Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (180)2022 02 11.
Article in English | MEDLINE | ID: mdl-35225280

ABSTRACT

Zebrafish exhibit remarkable life-long growth and regenerative abilities. For example, specialized stem cell niches established during embryogenesis support continuous growth of the entire visual system, both in the eye and the brain. Coordinated growth between the retinae and the optic tectum ensures accurate retinotopic mapping as new neurons are added in the eyes and brain. To address whether retinal axons provide crucial information for regulating tectal stem and progenitor cell behaviors such as survival, proliferation, and/or differentiation, it is necessary to be able to compare innervated and denervated tectal lobes within the same animal and across animals. Surgical removal of one eye from living larval zebrafish followed by observation of the optic tectum achieves this goal. The accompanying video demonstrates how to anesthetize larvae, electrolytically sharpen tungsten needles, and use them to remove one eye. It next shows how to dissect brains from fixed zebrafish larvae. Finally, the video provides an overview of the protocol for immunohistochemistry and a demonstration of how to mount stained embryos in low-melting-point agarose for microscopy.


Subject(s)
Visual Pathways , Zebrafish , Animals , Larva , Retina , Superior Colliculi , Visual Pathways/physiology , Zebrafish/physiology
2.
MicroPubl Biol ; 20212021.
Article in English | MEDLINE | ID: mdl-34723143

ABSTRACT

Cell behaviors such as survival, proliferation, and death are governed by a multitude of cues, both intrinsic and extrinsic. To test whether a wild-type environment could encourage the survival and/or differentiation of neuronal progenitor cells with impaired cell cycle progression, we transplanted cells from cdk1, dtl, slbp, fbxo5, ahctf1, gins2, hdac1, mcm5, ssrp1a, and rbbp6 mutant zebrafish embryos into wild-type embryos, creating chimeric zebrafish with mutant cells in the developing eye. We found that when cells from cdk1, dtl, slbp, gins2, mcm5, or rbbp6 mutants were transplanted into wild-type hosts, survival and/or differentiation was almost always compromised in a manner consistent with cell-autonomous cell death. Interestingly, we observed that fbxo5, ahctf1, hdac1, or ssrp1a mutant cells survived and sometimes exhibited signs of differentiation when grafted into wild-type eyes.

3.
Front Cell Dev Biol ; 8: 373, 2020.
Article in English | MEDLINE | ID: mdl-32548116

ABSTRACT

Efficient and accurate DNA replication is particularly critical in stem and progenitor cells for successful proliferation and survival. The replisome, an amalgam of protein complexes, is responsible for binding potential origins of replication, unwinding the double helix, and then synthesizing complimentary strands of DNA. According to current models, the initial steps of DNA unwinding and opening are facilitated by the CMG complex, which is composed of a GINS heterotetramer that connects Cdc45 with the mini-chromosome maintenance (Mcm) helicase. In this work, we provide evidence that in the absence of GINS function DNA replication is cell autonomously impaired, and we also show that gins1 and gins2 mutants exhibit elevated levels of apoptosis restricted to actively proliferating regions of the central nervous system (CNS). Intriguingly, our results also suggest that the rapid cell cycles during early embryonic development in zebrafish may not require the function of the canonical GINS complex as neither zygotic Gins1 nor Gins2 isoforms seem to be present during these stages.

4.
PLoS One ; 14(1): e0211073, 2019.
Article in English | MEDLINE | ID: mdl-30695021

ABSTRACT

Through forward genetic screening for mutations affecting visual system development, we identified prominent coloboma and cell-autonomous retinal neuron differentiation, lamination and retinal axon projection defects in eisspalte (ele) mutant zebrafish. Additional axonal deficits were present, most notably at midline axon commissures. Genetic mapping and cloning of the ele mutation showed that the affected gene is slbp, which encodes a conserved RNA stem-loop binding protein involved in replication dependent histone mRNA metabolism. Cells throughout the central nervous system remained in the cell cycle in ele mutant embryos at stages when, and locations where, post-mitotic cells have differentiated in wild-type siblings. Indeed, RNAseq analysis showed down-regulation of many genes associated with neuronal differentiation. This was coincident with changes in the levels and spatial localisation of expression of various genes implicated, for instance, in axon guidance, that likely underlie specific ele phenotypes. These results suggest that many of the cell and tissue specific phenotypes in ele mutant embryos are secondary to altered expression of modules of developmental regulatory genes that characterise, or promote transitions in, cell state and require the correct function of Slbp-dependent histone and chromatin regulatory genes.


