Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.568
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(12): 3141-3160.e23, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38759650

ABSTRACT

Systematic functional profiling of the gene set that directs embryonic development is an important challenge. To tackle this challenge, we used 4D imaging of C. elegans embryogenesis to capture the effects of 500 gene knockdowns and developed an automated approach to compare developmental phenotypes. The automated approach quantifies features-including germ layer cell numbers, tissue position, and tissue shape-to generate temporal curves whose parameterization yields numerical phenotypic signatures. In conjunction with a new similarity metric that operates across phenotypic space, these signatures enabled the generation of ranked lists of genes predicted to have similar functions, accessible in the PhenoBank web portal, for ∼25% of essential development genes. The approach identified new gene and pathway relationships in cell fate specification and morphogenesis and highlighted the utilization of specialized energy generation pathways during embryogenesis. Collectively, the effort establishes the foundation for comprehensive analysis of the gene set that builds a multicellular organism.


Subject(s)
Caenorhabditis elegans , Embryonic Development , Gene Expression Regulation, Developmental , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Profiling/methods , Gene Knockdown Techniques , Phenotype
2.
Cell ; 165(2): 396-409, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27020753

ABSTRACT

Multiple division cycles without growth are a characteristic feature of early embryogenesis. The female germline loads proteins and RNAs into oocytes to support these divisions, which lack many quality control mechanisms operating in somatic cells undergoing growth. Here, we describe a small RNA-Argonaute pathway that ensures early embryonic divisions in C. elegans by employing catalytic slicing activity to broadly tune, instead of silence, germline gene expression. Misregulation of one target, a kinesin-13 microtubule depolymerase, underlies a major phenotype associated with pathway loss. Tuning of target transcript levels is guided by the density of homologous small RNAs, whose generation must ultimately be related to target sequence. Thus, the tuning action of a small RNA-catalytic Argonaute pathway generates oocytes capable of supporting embryogenesis. We speculate that the specialized nature of germline chromatin led to the emergence of small RNA-catalytic Argonaute pathways in the female germline as a post-transcriptional control layer to optimize oocyte composition.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Embryo, Nonmammalian/metabolism , Metabolic Networks and Pathways , Oocytes/metabolism , Animals , Argonaute Proteins/metabolism , Base Sequence , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/metabolism , Cell Division , Embryo, Nonmammalian/cytology , Embryonic Development , Female , Kinesins/metabolism , Microtubules/metabolism , Molecular Sequence Data , RNA Processing, Post-Transcriptional
3.
Cell ; 163(6): 1333-47, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26607792

ABSTRACT

Interphase chromatin is organized in distinct nuclear sub-compartments, reflecting its degree of compaction and transcriptional status. In Caenorhabditis elegans embryos, H3K9 methylation is necessary to silence and to anchor repeat-rich heterochromatin at the nuclear periphery. In a screen for perinuclear anchors of heterochromatin, we identified a previously uncharacterized C. elegans chromodomain protein, CEC-4. CEC-4 binds preferentially mono-, di-, or tri-methylated H3K9 and localizes at the nuclear envelope independently of H3K9 methylation and nuclear lamin. CEC-4 is necessary for endogenous heterochromatin anchoring, but not for transcriptional repression, in contrast to other known H3K9 methyl-binders in worms, which mediate gene repression but not perinuclear anchoring. When we ectopically induce a muscle differentiation program in embryos, cec-4 mutants fail to commit fully to muscle cell fate. This suggests that perinuclear sequestration of chromatin during development helps restrict cell differentiation programs by stabilizing commitment to a specific cell fate. PAPERCLIP.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Chromosomal Proteins, Non-Histone/metabolism , Embryo, Nonmammalian/cytology , Heterochromatin , Histone Code , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Embryo, Nonmammalian/metabolism , Molecular Sequence Data , Sequence Alignment
4.
Nature ; 613(7943): 365-374, 2023 01.
Article in English | MEDLINE | ID: mdl-36544019

