 1 (MYCNOS1) on the activity of MYCN-amplified retinoblastoma. Methods: The profiles of MYCNOS variants and MYCN status were determined in 17 retinoblastoma tissues, cell lines, retinas, and retinal organoids. A functional study of MYCNOS1 expression was conducted in patient-derived tumor cells and in retinoblastoma cell lines via short hairpin RNA-mediated gene silencing. We carried out MYCN expression, cell viability, cell cycle, apoptosis, soft agar colony formation, and transwell assays to examine the role of MYCNOS1 in MYCN and cell behaviors. We analyzed a transcriptome of MYCN-amplified retinoblastoma cells deficient for MYCNOS1 and, finally, tested the responses of these cells to chemotherapeutic agents. Results: Expression of MYCNOS1 was associated with the expression and copy number of MYCN. Knockdown of MYCNOS1 caused instability of the MYCN protein, leading to cell cycle arrest and impaired proliferation and chemotaxis-directed migration in MYCN-amplified retinoblastoma cells in which RB1 was intact. MYCNOS1 expression was associated with gene signatures of photoreceptor cells and epithelial-mesenchymal transition. MYCNOS1 silencing enhanced the response of retinoblastoma cells to topotecan but not carboplatin. Conclusions: MYCNOS1 supports progression of retinoblastoma. Inhibition of MYCNOS1 expression may be necessary to suppress MYCN activity when treating MYCN-amplified cancers without RB1 mutation.


Subject(s)
Genes, Retinoblastoma/genetics , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Cell Line , Child , Female , Gene Knockdown Techniques , Gene Silencing , Humans , Male , Mutation/genetics , RNA, Small Interfering/genetics
6.
Sci Data ; 7(1): 326, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020484

ABSTRACT

Long non-coding RNA Knowledgebase (lncRNAKB) is an integrated resource for exploring lncRNA biology in the context of tissue-specificity and disease association. A systematic integration of annotations from six independent databases resulted in 77,199 human lncRNA (224,286 transcripts). The user-friendly knowledgebase covers a comprehensive breadth and depth of lncRNA annotation. lncRNAKB is a compendium of expression patterns, derived from analysis of RNA-seq data in thousands of samples across 31 solid human normal tissues (GTEx). Thousands of co-expression modules identified via network analysis and pathway enrichment to delineate lncRNA function are also accessible. Millions of expression quantitative trait loci (cis-eQTL) computed using whole genome sequence genotype data (GTEx) can be downloaded at lncRNAKB that also includes tissue-specificity, phylogenetic conservation and coding potential scores. Tissue-specific lncRNA-trait associations encompassing 323 GWAS (UK Biobank) are also provided. LncRNAKB is accessible at http://www.lncrnakb.org/ , and the data are freely available through Open Science Framework ( https://doi.org/10.17605/OSF.IO/RU4D2 ).


Subject(s)
Knowledge Bases , Organ Specificity , RNA, Long Noncoding/genetics , Humans , Molecular Sequence Annotation , Phylogeny , Quantitative Trait Loci
7.
Cancers (Basel) ; 12(2)2020 Feb 17.
Article in English | MEDLINE | ID: mdl-32079144

ABSTRACT

Melanoma is among the most malignant cutaneous cancers and when metastasized results in dramatically high mortality. Despite advances in high-throughput gene expression profiling in cancer transcriptomic studies, our understanding of mechanisms driving melanoma progression is still limited. We present here an in-depth bioinformatic analysis of the melanoma RNAseq, chromatin immunoprecipitation (ChIP)seq, and single-cell (sc)RNA seq data to understand cancer progression. Specifically, we have performed a consensus network analysis of RNA-seq data from clinically re-grouped melanoma samples to identify gene co-expression networks that are conserved in early (stage 1) and late (stage 4/invasive) stage melanoma. Overlaying the fold-change information on co-expression networks revealed several coordinately up or down-regulated subnetworks that may play a critical role in melanoma progression. Furthermore, by incorporating histone lysine-27 acetylation information and highly expressed genes identified from the single-cell RNA data from melanoma patient samples, we present a comprehensive list of pathways, putative protein-protein interactions (PPIs) and transcription factor (TF) networks that are driving cancer progression. From this analysis, we have identified Elk1, AP1 and E12 TF networks that coordinately change expression in late melanoma when compared to early melanoma, implicating these TFs in melanoma progression. Additionally, the sumoylation-associated interactome is upregulated in invasive melanoma. Together, this bioinformatic analysis potentially implicates a combination of TF networks and PPIs in melanoma progression, which if confirmed in the experimental systems, could be used as targets for drug intervention in melanoma.

