Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Elife ; 102021 11 16.
Article in English | MEDLINE | ID: mdl-34783657

ABSTRACT

During development, neurites and synapses segregate into specific neighborhoods or layers within nerve bundles. The developmental programs guiding placement of neurites in specific layers, and hence their incorporation into specific circuits, are not well understood. We implement novel imaging methods and quantitative models to document the embryonic development of the C. elegans brain neuropil, and discover that differential adhesion mechanisms control precise placement of single neurites onto specific layers. Differential adhesion is orchestrated via developmentally regulated expression of the IgCAM SYG-1, and its partner ligand SYG-2. Changes in SYG-1 expression across neuropil layers result in changes in adhesive forces, which sort SYG-2-expressing neurons. Sorting to layers occurs, not via outgrowth from the neurite tip, but via an alternate mechanism of retrograde zippering, involving interactions between neurite shafts. Our study indicates that biophysical principles from differential adhesion govern neurite placement and synaptic specificity in vivo in developing neuropil bundles.


Subject(s)
Brain/cytology , Brain/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/physiology , Cell Adhesion/genetics , Neurites/physiology , Animals , Caenorhabditis elegans Proteins/metabolism , Cell Adhesion/physiology , Gene Expression Regulation , Neurons/physiology , Synapses
2.
Nature ; 591(7848): 99-104, 2021 03.
Article in English | MEDLINE | ID: mdl-33627875

ABSTRACT

Neuropil is a fundamental form of tissue organization within the brain1, in which densely packed neurons synaptically interconnect into precise circuit architecture2,3. However, the structural and developmental principles that govern this nanoscale precision remain largely unknown4,5. Here we use an iterative data coarse-graining algorithm termed 'diffusion condensation'6 to identify nested circuit structures within the Caenorhabditis elegans neuropil, which is known as the nerve ring. We show that the nerve ring neuropil is largely organized into four strata that are composed of related behavioural circuits. The stratified architecture of the neuropil is a geometrical representation of the functional segregation of sensory information and motor outputs, with specific sensory organs and muscle quadrants mapping onto particular neuropil strata. We identify groups of neurons with unique morphologies that integrate information across strata and that create neural structures that cage the strata within the nerve ring. We use high resolution light-sheet microscopy7,8 coupled with lineage-tracing and cell-tracking algorithms9,10 to resolve the developmental sequence and reveal principles of cell position, migration and outgrowth that guide stratified neuropil organization. Our results uncover conserved structural design principles that underlie the architecture and function of the nerve ring neuropil, and reveal a temporal progression of outgrowth-based on pioneer neurons-that guides the hierarchical development of the layered neuropil. Our findings provide a systematic blueprint for using structural and developmental approaches to understand neuropil organization within the brain.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Neuropil/chemistry , Neuropil/metabolism , Algorithms , Animals , Brain/cytology , Brain/embryology , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/cytology , Cell Movement , Diffusion , Interneurons/metabolism , Motor Neurons/metabolism , Neurites/metabolism , Neuropil/cytology , Sensory Receptor Cells/metabolism
3.
Nat Biotechnol ; 38(11): 1337-1346, 2020 11.
Article in English | MEDLINE | ID: mdl-32601431

ABSTRACT

The contrast and resolution of images obtained with optical microscopes can be improved by deconvolution and computational fusion of multiple views of the same sample, but these methods are computationally expensive for large datasets. Here we describe theoretical and practical advances in algorithm and software design that result in image processing times that are tenfold to several thousand fold faster than with previous methods. First, we show that an 'unmatched back projector' accelerates deconvolution relative to the classic Richardson-Lucy algorithm by at least tenfold. Second, three-dimensional image-based registration with a graphics processing unit enhances processing speed 10- to 100-fold over CPU processing. Third, deep learning can provide further acceleration, particularly for deconvolution with spatially varying point spread functions. We illustrate our methods from the subcellular to millimeter spatial scale on diverse samples, including single cells, embryos and cleared tissue. Finally, we show performance enhancement on recently developed microscopes that have improved spatial resolution, including dual-view cleared-tissue light-sheet microscopes and reflective lattice light-sheet microscopes.


