Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.279
Filter
1.
Stem Cell Reports ; 17(2): 221-230, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35030319

ABSTRACT

Microglia are the resident immune cells of the central nervous system (CNS). They govern the immunogenicity of the retina, which is considered to be part of the CNS; however, it is not known how microglia develop in the eye. Here, we studied human-induced pluripotent stem cells (hiPSCs) that had been expanded into a self-formed ectodermal autonomous multi-zone (SEAM) of cells that partially mimics human eye development. Our results indicated that microglia-like cells, which have characteristics of yolk-sac-like linage cells, naturally develop in 2D eye-like SEAM organoids, which lack any vascular components. These cells are unique in that they are paired box protein 6 (PAX6)-positive, yet they possess some characteristics of mesoderm. Collectively, the data support the notion of the existence of an isolated, locally developing immune system in the eye, which is independent of the body's vasculature and general immune system.


Subject(s)
Microglia/metabolism , PAX6 Transcription Factor/metabolism , Cytokines/pharmacology , Eye/cytology , Eye/growth & development , Gene Expression Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lipopolysaccharides/pharmacology , Organoids/cytology , Organoids/metabolism , PAX6 Transcription Factor/genetics , Phagocytosis
2.
Life Sci ; 291: 120273, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35016877

ABSTRACT

AIM: Eye organoids are 3D models of the retina that provide new possibilities for studying retinal development, drug toxicity and the molecular mechanisms of diseases. Although there are several protocols that can be used to generate functional tissues, none have been used to assemble human retinal organoids containing mesenchymal stem cells (MSCs). MAIN METHODS: In this study we intend to assess the effective interactions of MSCs and human embryonic stem cells (hESCs) during retinal organoid formation. We evaluated the inducing activities of bone marrow MSCs (BM-MSCs), trabecular meshwork (TM), and stem cells from apical papilla (SCAP)-derived MSCs in differentiation of hESCs in a three-dimensional (3D) direct co-culture system. KEY FINDINGS: In comparison with the two other MSC sources, the induction potential of SCAP was confirmed in the co-culture system. Although the different SCAP cell ratios did not show any significant morphology changes during the first seven days, increasing the number of SCAPs improved formation of the optic vesicle (OV) structure, which was confirmed by assessment of specific markers. The OVs subsequently developed to an optic cup (OC), which was similar to the in vivo environment. These arrangements expressed MITF in the outer layer and CHX10 in the inner layer. SIGNIFICANCE: We assessed the inducing activity of SCAP during differentiation of hESCs towards a retinal fate in a 3D organoid system. However, future studies be conducted to gather additional details about the development of the eye field, retinal differentiation, and the molecular mechanisms of diseases.


Subject(s)
Cell Culture Techniques/methods , Gingiva/cytology , Retina/cytology , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Eye/cytology , Gingiva/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/cytology , Organoids/cytology , Organoids/growth & development , Organoids/metabolism , Retina/growth & development
3.
Nat Commun ; 12(1): 6974, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34848713

ABSTRACT

The phenomenon of tissue fluidity-cells' ability to rearrange relative to each other in confluent tissues-has been linked to several morphogenetic processes and diseases, yet few molecular regulators of tissue fluidity are known. Ommatidial rotation (OR), directed by planar cell polarity signaling, occurs during Drosophila eye morphogenesis and shares many features with polarized cellular migration in vertebrates. We utilize in vivo live imaging analysis tools to quantify dynamic cellular morphologies during OR, revealing that OR is driven autonomously by ommatidial cell clusters rotating in successive pulses within a permissive substrate. Through analysis of a rotation-specific nemo mutant, we demonstrate that precise regulation of junctional E-cadherin levels is critical for modulating the mechanical properties of the tissue to allow rotation to progress. Our study defines Nemo as a molecular tool to induce a transition from solid-like tissues to more viscoelastic tissues broadening our molecular understanding of tissue fluidity.