ABSTRACT

How paternal exposure to ionizing radiation affects genetic inheritance and disease risk in the offspring has been a long-standing question in radiation biology. In humans, nearly 80% of transmitted mutations arise in the paternal germline1, but the transgenerational effects of ionizing radiation exposure has remained controversial and the mechanisms are unknown. Here we show that in sex-separated Caenorhabditis elegans strains, paternal, but not maternal, exposure to ionizing radiation leads to transgenerational embryonic lethality. The offspring of irradiated males displayed various genome instability phenotypes, including DNA fragmentation, chromosomal rearrangement and aneuploidy. Paternal DNA double strand breaks were repaired by maternally provided error-prone polymerase theta-mediated end joining. Mechanistically, we show that depletion of an orthologue of human histone H1.0, HIS-24, or the heterochromatin protein HPL-1, could significantly reverse the transgenerational embryonic lethality. Removal of HIS-24 or HPL-1 reduced histone 3 lysine 9 dimethylation and enabled error-free homologous recombination repair in the germline of the F1 generation from ionizing radiation-treated P0 males, consequently improving the viability of the F2 generation. This work establishes the mechanistic underpinnings of the heritable consequences of paternal radiation exposure on the health of offspring, which may lead to congenital disorders and cancer in humans.


Subject(s)
Caenorhabditis elegans , DNA Damage , DNA Repair , Histones , Animals , Humans , Male , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/radiation effects , DNA Damage/radiation effects , Genomic Instability/radiation effects , Histones/metabolism , Mutation , Radiation, Ionizing , Embryo Loss/genetics , Female , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair , DNA Polymerase theta
5.
Mol Cell ; 81(3): 546-557.e5, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33378643

ABSTRACT

Eukaryotic cells regulate 5'-triphosphorylated RNAs (ppp-RNAs) to promote cellular functions and prevent recognition by antiviral RNA sensors. For example, RNA capping enzymes possess triphosphatase domains that remove the γ phosphates of ppp-RNAs during RNA capping. Members of the closely related PIR-1 (phosphatase that interacts with RNA and ribonucleoprotein particle 1) family of RNA polyphosphatases remove both the ß and γ phosphates from ppp-RNAs. Here, we show that C. elegans PIR-1 dephosphorylates ppp-RNAs made by cellular RNA-dependent RNA polymerases (RdRPs) and is required for the maturation of 26G-RNAs, Dicer-dependent small RNAs that regulate thousands of genes during spermatogenesis and embryogenesis. PIR-1 also regulates the CSR-1 22G-RNA pathway and has critical functions in both somatic and germline development. Our findings suggest that PIR-1 modulates both Dicer-dependent and Dicer-independent Argonaute pathways and provide insight into how cells and viruses use a conserved RNA phosphatase to regulate and respond to ppp-RNA species.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Phosphoric Monoester Hydrolases/metabolism , RNA Processing, Post-Transcriptional , RNA/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Gene Expression Regulation, Developmental , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , RNA/genetics , RNA Caps , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Spermatogenesis , Substrate Specificity
6.
Cell ; 154(2): 337-50, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870123

ABSTRACT

Synaptic contacts are largely established during embryogenesis and are then maintained during growth. To identify molecules involved in this process, we conducted a forward genetic screen in C. elegans and identified cima-1. In cima-1 mutants, synaptic contacts are correctly established during embryogenesis, but ectopic synapses emerge during postdevelopmental growth. cima-1 encodes a solute carrier in the SLC17 family of transporters that includes sialin, a protein that when mutated in humans results in neurological disorders. cima-1 does not function in neurons but rather functions in the nearby epidermal cells to correctly position glia during postlarval growth. Our findings indicate that CIMA-1 antagonizes the FGF receptor (FGFR), and does so most likely by inhibiting FGFR's role in epidermal-glia adhesion rather than signaling. Our data suggest that epidermal-glia crosstalk, in this case mediated by a transporter and the FGF receptor, is vital to preserve embryonically derived circuit architecture during postdevelopmental growth.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Neuroglia/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Sodium-Phosphate Cotransporter Proteins, Type I/metabolism , Synapses , Animals , Body Size , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , Embryonic Development , Epidermal Cells , Epidermis/metabolism , Mutation , Neurites/metabolism , Sodium-Phosphate Cotransporter Proteins, Type I/genetics
7.
Nature ; 609(7927): 597-604, 2022 09.
Article in English | MEDLINE | ID: mdl-35978196