Subject(s)
Algorithms , Image Processing, Computer-Assisted , Microscopy , Animals , Brain/diagnostic imaging , Caenorhabditis elegans/embryology , Cell Line , Deep Learning , Humans , Mice , Zebrafish/embryology
4.
J Vis Exp ; (148)2019 06 06.
Article in English | MEDLINE | ID: mdl-31233035

ABSTRACT

Caenorhabditis elegans (C. elegans) stands out as the only organism in which the challenge of understanding the cellular origins of an entire nervous system can be observed, with single cell resolution, in vivo. Here, we present an integrated protocol for the examination of neurodevelopment in C. elegans embryos. Our protocol combines imaging, lineaging and neuroanatomical tracing of single cells in developing embryos. We achieve long-term, four-dimensional (4D) imaging of living C. elegans embryos with nearly isotropic spatial resolution through the use of Dual-view Inverted Selective Plane Illumination Microscopy (diSPIM). Nuclei and neuronal structures in the nematode embryos are imaged and isotropically fused to yield images with resolution of ~330 nm in all three dimensions. These minute-by-minute high-resolution 4D data sets are then analyzed to correlate definitive cell-lineage identities with gene expression and morphological dynamics at single-cell and subcellular levels of detail. Our protocol is structured to enable modular implementation of each of the described steps and enhance studies on embryogenesis, gene expression, or neurodevelopment.


Subject(s)
Caenorhabditis elegans/embryology , Cell Lineage , Embryonic Development/physiology , Microscopy/methods , Animals , Cell Nucleus
5.
PLoS One ; 11(1): e0146874, 2016.
Article in English | MEDLINE | ID: mdl-26800457

ABSTRACT

BACKGROUND: Regulatory and biophysical mechanisms of cell-cell fusion are largely unknown despite the fundamental requirement for fused cells in eukaryotic development. Only two cellular fusogens that are not of clear recent viral origin have been identified to date, both in nematodes. One of these, EFF-1, is necessary for most cell fusions in Caenorhabditis elegans. Unregulated EFF-1 expression causes lethality due to ectopic fusion between cells not developmentally programmed to fuse, highlighting the necessity of tight fusogen regulation for proper development. Identifying factors that regulate EFF-1 and its paralog AFF-1 could lead to discovery of molecular mechanisms that control cell fusion upstream of the action of a membrane fusogen. Bioinformatic analysis of the EFF-1A isoform's predicted cytoplasmic domain (endodomain) previously revealed two motifs that have high probabilities of interacting with 14-3-3 proteins when phosphorylated. Mutation of predicted phosphorylation sites within these motifs caused measurable loss of eff-1 gene function in cell fusion in vivo. Moreover, a human 14-3-3 isoform bound to EFF-1::GFP in vitro. We hypothesized that the two 14-3-3 proteins in C. elegans, PAR-5 and FTT-2, may regulate either localization or fusion-inducing activity of EFF-1. METHODOLOGY/PRINCIPAL FINDINGS: Timing of fusion events was slightly but significantly delayed in animals unable to produce full-length EFF-1A. Yet, mutagenesis and live imaging showed that phosphoserines in putative 14-3-3 binding sites are not essential for EFF-1::GFP accumulation at the membrane contact between fusion partner cells. Moreover, although the EFF-1A endodomain was required for normal rates of eff-1-dependent epidermal cell fusions, reduced levels of FTT-2 and PAR-5 did not visibly affect the function of wild-type EFF-1 in the hypodermis. CONCLUSIONS/SIGNIFICANCE: Deletion of the EFF-1A endodomain noticeably affects the timing of hypodermal cell fusions in vivo. However, prohibiting phosphorylation of candidate 14-3-3-binding sites does not impact localization of the fusogen. Hypodermal membrane fusion activity persists when 14-3-3 expression levels are reduced.


Subject(s)
14-3-3 Proteins/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Membrane Glycoproteins/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Cell Fusion , Membrane Glycoproteins/genetics , Phosphorylation/physiology , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/genetics
6.
Elife ; 42015 Dec 03.
Article in English | MEDLINE | ID: mdl-26633880

ABSTRACT

The nematode Caenorhabditis elegans possesses a simple embryonic nervous system with few enough neurons that the growth of each cell could be followed to provide a systems-level view of development. However, studies of single cell development have largely been conducted in fixed or pre-twitching live embryos, because of technical difficulties associated with embryo movement in late embryogenesis. We present open-source untwisting and annotation software (http://mipav.cit.nih.gov/plugin_jws/mipav_worm_plugin.php) that allows the investigation of neurodevelopmental events in late embryogenesis and apply it to track the 3D positions of seam cell nuclei, neurons, and neurites in multiple elongating embryos. We also provide a tutorial describing how to use the software (Supplementary file 1) and a detailed description of the untwisting algorithm (Appendix). The detailed positional information we obtained enabled us to develop a composite model showing movement of these cells and neurites in an 'average' worm embryo. The untwisting and cell tracking capabilities of our method provide a foundation on which to catalog C. elegans neurodevelopment, allowing interrogation of developmental events in previously inaccessible periods of embryogenesis.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/physiology , Computational Biology/methods , Nervous System/cytology , Nervous System/embryology , Neurons/physiology , Software , Animals , Cell Tracking/methods , Data Curation
8.
BMC Bioinformatics ; 16: 189, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26051157

ABSTRACT

BACKGROUND: Imaging and image analysis advances are yielding increasingly complete and complicated records of cellular events in tissues and whole embryos. The ability to follow hundreds to thousands of cells at the individual level demands a spatio-temporal data infrastructure: tools to assemble and collate knowledge about development spatially in a manner analogous to geographic information systems (GIS). Just as GIS indexes items or events based on their spatio-temporal or 4D location on the Earth these tools would organize knowledge based on location within the tissues or embryos. Developmental processes are highly context-specific, but the complexity of the 4D environment in which they unfold is a barrier to assembling an understanding of any particular process from diverse sources of information. In the same way that GIS aids the understanding and use of geo-located large data sets, software can, with a proper frame of reference, allow large biological data sets to be understood spatially. Intuitive tools are needed to navigate the spatial structure of complex tissue, collate large data sets and existing knowledge with this spatial structure and help users derive hypotheses about developmental mechanisms. RESULTS: Toward this goal we have developed WormGUIDES, a mobile application that presents a 4D developmental atlas for Caenorhabditis elegans. The WormGUIDES mobile app enables users to navigate a 3D model depicting the nuclear positions of all cells in the developing embryo. The identity of each cell can be queried with a tap, and community databases searched for available information about that cell. Information about ancestry, fate and gene expression can be used to label cells and craft customized visualizations that highlight cells as potential players in an event of interest. Scenes are easily saved, shared and published to other WormGUIDES users. The mobile app is available for Android and iOS platforms. CONCLUSION: WormGUIDES provides an important tool for examining developmental processes and developing mechanistic hypotheses about their control. Critically, it provides the typical end user with an intuitive interface for developing and sharing custom visualizations of developmental processes. Equally important, because users can select cells based on their position and search for information about them, the app also serves as a spatially organized index into the large body of knowledge available to the C. elegans community online. Moreover, the app can be used to create and publish the result of exploration: interactive content that brings other researchers and students directly to the spatio-temporal point of insight. Ultimately the app will incorporate a detailed time lapse record of cell shape, beginning with neurons. This will add the key ability to navigate and understand the developmental events that result in the coordinated and precise emergence of anatomy, particularly the wiring of the nervous system.


Subject(s)
Caenorhabditis elegans/growth & development , Nervous System/cytology , Single-Cell Analysis/methods , Software , User-Computer Interface , Animals , Databases, Factual
9.
Cell Cycle ; 13(21): 3375-89, 2014.
Article in English | MEDLINE | ID: mdl-25485582

ABSTRACT

Drugs capable of specifically recognizing and killing cancer cells while sparing healthy cells are of great interest in anti-cancer therapy. An example of such a drug is edelfosine, the prototype molecule of a family of synthetic lipids collectively known as antitumor lipids (ATLs). A better understanding of the selectivity and the mechanism of action of these compounds would lead to better anticancer treatments. Using Caenorhabditis elegans, we modeled key features of the ATL selectivity against cancer cells. Edelfosine induced a selective and direct killing action on C. elegans embryos, which was dependent on cholesterol, without affecting adult worms and larvae. Distinct ATLs ranked differently in their embryonic lethal effect with edelfosine > perifosine > erucylphosphocholine >> miltefosine. Following a biased screening of 57 C. elegans mutants we found that inactivation of components of the insulin/IGF-1 signaling pathway led to resistance against the ATL edelfosine in both C. elegans and human tumor cells. This paper shows that C. elegans can be used as a rapid platform to facilitate ATL research and to further understand the mechanism of action of edelfosine and other synthetic ATLs.


Subject(s)
Antineoplastic Agents/pharmacology , Embryo, Nonmammalian/drug effects , Animals , Apoptosis/drug effects , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Cell Line, Tumor , Cholesterol/metabolism , Drug Resistance , Embryonic Development/drug effects , Humans , Insulin-Like Growth Factor I/metabolism , Larva/drug effects , Membrane Microdomains/metabolism , Phospholipid Ethers/pharmacology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology
10.
PLoS Genet ; 8(8): e1002863, 2012.
Article in English | MEDLINE | ID: mdl-22876199

ABSTRACT

Many cells in a developing embryo, including neurons and their axons and growth cones, must integrate multiple guidance cues to undergo directed growth and migration. The UNC-6/netrin, SLT-1/slit, and VAB-2/Ephrin guidance cues, and their receptors, UNC-40/DCC, SAX-3/Robo, and VAB-1/Eph, are known to be major regulators of cellular growth and migration. One important area of research is identifying the molecules that interpret this guidance information downstream of the guidance receptors to reorganize the actin cytoskeleton. However, how guidance cues regulate the actin cytoskeleton is not well understood. We report here that UNC-40/DCC, SAX-3/Robo, and VAB-1/Eph differentially regulate the abundance and subcellular localization of the WAVE/SCAR actin nucleation complex and its activator, Rac1/CED-10, in the Caenorhabditis elegans embryonic epidermis. Loss of any of these three pathways results in embryos that fail embryonic morphogenesis. Similar defects in epidermal enclosure have been observed when CED-10/Rac1 or the WAVE/SCAR actin nucleation complex are missing during embryonic development in C. elegans. Genetic and molecular experiments demonstrate that in fact, these three axonal guidance proteins differentially regulate the levels and membrane enrichment of the WAVE/SCAR complex and its activator, Rac1/CED-10, in the epidermis. Live imaging of filamentous actin (F-actin) in embryos developing in the absence of individual guidance receptors shows that high levels of F-actin are not essential for polarized cell migrations, but that properly polarized distribution of F-actin is essential. These results suggest that proper membrane recruitment and activation of CED-10/Rac1 and of WAVE/SCAR by signals at the plasma membrane result in polarized F-actin that permits directed movements and suggest how multiple guidance cues can result in distinct changes in actin nucleation during morphogenesis.


