Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.568
Filter
1.
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
2.
Elife ; 122024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900560

ABSTRACT

The paramount importance of mechanical forces in morphogenesis and embryogenesis is widely recognized, but understanding the mechanism at the cellular and molecular level remains challenging. Because of its simple internal organization, Caenorhabditis elegans is a rewarding system of study. As demonstrated experimentally, after an initial period of steady elongation driven by the actomyosin network, muscle contractions operate a quasi-periodic sequence of bending, rotation, and torsion, that leads to the final fourfold size of the embryos before hatching. How actomyosin and muscles contribute to embryonic elongation is investigated here theoretically. A filamentary elastic model that converts stimuli generated by biochemical signals in the tissue into driving forces, explains embryonic deformation under actin bundles and muscle activity, and dictates mechanisms of late elongation based on the effects of energy conversion and dissipation. We quantify this dynamic transformation by stretches applied to a cylindrical structure that mimics the body shape in finite elasticity, obtaining good agreement and understanding of both wild-type and mutant embryos at all stages.


Subject(s)
Actomyosin , Caenorhabditis elegans , Embryo, Nonmammalian , Muscle Contraction , Caenorhabditis elegans/embryology , Animals , Actomyosin/metabolism , Muscle Contraction/physiology , Embryo, Nonmammalian/physiology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Embryonic Development , Morphogenesis , Models, Biological , Biomechanical Phenomena
3.
PLoS Comput Biol ; 20(6): e1011882, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38838038

ABSTRACT

In embryonic development and organogenesis, cells sharing identical genetic codes acquire diverse gene expression states in a highly reproducible spatial distribution, crucial for multicellular formation and quantifiable through positional information. To understand the spontaneous growth of complexity, we constructed a one-dimensional division-decision model, simulating the growth of cells with identical genetic networks from a single cell. Our findings highlight the pivotal role of cell division in providing positional cues, escorting the system toward states rich in information. Moreover, we pinpointed lateral inhibition as a critical mechanism translating spatial contacts into gene expression. Our model demonstrates that the spatial arrangement resulting from cell division, combined with cell lineages, imparts positional information, specifying multiple cell states with increased complexity-illustrated through examples in C.elegans. This study constitutes a foundational step in comprehending developmental intricacies, paving the way for future quantitative formulations to construct synthetic multicellular patterns.


Subject(s)
Gene Regulatory Networks , Models, Biological , Animals , Gene Regulatory Networks/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/growth & development , Cell Division/physiology , Cell Division/genetics , Computational Biology , Embryonic Development/physiology , Embryonic Development/genetics , Cell Lineage , Computer Simulation , Gene Expression Regulation, Developmental/genetics
4.
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
5.
Mol Biol Cell ; 35(7): ar94, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38696255

ABSTRACT

Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formincyk-1(ts) mutant Caenorhabditis elegans 4-cell embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide with greatly reduced F-actin levels at the cell division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septinUNC-59 and anillinANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into the regulation of cytokinesis in other cell types, especially in stem cells with high potency.


Subject(s)
Actins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Division , Cytokinesis , Germ Cells , Septins , Animals , Cytokinesis/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Septins/metabolism , Septins/genetics , Germ Cells/metabolism , Germ Cells/cytology , Actins/metabolism , Contractile Proteins/metabolism , Actomyosin/metabolism
6.
Nat Commun ; 15(1): 4180, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755148

ABSTRACT

Computational super-resolution methods, including conventional analytical algorithms and deep learning models, have substantially improved optical microscopy. Among them, supervised deep neural networks have demonstrated outstanding performance, however, demanding abundant high-quality training data, which are laborious and even impractical to acquire due to the high dynamics of living cells. Here, we develop zero-shot deconvolution networks (ZS-DeconvNet) that instantly enhance the resolution of microscope images by more than 1.5-fold over the diffraction limit with 10-fold lower fluorescence than ordinary super-resolution imaging conditions, in an unsupervised manner without the need for either ground truths or additional data acquisition. We demonstrate the versatile applicability of ZS-DeconvNet on multiple imaging modalities, including total internal reflection fluorescence microscopy, three-dimensional wide-field microscopy, confocal microscopy, two-photon microscopy, lattice light-sheet microscopy, and multimodal structured illumination microscopy, which enables multi-color, long-term, super-resolution 2D/3D imaging of subcellular bioprocesses from mitotic single cells to multicellular embryos of mouse and C. elegans.


Subject(s)
Caenorhabditis elegans , Microscopy, Fluorescence , Animals , Caenorhabditis elegans/embryology , Microscopy, Fluorescence/methods , Mice , Imaging, Three-Dimensional/methods , Algorithms , Image Processing, Computer-Assisted/methods , Deep Learning
7.
Curr Biol ; 34(9): 1853-1865.e6, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38604167

ABSTRACT

Different signaling mechanisms concur to ensure robust tissue patterning and cell fate instruction during animal development. Most of these mechanisms rely on signaling proteins that are produced, transported, and detected. The spatiotemporal dynamics of signaling molecules are largely unknown, yet they determine signal activity's spatial range and time frame. Here, we use the Caenorhabditis elegans embryo to study how Wnt ligands, an evolutionarily conserved family of signaling proteins, dynamically organize to establish cell polarity in a developing tissue. We identify how Wnt ligands, produced in the posterior half of the embryos, spread extracellularly to transmit information to distant target cells in the anterior half. With quantitative live imaging and fluorescence correlation spectroscopy, we show that Wnt ligands diffuse through the embryo over a timescale shorter than the cell cycle, in the intercellular space, and outside the tissue below the eggshell. We extracted diffusion coefficients of Wnt ligands and their receptor Frizzled and characterized their co-localization. Integrating our different measurements and observations in a simple computational framework, we show how fast diffusion in the embryo can polarize individual cells through a time integration of the arrival of the ligands at the target cells. The polarity established at the tissue level by a posterior Wnt source can be transferred to the cellular level. Our results support a diffusion-based long-range Wnt signaling, which is consistent with the dynamics of developing processes.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Polarity , Embryo, Nonmammalian , Wnt Proteins , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Wnt Proteins/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Ligands , Wnt Signaling Pathway , Diffusion
8.
Dev Biol ; 511: 12-25, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38556137

ABSTRACT

During epithelial morphogenesis, the apical junctions connecting cells must remodel as cells change shape and make new connections with their neighbors. In the C. elegans embryo, new apical junctions form when epidermal cells migrate and seal with one another to encase the embryo in skin ('ventral enclosure'), and junctions remodel when epidermal cells change shape to squeeze the embryo into a worm shape ('elongation'). The junctional cadherin-catenin complex (CCC), which links epithelial cells to each other and to cortical actomyosin, is essential for C. elegans epidermal morphogenesis. RNAi genetic enhancement screens have identified several genes encoding proteins that interact with the CCC to promote epidermal morphogenesis, including the scaffolding protein Afadin (AFD-1), whose depletion alone results in only minor morphogenesis defects. Here, by creating a null mutation in afd-1, we show that afd-1 provides a significant contribution to ventral enclosure and elongation on its own. Unexpectedly, we find that afd-1 mutant phenotypes are strongly modified by diet, revealing a previously unappreciated parental nutritional input to morphogenesis. We identify functional interactions between AFD-1 and the CCC by demonstrating that E-cadherin is required for the polarized distribution of AFD-1 to cell contact sites in early embryos. Finally, we show that afd-1 promotes the enrichment of polarity regulator, and CCC-interacting protein, PAC-1/ARHGAP21 to cell contact sites, and we identify genetic interactions suggesting that afd-1 and pac-1 regulate epidermal morphogenesis at least in part through parallel mechanisms. Our findings reveal that C. elegans AFD-1 makes a significant contribution to epidermal morphogenesis and functionally interfaces with core and associated CCC proteins.


Subject(s)
Cadherins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Epidermis , Morphogenesis , Animals , Cadherins/metabolism , Cadherins/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Catenins/metabolism , Catenins/genetics , Epidermal Cells/metabolism , Epidermis/metabolism , Epidermis/embryology , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics
9.
Curr Biol ; 33(11): R447-R449, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37279667

ABSTRACT

Bub1 and Polo kinases are well-known multitasking regulators of mitosis. New work shows that they team up at kinetochores to determine the mitotic duration of embryonic divisions in nematodes. As is often the case, the key effector is Cdc20 activity.


Subject(s)
Cell Cycle , Embryo, Nonmammalian , Protein Serine-Threonine Kinases , Animals , Cdc20 Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Protein Serine-Threonine Kinases/metabolism , Mitosis , Time Factors , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism
10.
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
11.
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
12.
Proc Natl Acad Sci U S A ; 119(11): e2114205119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35259017

ABSTRACT

SignificanceIntracellular gradients have essential roles in cell and developmental biology, but their formation is not fully understood. We have developed a computational approach facilitating interpretation of protein dynamics and gradient formation. We have combined this computational approach with experiments to understand how Polo-Like Kinase 1 (PLK-1) forms a cytoplasmic gradient in Caenorhabditis elegans embryos. Although the PLK-1 gradient depends on the Muscle EXcess-5/6 (MEX-5/6) proteins, we reveal differences in PLK-1 and MEX-5 gradient formation that can be explained by a model with two components, PLK-1 bound to MEX-5 and unbound PLK-1. Our combined approach suggests that a weak coupling between PLK-1 and MEX-5 reaction-diffusion mechanisms dictates the dynamic exchange of PLK-1 with the cytoplasm, explaining PLK-1 high diffusivity and smooth gradient.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Proteome , Proteomics , Animals , Embryo, Nonmammalian , Models, Biological , Monte Carlo Method , Morphogenesis , Protein Serine-Threonine Kinases , Protein Transport , Proteomics/methods
13.
Nat Methods ; 19(2): 187-194, 2022 02.
Article in English | MEDLINE | ID: mdl-35115715

ABSTRACT

Single-guide RNAs can target exogenous CRISPR-Cas proteins to unique DNA locations, enabling genetic tools that are efficient, specific and scalable. Here we show that short synthetic guide Piwi-interacting RNAs (piRNAs) (21-nucleotide sg-piRNAs) expressed from extrachromosomal transgenes can, analogously, reprogram the endogenous piRNA pathway for gene-specific silencing in the hermaphrodite germline, sperm and embryos of Caenorhabditis elegans. piRNA-mediated interference ('piRNAi') is more efficient than RNAi and can be multiplexed, and auxin-mediated degradation of the piRNA-specific Argonaute PRG-1 allows conditional gene silencing. Target-specific silencing results in decreased messenger RNA levels, amplification of secondary small interfering RNAs and repressive chromatin modifications. Short (300 base pairs) piRNAi transgenes amplified from arrayed oligonucleotide pools also induce silencing, potentially making piRNAi highly scalable. We show that piRNAi can induce transgenerational epigenetic silencing of two endogenous genes (him-5 and him-8). Silencing is inherited for four to six generations after target-specific sg-piRNAs are lost, whereas depleting PRG-1 leads to essentially permanent epigenetic silencing.


Subject(s)
Animals, Genetically Modified/genetics , Caenorhabditis elegans/genetics , Gene Silencing , RNA Interference , RNA, Small Interfering/genetics , Animals , Argonaute Proteins/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/genetics , Embryo, Nonmammalian , Epigenesis, Genetic , Female , Male
14.
J Cell Biol ; 221(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-34994802

ABSTRACT

Contractile ring constriction during cytokinesis is thought to compact central spindle microtubules to form the midbody, an antiparallel microtubule bundle at the intercellular bridge. In Caenorhabditis elegans, central spindle microtubule assembly requires targeting of the CLASP family protein CLS-2 to the kinetochores in metaphase and spindle midzone in anaphase. CLS-2 targeting is mediated by the CENP-F-like HCP-1/2, but their roles in cytokinesis and midbody assembly are not known. We found that although HCP-1 and HCP-2 mostly function cooperatively, HCP-1 plays a more primary role in promoting CLS-2-dependent central spindle microtubule assembly. HCP-1/2 codisrupted embryos did not form central spindles but completed cytokinesis and formed functional midbodies capable of supporting abscission. These central spindle-independent midbodies appeared to form via contractile ring constriction-driven bundling of astral microtubules at the furrow tip. This work suggests that, in the absence of a central spindle, astral microtubules can support midbody assembly and that midbody assembly is more predictive of successful cytokinesis than central spindle assembly.


Subject(s)
Spindle Apparatus/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/metabolism , Embryo, Nonmammalian/metabolism , Green Fluorescent Proteins/metabolism , Microtubules/metabolism
15.
PLoS Comput Biol ; 18(1): e1009755, 2022 01.
Article in English | MEDLINE | ID: mdl-35030161

ABSTRACT

Morphogenesis is a precise and robust dynamic process during metazoan embryogenesis, consisting of both cell proliferation and cell migration. Despite the fact that much is known about specific regulations at molecular level, how cell proliferation and migration together drive the morphogenesis at cellular and organismic levels is not well understood. Using Caenorhabditis elegans as the model animal, we present a phase field model to compute early embryonic morphogenesis within a confined eggshell. With physical information about cell division obtained from three-dimensional time-lapse cellular imaging experiments, the model can precisely reproduce the early morphogenesis process as seen in vivo, including time evolution of location and morphology of each cell. Furthermore, the model can be used to reveal key cell-cell attractions critical to the development of C. elegans embryo. Our work demonstrates how genetic programming and physical forces collaborate to drive morphogenesis and provides a predictive model to decipher the underlying mechanism.


Subject(s)
Caenorhabditis elegans/embryology , Embryo, Nonmammalian/physiology , Embryonic Development/physiology , Models, Biological , Animals , Computational Biology
16.
Dev Biol ; 483: 13-21, 2022 03.
Article in English | MEDLINE | ID: mdl-34971598

ABSTRACT

Asymmetric cell division is an essential feature of normal development and certain pathologies. The process and its regulation have been studied extensively in the Caenorhabditis elegans embryo, particularly how symmetry of the actomyosin cortical cytoskeleton is broken by a sperm-derived signal at fertilization, upstream of polarity establishment. Diploscapter pachys is the closest parthenogenetic relative to C. elegans, and D. pachys one-cell embryos also divide asymmetrically. However how polarity is triggered in the absence of sperm remains unknown. In post-meiotic embryos, we find that the nucleus inhabits principally one embryo hemisphere, the future posterior pole. When forced to one pole by centrifugation, the nucleus returns to its preferred pole, although poles appear identical as concerns cortical ruffling and actin cytoskeleton. The location of the meiotic spindle also correlates with the future posterior pole and slight actin enrichment is observed at that pole in some early embryos along with microtubule structures emanating from the meiotic spindle. Polarized location of the nucleus is not observed in pre-meiotic D. pachys oocytes. All together our results are consistent with the idea that polarity of the D. pachys embryo is attained during meiosis, seemingly based on the location of the meiotic spindle, by a mechanism that may be present but suppressed in C. elegans.


Subject(s)
Asymmetric Cell Division/physiology , Meiosis/physiology , Oocytes/cytology , Oocytes/physiology , Parthenogenesis/physiology , Rhabditoidea/cytology , Rhabditoidea/embryology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Cell Nucleus/physiology , Female , Microtubules/physiology , Oviparity/physiology , Spindle Apparatus/physiology
17.
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
18.
Sci Rep ; 11(1): 20359, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645931

ABSTRACT

Previous studies indicated that the P-body components, CGH-1 and EDC-3 may play a crucial role in the regulation of lifespan in Caenorhabditis elegans. Homo sapiens DDX6 or Saccharomyces cerevisiae Dhh1p (CGH-1 in C. elegans) could form complexes with EDC3 (Edc3p in yeast), respectively, which is significant for translation inhibition and mRNA decay. However, it is currently unclear how CGH-1 can be recognized by EDC-3 in C. elegans. Here, we provided structural and biochemical insights into the interaction between CGH-1 and EDC-3. Combined with homology modeling, mutation, and ITC assays, we uncovered an interface between CGH-1 RecA2 domain and EDC-3 FDF-FEK. Additionally, GST-pulldown and co-localization experiments confirmed the interaction between CGH-1 and EDC-3 in vitro and in vivo. We also analyzed PATR-1-binding interface on CGH-1 RecA2 by ITC assays. Moreover, we unveiled the similarity and differences of the binding mode between EDC-3 and CAR-1 or PATR-1. Taken together, these findings provide insights into the recognition of DEAD-box protein CGH-1 by EDC-3 FDF-FEK motif, suggesting important functional implications.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans/embryology , RNA Helicases/chemistry , RNA Nucleotidyltransferases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Humans , Protein Binding , RNA Helicases/genetics , RNA Nucleotidyltransferases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
19.
Elife ; 102021 07 05.
Article in English | MEDLINE | ID: mdl-34223818

ABSTRACT

We describe MIP-1 and MIP-2, novel paralogous C. elegans germ granule components that interact with the intrinsically disordered MEG-3 protein. These proteins promote P granule condensation, form granules independently of MEG-3 in the postembryonic germ line, and balance each other in regulating P granule growth and localization. MIP-1 and MIP-2 each contain two LOTUS domains and intrinsically disordered regions and form homo- and heterodimers. They bind and anchor the Vasa homolog GLH-1 within P granules and are jointly required for coalescence of MEG-3, GLH-1, and PGL proteins. Animals lacking MIP-1 and MIP-2 show temperature-sensitive embryonic lethality, sterility, and mortal germ lines. Germline phenotypes include defects in stem cell self-renewal, meiotic progression, and gamete differentiation. We propose that these proteins serve as scaffolds and organizing centers for ribonucleoprotein networks within P granules that help recruit and balance essential RNA processing machinery to regulate key developmental transitions in the germ line.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Germ Cells/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/genetics
20.
Cell Rep ; 36(1): 109326, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233197

ABSTRACT

Coordination between cell differentiation and proliferation during development requires the balance between asymmetric and symmetric modes of cell division. However, the cellular intrinsic cue underlying the choice between these two division modes remains elusive. Here, we show evidence in Caenorhabditis elegans that the invariable lineage of the division modes is specified by the balance between antagonizing complexes of partitioning-defective (PAR) proteins. By uncoupling unequal inheritance of PAR proteins from that of fate determinants during cell division, we demonstrate that changes in the balance between PAR-2 and PAR-6 can be sufficient to re-program the division modes from symmetric to asymmetric and vice versa in two daughter cells. The division mode adopted occurs independently of asymmetry in cytoplasmic fate determinants, cell-size asymmetry, and cell-cycle asynchrony between sister cells. We propose that the balance between PAR proteins represents an intrinsic self-organizing cue for the specification of the two division modes during development.


Subject(s)
Asymmetric Cell Division , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Embryo, Nonmammalian/cytology , Embryonic Development , Animals , Cell Lineage , Cell Polarity , Computer Simulation , Embryo, Nonmammalian/metabolism , Models, Biological , Zygote/cytology , Zygote/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...