ABSTRACT

A key event at the onset of development is the activation of a contractile actomyosin cortex during the oocyte-to-embryo transition1-3. Here we report on the discovery that, in Caenorhabditis elegans oocytes, actomyosin cortex activation is supported by the emergence of thousands of short-lived protein condensates rich in F-actin, N-WASP and the ARP2/3 complex4-8 that form an active micro-emulsion. A phase portrait analysis of the dynamics of individual cortical condensates reveals that condensates initially grow and then transition to disassembly before dissolving completely. We find that, in contrast to condensate growth through diffusion9, the growth dynamics of cortical condensates are chemically driven. Notably, the associated chemical reactions obey mass action kinetics that govern both composition and size. We suggest that the resultant condensate dynamic instability10 suppresses coarsening of the active micro-emulsion11, ensures reaction kinetics that are independent of condensate size and prevents runaway F-actin nucleation during the formation of the first cortical actin meshwork.


Subject(s)
Actomyosin , Biomolecular Condensates , Caenorhabditis elegans , Oocytes , Actin Cytoskeleton/metabolism , Actin-Related Protein 2/metabolism , Actin-Related Protein 3/metabolism , Actins/metabolism , Actomyosin/chemistry , Actomyosin/metabolism , Animals , Biomolecular Condensates/chemistry , Biomolecular Condensates/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Emulsions/chemistry , Emulsions/metabolism , Oocytes/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
8.
Mol Cell ; 78(1): 96-111.e6, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32105612

ABSTRACT

Current models suggest that chromosome domains segregate into either an active (A) or inactive (B) compartment. B-compartment chromatin is physically separated from the A compartment and compacted by the nuclear lamina. To examine these models in the developmental context of C. elegans embryogenesis, we undertook chromosome tracing to map the trajectories of entire autosomes. Early embryonic chromosomes organized into an unconventional barbell-like configuration, with two densely folded B compartments separated by a central A compartment. Upon gastrulation, this conformation matured into conventional A/B compartments. We used unsupervised clustering to uncover subpopulations with differing folding properties and variable positioning of compartment boundaries. These conformations relied on tethering to the lamina to stretch the chromosome; detachment from the lamina compacted, and allowed intermingling between, A/B compartments. These findings reveal the diverse conformations of early embryonic chromosomes and uncover a previously unappreciated role for the lamina in systemic chromosome stretching.


Subject(s)
Caenorhabditis elegans/genetics , Chromosomes/chemistry , Nuclear Lamina/physiology , Animals , Caenorhabditis elegans/embryology , Chromosomes/ultrastructure , Embryo, Nonmammalian/ultrastructure , Gastrulation/genetics , In Situ Hybridization, Fluorescence , Molecular Conformation
9.
Genes Dev ; 34(9-10): 678-687, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32217667

ABSTRACT

The proteostasis network is regulated by transcellular communication to promote health and fitness in metazoans. In Caenorhabditis elegans, signals from the germline initiate the decline of proteostasis and repression of cell stress responses at reproductive maturity, indicating that commitment to reproduction is detrimental to somatic health. Here we show that proteostasis and stress resilience are also regulated by embryo-to-mother communication in reproductive adults. To identify genes that act directly in the reproductive system to regulate somatic proteostasis, we performed a tissue targeted genetic screen for germline modifiers of polyglutamine aggregation in muscle cells. We found that inhibiting the formation of the extracellular vitelline layer of the fertilized embryo inside the uterus suppresses aggregation, improves stress resilience in an HSF-1-dependent manner, and restores the heat-shock response in the somatic tissues of the parent. This pathway relies on DAF-16/FOXO activation in vulval tissues to maintain stress resilience in the mother, suggesting that the integrity of the embryo is monitored by the vulva to detect damage and initiate an organismal protective response. Our findings reveal a previously undescribed transcellular pathway that links the integrity of the developing progeny to proteostasis regulation in the parent.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Proteostasis/genetics , Stress, Physiological/physiology , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , Cell Communication , Embryo, Nonmammalian , Female , Forkhead Transcription Factors/genetics , Helminth Proteins/genetics , Helminth Proteins/metabolism , Transcriptional Activation/genetics
10.
Cell ; 151(6): 1229-42, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23201143

ABSTRACT

The mechanisms by which functional left/right asymmetry arises in morphologically symmetric nervous systems are poorly understood. Here, we provide a mechanistic framework for how functional asymmetry in a postmitotic neuron pair is specified in C. elegans. A key feature of this mechanism is a temporally separated, two-step activation of the lsy-6 miRNA locus. The lsy-6 locus is first "primed" by chromatin decompaction in the precursor for the left neuron, but not the right neuron, several divisions before the neurons are born. lsy-6 expression is then "boosted" to functionally relevant levels several divisions later in the mother of the left neuron, through the activity of a bilaterally expressed transcription factor that can only activate lsy-6 in the primed neuron. This study shows how cells can become committed during early developmental stages to execute a specific fate much later in development and provides a conceptual framework for understanding the generation of neuronal diversity.