Subject(s)
Animals, Genetically Modified , Axon Guidance/genetics , Cell Differentiation , Cell Proliferation , Coloboma , Retinal Diseases , Zebrafish Proteins/deficiency , Zebrafish , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Coloboma/embryology , Coloboma/genetics , Coloboma/pathology , Histones/genetics , Histones/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Retinal Diseases/embryology , Retinal Diseases/genetics , Retinal Diseases/pathology , Zebrafish/embryology , Zebrafish/genetics
5.
Health Phys ; 114(4): 421-428, 2018 04.
Article in English | MEDLINE | ID: mdl-29481533

ABSTRACT

Ionizing radiation (IR) has been linked to multiple types of cellular responses, but its effects on developing organisms are still poorly understood. The authors investigated whether zebrafish embryos exhibit differential responses relative to IR dose and developmental age at time of exposure. Early-stage zebrafish embryos were exposed to different levels of gamma radiation and then, at varying points after irradiation, assayed for morphological defects and levels of cell death. To quantify in vivo cellular responses to low-dose IR exposure and explore how tissue-specific cell functions affect radiation response, apoptotic cells were counted in three regions: the tail, urogenital papilla, and left eye. The authors found that increased gamma radiation doses correlated with increased levels of apoptosis in the developing tail and eye, whereas cells of the urogenital papilla appeared to undergo apoptosis independently of radiation dose. This suggests that the linear-no-threshold model may not be appropriate in all contexts. Grouping embryos by age at IR exposure revealed that gamma radiation exposure resulted in higher levels of apoptosis in embryos irradiated at 2 d post fertilization (dpf), suggesting a radiosensitive stage of development. Moreover, levels of apoptosis were statistically influenced by days grown after irradiation, with embryos fixed at later stages showing more dramatic apoptotic responses to radiation exposure. This latency to effect suggests potential competition between DNA repair and apoptosis pathways, which may lead to the accumulation of apoptotic cells only after an initial lag period.


Subject(s)
Apoptosis/radiation effects , Embryo, Nonmammalian/pathology , Embryonic Development/radiation effects , Zebrafish/embryology , Animals , DNA Damage/radiation effects , Dose-Response Relationship, Radiation , Embryo, Nonmammalian/radiation effects , Gamma Rays , Radiation Exposure
6.
Article in English | MEDLINE | ID: mdl-28383800

ABSTRACT

A recent outbreak of Zika virus (ZIKV) in Brazil is associated with microcephaly in infants born of infected mothers. As this pandemic spreads, rapid scientific investigation is shedding new light on how prenatal infection with ZIKV causes microcephaly. In this analysis we provide an overview of both microcephaly and ZIKV, explore the connection between prenatal ZIKV infection and microcephaly, and highlight recent insights into how prenatal ZIKV infection depletes the pool of neural progenitors in the developing brain. WIREs Dev Biol 2017, 6:e273. doi: 10.1002/wdev.273 For further resources related to this article, please visit the WIREs website.


Subject(s)
Microcephaly/etiology , Zika Virus Infection/complications , Zika Virus/pathogenicity , Animals , Humans , Microcephaly/pathology
7.
Mol Cell ; 63(6): 1034-43, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27635761

ABSTRACT

Mitochondria divide to control their size, distribution, turnover, and function. Dynamin-related protein 1 (Drp1) is a critical mechanochemical GTPase that drives constriction during mitochondrial division. It is generally believed that mitochondrial division is regulated during recruitment of Drp1 to mitochondria and its oligomerization into a division apparatus. Here, we report an unforeseen mechanism that regulates mitochondrial division by coincident interactions of Drp1 with the head group and acyl chains of phospholipids. Drp1 recognizes the head group of phosphatidic acid (PA) and two saturated acyl chains of another phospholipid by penetrating into the hydrophobic core of the membrane. The dual phospholipid interactions restrain Drp1 via inhibition of oligomerization-stimulated GTP hydrolysis that promotes membrane constriction. Moreover, a PA-producing phospholipase, MitoPLD, binds Drp1, creating a PA-rich microenvironment in the vicinity of a division apparatus. Thus, PA controls the activation of Drp1 after the formation of the division apparatus.


Subject(s)
Dynamins/genetics , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Phosphatidic Acids/metabolism , Phospholipase D/genetics , Testis/metabolism , Animals , Binding Sites , Dynamins/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Expression Regulation , Guanosine Triphosphate/metabolism , Male , Mice , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins/metabolism , Phospholipase D/metabolism , Protein Binding , Signal Transduction , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Testis/ultrastructure
8.
Development ; 143(7): 1087-98, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26893342

ABSTRACT

Maintaining neurogenesis in growing tissues requires a tight balance between progenitor cell proliferation and differentiation. In the zebrafish retina, neuronal differentiation proceeds in two stages with embryonic retinal progenitor cells (RPCs) of the central retina accounting for the first rounds of differentiation, and stem cells from the ciliary marginal zone (CMZ) being responsible for late neurogenesis and growth of the eye. In this study, we analyse two mutants with small eyes that display defects during both early and late phases of retinal neurogenesis. These mutants carry lesions in gdf6a, a gene encoding a BMP family member previously implicated in dorsoventral patterning of the eye. We show that gdf6a mutant eyes exhibit expanded retinoic acid (RA) signalling and demonstrate that exogenous activation of this pathway in wild-type eyes inhibits retinal growth, generating small eyes with a reduced CMZ and fewer proliferating progenitors, similar to gdf6a mutants. We provide evidence that RA regulates the timing of RPC differentiation by promoting cell cycle exit. Furthermore, reducing RA signalling in gdf6a mutants re-establishes appropriate timing of embryonic retinal neurogenesis and restores putative stem and progenitor cell populations in the CMZ. Together, our results support a model in which dorsally expressed gdf6a limits RA pathway activity to control the transition from proliferation to differentiation in the growing eye.


Subject(s)
Growth Differentiation Factor 6/genetics , Neurogenesis/genetics , Retina/embryology , Tretinoin/metabolism , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Cycle/genetics , Cell Proliferation , Embryo, Nonmammalian/embryology , Neurogenesis/physiology , Signal Transduction/genetics , Stem Cells/cytology
9.
J Clin Invest ; 124(5): 2260-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24743148

ABSTRACT

Downmodulation or loss-of-function mutations of the gene encoding NOTCH1 are associated with dysfunctional squamous cell differentiation and development of squamous cell carcinoma (SCC) in skin and internal organs. While NOTCH1 receptor activation has been well characterized, little is known about how NOTCH1 gene transcription is regulated. Using bioinformatics and functional screening approaches, we identified several regulators of the NOTCH1 gene in keratinocytes, with the transcription factors DLX5 and EGR3 and estrogen receptor ß (ERß) directly controlling its expression in differentiation. DLX5 and ERG3 are required for RNA polymerase II (PolII) recruitment to the NOTCH1 locus, while ERß controls NOTCH1 transcription through RNA PolII pause release. Expression of several identified NOTCH1 regulators, including ERß, is frequently compromised in skin, head and neck, and lung SCCs and SCC-derived cell lines. Furthermore, a keratinocyte ERß-dependent program of gene expression is subverted in SCCs from various body sites, and there are consistent differences in mutation and gene-expression signatures of head and neck and lung SCCs in female versus male patients. Experimentally increased ERß expression or treatment with ERß agonists inhibited proliferation of SCC cells and promoted NOTCH1 expression and squamous differentiation both in vitro and in mouse xenotransplants. Our data identify a link between transcriptional control of NOTCH1 expression and the estrogen response in keratinocytes, with implications for differentiation therapy of squamous cancer.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Receptor, Notch1/biosynthesis , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Estrogen Receptor beta/genetics , Female , Genetic Loci , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Transplantation , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Receptor, Notch1/genetics , Transcription, Genetic/genetics
10.
Dev Neurobiol ; 72(3): 328-45, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21563317

ABSTRACT

Fish and amphibia are capable of lifelong growth and regeneration. The two core components of their visual system, the retina and tectum both maintain small populations of stem cells that contribute new neurons and glia to these tissues as they grow. As the animals age, the initial retinal projections onto the tectum are continuously remodeled to maintain retinotopy. These properties raise several biological challenges related to the control of proliferation and differentiation of retinal and tectal stem cells. For instance, how do stem and progenitor cells integrate intrinsic and extrinsic cues to produce the appropriate type and number of cells needed by the growing tissue. Does retinal growth or neuronal activity influence tectal growth? What are the cellular and molecular mechanisms that enable retinal axons to shift their tectal connections as these two tissues grow in incongruent patterns? While we cannot yet provide answers to these questions, this review attempts to supply background and context, laying the ground work for new investigations.


Subject(s)
Amphibians/growth & development , Fishes/growth & development , Nerve Net/growth & development , Visual Pathways/growth & development , Amphibians/physiology , Animals , Fishes/physiology , Humans , Nerve Net/physiology , Retina/growth & development , Retina/physiology , Tectum Mesencephali/growth & development , Tectum Mesencephali/physiology , Visual Pathways/physiology
11.
PLoS One ; 5(11): e15427, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-21079775

ABSTRACT

Funduscopy is one of the most commonly used diagnostic tools in the ophthalmic practice, allowing for a ready assessment of pathological changes in the retinal vasculature and the outer retina. This non-invasive technique has so far been rarely used in animal model for ophthalmic diseases, albeit its potential as a screening assay in genetic screens. The zebrafish (Danio rerio) is well suited for such genetic screens for ocular alterations. Therefore we developed funduscopy in adult zebrafish and employed it as a screening tool to find alterations in the anterior segment and the fundus of the eye of genetically modified adult animals.A stereomicroscope with coaxial reflected light illumination was used to obtain fundus color images of the zebrafish. In order to find lens and retinal alterations, a pilot screen of 299 families of the F3 generation of ENU-treated adult zebrafish was carried out.Images of the fundus of the eye and the anterior segment can be rapidly obtained and be used to identify alterations in genetically modified animals. A number of putative mutants with cataracts, defects in the cornea, eye pigmentation, ocular vessels and retina were identified. This easily implemented method can also be used to obtain fundus images from rodent retinas.In summary, we present funduscopy as a valuable tool to analyse ocular abnormalities in adult zebrafish and other small animal models. A proof of principle screen identified a number of putative mutants, making funduscopy based screens in zebrafish feasible.


Subject(s)
Eye Diseases/diagnosis , Fish Diseases/diagnosis , Mutation , Ophthalmoscopes , Zebrafish/genetics , Animals , Cataract/diagnosis , Cataract/genetics , Corneal Diseases/diagnosis , Corneal Diseases/genetics , Eye Diseases/genetics , Fish Diseases/genetics , Mice , Ophthalmoscopy/methods , Reproducibility of Results , Retinal Diseases/diagnosis , Retinal Diseases/genetics , Sensitivity and Specificity
12.
Development ; 137(13): 2107-15, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20504962

ABSTRACT

It is currently unclear how intrinsic and extrinsic mechanisms cooperate to control the progression from self-renewing to neurogenic divisions in retinal precursor cells. Here, we use the zebrafish flotte lotte (flo) mutant, which carries a mutation in the elys (ahctf1) gene, to study the relationship between cell cycle progression and neuronal differentiation by investigating how proliferating progenitor cells transition towards differentiation in a retinal stem cell niche termed the ciliary marginal zone (CMZ). In zebrafish embryos without Elys, CMZ cells retain the capacity to proliferate but lose the ability to enter their final neurogenic divisions to differentiate as neurons. However, mosaic retinae composed of wild-type and flo cells show that despite inherent cell cycle defects, flo mutant cells progress from proliferation to differentiation when in the vicinity of wild-type retinal neurons. We propose that the differentiated retinal environment limits the proliferation of precursors emerging from the CMZ in a manner that explains the spatial organisation of cells in the CMZ and ensures that proliferative retinal progenitors are driven towards differentiation.