8.
Stem Cell Reports ; 13(5): 891-905, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31631019

ABSTRACT

Stem cell-derived retinal organoids recapitulate many landmarks of in vivo differentiation but lack functional maturation of distinct cell types, especially photoreceptors. Using comprehensive temporal transcriptome analyses, we show that transcriptome shift from postnatal day 6 (P6) to P10, associated with morphogenesis and synapse formation during mouse retina development, was not evident in organoids, and co-expression clusters with similar patterns included different sets of genes. Furthermore, network analysis identified divergent regulatory dynamics between developing retina in vivo and in organoids, with temporal dysregulation of specific signaling pathways and delayed or reduced expression of genes involved in photoreceptor function(s) and survival. Accordingly, addition of docosahexaenoic acid and fibroblast growth factor 1 to organoid cultures specifically promoted the maturation of photoreceptors, including cones. Our study thus identifies regulatory signals deficient in developing retinal organoids and provides experimental validation by producing a more mature retina in vitro, thereby facilitating investigations in disease modeling and therapies.


Subject(s)
Organoids/cytology , Retina/cytology , Transcriptome , Animals , Cell Differentiation , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Organoids/growth & development , Organoids/metabolism , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Retina/growth & development , Retina/metabolism , Signal Transduction
9.
Elife ; 82019 10 15.
Article in English | MEDLINE | ID: mdl-31612862

ABSTRACT

Oogenesis features an enormous increase in mitochondrial mass and mtDNA copy number, which are required to furnish mature eggs with an adequate supply of mitochondria and to curb the transmission of deleterious mtDNA variants. Quiescent in dividing germ cells, mtDNA replication initiates upon oocyte determination in the Drosophila ovary, which necessitates active mitochondrial respiration. However, the underlying mechanism for this dynamic regulation remains unclear. Here, we show that an feedforward insulin-Myc loop promotes mitochondrial respiration and biogenesis by boosting the expression of electron transport chain subunits and of factors essential for mtDNA replication and expression, and for the import of mitochondrial proteins. We further reveal that transient activation of JNK enhances the expression of the insulin receptor and initiates the insulin-Myc signaling loop. This signaling relay promotes mitochondrial biogenesis in the ovary, and thereby plays a role in limiting the transmission of deleterious mtDNA mutations. Our study demonstrates cellular mechanisms that couple mitochondrial biogenesis and inheritance with oocyte development.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Electron Transport Chain Complex Proteins/biosynthesis , Genes, Mitochondrial , Insulin/metabolism , MAP Kinase Kinase 4/metabolism , Mitochondria/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/metabolism , Animals , Drosophila , Mitochondria/enzymology , Organelle Biogenesis , Signal Transduction
10.
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
11.
BMC Genomics ; 19(1): 877, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30518325

ABSTRACT

BACKGROUND: The health and resilience of species in natural environments is increasingly challenged by complex anthropogenic stressor combinations including climate change, habitat encroachment, and chemical contamination. To better understand impacts of these stressors we examined the individual- and combined-stressor impacts of malaria infection, food limitation, and 2,4,6-trinitrotoluene (TNT) exposures on gene expression in livers of Western fence lizards (WFL, Sceloporus occidentalis) using custom WFL transcriptome-based microarrays. RESULTS: Computational analysis including annotation enrichment and correlation analysis identified putative functional mechanisms linking transcript expression and toxicological phenotypes. TNT exposure increased transcript expression for genes involved in erythropoiesis, potentially in response to TNT-induced anemia and/or methemoglobinemia and caused dose-specific effects on genes involved in lipid and overall energy metabolism consistent with a hormesis response of growth stimulation at low doses and adverse decreases in lizard growth at high doses. Functional enrichment results were indicative of inhibited potential for lipid mobilization and catabolism in TNT exposures which corresponded with increased inguinal fat weights and was suggestive of a decreased overall energy budget. Malaria infection elicited enriched expression of multiple immune-related functions likely corresponding to increased white blood cell (WBC) counts. Food limitation alone enriched functions related to cellular energy production and decreased expression of immune responses consistent with a decrease in WBC levels. CONCLUSIONS: Despite these findings, the lizards demonstrated immune resilience to malaria infection under food limitation with transcriptional results indicating a fully competent immune response to malaria, even under bio-energetic constraints. Interestingly, both TNT and malaria individually increased transcriptional expression of immune-related genes and increased overall WBC concentrations in blood; responses that were retained in the TNT x malaria combined exposure. The results demonstrate complex and sometimes unexpected responses to multiple stressors where the lizards displayed remarkable resiliency to the stressor combinations investigated.