Subject(s)
Body Patterning , Caenorhabditis elegans/embryology , Neurons/cytology , Animals , Base Sequence , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Cell Lineage , Embryo, Nonmammalian/metabolism , Gene Expression , MicroRNAs/metabolism , Neurons/metabolism , Receptors, Notch/metabolism , Sequence Alignment
11.
Cell ; 150(5): 934-47, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22939621

ABSTRACT

The factors that sequester transcriptionally repressed heterochromatin at the nuclear periphery are currently unknown. In a genome-wide RNAi screen, we found that depletion of S-adenosylmethionine (SAM) synthetase reduces histone methylation globally and causes derepression and release of heterochromatin from the nuclear periphery in Caenorhabditis elegans embryos. Analysis of histone methyltransferases (HMTs) showed that elimination of two HMTs, MET-2 and SET-25, mimics the loss of SAM synthetase, abrogating the perinuclear attachment of heterochromatic transgenes and of native chromosomal arms rich in histone H3 lysine 9 methylation. The two HMTs target H3K9 in a consecutive fashion: MET-2, a SETDB1 homolog, mediates mono- and dimethylation, and SET-25, a previously uncharacterized HMT, deposits H3K9me3. SET-25 colocalizes with its own product in perinuclear foci, in a manner dependent on H3K9me3, but not on its catalytic domain. This colocalization suggests an autonomous, self-reinforcing mechanism for the establishment and propagation of repeat-rich heterochromatin.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Cell Nucleus/chemistry , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/genetics , Chromosomes/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Genome, Helminth , Histone-Lysine N-Methyltransferase/analysis , Histone-Lysine N-Methyltransferase/genetics , Lamins/metabolism , Methionine Adenosyltransferase/metabolism , Methylation , Mutation
12.
Nature ; 600(7888): 279-284, 2021 12.
Article in English | MEDLINE | ID: mdl-34837071

ABSTRACT

Confocal microscopy1 remains a major workhorse in biomedical optical microscopy owing to its reliability and flexibility in imaging various samples, but suffers from substantial point spread function anisotropy, diffraction-limited resolution, depth-dependent degradation in scattering samples and volumetric bleaching2. Here we address these problems, enhancing confocal microscopy performance from the sub-micrometre to millimetre spatial scale and the millisecond to hour temporal scale, improving both lateral and axial resolution more than twofold while simultaneously reducing phototoxicity. We achieve these gains using an integrated, four-pronged approach: (1) developing compact line scanners that enable sensitive, rapid, diffraction-limited imaging over large areas; (2) combining line-scanning with multiview imaging, developing reconstruction algorithms that improve resolution isotropy and recover signal otherwise lost to scattering; (3) adapting techniques from structured illumination microscopy, achieving super-resolution imaging in densely labelled, thick samples; (4) synergizing deep learning with these advances, further improving imaging speed, resolution and duration. We demonstrate these capabilities on more than 20 distinct fixed and live samples, including protein distributions in single cells; nuclei and developing neurons in Caenorhabditis elegans embryos, larvae and adults; myoblasts in imaginal disks of Drosophila wings; and mouse renal, oesophageal, cardiac and brain tissues.


Subject(s)
Deep Learning , Microscopy, Confocal/methods , Microscopy, Confocal/standards , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Caenorhabditis elegans/growth & development , Cell Line, Tumor , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Humans , Imaginal Discs/cytology , Mice , Myoblasts/cytology , Organ Specificity , Single-Cell Analysis , Tissue Fixation
13.
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
14.
Proc Natl Acad Sci U S A ; 121(25): e2318838121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38870057

ABSTRACT

Hertwig's rule states that cells divide along their longest axis, usually driven by forces acting on the mitotic spindle. Here, we show that in contrast to this rule, microtubule-based pulling forces in early Caenorhabditis elegans embryos align the spindle with the short axis of the cell. We combine theory with experiments to reveal that in order to correct this misalignment, inward forces generated by the constricting cytokinetic ring rotate the entire cell until the spindle is aligned with the cell's long axis. Experiments with slightly compressed mouse zygotes indicate that this cytokinetic ring-driven mechanism of ensuring Hertwig's rule is general for cells capable of rotating inside a confining shell, a scenario that applies to early cell divisions of many systems.