Subject(s)
Actins/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Immunologic/metabolism , rac GTP-Binding Proteins/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actins/genetics , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Adhesion Molecules/genetics , Cell Cycle Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Movement/genetics , Cell Polarity/genetics , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Morphogenesis/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Immunologic/genetics , Signal Transduction , Video Recording , rac GTP-Binding Proteins/genetics , Roundabout Proteins
11.
J Cell Biol ; 197(5): 643-58, 2012 May 28.
Article in English | MEDLINE | ID: mdl-22613834

ABSTRACT

Regulation of actin dynamics through the Nck/N-WASp (neural Wiskott-Aldrich syndrome protein)/Arp2/3 pathway is essential for organogenesis, cell invasiveness, and pathogen infection. Although many of the proteins involved in this pathway are known, the detailed mechanism by which it functions remains undetermined. To examine the signaling mechanism, we used a two-pronged strategy involving computational modeling and quantitative experimentation. We developed predictions for Nck-dependent actin polymerization using the Virtual Cell software system. In addition, we used antibody-induced aggregation of membrane-targeted Nck SH3 domains to test these predictions and to determine how the number of molecules in Nck aggregates and the density of aggregates affected localized actin polymerization in living cells. Our results indicate that the density of Nck molecules in aggregates is a critical determinant of actin polymerization. Furthermore, results from both computational simulations and experimentation support a model in which the Nck/N-WASp/Arp2/3 stoichiometry is 4:2:1. These results provide new insight into activities involving localized actin polymerization, including tumor cell invasion, microbial pathogenesis, and T cell activation.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Oncogene Proteins/metabolism , Polymerization , Actins/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Cell Survival , Computer Simulation , HEK293 Cells , Humans , Oncogene Proteins/chemistry , Signal Transduction , src Homology Domains
12.
Biophys J ; 103(3): 627, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-33049908
13.
Dev Biol ; 357(2): 356-69, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21798253

ABSTRACT

Regulated movements of the nucleus are essential during zygote formation, cell migrations, and differentiation of neurons. The nucleus moves along microtubules (MTs) and is repositioned on F-actin at the cellular cortex. Two families of nuclear envelope proteins, SUN and KASH, link the nucleus to the actin and MT cytoskeletons during nuclear movements. However, the role of actin nucleators in nuclear migration and positioning is poorly understood. We show that the branched actin nucleator, Arp2/3, affects nuclear movements throughout embryonic and larval development in C. elegans, including nuclear migrations in epidermal cells and neuronal precursors. In one-cell embryos the migration of the male pronucleus to meet the female pronucleus after fertilization requires Arp2/3. Loss of Arp2/3 or its activators changes the dynamics of non-muscle myosin, NMY-2, and alters the cortical accumulation of posterior PAR proteins. Reduced establishment of the posterior microtubule cytoskeleton in Arp2/3 mutants correlates with reduced male pronuclear migration. The UNC-84/SUN nuclear envelope protein that links the nucleus to the MT and actin cytoskeleton is known to regulate later nuclear migrations. We show here it also positions the male pronucleus. These studies demonstrate a global role for Arp2/3 in nuclear migrations. In the C. elegans one-cell embryo Arp2/3 promotes the establishment of anterior/posterior polarity and promotes MT growth that propels the anterior migration of the male pronucleus. In contrast with previous studies emphasizing pulling forces on the male pronucleus, we propose that robust MT nucleation pushes the male pronucleus anteriorly to join the female pronucleus.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Cell Nucleus/metabolism , Cell Polarity , Movement , Zygote/cytology , Actins/metabolism , Actomyosin/metabolism , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Centrosome/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Epidermal Cells , Epidermis/embryology , Epidermis/metabolism , Female , Male , Microtubules/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Envelope/metabolism , Zygote/metabolism
14.
Int Rev Cell Mol Biol ; 289: 149-209, 2011.
Article in English | MEDLINE | ID: mdl-21749901

ABSTRACT

Many types of eukaryotic cells can fuse together as part of their normal developmental program or life cycle. This review describes a diverse set of examples of such cell types and focuses attention on several molecules that appear intimately involved in the process of plasma membrane merger that lies at the crux of every cell-fusion event. Some of these examples come from experimental systems where the discovery of molecules essential for cell fusion is sped by the approachability of the experimental organism itself. In other cases, especially in the many fusing human cell types, the molecular players in cell-cell membrane fusion are still to be discovered.


Subject(s)
Cell Fusion , Amino Acid Sequence , Animals , Body Patterning , Cell Communication , Humans , Membrane Fusion , Molecular Sequence Data , Reproduction , Viruses/metabolism
15.
EMBO J ; 29(18): 3184-95, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20700105

ABSTRACT

The BK channel, a voltage- and Ca(2+)-gated large-conductance potassium channel with many important functions, is often localized at specific subcellular domains. Although proper subcellular localization is likely a prerequisite for the channel to perform its physiological functions, little is known about the molecular basis of localization. Here, we show that CTN-1, a homologue of mammalian α-catulin, is required for subcellular localization of SLO-1, the Caenorhabditis elegans BK channel α-subunit, in body-wall muscle cells. CTN-1 was identified in a genetic screen for mutants that suppressed a lethargic phenotype caused by expressing a gain-of-function (gf) isoform of SLO-1. In body-wall muscle cells, CTN-1 coclusters with SLO-1 at regions of dense bodies, which are Z-disk analogs of mammalian skeletal muscle. In ctn-1 loss-of-function (lf) mutants, SLO-1 was mislocalized in body-wall muscle but its transcription and protein level were unchanged. Targeted rescue of ctn-1(lf) in muscle was sufficient to reinstate the lethargic phenotype in slo-1(gf);ctn-1(lf). These results suggest that CTN-1 plays an important role in BK channel function by mediating channel subcellular localization.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle Cells/metabolism , alpha Catenin/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Female , Large-Conductance Calcium-Activated Potassium Channels/genetics , Molecular Sequence Data , Oocytes/metabolism , Phenotype , Sequence Homology, Amino Acid , Subcellular Fractions , Xenopus laevis , alpha Catenin/genetics
16.
Cold Spring Harb Protoc ; 2010(3): pdb.prot5388, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20194457

ABSTRACT

This protocol describes the preparation of Caenorhabditis elegans embryos for four-dimensional (4D) imaging. Embryos are excised from gravid hermaphrodites and allowed to adhere directly to a poly-L-lysine-coated coverslip. The coverslip is then used to create a slide chamber in which the embryos are suspended in buffer. These suspended mounts are better for 4D recordings than agar mounts because, although agar mounts produce more uniformly oriented embryos, evaporation from the agar pads tends to shift the embryos gradually over time. 4D spatial registration with suspended mounts is quite stable, and it is possible to digitally reorient embryos within the recorded volume of the three-dimensional (3D) stack during post-processing of the data.


Subject(s)
Caenorhabditis elegans/embryology , Embryology/methods , Image Processing, Computer-Assisted/methods , Animals , Embryo, Nonmammalian/cytology
17.
Cold Spring Harb Protoc ; 2010(3): pdb.top71, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20194475

ABSTRACT

Embryos are remarkable for their combination of pluripotency, three-dimensionality, and swiftness of subcellular and developmental rearrangements. Embryogenesis in the nematode Caenorhabditis elegans is uniquely suited among model systems to high-resolution dynamic imaging. Within a single high-magnification, high-numerical aperture (NA) microscope field, at submicrometer resolution, it is possible to observe several entire animals taking form. The full approximately 14-h course of embryonic cleavage and morphogenesis of this transparent, free-living worm is essentially invariant. Observing specific fluorescently labeled components during embryonic development promises to reveal the roles of organelles and molecules in an extremely diverse and reproducible set of contexts. The C. elegans community has created a growing collection of hundreds of transgenic strains expressing green fluorescent protein (GFP)-labeled versions of distinct endogenously expressed genes. The task of correlating the resulting expression and localization patterns in space and time is simultaneously alluring and technically demanding. This article describes the use of four-dimensional (4D) laser-scanning microscopy and subsequent data processing to record, portray, analyze, and compare the expression of fluorescently tagged gene products during development of the nematode embryo.


Subject(s)
Caenorhabditis elegans/embryology , Image Processing, Computer-Assisted/methods , Microscopy, Confocal/methods , Animals , Artificial Gene Fusion , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Staining and Labeling/methods
18.
BMC Neurosci ; 11: 32, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20202202

ABSTRACT

BACKGROUND: Peptidergic neurons store and secrete the contents of large dense core vesicles (LDCVs) from axon terminals and from dendrites. Secretion of peptides requires a highly regulated exocytotic mechanism, plus coordinated synthesis and transport of LDCVs to their sites of release. Although these trafficking events are critical to function, little is known regarding the dynamic behavior of LDCVs and the mechanisms by which their transport is regulated. Sensory neurons also package opiate receptors in peptide-containing LDCVs, which is thought to be important in pain sensation. Since peptide granules cannot be refilled locally after their contents are secreted, it is particularly important to understand how neurons support regulated release of peptides. RESULTS: A vector encoding soluble peptidylglycine alpha-hydroxylating monooxygenase fused to green fluorescent protein was constructed to address these questions in cultured primary peptidergic neurons of the trigeminal ganglion using time lapse confocal microscopy. The time course of release differs with secretagogue; the secretory response to depolarization with K+ is rapid and terminates within 15 minutes, while phorbol ester stimulation of secretion is maintained over a longer period. The data demonstrate fundamental differences between LDCV dynamics in axons and growth cones under basal conditions. CONCLUSIONS: Under basal conditions, LDCVs move faster away from the soma than toward the soma, but fewer LDCVs travel anterograde than retrograde. Stimulation decreased average anterograde velocity and increases granule pausing. Data from antibody uptake, quantification of enzyme secretion and appearance of pHluorin fluorescence demonstrate distributed release of peptides all along the axon, not just at terminals.


Subject(s)
Neurons/physiology , Secretory Pathway/physiology , Secretory Vesicles/physiology , Trigeminal Ganglion/physiology , Actins/metabolism , Animals , Axons/drug effects , Axons/physiology , Cells, Cultured , Cytoskeleton/physiology , Green Fluorescent Proteins/metabolism , Growth Cones/drug effects , Growth Cones/physiology , Mixed Function Oxygenases/metabolism , Motion , Neurons/drug effects , Peripheral Nervous System Agents/pharmacology , Phorbol Esters/pharmacology , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Secretory Pathway/drug effects , Secretory Vesicles/drug effects , Secretory Vesicles/metabolism , Time Factors , Trigeminal Ganglion/drug effects
20.
Methods Mol Biol ; 475: 223-44, 2008.
Article in English | MEDLINE | ID: mdl-18979247

ABSTRACT

Cell fusion is a very dynamic process in which the entire membrane and cellular contents of two or more cells merge into one. Strategies developed to understand the component processes that make up a full fusion event require imaging to be performed over a range of space and time scales. These strategies must cover detection of nanometer-sized pores, monitoring cytoplasmic diffusion and the dynamic localization of proteins that induce fusion competence, and three-dimensional reconstruction of multinucleated cells. Caenorhabditis elegans' small size, predictable development, and transparent body make this organism optimal for microscopic investigations. In this chapter, focus is placed on light microscopy techniques that have been used thus far to study developmental fusion events in C. elegans and the insights that have been gained from them. There is also a general overview of the developmental timing of the cell fusion events. Additionally, several protocols are described for preparing both fixed and live specimens at various developmental stages of C. elegans for examination via optical microscopy.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Cell Fusion/methods , Microscopy/methods , Animals , Antibodies , Caenorhabditis elegans/embryology , Embryo, Nonmammalian/cytology , Epidermal Cells , Freezing , Permeability , Porosity , Tissue Fixation
SELECTION OF CITATIONS
SEARCH DETAIL
...