Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.446
Filter
1.
PLoS One ; 19(5): e0304064, 2024.
Article in English | MEDLINE | ID: mdl-38787850

ABSTRACT

Asymmetric cell division is an important mechanism that generates cellular diversity during development. Not only do asymmetric cell divisions produce daughter cells of different fates, but many can also produce daughters of different sizes, which we refer to as Daughter Cell Size Asymmetry (DCSA). In Caenorhabditis elegans, apoptotic cells are frequently produced by asymmetric divisions that exhibit DCSA, where the smaller daughter dies. We focus here on the divisions of the Q.a and Q.p neuroblasts, which produce larger surviving cells and smaller apoptotic cells and divide with opposite polarity using both distinct and overlapping mechanisms. Several proteins regulate DCSA in these divisions. Previous studies showed that the PIG-1/MELK and TOE-2 proteins regulate DCSA in both the Q.a and Q.p divisions, and the non-muscle myosin NMY-2 regulates DCSA in the Q.a division but not the Q.p division. In this study, we examined endogenously tagged NMY-2, TOE-2, and PIG-1 reporters and characterized their distribution at the cortex during the Q.a and Q.p divisions. In both divisions, TOE-2 localized toward the side of the dividing cell that produced the smaller daughter, whereas PIG-1 localized toward the side that produced the larger daughter. As previously reported, NMY-2 localized to the side of Q.a that produced the smaller daughter and did not localize asymmetrically in Q.p. We used temperature-sensitive nmy-2 mutants to determine the role of nmy-2 in these divisions and were surprised to find that these mutants only displayed DCSA defects in the Q.p division. We generated double mutant combinations between the nmy-2 mutations and mutations in toe-2 and pig-1. Because previous studies indicate that DCSA defects result in the transformation of cells fated to die into their sister cells, the finding that the nmy-2 mutations did not significantly alter the Q.a and Q.p DCSA defects of toe-2 and pig-1 mutants but did alter the number of daughter cells produced by Q.a and Q.p suggests that nmy-2 plays a role in specifying the fates of the Q.a and Q.p that is independent of its role in DCSA.


Subject(s)
Asymmetric Cell Division , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Cell Size , Myosins/metabolism , Myosins/genetics , Protein Serine-Threonine Kinases
2.
Bioinformatics ; 40(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38775410

ABSTRACT

MOTIVATION: Accurate segmentation and recognition of C.elegans cells are critical for various biological studies, including gene expression, cell lineages, and cell fates analysis at single-cell level. However, the highly dense distribution, similar shapes, and inhomogeneous intensity profiles of whole-body cells in 3D fluorescence microscopy images make automatic cell segmentation and recognition a challenging task. Existing methods either rely on additional fiducial markers or only handle a subset of cells. Given the difficulty or expense associated with generating fiducial features in many experimental settings, a marker-free approach capable of reliably segmenting and recognizing C.elegans whole-body cells is highly desirable. RESULTS: We report a new pipeline, called automated segmentation and recognition (ASR) of cells, and applied it to 3D fluorescent microscopy images of L1-stage C.elegans with 558 whole-body cells. A novel displacement vector field based deep learning model is proposed to address the problem of reliable segmentation of highly crowded cells with blurred boundary. We then realize the cell recognition by encoding and exploiting statistical priors on cell positions and structural similarities of neighboring cells. To the best of our knowledge, this is the first method successfully applied to the segmentation and recognition of C.elegans whole-body cells. The ASR-segmentation module achieves an F1-score of 0.8956 on a dataset of 116 C.elegans image stacks with 64 728 cells (accuracy 0.9880, AJI 0.7813). Based on the segmentation results, the ASR recognition module achieved an average accuracy of 0.8879. We also show ASR's applicability to other cell types, e.g. platynereis and rat kidney cells. AVAILABILITY AND IMPLEMENTATION: The code is available at https://github.com/reaneyli/ASR.


Subject(s)
Caenorhabditis elegans , Caenorhabditis elegans/cytology , Animals , Microscopy, Fluorescence/methods , Imaging, Three-Dimensional/methods , Image Processing, Computer-Assisted/methods , Algorithms , Deep Learning
3.
Genetics ; 227(2)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38785371

ABSTRACT

Since the days of Ramón y Cajal, the vast diversity of neuronal and particularly dendrite morphology has been used to catalog neurons into different classes. Dendrite morphology varies greatly and reflects the different functions performed by different types of neurons. Significant progress has been made in our understanding of how dendrites form and the molecular factors and forces that shape these often elaborately sculpted structures. Here, we review work in the nematode Caenorhabditis elegans that has shed light on the developmental mechanisms that mediate dendrite morphogenesis with a focus on studies investigating ciliated sensory neurons and the highly elaborated dendritic trees of somatosensory neurons. These studies, which combine time-lapse imaging, genetics, and biochemistry, reveal an intricate network of factors that function both intrinsically in dendrites and extrinsically from surrounding tissues. Therefore, dendrite morphogenesis is the result of multiple tissue interactions, which ultimately determine the shape of dendritic arbors.