Subject(s)
Neurogenesis , Nuclear Pore Complex Proteins/metabolism , Retina/cytology , Stem Cells/cytology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Apoptosis , Feedback , Nuclear Pore Complex Proteins/genetics , Organ Size , Retina/metabolism , Zebrafish Proteins/genetics
13.
Trends Cell Biol ; 17(11): 563-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17959383

ABSTRACT

In many organisms, ranging from yeast to humans, mitochondria fuse and divide to change their morphology in response to a multitude of signals. During the past decade, work using yeast and mammalian cells has identified much of the machinery required for fusion and division, including the dynamin-related GTPases--mitofusins (Fzo1p in yeast) and OPA1 (Mgm1p in yeast) for fusion and Drp1 (Dnm1p) for division. However, the mechanisms by which cells regulate these dynamic processes have remained largely unknown. Recent studies have uncovered regulatory mechanisms that control the activity, assembly, distribution and stability of the key components for mitochondrial fusion and division. In this review, we discuss how mitochondrial dynamics are controlled and how these events are coordinated with cell growth, mitosis, apoptosis and human diseases.


Subject(s)
Genes, Mitochondrial/physiology , Membrane Fusion/physiology , Mitochondrial Membranes/physiology , Mitochondrial Proteins/physiology , Animals , Humans , Membrane Fusion/genetics , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism
14.
Dev Cell ; 12(3): 363-75, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17336903

ABSTRACT

Yeast mitochondrial division requires the dynamin-related Dnm1 protein. By isolating high-copy suppressors of a dominant-negative Dnm1p mutant, we uncovered an unexpected role in mitochondrial division and inheritance for Num1p, a protein previously shown to facilitate nuclear migration. num1 mutants contain an interconnected network of mitochondrial tubules, remarkably similar to cells lacking Dnm1p, and time-lapse microscopy confirms that mitochondrial fission is greatly reduced in num1Delta cells. We also find that Num1p assembles into punctate structures, which often colocalize with mitochondrial-bound Dnm1p particles. Suggesting a role for both Num1p and Dnm1p in mitochondrial inheritance, we find that num1 dnm1 double mutants accumulate mitochondria in daughter buds and that mother cells are frequently devoid of all mitochondria. Thus, our studies have revealed an additional role for Dnm1p in mitochondrial transmission through its interaction with Num1p, thereby providing a link between mitochondrial division and inheritance.


Subject(s)
Calcium-Binding Proteins/metabolism , Cytokinesis/physiology , Extrachromosomal Inheritance/genetics , GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Binding Sites/physiology , Calcium-Binding Proteins/genetics , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Cytoskeletal Proteins , DNA, Mitochondrial/genetics , GTP Phosphohydrolases/genetics , Mitochondria/genetics , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins , Protein Transport/physiology , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics
15.
Mol Biol Cell ; 14(10): 4126-39, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14517324

ABSTRACT

The Net2, Fis1, and Dnm1 proteins are required for the division of mitochondria in the yeast Saccharomyces cerevisiae. Net2p has an amino-terminal region that contains predicted coiled-coil motifs and a carboxyl-terminal domain composed of WD-40 repeats. We found that the amino-terminal part of Net2p interacts with Fis1p, whereas the carboxyl-terminal region interacts with both Dnm1p and Fis1p. Overproduction of either domain of Net2p in yeast cells poisons mitochondrial fission, and the dominant-negative effect caused by the WD-repeats of Net2p is suppressed by increased levels of Dnm1p. Point mutations in the WD-region of Net2p or in the GTPase region of Dnm1p disrupt the normal Net2p-Dnm1p interaction, causing Net2p to lose its normal punctate distribution. Our results suggest that Dnm1p interacts with the WD-repeats of Net2p and in a GTP-dependent manner recruits Net2p to sites of mitochondrial division. Furthermore, our results indicate that Net2p is required for proper assembly of the mitochondrial fission components to regulate organelle division.


Subject(s)
Carrier Proteins/metabolism , GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Carrier Proteins/genetics , Cell Compartmentation/physiology , GTP Phosphohydrolases/genetics , Microscopy, Fluorescence , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Molecular Sequence Data , Plasmids/genetics , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...