Subject(s)
Adherens Junctions , Cell Polarity , Extracellular Fluid , Adherens Junctions/genetics , Adherens Junctions/metabolism , Animals , Cadherins , Cell Polarity/genetics , Cell Polarity/physiology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Ectoderm , Eye/cytology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis , Wings, Animal/cytology
4.
Biomed Res Int ; 2021: 6548554, 2021.
Article in English | MEDLINE | ID: mdl-34840978

ABSTRACT

Tissue engineering is biomedical engineering that uses suitable biochemical and physicochemical factors to assemble functional constructs that restore or improve damaged tissues. Recently, cell therapies as a subset of tissue engineering have been very promising in the treatment of ocular diseases. One of the most important biophysical factors to make this happen is noninvasive electrical stimulation (ES) to target ocular cells that may preserve vision in multiple retinal and optic nerve diseases. The science of cellular and biophysical interactions is very exciting in regenerative medicine now. Although the exact effect of ES on cells is unknown, multiple mechanisms are considered to underlie the effects of ES, including increased production of neurotrophic agents, improved cell migration, and inhibition of proinflammatory cytokines and cellular apoptosis. In this review, we highlighted the effects of ES on ocular cells, especially on the corneal, retinal, and optic nerve cells. Initially, we summarized the current literature on the in vitro and in vivo effects of ES on ocular cells and then we provided the clinical studies describing the effect of ES on ocular complications. For each area, we used some of the most impactful articles to show the important concepts and results that advanced the state of these interactions. We conclude with reflections on emerging new areas and perspectives for future development in this field.


Subject(s)
Electric Stimulation Therapy/methods , Eye Diseases/therapy , Eye/cytology , Tissue Engineering/methods , Animals , Cell- and Tissue-Based Therapy/methods , Eye Diseases/physiopathology , Humans , In Vitro Techniques , Regenerative Medicine/methods , Stem Cells/cytology
5.
Biomed Res Int ; 2021: 4982227, 2021.
Article in English | MEDLINE | ID: mdl-34285913

ABSTRACT

PDGFRα signaling is critically important in ocular development. Previous data on PDGFRα lacks an expression map with high spatial and temporal resolution and lineage information. In this study, we aim to present a detailed PDGFRα expression and lineage map from early embryogenesis to adulthood. PDGFRα-CreER; mT/mG reporter mice were analyzed. mEGFP-positive cells contributed to multiple ocular lineages in a spatiotemporally regulated manner. A dynamic PDGFRα expression was identified in corneal stromal cells, lens epithelial cells, lens fiber cells, and retinal astrocytes during the entire period of eye development, while PDGFRα expression in retinal astrocytes from E17.5 onwards and in Müller glial cells was identified within two weeks after birth. By revealing detailed characterization of gene expression and function, we present a comprehensive map of PDGFRα-expressing cells in the eye for a better understanding of PDGFRα signaling's role during eye development.


Subject(s)
Cell Lineage , Eye/cytology , Eye/embryology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Animals, Newborn , Cell Lineage/genetics , Cornea/cytology , Cornea/embryology , Embryo, Mammalian/cytology , Eye/metabolism , Gene Expression Regulation, Developmental , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Retina/cytology
6.
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
7.
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
8.
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
9.
Stem Cell Res ; 51: 102200, 2021 03.
Article in English | MEDLINE | ID: mdl-33535156

ABSTRACT

Recently, coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 has spread around the world and is receiving worldwide attention. Approximately 20% of infected patients are suffering from severe disease of multiple systems and in danger of death, while the ocular complications of SARS-CoV-2-infected patients have not been reported generally. Herein, we focus on two major receptors of SARS-CoV-2, ACE2 and CD147 (BSG), in human ocular cells, and interpret the potential roles of coronaviruses in human ocular tissues and diseases.


Subject(s)
COVID-19/pathology , Eye/virology , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antirheumatic Agents/therapeutic use , Basigin/metabolism , COVID-19/transmission , COVID-19/virology , Dexamethasone/therapeutic use , Eye/cytology , Eye/metabolism , Eye Diseases/pathology , Eye Diseases/virology , Glucocorticoids/therapeutic use , Humans , Renin-Angiotensin System/physiology , SARS-CoV-2/isolation & purification , COVID-19 Drug Treatment
10.
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
11.
J Comp Neurol ; 529(3): 616-634, 2021 02.
Article in English | MEDLINE | ID: mdl-32592497

ABSTRACT

A great diversity of adaptations is found among animals with compound eyes and even closely related taxa can show variation in their light-adaptation strategies. A prime example of a visual system evolved to function in specific light environments is the fiddler crab, used widely as a model to research aspects of crustacean vision and neural pathways. However, questions remain regarding how their eyes respond to the changes in brightness spanning many orders of magnitude, associated with their habitat and ecology. The fiddler crab Afruca tangeri forages at low tide on tropical and semi-tropical mudflats, under bright sunlight and on moonless nights, suggesting that their eyes undergo effective light adaptation. Using synchrotron X-ray tomography, light and transmission electron microscopy and in vivo ophthalmoscopy, we describe the ultrastructural changes in the eye between day and night. Dark adaptation at dusk triggered extensive widening of the rhabdoms and crystalline cone tips. This doubled the ommatidial acceptance angles and increased microvillar surface area for light capture in the rhabdom, theoretically boosting optical sensitivity 7.4 times. During daytime, only partial dark-adaptation was achieved and rhabdoms remained narrow, indicating strong circadian control on the process. Bright light did not evoke changes in screening pigment distributions, suggesting a structural inability to adapt rapidly to the light level fluctuations frequently experienced when entering their burrow to escape predators. This should enable fiddler crabs to shelter for several minutes without undergoing significant dark-adaptation, their vision remaining effectively adapted for predator detection when surfacing again in bright light.


Subject(s)
Adaptation, Ocular/physiology , Eye/chemistry , Eye/cytology , Ocular Physiological Phenomena , Animals , Brachyura , Eye/metabolism , Female , Male , Microscopy, Electron, Transmission/methods
12.
Wiley Interdiscip Rev Dev Biol ; 10(2): e380, 2021 03.
Article in English | MEDLINE | ID: mdl-32400100

ABSTRACT

The compound eyes of flies exhibit striking variation in size, which has contributed to the adaptation of these animals to different habitats and their evolution of specialist behaviors. These differences in size are caused by differences in the number and/or size of ommatidia, which are specified during the development of the retinal field in the eye imaginal disc. While the genes and developmental mechanisms that regulate the formation of compound eyes are understood in great detail in the fruit fly Drosophila melanogaster, we know very little about the genetic changes and mechanistic alterations that lead to natural variation in ommatidia number and/or size, and thus overall eye size, within and between fly species. Understanding the genetic and developmental bases for this natural variation in eye size not only has great potential to help us understand adaptations in fly vision but also determine how eye size and organ size more generally are regulated. Here we explore the genetic and developmental mechanisms that could underlie natural differences in compound eye size within and among fly species based on our knowledge of eye development in D. melanogaster and the few cases where the causative genes and mechanisms have already been identified. We suggest that the fly eye provides an evolutionary and developmental framework to better understand the regulation and diversification of this crucial sensory organ globally at a systems level as well as the gene regulatory networks and mechanisms acting at the tissue, cellular and molecular levels. This article is categorized under: Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Invertebrate Organogenesis > Flies Comparative Development and Evolution > Regulation of Organ Diversity.


Subject(s)
Biological Evolution , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Eye/cytology , Organogenesis , Animals , Eye/metabolism , Organ Size
13.
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
14.
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
15.
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
16.
J Ocul Pharmacol Ther ; 36(10): 715-724, 2020 12.
Article in English | MEDLINE | ID: mdl-33121321