Subject(s)
Caenorhabditis elegans , Dendrites , Morphogenesis , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/cytology , Dendrites/metabolism , Morphogenesis/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/cytology
4.
J Cell Biol ; 223(5)2024 May 06.
Article in English | MEDLINE | ID: mdl-38470363

ABSTRACT

Mitochondria transport is crucial for axonal mitochondria distribution and is mediated by kinesin-1-based anterograde and dynein-based retrograde motor complexes. While Miro and Milton/TRAK were identified as key adaptors between mitochondria and kinesin-1, recent studies suggest the presence of additional mechanisms. In C. elegans, ric-7 is the only single gene described so far, other than kinesin-1, that is absolutely required for axonal mitochondria localization. Using CRISPR engineering in C. elegans, we find that Miro is important but is not essential for anterograde traffic, whereas it is required for retrograde traffic. Both the endogenous RIC-7 and kinesin-1 act at the leading end to transport mitochondria anterogradely. RIC-7 binding to mitochondria requires its N-terminal domain and partially relies on MIRO-1, whereas RIC-7 accumulation at the leading end depends on its disordered region, kinesin-1, and metaxin2. We conclude that transport complexes containing kinesin-1 and RIC-7 polarize at the leading edge of mitochondria and are required for anterograde axonal transport in C. elegans.


Subject(s)
Axonal Transport , Kinesins , Animals , Axons , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Kinesins/metabolism , Mitochondria/metabolism
5.
Nature ; 628(8008): 630-638, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538795

ABSTRACT

Lysosomes are degradation and signalling centres crucial for homeostasis, development and ageing1. To meet diverse cellular demands, lysosomes remodel their morphology and function through constant fusion and fission2,3. Little is known about the molecular basis of fission. Here we identify HPO-27, a conserved HEAT repeat protein, as a lysosome scission factor in Caenorhabditis elegans. Loss of HPO-27 impairs lysosome fission and leads to an excessive tubular network that ultimately collapses. HPO-27 and its human homologue MROH1 are recruited to lysosomes by RAB-7 and enriched at scission sites. Super-resolution imaging, negative-staining electron microscopy and in vitro reconstitution assays reveal that HPO-27 and MROH1 self-assemble to mediate the constriction and scission of lysosomal tubules in worms and mammalian cells, respectively, and assemble to sever supported membrane tubes in vitro. Loss of HPO-27 affects lysosomal morphology, integrity and degradation activity, which impairs animal development and longevity. Thus, HPO-27 and MROH1 act as self-assembling scission factors to maintain lysosomal homeostasis and function.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Lysosomes , Animals , Humans , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/ultrastructure , Homeostasis , Longevity , Lysosomes/metabolism , Lysosomes/ultrastructure , Amino Acid Motifs , Microscopy, Electron
6.
Genetics ; 227(1)2024 May 07.
Article in English | MEDLINE | ID: mdl-38427913

ABSTRACT

RNA-binding proteins FBF-1 and FBF-2 (FBFs) are required for germline stem cell maintenance and the sperm/oocyte switch in Caenorhabditis elegans, although the mechanisms controlling FBF protein levels remain unknown. We identified an interaction between both FBFs and CSN-5), a component of the constitutive photomorphogenesis 9 (COP9) signalosome best known for its role in regulating protein degradation. Here, we find that the Mpr1/Pad1 N-terminal metalloprotease domain of CSN-5 interacts with the Pumilio and FBF RNA-binding domain of FBFs and the interaction is conserved for human homologs CSN5 and PUM1. The interaction between FBF-2 and CSN-5 can be detected in vivo by proximity ligation. csn-5 mutation results in the destabilization of FBF proteins, which may explain previously observed decrease in the numbers of germline stem and progenitor cells, and disruption of oogenesis. The loss of csn-5 does not decrease the levels of a related PUF protein PUF-3, and csn-5(lf) phenotype is not enhanced by fbf-1/2 knockdown, suggesting that the effect is specific to FBFs. The effect of csn-5 on oogenesis is largely independent of the COP9 signalosome and is cell autonomous. Surprisingly, the regulation of FBF protein levels involves a combination of COP9-dependent and COP9-independent mechanisms differentially affecting FBF-1 and FBF-2. This work supports a previously unappreciated role for CSN-5 in the stabilization of germline stem cell regulatory proteins FBF-1 and FBF-2.