Subject(s)
Environmental Pollutants/toxicity , Lizards/metabolism , Transcriptome/drug effects , Animals , Body Weight/drug effects , Climate Change , Cluster Analysis , Ecosystem , Energy Metabolism/drug effects , Erythropoiesis/drug effects , Hemolysis/drug effects , Liver/drug effects , Liver/metabolism , Lizards/genetics , Lizards/parasitology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Plasmodium/pathogenicity , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Sequence Analysis, RNA , Spleen/parasitology , Spleen/physiology , Trinitrotoluene/toxicity
12.
Sci Rep ; 8(1): 15664, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30353124

ABSTRACT

Persistent or recurrent retinoblastoma (RB) is associated with the presence of vitreous or/and subretinal seeds in advanced RB and represents a major cause of therapeutic failure. This necessitates the development of novel therapies and thus requires a model of advanced RB for testing candidate therapeutics. To this aim, we established and characterized a three-dimensional, self-organizing organoid model derived from chemotherapy-naïve tumors. The responses of organoids to drugs were determined and compared to relate organoid model to advanced RB, in terms of drug sensitivities. We found that organoids had histological features resembling retinal tumors and seeds and retained DNA copy-number alterations as well as gene and protein expression of the parental tissue. Cone signal circuitry (M/L+ cells) and glial tumor microenvironment (GFAP+ cells) were primarily present in organoids. Topotecan alone or the combined drug regimen of topotecan and melphalan effectively targeted proliferative tumor cones (RXRγ+ Ki67+) in organoids after 24-h drug exposure, blocking mitotic entry. In contrast, methotrexate showed the least efficacy against tumor cells. The drug responses of organoids were consistent with those of tumor cells in advanced disease. Patient-derived organoids enable the creation of a faithful model to use in examining novel therapeutics for RB.


Subject(s)
Carcinogenesis/pathology , Models, Biological , Organoids/pathology , Retinoblastoma/drug therapy , Retinoblastoma/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Child, Preschool , DNA Copy Number Variations/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Infant , Melphalan/pharmacology , Melphalan/therapeutic use , Neoplasm Staging , Organoids/drug effects , Retinal Neoplasms/drug therapy , Retinal Neoplasms/pathology , Retinoblastoma/genetics , Topotecan/pharmacology , Topotecan/therapeutic use
13.
Prog Retin Eye Res ; 65: 1-27, 2018 07.
Article in English | MEDLINE | ID: mdl-29544768

ABSTRACT

Complex biological processes, such as organogenesis and homeostasis, are stringently regulated by genetic programs that are fine-tuned by epigenetic factors to establish cell fates and/or to respond to the microenvironment. Gene regulatory networks that guide cell differentiation and function are modulated and stabilized by modifications to DNA, RNA and proteins. In this review, we focus on two key epigenetic changes - DNA methylation and histone modifications - and discuss their contribution to retinal development, aging and disease, especially in the context of age-related macular degeneration (AMD) and diabetic retinopathy. We highlight less-studied roles of DNA methylation and provide the RNA expression profiles of epigenetic enzymes in human and mouse retina in comparison to other tissues. We also review computational tools and emergent technologies to profile, analyze and integrate epigenetic information. We suggest implementation of editing tools and single-cell technologies to trace and perturb the epigenome for delineating its role in transcriptional regulation. Finally, we present our thoughts on exciting avenues for exploring epigenome in retinal metabolism, disease modeling, and regeneration.


Subject(s)
Epigenesis, Genetic/physiology , Epigenomics , Gene Expression Regulation/physiology , Macular Degeneration/genetics , Retina , DNA Methylation/physiology , Gene Regulatory Networks/physiology , Humans , RNA/metabolism , Retina/embryology , Retina/physiology
14.
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
15.
Invest Ophthalmol Vis Sci ; 58(11): 4422-4435, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28863214

ABSTRACT

Purpose: Transcriptome analysis by next generation sequencing allows qualitative and quantitative profiling of expression patterns associated with development and disease. However, most transcribed sequences do not encode proteins, and little is known about the functional relevance of noncoding (nc) transcriptome in neuronal subtypes. The goal of this study was to perform a comprehensive analysis of long noncoding (lncRNAs) and antisense (asRNAs) RNAs expressed in mouse retinal photoreceptors. Methods: Transcriptomic profiles were generated at six developmental time points from flow-sorted Nrlp-GFP (rods) and Nrlp-GFP;Nrl-/- (S-cone like) mouse photoreceptors. Bioinformatic analysis was performed to identify novel noncoding transcripts and assess their regulation by rod differentiation factor neural retina leucine zipper (NRL). In situ hybridization (ISH) was used for validation and cellular localization. Results: NcRNA profiles demonstrated dynamic yet specific expression signature and coexpression clusters during rod development. In addition to currently annotated 586 lncRNAs and 454 asRNAs, we identified 1037 lncRNAs and 243 asRNAs by de novo assembly. Of these, 119 lncRNAs showed altered expression in the absence of NRL and included NRL binding sites in their promoter/enhancer regions. ISH studies validated the expression of 24 lncRNAs (including 12 previously unannotated) and 4 asRNAs in photoreceptors. Coexpression analysis demonstrated 63 functional modules and 209 significant antisense-gene correlations, allowing us to predict possible role of these lncRNAs in rods. Conclusions: Our studies reveal coregulation of coding and noncoding transcripts in rod photoreceptors by NRL and establish the framework for deciphering the function of ncRNAs during retinal development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Eye Proteins/metabolism , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , Retina/growth & development , Retinal Rod Photoreceptor Cells/metabolism , Transcriptome/genetics , Animals , Cell Differentiation/physiology , Flow Cytometry , Gene Expression Regulation/physiology , High-Throughput Nucleotide Sequencing , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism
16.
Nat Commun ; 8: 14716, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28291770

ABSTRACT

In retinitis pigmentosa, loss of cone photoreceptors leads to blindness, and preservation of cone function is a major therapeutic goal. However, cone loss is thought to occur as a secondary event resulting from degeneration of rod photoreceptors. Here we report a genome editing approach in which adeno-associated virus (AAV)-mediated CRISPR/Cas9 delivery to postmitotic photoreceptors is used to target the Nrl gene, encoding for Neural retina-specific leucine zipper protein, a rod fate determinant during photoreceptor development. Following Nrl disruption, rods gain partial features of cones and present with improved survival in the presence of mutations in rod-specific genes, consequently preventing secondary cone degeneration. In three different mouse models of retinal degeneration, the treatment substantially improves rod survival and preserves cone function. Our data suggest that CRISPR/Cas9-mediated NRL disruption in rods may be a promising treatment option for patients with retinitis pigmentosa.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Cell Survival/genetics , Eye Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/metabolism , Animals , CRISPR-Cas Systems , Dependovirus , Gene Editing/methods , Gene Knockdown Techniques , Mice , Retinal Cone Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/cytology , Retinitis Pigmentosa/genetics
17.
Cell Rep ; 17(9): 2460-2473, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27880916

ABSTRACT

Gene regulatory networks (GRNs) guiding differentiation of cell types and cell assemblies in the nervous system are poorly understood because of inherent complexities and interdependence of signaling pathways. Here, we report transcriptome dynamics of differentiating rod photoreceptors in the mammalian retina. Given that the transcription factor NRL determines rod cell fate, we performed expression profiling of developing NRL-positive (rods) and NRL-negative (S-cone-like) mouse photoreceptors. We identified a large-scale, sharp transition in the transcriptome landscape between postnatal days 6 and 10 concordant with rod morphogenesis. Rod-specific temporal DNA methylation corroborated gene expression patterns. De novo assembly and alternative splicing analyses revealed previously unannotated rod-enriched transcripts and the role of NRL in transcript maturation. Furthermore, we defined the relationship of NRL with other transcriptional regulators and downstream cognate effectors. Our studies provide the framework for comprehensive system-level analysis of the GRN underlying the development of a single sensory neuron, the rod photoreceptor.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Retinal Cone Photoreceptor Cells/metabolism , Transcriptome/genetics , Alternative Splicing/genetics , Animals , Animals, Newborn , Cell Differentiation/genetics , Computer Simulation , DNA Methylation/genetics , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Annotation , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Invest Ophthalmol Vis Sci ; 57(14): 6374-6381, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27898983

ABSTRACT

Purpose: A majority of genes associated with inherited retinal diseases (IRDs) have been identified in patients of European origin. Indigenous African populations exhibit rich genomic diversity, and evaluation of reported genetic mutations has yielded low returns so far. Our goal was to perform whole-exome sequencing (WES) to examine variants in known IRD genes in underrepresented African cohorts. Methods: Whole-exome sequencing was performed on 56 samples from 16 families with diverse IRD phenotypes that had remained undiagnosed after screening for known mutations using genotyping-based microarrays (Asper Ophthalmics). Variants in reported IRD genes were identified using WES and validated by Sanger sequencing. Custom TaqMan assays were used to screen for identified mutations in 193 unrelated indigenous Africans with IRDs. Results: A total of 3494 variants were identified in 217 known IRD genes, leading to the identification of seven different mutations (including six novel) in six genes (RHO, PRPF3, PRPF31, ABCA4, CERKL, and PDE6B) in six distinct families. TaqMan screening in additional probands revealed identical homozygous CERKL and PDE6B variants in four more patients. Conclusions: This is the first report of WES of patients with IRDs in indigenous African populations. Our study identified genetic defects in almost 40% of the families analyzed, significantly enhancing the molecular diagnosis of IRD in South Africa. Thus, WES of understudied cohorts seems to present an effective strategy for determining novel mutations in heterogeneous retinal diseases.


Subject(s)
DNA/genetics , Exome/genetics , Eye Proteins/genetics , Genome-Wide Association Study/methods , Molecular Diagnostic Techniques/methods , Mutation , Retinal Diseases/diagnosis , DNA Mutational Analysis , Eye Proteins/metabolism , Female , Genotype , Humans , Incidence , Male , Pedigree , Phenotype , Retinal Diseases/epidemiology , Retinal Diseases/genetics , Risk Factors , South Africa/epidemiology , Young Adult
19.
Prog Retin Eye Res ; 55: 1-31, 2016 11.
Article in English | MEDLINE | ID: mdl-27297499

ABSTRACT

The advent of high throughput next generation sequencing (NGS) has accelerated the pace of discovery of disease-associated genetic variants and genomewide profiling of expressed sequences and epigenetic marks, thereby permitting systems-based analyses of ocular development and disease. Rapid evolution of NGS and associated methodologies presents significant challenges in acquisition, management, and analysis of large data sets and for extracting biologically or clinically relevant information. Here we illustrate the basic design of commonly used NGS-based methods, specifically whole exome sequencing, transcriptome, and epigenome profiling, and provide recommendations for data analyses. We briefly discuss systems biology approaches for integrating multiple data sets to elucidate gene regulatory or disease networks. While we provide examples from the retina, the NGS guidelines reviewed here are applicable to other tissues/cell types as well.


Subject(s)
Computational Biology/methods , Genome-Wide Association Study , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Retinal Diseases/genetics , Humans
20.
Invest Ophthalmol Vis Sci ; 57(5): ORSFl1-ORSFl11, 2016 04 01.
Article in English | MEDLINE | ID: mdl-27116668

ABSTRACT

We discuss the use of pluripotent stem cell lines carrying fluorescent reporters driven by retinal promoters to derive three-dimensional (3-D) retina in culture and how this system can be exploited for elucidating human retinal biology, creating disease models in a dish, and designing targeted drug screens for retinal and macular degeneration. Furthermore, we realize that stem cell investigations are labor-intensive and require extensive resources. To expedite scientific discovery by sharing of resources and to avoid duplication of efforts, we propose the formation of a Retinal Stem Cell Consortium. In the field of vision, such collaborative approaches have been enormously successful in elucidating genetic susceptibility associated with age-related macular degeneration.


Subject(s)
Pluripotent Stem Cells , Retina/cytology , Retinal Degeneration , Tissue Culture Techniques/methods , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Disease Models, Animal , Gene Expression Profiling , Genes, Reporter/physiology , Humans , Macular Degeneration/metabolism , Macular Degeneration/therapy , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/therapy , Retinal Neurons/cytology , Retinal Pigment Epithelium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...