Subject(s)
Caenorhabditis elegans , Spindle Apparatus , Animals , Caenorhabditis elegans/embryology , Mice , Spindle Apparatus/metabolism , Microtubules/metabolism , Cytokinesis/physiology , Rotation , Zygote/metabolism , Zygote/cytology , Zygote/growth & development , Embryo, Nonmammalian/cytology , Embryonic Development/physiology , Models, Biological
15.
Genes Dev ; 33(7-8): 436-451, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30804228

ABSTRACT

Caenorhabditis elegans has two histone H3 Lys9 methyltransferases, MET-2 (SETDB1 homolog) and SET-25 (G9a/SUV39H1 related). In worms, we found simple repeat sequences primarily marked by H3K9me2, while transposable elements and silent tissue-specific genes bear H3K9me3. RNA sequencing (RNA-seq) in histone methyltransferase (HMT) mutants shows that MET-2-mediated H3K9me2 is necessary for satellite repeat repression, while SET-25 silences a subset of transposable elements and tissue-specific genes through H3K9me3. A genome-wide synthetic lethality screen showed that RNA processing, nuclear RNA degradation, the BRCA1/BARD1 complex, and factors mediating replication stress survival are necessary for germline viability in worms lacking MET-2 but not SET-25. Unlike set-25 mutants, met-2-null worms accumulated satellite repeat transcripts, which form RNA:DNA hybrids on repetitive sequences, additively with the loss of BRCA1 or BARD1. BRCA1/BARD1-mediated H2A ubiquitination and MET-2 deposited H3K9me2 on satellite repeats are partially interdependent, suggesting both that the loss of silencing generates BRCA-recruiting DNA damage and that BRCA1 recruitment by damage helps silence repeats. The artificial induction of MSAT1 transcripts can itself trigger damage-induced germline lethality in a wild-type background, arguing that the synthetic sterility upon BRCA1/BARD1 and H3K9me2 loss is directly linked to the DNA damage provoked by unscheduled satellite repeat transcription.


Subject(s)
BRCA1 Protein/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental/genetics , Histones/genetics , Animals , BRCA1 Protein/metabolism , Caenorhabditis elegans Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Transposable Elements/genetics , Embryo, Nonmammalian , Fertility/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Microsatellite Repeats/genetics , Mutation , RNA Processing, Post-Transcriptional/genetics , Temperature
16.
Nat Rev Mol Cell Biol ; 15(10): 634-46, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25237825

ABSTRACT

During cell division, it is critical to properly partition functional sets of organelles to each daughter cell. The partitioning of mitochondria shares some common features with that of other organelles, particularly in the use of interactions with cytoskeletal elements to facilitate delivery to the daughter cells. However, mitochondria have unique features - including their own genome and a maternal mode of germline transmission - that place additional demands on this process. Consequently, mechanisms have evolved to regulate mitochondrial segregation during cell division, oogenesis, fertilization and tissue development, as well as to ensure the integrity of these organelles and their DNA, including fusion-fission dynamics, organelle transport, mitophagy and genetic selection of functional genomes. Defects in these processes can lead to cell and tissue pathologies.


Subject(s)
Cell Division/genetics , Cell Division/physiology , Mitochondria/genetics , Mitochondria/physiology , Mitochondrial Dynamics/physiology , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/physiology , Embryonic Development , Fertilization/genetics , Humans , Mitophagy , Oogenesis/genetics , Saccharomyces cerevisiae/growth & development
17.
Nat Rev Mol Cell Biol ; 15(5): 327-39, 2014 May.
Article in English | MEDLINE | ID: mdl-24739741

ABSTRACT

With the advent of imaging probes and live microscopy, developmental biologists have markedly extended our understanding of the molecular and cellular details of embryonic development. To fully comprehend the complex mechanistic framework that forms the developing organism, quantitative studies with high fidelity in space and time are now required. We discuss how integrating established, newly introduced and future imaging tools with quantitative analysis will ensure that imaging can fulfil its promise to elucidate how new life begins.