ABSTRACT

Purpose: The human eye is a sophisticated and sensitive sensory organ. Because of the existence of the blood-ocular barrier and corneal-scleral barrier, safe and efficient ocular drug delivery system is highly desired; yet, it remains an unsolved issue. Due to the unique structure and drug loading property, Poly(amidoamine) (PAMAM) has received much attention in the ocular drug delivery investigation. Herein, we evaluated the ocular cytotoxicity and biosafety of PAMAM dendrimers. Methods: The ocular cytotoxicity and biosafety of PAMAM dendrimers were evaluated by conducting in vitro and in vivo experiments on ocular systems. The in vitro effect of PAMAM dendrimer of different generations (G4.0, G5.0, and G6.0) and concentrations on ocular cell metabolism, apoptosis, and oxidative damage were quantitatively assessed. In vivo biosafety of PAMAM dendrimers were further investigated on intraocular tissue by ocular irritation and intravitreal injection approaches. Results: It is found that that the cytotoxicity of PAMAM was time and generation dependent. PAMAM at a concentration below 50 µg/mL had minimal impact on the ocular tissue, whereas it caused apparent damage when above 50 µg/mL in the investigated situation. Further, our in vivo results showed that higher concentration of dendrimer (100 µg/mL) was associated with functional impairment demonstrated via optical coherence tomography and electroretinogram, although macroscopic structural changes were absent in fundus and histopathological studies. Overall, a higher concentration of PAMAM, such as above 50 µg/mL, may cause ocular functional damage. Conclusion: The PAMAM at the concentrations lower than 50 µg/mL showed good biocompatibility and biosafety in human ocular cells and tissues.


Subject(s)
Dendrimers/adverse effects , Eye/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Dendrimers/administration & dosage , Dendrimers/chemistry , Drug Delivery Systems , Electroretinography , Eye/cytology , Eye/metabolism , Humans , Male , Oxidative Stress/drug effects , Rabbits , Time Factors , Tomography, Optical Coherence
17.
Cells ; 9(8)2020 08 08.
Article in English | MEDLINE | ID: mdl-32784474

ABSTRACT

Many disorders of aging, including blinding-diseases, are associated with deficiency of brain and muscle arnt-like protein 1 (Bmal1) and, thereby, dysregulation of antioxidant-defense pathway. However, knowledge is limited regarding the role of Bmal1 regulation of antioxidant-pathway in the eye lens/lens epithelial cells (LECs) at the molecular level. We found that, in aging human (h)LECs, a progressive decline of nuclear factor erythroid 2-related factor 2 (Nrf2)/ARE (antioxidant response element)-mediated antioxidant genes was connected to Bmal1-deficiency, leading to accumulation of reactive oxygen species (ROS) and cell-death. Bmal1-depletion disrupted Nrf2 and expression of its target antioxidant genes, like Peroxiredoxin 6 (Prdx6). DNA binding and transcription assays showed that Bmal1 controlled expression by direct binding to E-Box in Prdx6 promoter to regulate its transcription. Mutation at E-Box or ARE reduced promoter activity, while disruption of both sites diminished the activity, suggesting that both sites were required for peak Prdx6-transcription. As in aging hLECs, ROS accumulation was increased in Bmal1-deficient cells and the cells were vulnerable to death. Intriguingly, Bmal1/Nrf2/Prdx6 and PhaseII antioxidants showed rhythmic expression in mouse lenses in vivo and were reciprocally linked to ROS levels. We propose that Bmal1 is pivotal for regulating oxidative responses. Findings also reveal a circadian control of antioxidant-pathway, which is important in combating lens/LECs damage induced by aging or oxidative stress.


Subject(s)
ARNTL Transcription Factors/metabolism , Aging/metabolism , Circadian Rhythm , Epithelial Cells/metabolism , Eye/metabolism , NF-E2-Related Factor 2/metabolism , Peroxiredoxin VI/metabolism , Animals , Cell Line , Epithelial Cells/cytology , Epithelial Cells/pathology , Eye/cytology , Eye/pathology , Female , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Oxidative Stress , Reactive Oxygen Species/metabolism
18.
Res Vet Sci ; 132: 393-399, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32758722