Subject(s)
COP9 Signalosome Complex , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , COP9 Signalosome Complex/metabolism , COP9 Signalosome Complex/genetics , Germ Cells/metabolism , Oogenesis/genetics , Protein Stability , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Stem Cells/metabolism , Stem Cells/cytology
7.
Nature ; 623(7986): 406-414, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37914938

ABSTRACT

Establishing how neural function emerges from network properties is a fundamental problem in neuroscience1. Here, to better understand the relationship between the structure and the function of a nervous system, we systematically measure signal propagation in 23,433 pairs of neurons across the head of the nematode Caenorhabditis elegans by direct optogenetic activation and simultaneous whole-brain calcium imaging. We measure the sign (excitatory or inhibitory), strength, temporal properties and causal direction of signal propagation between these neurons to create a functional atlas. We find that signal propagation differs from model predictions that are based on anatomy. Using mutants, we show that extrasynaptic signalling not visible from anatomy contributes to this difference. We identify many instances of dense-core-vesicle-dependent signalling, including on timescales of less than a second, that evoke acute calcium transients-often where no direct wired connection exists but where relevant neuropeptides and receptors are expressed. We propose that, in such cases, extrasynaptically released neuropeptides serve a similar function to that of classical neurotransmitters. Finally, our measured signal propagation atlas better predicts the neural dynamics of spontaneous activity than do models based on anatomy. We conclude that both synaptic and extrasynaptic signalling drive neural dynamics on short timescales, and that measurements of evoked signal propagation are crucial for interpreting neural function.


Subject(s)
Caenorhabditis elegans , Neural Pathways , Neurons , Animals , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Calcium/analysis , Calcium/metabolism , Models, Neurological , Mutation , Neural Pathways/physiology , Neurons/metabolism , Neurons/physiology , Neuropeptides/metabolism , Synapses/metabolism , Signal Transduction/physiology
9.
Cell ; 186(20): 4310-4324.e23, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37703874

ABSTRACT

Cellular homeostasis requires the robust control of biomolecule concentrations, but how do millions of mRNAs coordinate their stoichiometries in the face of dynamic translational changes? Here, we identified a two-tiered mechanism controlling mRNA:mRNA and mRNA:protein stoichiometries where mRNAs super-assemble into condensates with buffering capacity and sorting selectivity through phase-transition mechanisms. Using C. elegans oogenesis arrest as a model, we investigated the transcriptome cytosolic reorganization through the sequencing of RNA super-assemblies coupled with single mRNA imaging. Tightly repressed mRNAs self-assembled into same-sequence nanoclusters that further co-assembled into multiphase condensates. mRNA self-sorting was concentration dependent, providing a self-buffering mechanism that is selective to sequence identity and controls mRNA:mRNA stoichiometries. The cooperative sharing of limiting translation repressors between clustered mRNAs prevented the disruption of mRNA:repressor stoichiometries in the cytosol. Robust control of mRNA:mRNA and mRNA:protein stoichiometries emerges from mRNA self-demixing and cooperative super-assembly into multiphase multiscale condensates with dynamic storage capacity.


Subject(s)
Biomolecular Condensates , Caenorhabditis elegans , RNA, Messenger , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Oogenesis , Protein Biosynthesis , RNA Transport , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Proteins/chemistry , Proteins/metabolism , Biomolecular Condensates/chemistry , Biomolecular Condensates/metabolism
10.
Cell Rep ; 42(8): 112902, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37531250

ABSTRACT

Aging is characterized by a global decline in physiological function. However, by constructing a complete single-cell gene expression atlas, we find that Caenorhabditis elegans aging is not random in nature but instead is characterized by coordinated changes in functionally related metabolic, proteostasis, and stress-response genes in a cell-type-specific fashion, with downregulation of energy metabolism being the only nearly universal change. Similarly, the rates at which cells age differ significantly between cell types. In some cell types, aging is characterized by an increase in cell-to-cell variance, whereas in others, variance actually decreases. Remarkably, multiple resilience-enhancing transcription factors known to extend lifespan are activated across many cell types with age; we discovered new longevity candidates, such as GEI-3, among these. Together, our findings suggest that cells do not age passively but instead react strongly, and individualistically, to events that occur during aging. This atlas can be queried through a public interface.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Gene Expression Regulation, Developmental , Transcription Factors , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Aging , Cellular Senescence , Energy Metabolism , Longevity , Transcription Factors/genetics , Transcription Factors/metabolism , Homeostasis , Single-Cell Gene Expression Analysis , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Physiological Phenomena
11.
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
12.
J Biol Chem ; 299(6): 104799, 2023 06.
Article in English | MEDLINE | ID: mdl-37164154

ABSTRACT

The human AdipoR2 and its Caenorhabditis elegans homolog PAQR-2 are multipass plasma membrane proteins that protect cells against membrane rigidification. However, how AdipoR2 promotes membrane fluidity mechanistically is not clear. Using 13C-labeled fatty acids, we show that AdipoR2 can promote the elongation and incorporation of membrane-fluidizing polyunsaturated fatty acids into phospholipids. To elucidate the molecular basis of these activities, we performed immunoprecipitations of tagged AdipoR2 and PAQR-2 expressed in HEK293 cells or whole C. elegans, respectively, and identified coimmunoprecipitated proteins using mass spectrometry. We found that several of the evolutionarily conserved AdipoR2/PAQR-2 interactors are important for fatty acid elongation and incorporation into phospholipids. We experimentally verified some of these interactions, namely, with the dehydratase HACD3 that is essential for the third of four steps in long-chain fatty acid elongation and ACSL4 that is important for activation of unsaturated fatty acids and their channeling into phospholipids. We conclude that AdipoR2 and PAQR-2 can recruit protein interactors to promote the production and incorporation of unsaturated fatty acids into phospholipids.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Membrane , Fatty Acids , Membrane Fluidity , Receptors, Adiponectin , Animals , Humans , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Membrane/metabolism , Fatty Acids/metabolism , HEK293 Cells , Membrane Fluidity/physiology , Phospholipids/metabolism , Receptors, Adiponectin/metabolism , Protein Binding
13.
J Cell Biol ; 222(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36219157

ABSTRACT

Cytokinesis requires the constriction of an actomyosin-based contractile ring and involves multiple F-actin crosslinkers. We show that partial depletion of the C. elegans cytokinetic formin generates contractile rings with low F-actin levels that constrict but are structurally fragile, and we use this background to investigate the roles of the crosslinkers plastin/PLST-1 and ß-heavy-spectrin/SMA-1 during ring constriction. We show that the removal of PLST-1 or SMA-1 has opposite effects on the structural integrity of fragile rings. PLST-1 loss reduces cortical tension that resists ring constriction and makes fragile rings less prone to ruptures and regressions, whereas SMA-1 loss exacerbates structural defects, leading to frequent ruptures and cytokinesis failure. Fragile rings without SMA-1 or containing a shorter SMA-1, repeatedly rupture at the same site, and SMA-1::GFP accumulates at repair sites in fragile rings and in rings cut by laser microsurgery. These results establish that ß-heavy-spectrin stabilizes the constricting ring and reveals the importance of ß-heavy-spectrin size for network connectivity at low F-actin density.


Subject(s)
Actin Cytoskeleton , Cytokinesis , Spectrin , Actins , Actomyosin , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/metabolism , Formins , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Spectrin/metabolism
14.
Nature ; 610(7933): 768-774, 2022 10.
Article in English | MEDLINE | ID: mdl-36261532

ABSTRACT

Haem is an iron-containing tetrapyrrole that is critical for a variety of cellular and physiological processes1-3. Haem binding proteins are present in almost all cellular compartments, but the molecular mechanisms that regulate the transport and use of haem within the cell remain poorly understood2,3. Here we show that haem-responsive gene 9 (HRG-9) (also known as transport and Golgi organization 2 (TANGO2)) is an evolutionarily conserved haem chaperone with a crucial role in trafficking haem out of haem storage or synthesis sites in eukaryotic cells. Loss of Caenorhabditis elegans hrg-9 and its paralogue hrg-10 results in the accumulation of haem in lysosome-related organelles, the haem storage site in worms. Similarly, deletion of the hrg-9 homologue TANGO2 in yeast and mammalian cells induces haem overload in mitochondria, the site of haem synthesis. We demonstrate that TANGO2 binds haem and transfers it from cellular membranes to apo-haemoproteins. Notably, homozygous tango2-/- zebrafish larvae develop pleiotropic symptoms including encephalopathy, cardiac arrhythmia and myopathy, and die during early development. These defects partially resemble the symptoms of human TANGO2-related metabolic encephalopathy and arrhythmias, a hereditary disease caused by mutations in TANGO24-8. Thus, the identification of HRG-9 as an intracellular haem chaperone provides a biological basis for exploring the aetiology and treatment of TANGO2-related disorders.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Heme , Animals , Humans , Arrhythmias, Cardiac/metabolism , Brain Diseases/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Heme/metabolism , Mitochondria/metabolism , Molecular Chaperones/metabolism , Zebrafish/genetics , Zebrafish/metabolism
15.
Proc Natl Acad Sci U S A ; 119(37): e2206817119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36067313

ABSTRACT

The classification of neurons into distinct types reveals hierarchical taxonomic relationships that reflect the extent of similarity between neuronal cell types. At the base of such taxonomies are neuronal cells that are very similar to one another but differ in a small number of reproducible and select features. How are very similar members of a neuron class that share many features instructed to diversify into distinct subclasses? We show here that the six very similar members of the Caenorhabditis elegans IL2 sensory neuron class, which are all specified by a homeobox terminal selector, unc-86/BRN3, differentiate into two subtly distinct subclasses, a dorsoventral subclass and a lateral subclass, by the toggle switch-like action of the sine oculis/SIX homeobox gene unc-39. unc-39 is expressed only in the lateral IL2 neurons, and loss of unc-39 leads to a homeotic transformation of the lateral into the dorsoventral class; conversely, ectopic unc-39 expression converts the dorsoventral subclass into the lateral subclass. Hence, a terminal selector homeobox gene controls both class- as well as subclass-specific features, while a subordinate homeobox gene determines the ability of the class-specific homeobox gene to activate subtype-specific target genes. We find a similar regulatory mechanism operating in a distinct class of six motor neurons. Our findings underscore the importance of homeobox genes in neuronal identity control and invite speculations about homeotic identity transformations as potential drivers of evolutionary novelty during cell-type evolution in the brain.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Genes, Homeobox , Homeodomain Proteins , Sensory Receptor Cells , Transcription Factors , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Motor Neurons/classification , Motor Neurons/cytology , Sensory Receptor Cells/classification , Sensory Receptor Cells/cytology , Transcription Factors/genetics , Transcription Factors/physiology
16.
J Cell Biol ; 221(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-36121422

ABSTRACT

How cells spatially organize their plasma membrane, cytoskeleton, and cytoplasm remains a central question for cell biologists. In this issue of JCB, Calvi et al. (2022. J. Cell Biol.https://doi.org/10.1083/jcb.202201048) identify PP1 phosphatases as key regulators of C. elegans anterior-posterior polarity, by counterbalancing aPKC-mediated phosphorylation of PAR-2.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Polarity , Phosphoprotein Phosphatases , Protein Kinase C , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Caenorhabditis elegans Proteins/metabolism , Cell Membrane , Cytoplasm , Microtubules/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinase C/metabolism
17.
Development ; 149(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35394007

ABSTRACT

A long-standing biological question is how DNA cis-regulatory elements shape transcriptional patterns during metazoan development. Reporter constructs, cell culture assays and computational modeling have made major contributions to answering this question, but analysis of elements in their natural context is an important complement. Here, we mutate Notch-dependent LAG-1 binding sites (LBSs) in the endogenous Caenorhabditis elegans sygl-1 gene, which encodes a key stem cell regulator, and analyze the consequences on sygl-1 expression (nascent transcripts, mRNA, protein) and stem cell maintenance. Mutation of one LBS in a three-element cluster approximately halved both expression and stem cell pool size, whereas mutation of two LBSs essentially abolished them. Heterozygous LBS mutant clusters provided intermediate values. Our results lead to two major conclusions. First, both LBS number and configuration impact cluster activity: LBSs act additively in trans and synergistically in cis. Second, the SYGL-1 gradient promotes self-renewal above its functional threshold and triggers differentiation below the threshold. Our approach of coupling CRISPR/Cas9 LBS mutations with effects on both molecular and biological readouts establishes a powerful model for in vivo analyses of DNA cis-regulatory elements.


Subject(s)
Caenorhabditis elegans , Regulatory Elements, Transcriptional , Stem Cells , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Cell Self Renewal , DNA/metabolism , DNA-Binding Proteins/genetics , Receptors, Notch , Stem Cells/cytology
18.
Opt Express ; 30(2): 1546-1554, 2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35209312

ABSTRACT

Deep-brain microscopy is strongly limited by the size of the imaging probe, both in terms of achievable resolution and potential trauma due to surgery. Here, we show that a segment of an ultra-thin multi-mode fiber (cannula) can replace the bulky microscope objective inside the brain. By creating a self-consistent deep neural network that is trained to reconstruct anthropocentric images from the raw signal transported by the cannula, we demonstrate a single-cell resolution (< 10µm), depth sectioning resolution of 40 µm, and field of view of 200 µm, all with green-fluorescent-protein labelled neurons imaged at depths as large as 1.4 mm from the brain surface. Since ground-truth images at these depths are challenging to obtain in vivo, we propose a novel ensemble method that averages the reconstructed images from disparate deep-neural-network architectures. Finally, we demonstrate dynamic imaging of moving GCaMp-labelled C. elegans worms. Our approach dramatically simplifies deep-brain microscopy.


Subject(s)
Brain/diagnostic imaging , Machine Learning , Microscopy, Fluorescence/methods , Neuroimaging/methods , Animals , Caenorhabditis elegans/cytology , Cells, Cultured , Green Fluorescent Proteins/metabolism , Image Processing, Computer-Assisted/methods , Mice , Minimally Invasive Surgical Procedures , Neural Networks, Computer , Neurons/cytology , Neurons/metabolism
19.
Elife ; 112022 02 04.
Article in English | MEDLINE | ID: mdl-35119366

ABSTRACT

Cell identity is characterized by a distinct combination of gene expression, cell morphology, and cellular function established as progenitor cells divide and differentiate. Following establishment, cell identities can be unstable and require active and continuous maintenance throughout the remaining life of a cell. Mechanisms underlying the maintenance of cell identities are incompletely understood. Here, we show that the gene ctbp-1, which encodes the transcriptional corepressor C-terminal binding protein-1 (CTBP-1), is essential for the maintenance of the identities of the two AIA interneurons in the nematode Caenorhabditis elegans. ctbp-1 is not required for the establishment of the AIA cell fate but rather functions cell-autonomously and can act in later larval stage and adult worms to maintain proper AIA gene expression, morphology and function. From a screen for suppressors of the ctbp-1 mutant phenotype, we identified the gene egl-13, which encodes a SOX family transcription factor. We found that egl-13 regulates AIA function and aspects of AIA gene expression, but not AIA morphology. We conclude that the CTBP-1 protein maintains AIA cell identity in part by utilizing EGL-13 to repress transcriptional activity in the AIAs. More generally, we propose that transcriptional corepressors like CTBP-1 might be critical factors in the maintenance of cell identities, harnessing the DNA-binding specificity of transcription factors like EGL-13 to selectively regulate gene expression in a cell-specific manner.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Interneurons/cytology , Repressor Proteins/metabolism , SOXD Transcription Factors/metabolism , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Gene Expression Regulation, Developmental , Interneurons/physiology , Repressor Proteins/genetics , SOXD Transcription Factors/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
20.
PLoS Biol ; 20(2): e3001317, 2022 02.
Article in English | MEDLINE | ID: mdl-35192608

ABSTRACT

Cell invasion is an initiating event during tumor cell metastasis and an essential process during development. A screen of C. elegans orthologs of genes overexpressed in invasive human melanoma cells has identified several components of the conserved DNA pre-replication complex (pre-RC) as positive regulators of anchor cell (AC) invasion. The pre-RC genes function cell-autonomously in the G1-arrested AC to promote invasion, independently of their role in licensing DNA replication origins in proliferating cells. While the helicase activity of the pre-RC is necessary for AC invasion, the downstream acting DNA replication initiation factors are not required. The pre-RC promotes the invasive fate by regulating the expression of extracellular matrix genes and components of the PI3K signaling pathway. Increasing PI3K pathway activity partially suppressed the AC invasion defects caused by pre-RC depletion, suggesting that the PI3K pathway is one critical pre-RC target. We propose that the pre-RC, or a part of it, acts in the postmitotic AC as a transcriptional regulator that facilitates the switch to an invasive phenotype.


Subject(s)
Caenorhabditis elegans/genetics , Cell Cycle/genetics , Cell Movement/genetics , DNA Replication/genetics , Replication Origin/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Female , Gene Expression Profiling/methods , Gene Ontology , Larva/cytology , Larva/genetics , Larva/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Vulva/cytology , Vulva/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...