Subject(s)
Caenorhabditis elegans/anatomy & histology , Drosophila melanogaster/anatomy & histology , Image Processing, Computer-Assisted/methods , Molecular Imaging/methods , Zebrafish/anatomy & histology , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Embryonic Development , Genes, Reporter , Green Fluorescent Proteins/genetics , Image Processing, Computer-Assisted/instrumentation , Microscopy, Fluorescence, Multiphoton , Molecular Imaging/instrumentation , Molecular Probes , Spectrometry, Fluorescence , Zebrafish/embryology , Zebrafish/genetics
18.
Cell ; 145(3): 470-82, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529718

ABSTRACT

High-content screening for gene profiling has generally been limited to single cells. Here, we explore an alternative approach-profiling gene function by analyzing effects of gene knockdowns on the architecture of a complex tissue in a multicellular organism. We profile 554 essential C. elegans genes by imaging gonad architecture and scoring 94 phenotypic features. To generate a reference for evaluating methods for network construction, genes were manually partitioned into 102 phenotypic classes, predicting functions for uncharacterized genes across diverse cellular processes. Using this classification as a benchmark, we developed a robust computational method for constructing gene networks from high-content profiles based on a network context-dependent measure that ranks the significance of links between genes. Our analysis reveals that multi-parametric profiling in a complex tissue yields functional maps with a resolution similar to genetic interaction-based profiling in unicellular eukaryotes-pinpointing subunits of macromolecular complexes and components functioning in common cellular processes.


Subject(s)
Caenorhabditis elegans/genetics , Computational Biology/methods , Gene Regulatory Networks , Genetic Techniques , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Embryo, Nonmammalian/metabolism , Gene Knockdown Techniques , Gonads/embryology , Phenotype
19.
Cell ; 145(5): 720-31, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21620137

ABSTRACT

Extracellular free heme can intercalate into membranes and promote damage to cellular macromolecules. Thus it is likely that specific intercellular pathways exist for the directed transport, trafficking, and delivery of heme to cellular destinations, although none have been found to date. Here we show that Caenorhabditis elegans HRG-3 is required for the delivery of maternal heme to developing embryos. HRG-3 binds heme and is exclusively secreted by maternal intestinal cells into the interstitial fluid for transport of heme to extraintestinal cells, including oocytes. HRG-3 deficiency results either in death during embryogenesis or in developmental arrest immediately post-hatching-phenotypes that are fully suppressed by maternal but not zygotic hrg-3 expression. Our results establish a role for HRG-3 as an intercellular heme-trafficking protein.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Heme/metabolism , Hemeproteins/metabolism , Animals , Animals, Genetically Modified , Gene Expression Regulation, Developmental , Genes, Reporter , Heme/deficiency , Hemeproteins/chemistry , Hemeproteins/genetics , Intestinal Mucosa/metabolism , Mutation , Phenotype , Protein Transport , Secretory Pathway
20.
Cell ; 146(6): 942-54, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925317

ABSTRACT

Extrinsic signals received by a cell can induce remodeling of the cytoskeleton, but the downstream effects of cytoskeletal changes on gene expression have not been well studied. Here, we show that during telophase of an asymmetric division in C. elegans, extrinsic Wnt signaling modulates spindle structures through APR-1/APC, which in turn promotes asymmetrical nuclear localization of WRM-1/ß-catenin and POP-1/TCF. APR-1 that localized asymmetrically along the cortex established asymmetric distribution of astral microtubules, with more microtubules found on the anterior side. Perturbation of the Wnt signaling pathway altered this microtubule asymmetry and led to changes in nuclear WRM-1 asymmetry, gene expression, and cell-fate determination. Direct manipulation of spindle asymmetry by laser irradiation altered the asymmetric distribution of nuclear WRM-1. Moreover, laser manipulation of the spindles rescued defects in nuclear POP-1 asymmetry in wnt mutants. Our results reveal a mechanism in which the nuclear localization of proteins is regulated through the modulation of microtubules.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Cytoskeletal Proteins/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , Caenorhabditis elegans/embryology , Cell Nucleus/metabolism , Kinesins/metabolism , Microtubules/metabolism , Spindle Apparatus , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL