Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.276
Filter
Add more filters

Publication year range
1.
Cell ; 178(4): 980-992.e17, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353220

ABSTRACT

Metabolic conditions affect the developmental tempo of animals. Developmental gene regulatory networks (GRNs) must therefore synchronize their dynamics with a variable timescale. We find that layered repression of genes couples GRN output with variable metabolism. When repressors of transcription or mRNA and protein stability are lost, fewer errors in Drosophila development occur when metabolism is lowered. We demonstrate the universality of this phenomenon by eliminating the entire microRNA family of repressors and find that development to maturity can be largely rescued when metabolism is reduced. Using a mathematical model that replicates GRN dynamics, we find that lowering metabolism suppresses the emergence of developmental errors by curtailing the influence of auxiliary repressors on GRN output. We experimentally show that gene expression dynamics are less affected by loss of repressors when metabolism is reduced. Thus, layered repression provides robustness through error suppression and may provide an evolutionary route to a shorter reproductive cycle.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Neurons/metabolism , Animals , Animals, Genetically Modified , Brain/cytology , Drosophila melanogaster/growth & development , Eye/cytology , Female , Insulin/metabolism , Loss of Function Mutation , MicroRNAs/metabolism , Models, Theoretical , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic
2.
Genes Dev ; 35(9-10): 677-691, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33888564

ABSTRACT

During the development of the vertebrate nervous systems, genetic programs assemble an immature circuit that is subsequently refined by neuronal activity evoked by external stimuli. However, prior to sensory experience, the intrinsic property of the developing nervous system also triggers correlated network-level neuronal activity, with retinal waves in the developing vertebrate retina being the best documented example. Spontaneous activity has also been found in the visual system of Drosophila Here, we compare the spontaneous activity of the developing visual system between mammalian and Drosophila and suggest that Drosophila is an emerging model for mechanistic and functional studies of correlated spontaneous activity.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Retina/cytology , Retina/embryology , Sensory Receptor Cells/physiology , Animals , Drosophila melanogaster/physiology , Eye/cytology , Eye/growth & development , Humans , Models, Animal , Retina/physiology , Sensory Receptor Cells/cytology
3.
Nature ; 588(7836): 124-129, 2020 12.
Article in English | MEDLINE | ID: mdl-33268865

ABSTRACT

Ageing is a degenerative process that leads to tissue dysfunction and death. A proposed cause of ageing is the accumulation of epigenetic noise that disrupts gene expression patterns, leading to decreases in tissue function and regenerative capacity1-3. Changes to DNA methylation patterns over time form the basis of ageing clocks4, but whether older individuals retain the information needed to restore these patterns-and, if so, whether this could improve tissue function-is not known. Over time, the central nervous system (CNS) loses function and regenerative capacity5-7. Using the eye as a model CNS tissue, here we show that ectopic expression of Oct4 (also known as Pou5f1), Sox2 and Klf4 genes (OSK) in mouse retinal ganglion cells restores youthful DNA methylation patterns and transcriptomes, promotes axon regeneration after injury, and reverses vision loss in a mouse model of glaucoma and in aged mice. The beneficial effects of OSK-induced reprogramming in axon regeneration and vision require the DNA demethylases TET1 and TET2. These data indicate that mammalian tissues retain a record of youthful epigenetic information-encoded in part by DNA methylation-that can be accessed to improve tissue function and promote regeneration in vivo.


Subject(s)
Aging/genetics , Cellular Reprogramming/genetics , DNA Methylation , Epigenesis, Genetic , Eye , Nerve Regeneration/genetics , Vision, Ocular/genetics , Vision, Ocular/physiology , Aging/physiology , Animals , Axons/physiology , Cell Line, Tumor , Cell Survival , DNA-Binding Proteins/genetics , Dependovirus/genetics , Dioxygenases , Disease Models, Animal , Eye/cytology , Eye/innervation , Eye/pathology , Female , Genetic Vectors/genetics , Glaucoma/genetics , Glaucoma/pathology , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Optic Nerve Injuries/genetics , Proto-Oncogene Proteins/genetics , Retinal Ganglion Cells/cytology , SOXB1 Transcription Factors/genetics , Transcriptome/genetics
4.
EMBO J ; 40(6): e105123, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33555074