ABSTRACT

The main aims of this study were to determine whether CD4+ and CD8+ cells are present in the normal chambers of the eye in dogs and to verify the hypothesis that uncomplicated cataract may be associated with the local recruitment of CD4+ and CD8+ cells. The presence of CD4+ and CD8+ cells was detected in aqueous humor (AH) of normal and cataract eyes. The study did not reveal differences in the percentage and absolute number of CD4+ cells between normal and cataract eyes. However, the values of these parameters in AH from cataract eyes were approximately 2- and 3-fold higher than in normal eyes, respectively. The mean percentage and absolute count of CD8+ cells increased approximately by 2.7- and 6-fold, respectively, in AH samples from cataract eyes compared to normal ones. The absolute count of CD4+ and CD8+ cells in AH of uveitic eyes was approximately 5- and 3-fold higher than in cataract eyes. The results indicate that CD4+ and CD8+ cells occur constitutively in the normal chambers of the eye in dogs. However, it should be pointed out that both of these cell populations appeared in trace amounts. The development of uncomplicated cataract in dogs may not be immunologically neutral in terms of the local immune response, but it may be associated with the recruitment of CD8+ cells into the eye chambers. This event does not seem to be of an inflammatory nature because it appears on a scale a few times smaller than in the course of uveitis.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cataract/veterinary , Dog Diseases/physiopathology , Dogs/physiology , Eye/cytology , Uveitis/veterinary , Animals , Aqueous Humor/physiology , Cataract/physiopathology , Female , Male , Uveitis/physiopathology
19.
J Vis Exp ; (160)2020 06 17.
Article in English | MEDLINE | ID: mdl-32628177

ABSTRACT

The innate immune system plays important roles in ocular pathophysiology including uveitis, diabetic retinopathy, and age-related macular degeneration. Innate immune cells, specifically mononuclear phagocytes, express overlapping cell surface markers, which makes identifying these populations a challenge. Multi-parameter flow cytometry allows for the simultaneous, quantitative analysis of multiple cell surface markers in order to differentiate monocytes, macrophages, microglia, and dendritic cells in mouse eyes. This protocol describes the enucleation of whole mouse eyes, ocular dissection, digestion into a single cell suspension, and staining of the single cell suspension for myeloid cell markers. Additionally, we explain the proper methods for determining voltages using single color controls and for delineating positive gates using fluorescence minus one controls. The major limitation of multi-parameter flow cytometry is the absence of tissue architecture. This limitation can be overcome by multi-parameter flow cytometry of individual ocular compartments or complimentary immunofluorescence staining. However, immunofluorescence is limited by its lack of quantitative analysis and reduced number of fluorophores on most microscopes. We describe the use of multi-parametric flow cytometry to provide highly quantitative analysis of mononuclear phagocytes in laser-induced choroidal neovascularization. Additionally, multi-parameter flow cytometry can be used for the identification of macrophage subsets, fate mapping, and cell sorting for transcriptomic or proteomic studies.


Subject(s)
Eye/cytology , Eye/diagnostic imaging , Flow Cytometry , Phagocytes/cytology , Animals , Antibodies/metabolism , Dendritic Cells/cytology , Female , Fluorescent Dyes/metabolism , Lasers , Macrophages/cytology , Male , Mice, Inbred C57BL , Microglia/cytology , Monocytes/cytology , Phagocytes/immunology
20.
Nat Commun ; 11(1): 2772, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32487986

ABSTRACT

Vestigial structures are key indicators of evolutionary descent, but the mechanisms underlying their development are poorly understood. This study examines vestigial eye formation in the teleost Astyanax mexicanus, which consists of a sighted surface-dwelling morph and multiple populations of blind cave morphs. Cavefish embryos initially develop eyes, but they subsequently degenerate and become vestigial structures embedded in the head. The mutated genes involved in cavefish vestigial eye formation have not been characterized. Here we identify cystathionine ß-synthase a (cbsa), which encodes the key enzyme of the transsulfuration pathway, as one of the mutated genes responsible for eye degeneration in multiple cavefish populations. The inactivation of cbsa affects eye development by increasing the transsulfuration intermediate homocysteine and inducing defects in optic vasculature, which result in aneurysms and eye hemorrhages. Our findings suggest that localized modifications in the circulatory system may have contributed to the evolution of vestigial eyes in cavefish.


Subject(s)
Cystathionine beta-Synthase/genetics , Cystathionine/metabolism , Eye/embryology , Eye/metabolism , Fishes/physiology , Animals , Apoptosis , Biological Evolution , Brain/embryology , Cardiovascular System , Cystathionine beta-Synthase/metabolism , Developmental Biology , Eye/cytology , Eye/growth & development , Female , Fishes/embryology , Fishes/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Head , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Male , Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...