ABSTRACT

Similar to the brain, the eye is considered an immune-privileged organ where tissue-resident macrophages provide the major immune cell constituents. However, little is known about spatially restricted macrophage subsets within different eye compartments with regard to their origin, function, and fate during health and disease. Here, we combined single-cell analysis, fate mapping, parabiosis, and computational modeling to comprehensively examine myeloid subsets in distinct parts of the eye during homeostasis. This approach allowed us to identify myeloid subsets displaying diverse transcriptional states. During choroidal neovascularization, a typical hallmark of neovascular age-related macular degeneration (AMD), we recognized disease-specific macrophage subpopulations with distinct molecular signatures. Our results highlight the heterogeneity of myeloid subsets and their dynamics in the eye that provide new insights into the innate immune system in this organ which may offer new therapeutic targets for ophthalmological diseases.


Subject(s)
Choroid/blood supply , Eye/immunology , Macrophages/immunology , Myeloid Cells/immunology , Neovascularization, Physiologic/physiology , Animals , Choroid/embryology , Computational Biology , Computer Simulation , Eye/cytology , Eye/metabolism , Female , Homeostasis/immunology , Humans , Immunity, Innate/immunology , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/physiology , Myeloid Cells/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic/genetics
5.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33982759

ABSTRACT

Genetic screens are designed to target individual genes for the practical reason of establishing a clear association between a mutant phenotype and a single genetic locus. This allows for a developmental or physiological role to be assigned to the wild-type gene. We previously observed that the concurrent loss of Pax6 and Polycomb epigenetic repressors in Drosophila leads the eye to transform into a wing. This fate change is not seen when either factor is disrupted separately. An implication of this finding is that standard screens may miss the roles that combinations of genes play in development. Here, we show that this phenomenon is not limited to Pax6 and Polycomb but rather applies more generally. We demonstrate that in the Drosophila eye-antennal disc, the simultaneous downregulation of Pax6 with either the NURF nucleosome remodeling complex or the Pointed transcription factor transforms the head epidermis into an antenna. This is a previously unidentified fate change that is also not observed with the loss of individual genes. We propose that the use of multi-gene knockdowns is an essential tool for unraveling the complexity of development.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/physiology , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Animals , Epidermis , Eye/cytology , Eye/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Larva , Nucleosomes , Polycomb-Group Proteins/genetics , Transcription Factors/metabolism
6.
Nature ; 556(7699): 108-112, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29590089

ABSTRACT

Embryonic development is a crucial period in the life of a multicellular organism, during which limited sets of embryonic progenitors produce all cells in the adult body. Determining which fate these progenitors acquire in adult tissues requires the simultaneous measurement of clonal history and cell identity at single-cell resolution, which has been a major challenge. Clonal history has traditionally been investigated by microscopically tracking cells during development, monitoring the heritable expression of genetically encoded fluorescent proteins and, more recently, using next-generation sequencing technologies that exploit somatic mutations, microsatellite instability, transposon tagging, viral barcoding, CRISPR-Cas9 genome editing and Cre-loxP recombination. Single-cell transcriptomics provides a powerful platform for unbiased cell-type classification. Here we present ScarTrace, a single-cell sequencing strategy that enables the simultaneous quantification of clonal history and cell type for thousands of cells obtained from different organs of the adult zebrafish. Using ScarTrace, we show that a small set of multipotent embryonic progenitors generate all haematopoietic cells in the kidney marrow, and that many progenitors produce specific cell types in the eyes and brain. In addition, we study when embryonic progenitors commit to the left or right eye. ScarTrace reveals that epidermal and mesenchymal cells in the caudal fin arise from the same progenitors, and that osteoblast-restricted precursors can produce mesenchymal cells during regeneration. Furthermore, we identify resident immune cells in the fin with a distinct clonal origin from other blood cell types. We envision that similar approaches will have major applications in other experimental systems, in which the matching of embryonic clonal origin to adult cell type will ultimately allow reconstruction of how the adult body is built from a single cell.


Subject(s)
Cell Lineage , Cell Tracking/methods , Clone Cells/cytology , Clone Cells/metabolism , Sequence Analysis/methods , Single-Cell Analysis , Zebrafish/anatomy & histology , Animal Fins/cytology , Animals , Brain/cytology , CRISPR-Cas Systems/genetics , Cell Lineage/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Eye/cytology , Female , Genes, Reporter/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Male , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Organ Specificity , Regeneration , Transcriptome , Whole Body Imaging , Zebrafish/embryology , Zebrafish/genetics
7.
PLoS Genet ; 17(6): e1009146, 2021 06.
Article in English | MEDLINE | ID: mdl-34097697

ABSTRACT

The Hippo pathway is an important regulator of organ growth and cell fate. In the R8 photoreceptor cells of the Drosophila melanogaster eye, the Hippo pathway controls the fate choice between one of two subtypes that express either the blue light-sensitive Rhodopsin 5 (Hippo inactive R8 subtype) or the green light-sensitive Rhodopsin 6 (Hippo active R8 subtype). The degree to which the mechanism of Hippo signal transduction and the proteins that mediate it are conserved in organ growth and R8 cell fate choice is currently unclear. Here, we identify Crumbs and the apical spectrin cytoskeleton as regulators of R8 cell fate. By contrast, other proteins that influence Hippo-dependent organ growth, such as the basolateral spectrin cytoskeleton and Ajuba, are dispensable for the R8 cell fate choice. Surprisingly, Crumbs promotes the Rhodopsin 5 cell fate, which is driven by Yorkie, rather than the Rhodopsin 6 cell fate, which is driven by Warts and the Hippo pathway, which contrasts with its impact on Hippo activity in organ growth. Furthermore, neither the apical spectrin cytoskeleton nor Crumbs appear to regulate the Hippo pathway through mechanisms that have been observed in growing organs. Together, these results show that only a subset of Hippo pathway proteins regulate the R8 binary cell fate decision and that aspects of Hippo signalling differ between growing organs and post-mitotic R8 cells.


Subject(s)
Cell Lineage/physiology , Drosophila Proteins/physiology , Eye Proteins/physiology , Eye/cytology , Membrane Proteins/physiology , Rhodopsin/physiology , Spectrin/physiology , Animals , Cytoskeleton/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster , Eye/growth & development , Intracellular Signaling Peptides and Proteins/metabolism , Photoreceptor Cells, Invertebrate/physiology , Protein Serine-Threonine Kinases/metabolism
8.
Proc Natl Acad Sci U S A ; 117(19): 10339-10349, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32341164

ABSTRACT

Increased intraocular pressure (IOP) represents a major risk factor for glaucoma, a prevalent eye disease characterized by death of retinal ganglion cells; lowering IOP is the only proven treatment strategy to delay disease progression. The main determinant of IOP is the equilibrium between production and drainage of aqueous humor, with compromised drainage generally viewed as the primary contributor to dangerous IOP elevations. Drainage occurs through two pathways in the anterior segment of the eye called conventional and uveoscleral. To gain insights into the cell types that comprise these pathways, we used high-throughput single-cell RNA sequencing (scRNAseq). From ∼24,000 single-cell transcriptomes, we identified 19 cell types with molecular markers for each and used histological methods to localize each type. We then performed similar analyses on four organisms used for experimental studies of IOP dynamics and glaucoma: cynomolgus macaque (Macaca fascicularis), rhesus macaque (Macaca mulatta), pig (Sus scrofa), and mouse (Mus musculus). Many human cell types had counterparts in these models, but differences in cell types and gene expression were evident. Finally, we identified the cell types that express genes implicated in glaucoma in all five species. Together, our results provide foundations for investigating the pathogenesis of glaucoma and for using model systems to assess mechanisms and potential interventions.


Subject(s)
Aqueous Humor/metabolism , Disease Models, Animal , Eye/metabolism , Glaucoma/pathology , Intraocular Pressure , Trabecular Meshwork/metabolism , Transcriptome , Animals , Biomarkers/analysis , Eye/cytology , Glaucoma/metabolism , Humans , Macaca fascicularis , Macaca mulatta , Mice , Species Specificity , Swine
9.
Proc Natl Acad Sci U S A ; 117(51): 32545-32556, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33288705

ABSTRACT

Apoptosis, a conserved form of programmed cell death, shows interspecies differences that may reflect evolutionary diversification and adaptation, a notion that remains largely untested. Among insects, the most speciose animal group, the apoptotic pathway has only been fully characterized in Drosophila melanogaster, and apoptosis-related proteins have been studied in a few other dipteran and lepidopteran species. Here, we studied the apoptotic pathway in the aphid Acyrthosiphon pisum, an insect pest belonging to the Hemiptera, an earlier-diverging and distantly related order. We combined phylogenetic analyses and conserved domain identification to annotate the apoptotic pathway in A. pisum and found low caspase diversity and a large expansion of its inhibitory part, with 28 inhibitors of apoptosis (IAPs). We analyzed the spatiotemporal expression of a selected set of pea aphid IAPs and showed that they are differentially expressed in different life stages and tissues, suggesting functional diversification. Five IAPs are specifically induced in bacteriocytes, the specialized cells housing symbiotic bacteria, during their cell death. We demonstrated the antiapoptotic role of these five IAPs using heterologous expression in a tractable in vivo model, the Drosophila melanogaster developing eye. Interestingly, IAPs with the strongest antiapoptotic potential contain two BIR and two RING domains, a domain association that has not been observed in any other species. We finally analyzed all available aphid genomes and found that they all show large IAP expansion, with new combinations of protein domains, suggestive of evolutionarily novel aphid-specific functions.


Subject(s)
Aphids/cytology , Aphids/physiology , Apoptosis/physiology , Insect Proteins/chemistry , Insect Proteins/metabolism , Animals , Animals, Genetically Modified , Caspases/chemistry , Caspases/metabolism , Drosophila melanogaster/genetics , Eye/cytology , Eye/pathology , Gene Expression Regulation , Genome, Insect , Inhibitor of Apoptosis Proteins/metabolism , Insect Proteins/genetics , Phylogeny , Protein Domains
10.
Nature ; 532(7600): 500-3, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-27074512

ABSTRACT

Tullimonstrum gregarium is an iconic soft-bodied fossil from the Carboniferous Mazon Creek Lagerstätte (Illinois, USA). Despite a large number of specimens and distinct anatomy, various analyses over the past five decades have failed to determine the phylogenetic affinities of the 'Tully monster', and although it has been allied to such disparate phyla as the Mollusca, Annelida or Chordata, it remains enigmatic. The nature and phylogenetic affinities of Tullimonstrum have defied confident systematic placement because none of its preserved anatomy provides unequivocal evidence of homology, without which comparative analysis fails. Here we show that the eyes of Tullimonstrum possess ultrastructural details indicating homology with vertebrate eyes. Anatomical analysis using scanning electron microscopy reveals that the eyes of Tullimonstrum preserve a retina defined by a thick sheet comprising distinct layers of spheroidal and cylindrical melanosomes. Time-of-flight secondary ion mass spectrometry and multivariate statistics provide further evidence that these microbodies are melanosomes. A range of animals have melanin in their eyes, but the possession of melanosomes of two distinct morphologies arranged in layers, forming retinal pigment epithelium, is a synapomorphy of vertebrates. Our analysis indicates that in addition to evidence of colour patterning, ecology and thermoregulation, fossil melanosomes can also carry a phylogenetic signal. Identification in Tullimonstrum of spheroidal and cylindrical melanosomes forming the remains of retinal pigment epithelium indicates that it is a vertebrate; considering its body parts in this new light suggests it was an anatomically unusual member of total group Vertebrata.


Subject(s)
Eye , Fossils , Phylogeny , Vertebrates/classification , Animals , Eye/chemistry , Eye/cytology , Eye/ultrastructure , Illinois , Melanosomes/ultrastructure , Microscopy, Electron, Scanning , Retinal Pigment Epithelium/chemistry , Retinal Pigment Epithelium/ultrastructure , Vertebrates/anatomy & histology
11.
Proc Natl Acad Sci U S A ; 116(38): 19046-19054, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31484764

ABSTRACT

Naturalists have been fascinated for centuries by animal colors and color patterns. While widely studied at the adult stage, we know little about color patterns in the embryo. Here, we study a trait consisting of coloration that is specific to the embryo and absent from postembryonic stages in water striders (Gerromorpha). By combining developmental genetics with chemical and phylogenetic analyses across a broad sample of species, we uncovered the mechanisms underlying the emergence and diversification of embryonic colors in this group of insects. We show that the pteridine biosynthesis pathway, which ancestrally produces red pigment in the eyes, has been recruited during embryogenesis in various extraocular tissues including antennae and legs. In addition, we discovered that this cooption is common to all water striders and initially resulted in the production of yellow extraocular color. Subsequently, 6 lineages evolved bright red color and 2 lineages lost the color independently. Despite the high diversity in colors and color patterns, we show that the underlying biosynthesis pathway remained stable throughout the 200 million years of Gerromorpha evolutionary time. Finally, we identified erythropterin and xanthopterin as the pigments responsible for these colors in the embryo of various species. These findings demonstrate how traits can emerge through the activation of a biosynthesis pathway in new developmental contexts.


Subject(s)
Color , Embryo, Nonmammalian/metabolism , Heteroptera/physiology , Pigmentation/physiology , Pigments, Biological/metabolism , Pteridines/metabolism , Signal Transduction , Animals , Biological Evolution , Embryo, Nonmammalian/cytology , Eye/cytology , Eye/metabolism , Heteroptera/classification , Phenotype , Phylogeny
12.
Dev Biol ; 464(1): 53-70, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32464117

ABSTRACT

Hippo signaling is an important regulator of tissue size, but it also has a lesser-known role in tissue morphogenesis. Here we use the Drosophila pupal eye to explore the role of the Hippo effector Yki and its cofactor Mask in morphogenesis. We found that Mask is required for the correct distribution and accumulation of adherens junctions and appropriate organization of the cytoskeleton. Accordingly, disrupting mask expression led to severe mis-patterning and similar defects were observed when yki was reduced or in response to ectopic wts. Further, the patterning defects generated by reducing mask expression were modified by Hippo pathway activity. RNA-sequencing revealed a requirement for Mask for appropriate expression of numerous genes during eye morphogenesis. These included genes implicated in cell adhesion and cytoskeletal organization, a comprehensive set of genes that promote cell survival, and numerous signal transduction genes. To validate our transcriptome analyses, we then considered two loci that were modified by Mask activity: FER and Vinc, which have established roles in regulating adhesion. Modulating the expression of either locus modified mask mis-patterning and adhesion phenotypes. Further, expression of FER and Vinc was modified by Yki. It is well-established that the Hippo pathway is responsive to changes in cell adhesion and the cytoskeleton, but our data indicate that Hippo signaling also regulates these structures.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Eye/embryology , Gene Expression Regulation, Developmental/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Organogenesis/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Eye/cytology , Eye Proteins/genetics , Eye Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , YAP-Signaling Proteins
13.
J Biol Chem ; 295(11): 3456-3465, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32034090

ABSTRACT

PITX2 (Paired-like homeodomain transcription factor 2) plays important roles in asymmetric development of the internal organs and symmetric development of eye tissues. During eye development, cranial neural crest cells migrate from the neural tube and form the periocular mesenchyme (POM). POM cells differentiate into several ocular cell types, such as corneal endothelial cells, keratocytes, and some ocular mesenchymal cells. In this study, we used transcription activator-like effector nuclease technology to establish a human induced pluripotent stem cell (hiPSC) line expressing a fluorescent reporter gene from the PITX2 promoter. Using homologous recombination, we heterozygously inserted a PITX2-IRES2-EGFP sequence downstream of the stop codon in exon 8 of PITX2 Cellular pluripotency was monitored with alkaline phosphatase and immunofluorescence staining of pluripotency markers, and the hiPSC line formed normal self-formed ectodermal autonomous multizones. Using a combination of previously reported methods, we induced PITX2 in the hiPSC line and observed simultaneous EGFP and PITX2 expression, as indicated by immunoblotting and immunofluorescence staining. PITX2 mRNA levels were increased in EGFP-positive cells, which were collected by cell sorting, and marker gene expression analysis of EGFP-positive cells induced in self-formed ectodermal autonomous multizones revealed that they were genuine POM cells. Moreover, after 2 days of culture, EGFP-positive cells expressed the PITX2 protein, which co-localized with forkhead box C1 (FOXC1) protein in the nucleus. We anticipate that the PITX2-EGFP hiPSC reporter cell line established and validated here can be utilized to isolate POM cells and to analyze PITX2 expression during POM cell induction.


Subject(s)
Cell Separation , Eye/cytology , Genes, Reporter , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Clone Cells , Ectoderm/cytology , Embryo, Mammalian/cytology , Fluorescence , Humans , Mice, Inbred ICR , Phenotype , Promoter Regions, Genetic/genetics , RNA Splicing/genetics , Reproducibility of Results , Homeobox Protein PITX2
14.
Development ; 145(22)2018 11 19.
Article in English | MEDLINE | ID: mdl-30333214

ABSTRACT

Establishment of precise three-dimensional tissue structure is vital for organ function. In the visual system, optic fissure and stalk morphogenesis is a crucial yet poorly understood process, disruptions of which can lead to coloboma, a birth defect causing visual impairment. Here, we use four-dimensional imaging, cell tracking, and molecular genetics in zebrafish to define the cell movements underlying normal optic fissure and stalk formation. We determine how these events are disrupted in a coloboma model in which the Hedgehog (Hh) receptor ptch2 is lost, resulting in overactive Hh signaling. In the ptch2 mutant, cells exhibit defective motile behaviors and morphology. Cells that should contribute to the fissure do not arrive at their correct position, and instead contribute to an ectopically large optic stalk. Our results suggest that overactive Hh signaling, through overexpression of downstream transcriptional targets, impairs cell motility underlying optic fissure and stalk formation, via non-cell-autonomous and cell-autonomous mechanisms. More broadly, our cell motility and morphology analyses provide a new framework for studying other coloboma-causing mutations that disrupt optic fissure or stalk formation.


Subject(s)
Cell Movement , Eye/cytology , Eye/growth & development , Hedgehog Proteins/metabolism , Morphogenesis , Signal Transduction , Zebrafish/growth & development , Zebrafish/metabolism , Animals , Eye/anatomy & histology , Models, Biological , Mutation/genetics , Transcription, Genetic , Zebrafish Proteins/metabolism
15.
PLoS Genet ; 14(1): e1007173, 2018 01.
Article in English | MEDLINE | ID: mdl-29324767

ABSTRACT

Transcriptional regulators can specify different cell types from a pool of equivalent progenitors by activating distinct developmental programs. The Glass transcription factor is expressed in all progenitors in the developing Drosophila eye, and is maintained in both neuronal and non-neuronal cell types. Glass is required for neuronal progenitors to differentiate as photoreceptors, but its role in non-neuronal cone and pigment cells is unknown. To determine whether Glass activity is limited to neuronal lineages, we compared the effects of misexpressing it in neuroblasts of the larval brain and in epithelial cells of the wing disc. Glass activated overlapping but distinct sets of genes in these neuronal and non-neuronal contexts, including markers of photoreceptors, cone cells and pigment cells. Coexpression of other transcription factors such as Pax2, Eyes absent, Lozenge and Escargot enabled Glass to induce additional genes characteristic of the non-neuronal cell types. Cell type-specific glass mutations generated in cone or pigment cells using somatic CRISPR revealed autonomous developmental defects, and expressing Glass specifically in these cells partially rescued glass mutant phenotypes. These results indicate that Glass is a determinant of organ identity that acts in both neuronal and non-neuronal cells to promote their differentiation into functional components of the eye.


Subject(s)
Cell Differentiation/genetics , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster , Eye/embryology , Eye/metabolism , Neural Stem Cells/physiology , Stem Cells/physiology , Animals , Animals, Genetically Modified , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Eye/cytology , Female , Gene Expression Regulation, Developmental , Organogenesis/genetics
16.
PLoS Genet ; 14(3): e1007244, 2018 03.
Article in English | MEDLINE | ID: mdl-29529029

ABSTRACT

A mismatch between optical power and ocular axial length results in refractive errors. Uncorrected refractive errors constitute the most common cause of vision loss and second leading cause of blindness worldwide. Although the retina is known to play a critical role in regulating ocular growth and refractive development, the precise factors and mechanisms involved are poorly defined. We have previously identified a role for the secreted serine protease PRSS56 in ocular size determination and PRSS56 variants have been implicated in the etiology of both hyperopia and myopia, highlighting its importance in refractive development. Here, we use a combination of genetic mouse models to demonstrate that Prss56 mutations leading to reduced ocular size and hyperopia act via a loss of function mechanism. Using a conditional gene targeting strategy, we show that PRSS56 derived from Müller glia contributes to ocular growth, implicating a new retinal cell type in ocular size determination. Importantly, we demonstrate that persistent activity of PRSS56 is required during distinct developmental stages spanning the pre- and post-eye opening periods to ensure optimal ocular growth. Thus, our mouse data provide evidence for the existence of a molecule contributing to both the prenatal and postnatal stages of human ocular growth. Finally, we demonstrate that genetic inactivation of Prss56 rescues axial elongation in a mouse model of myopia caused by a null mutation in Egr1. Overall, our findings identify PRSS56 as a potential therapeutic target for modulating ocular growth aimed at preventing or slowing down myopia, which is reaching epidemic proportions.


Subject(s)
Eye/growth & development , Gene Expression Regulation, Developmental , Refractive Errors/genetics , Serine Proteases/metabolism , Animals , Disease Models, Animal , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Eye/cytology , Eye/embryology , Female , Humans , Hyperopia/genetics , Male , Mice, Mutant Strains , Mice, Transgenic , Myopia/genetics , Myopia/pathology , Neuroglia/metabolism , Refraction, Ocular/genetics , Refraction, Ocular/physiology , Refractive Errors/prevention & control , Serine Proteases/genetics
17.
PLoS Genet ; 14(5): e1007391, 2018 05.
Article in English | MEDLINE | ID: mdl-29758044

ABSTRACT

Planar cell polarity (PCP) instructs tissue patterning in a wide range of organisms from fruit flies to humans. PCP signaling coordinates cell behavior across tissues and is integrated by cells to couple cell fate identity with position in a developing tissue. In the fly eye, PCP signaling is required for the specification of R3 and R4 photoreceptors based upon their positioning relative to the dorso-ventral axis. The 'core' PCP pathway involves the asymmetric localization of two distinct membrane-bound complexes, one containing Frizzled (Fz, required in R3) and the other Van Gogh (Vang, required in R4). Inhibitory interactions between the cytosolic components of each complex reinforce asymmetric localization. Prickle (Pk) and Spiny-legs (Pk-Sple) are two antagonistic isoforms of the prickle (pk) gene and are cytoplasmic components of the Vang complex. The balance between their levels is critical for tissue patterning, with Pk-Sple being the major functional isoform in the eye. Here we uncover a post-translational role for Nemo kinase in limiting the amount of the minor isoform Pk. We identified Pk as a Nemo substrate in a genome-wide in vitro band-shift screen. In vivo, nemo genetically interacts with pkpk but not pksple and enhances PCP defects in the eye and leg. Nemo phosphorylation limits Pk levels and is required specifically in the R4 photoreceptor like the major isoform, Pk-Sple. Genetic interaction and biochemical data suggest that Nemo phosphorylation of Pk leads to its proteasomal degradation via the Cullin1/SkpA/Slmb complex. dTAK and Homeodomain interacting protein kinase (Hipk) may also act together with Nemo to target Pk for degradation, consistent with similar observations in mammalian studies. Our results therefore demonstrate a mechanism to maintain low levels of the minor Pk isoform, allowing PCP complexes to form correctly and specify cell fate.


Subject(s)
Cell Polarity/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , LIM Domain Proteins/genetics , Mitogen-Activated Protein Kinases/genetics , Animals , Animals, Genetically Modified , Cell Line , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye/cytology , Eye/metabolism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , LIM Domain Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis , Signal Transduction/genetics , Substrate Specificity , Wings, Animal/cytology , Wings, Animal/metabolism
18.
Mar Drugs ; 18(6)2020 May 26.
Article in English | MEDLINE | ID: mdl-32466624

ABSTRACT

Fucoidans from brown seaweeds are promising substances as potential drugs against age-related macular degeneration (AMD). The heterogeneity of fucoidans requires intensive research in order to find suitable species and extraction methods. Ten different fucoidan samples extracted enzymatically from Laminaria digitata (LD), Saccharina latissima (SL) and Fucus distichus subsp. evanescens (FE) were tested for toxicity, oxidative stress protection and VEGF (vascular endothelial growth factor) inhibition. For this study crude fucoidans were extracted from seaweeds using different enzymes and SL fucoidans were further separated into three fractions (SL_F1-F3) by ion-exchange chromatography (IEX). Fucoidan composition was analyzed by high performance anion exchange chromatography (HPAEC) after acid hydrolysis. The crude extracts contained alginate, while two of the fractionated SL fucoidans SL_F2 and SL_F3 were highly pure. Cell viability was assessed with an 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay in OMM-1 and ARPE-19. Protective effects were investigated after 24 h of stress insult in OMM-1 and ARPE-19. Secreted VEGF was analyzed via ELISA (enzyme-linked immunosorbent assay) in ARPE-19 cells. Fucoidans showed no toxic effects. In OMM-1 SL_F2 and several FE fucoidans were protective. LD_SiAT2 (Cellic®CTec2 + Sigma-Aldrich alginate lyase), FE_SiAT3 (Cellic® CTec3 + Sigma-Aldrich alginate lyase), SL_F2 and SL_F3 inhibited VEGF with the latter two as the most effective. We could show that enzyme treated fucoidans in general and the fractionated SL fucoidans SL_F2 and SL_F3 are very promising for beneficial AMD relevant biological activities.


Subject(s)
Cell Survival/drug effects , Eye/cytology , Macular Degeneration/prevention & control , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Protective Agents/pharmacology , Seaweed/chemistry , Humans , Ophthalmic Solutions
19.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233821

ABSTRACT

During development, the precise control of tissue morphogenesis requires changes in the cell number, size, shape, position, and gene expression, which are driven by both chemical and mechanical cues from the surrounding microenvironment. Such physical and architectural features inform cells about their proliferative and migratory capacity, enabling the formation and maintenance of complex tissue architecture. In polarised epithelia, the apical cell cortex, a thin actomyosin network that lies directly underneath the apical plasma membrane, functions as a platform to facilitate signal transmission between the external environment and downstream signalling pathways. One such signalling pathway culminates in the regulation of YES-associated protein (YAP) and TAZ transcriptional co-activators and their sole Drosophila homolog, Yorkie, to drive proliferation and differentiation. Recent studies have demonstrated that YAP/Yorkie exhibit a distinct function at the apical cell cortex. Here, we review recent efforts to understand the mechanisms that regulate YAP/Yki at the apical cell cortex of epithelial cells and how normal and disturbed YAP-actomyosin networks are involved in eye development and disease.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drosophila Proteins/physiology , Epithelial Cells , Eye , Nuclear Proteins/physiology , Organogenesis , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Proliferation , Drosophila , Epithelial Cells/cytology , Epithelial Cells/metabolism , Eye/cytology , Eye/embryology , Gene Expression Regulation, Developmental , Humans , Mice , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
20.
Dev Biol ; 434(1): 24-35, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29166577

ABSTRACT

Cell cycle-related kinase (CCRK) is a conserved regulator of ciliogenesis whose loss in mice leads to a wide range of developmental defects, including exencephaly, preaxial polydactyly, skeletal abnormalities, and microphthalmia. Here, we investigate the role of CCRK in mouse eye development. Ccrk mutants show dramatic patterning defects, with an expansion of the optic stalk domain into the optic cup, as well as an expansion of the retinal pigment epithelium (RPE) into neural retina (NR) territory. In addition, Ccrk mutants display a shortened optic stalk. These defects are associated with bimodal changes in Hedgehog (Hh) pathway activity within the eye, including the loss of proximal, high level responses but a gain in distal, low level responses. We simultaneously removed the Hh activator GLI2 in Ccrk mutants (Ccrk-/-;Gli2-/-), which resulted in rescue of optic cup patterning and exacerbation of optic stalk length defects. Next, we disrupted the Hh pathway antagonist GLI3 in mutants lacking CCRK (Ccrk-/-;Gli3-/-), which lead to even greater expansion of the RPE markers into the NR domain and a complete loss of NR specification within the optic cup. These results indicate that CCRK functions in eye development by both positively and negatively regulating the Hh pathway, and they reveal distinct requirements for Hh signaling in patterning and morphogenesis of the eyes.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Embryo, Mammalian/embryology , Eye/embryology , Hedgehog Proteins/metabolism , Organogenesis/physiology , Signal Transduction/physiology , Zinc Finger Protein Gli2/metabolism , Animals , Cyclin-Dependent Kinases/genetics , Embryo, Mammalian/cytology , Eye/cytology , Female , Hedgehog Proteins/genetics , Male , Mice , Mice, Mutant Strains , Zinc Finger Protein Gli2/genetics , Cyclin-Dependent Kinase-Activating Kinase
SELECTION OF CITATIONS
SEARCH DETAIL