Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.089
Filter
Add more filters

Publication year range
1.
Cell ; 184(4): 899-911.e13, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33545089

ABSTRACT

Changes in appendage structure underlie key transitions in vertebrate evolution. Addition of skeletal elements along the proximal-distal axis facilitated critical transformations, including the fin-to-limb transition that permitted generation of diverse modes of locomotion. Here, we identify zebrafish mutants that form supernumerary long bones in their pectoral fins. These new bones integrate into musculature, form joints, and articulate with neighboring elements. This phenotype is caused by activating mutations in previously unrecognized regulators of appendage patterning, vav2 and waslb, that function in a common pathway. This pathway is required for appendage development across vertebrates, and loss of Wasl in mice causes defects similar to those seen in murine Hox mutants. Concordantly, formation of supernumerary bones requires Hox11 function, and mutations in the vav2/wasl pathway drive enhanced expression of hoxa11b, indicating developmental homology with the forearm. Our findings reveal a latent, limb-like pattern ability in fins that is activated by simple genetic perturbation.


Subject(s)
Bone and Bones/embryology , Extremities/embryology , Zebrafish/embryology , Actins/metabolism , Animal Fins/embryology , Animals , Base Sequence , Body Patterning , CRISPR-Cas Systems/genetics , Cell Lineage , Epistasis, Genetic , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Genes, Reporter , HeLa Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mutation/genetics , Phenotype , Phylogeny , Signal Transduction/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
2.
Cell ; 184(26): 6313-6325.e18, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34942099

ABSTRACT

How tissues acquire complex shapes is a fundamental question in biology and regenerative medicine. Zebrafish semicircular canals form from invaginations in the otic epithelium (buds) that extend and fuse to form the hubs of each canal. We find that conventional actomyosin-driven behaviors are not required. Instead, local secretion of hyaluronan, made by the enzymes uridine 5'-diphosphate dehydrogenase (ugdh) and hyaluronan synthase 3 (has3), drives canal morphogenesis. Charged hyaluronate polymers osmotically swell with water and generate isotropic extracellular pressure to deform the overlying epithelium into buds. The mechanical anisotropy needed to shape buds into tubes is conferred by a polarized distribution of actomyosin and E-cadherin-rich membrane tethers, which we term cytocinches. Most work on tissue morphogenesis ascribes actomyosin contractility as the driving force, while the extracellular matrix shapes tissues through differential stiffness. Our work inverts this expectation. Hyaluronate pressure shaped by anisotropic tissue stiffness may be a widespread mechanism for powering morphological change in organogenesis and tissue engineering.


Subject(s)
Extracellular Space/chemistry , Hyaluronic Acid/pharmacology , Morphogenesis , Organ Specificity , Pressure , Semicircular Canals/cytology , Semicircular Canals/embryology , Actomyosin/metabolism , Animals , Anisotropy , Behavior, Animal , Extracellular Matrix/metabolism , Hyaluronic Acid/biosynthesis , Models, Biological , Morphogenesis/drug effects , Organ Specificity/drug effects , Osmotic Pressure , Semicircular Canals/diagnostic imaging , Stereotyped Behavior , Zebrafish/embryology , Zebrafish Proteins/metabolism
3.
Cell ; 179(1): 74-89.e10, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31495570

ABSTRACT

During neural tube closure and spinal cord development, many cells die in both the central and peripheral nervous systems (CNS and PNS, respectively). However, myeloid-derived professional phagocytes have not yet colonized the trunk region during early neurogenesis. How apoptotic cells are removed from this region during these stages remains largely unknown. Using live imaging in zebrafish, we demonstrate that neural crest cells (NCCs) respond rapidly to dying cells and phagocytose cellular debris around the neural tube. Additionally, NCCs have the ability to enter the CNS through motor exit point transition zones and clear debris in the spinal cord. Surprisingly, NCCs phagocytosis mechanistically resembles macrophage phagocytosis and their recruitment toward cellular debris is mediated by interleukin-1ß. Taken together, our results reveal a role for NCCs in phagocytosis of debris in the developing nervous system before the presence of professional phagocytes.


Subject(s)
Cell Movement/physiology , Neural Crest/physiology , Neurogenesis/physiology , Peripheral Nervous System/growth & development , Phagocytosis/physiology , Spinal Cord/growth & development , Animals , Animals, Genetically Modified , Apoptosis/physiology , Cell Differentiation/physiology , Interleukin-1beta/metabolism , Phagocytes/physiology , Phagosomes/physiology , Zebrafish/embryology
4.
Cell ; 177(6): 1463-1479.e18, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31080065

ABSTRACT

Segregation of maternal determinants within the oocyte constitutes the first step in embryo patterning. In zebrafish oocytes, extensive ooplasmic streaming leads to the segregation of ooplasm from yolk granules along the animal-vegetal axis of the oocyte. Here, we show that this process does not rely on cortical actin reorganization, as previously thought, but instead on a cell-cycle-dependent bulk actin polymerization wave traveling from the animal to the vegetal pole of the oocyte. This wave functions in segregation by both pulling ooplasm animally and pushing yolk granules vegetally. Using biophysical experimentation and theory, we show that ooplasm pulling is mediated by bulk actin network flows exerting friction forces on the ooplasm, while yolk granule pushing is achieved by a mechanism closely resembling actin comet formation on yolk granules. Our study defines a novel role of cell-cycle-controlled bulk actin polymerization waves in oocyte polarization via ooplasmic segregation.


Subject(s)
Actins/metabolism , Cell Cycle/physiology , Oocytes/metabolism , Actins/physiology , Animals , Cell Polarity/physiology , Cytoplasm/metabolism , Egg Yolk/physiology , Polymerization , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zygote
5.
Cell ; 160(4): 581-582, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679757

ABSTRACT

Migrating cells exhibit distinct motility modes and can switch between modes based on chemical or physical cues. Liu et al. and Ruprecht et al. now describe how confinement and contractility influence motility mode plasticity and instigate a mode termed stable bleb migration in embryonic and tumor cells.


Subject(s)
Cell Movement , Embryo, Nonmammalian/cytology , Gastrula/cytology , Mesoderm/cytology , Stem Cells/cytology , Zebrafish/embryology , Animals , Humans
6.
Cell ; 160(4): 673-685, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679761

ABSTRACT

3D amoeboid cell migration is central to many developmental and disease-related processes such as cancer metastasis. Here, we identify a unique prototypic amoeboid cell migration mode in early zebrafish embryos, termed stable-bleb migration. Stable-bleb cells display an invariant polarized balloon-like shape with exceptional migration speed and persistence. Progenitor cells can be reversibly transformed into stable-bleb cells irrespective of their primary fate and motile characteristics by increasing myosin II activity through biochemical or mechanical stimuli. Using a combination of theory and experiments, we show that, in stable-bleb cells, cortical contractility fluctuations trigger a stochastic switch into amoeboid motility, and a positive feedback between cortical flows and gradients in contractility maintains stable-bleb cell polarization. We further show that rearward cortical flows drive stable-bleb cell migration in various adhesive and non-adhesive environments, unraveling a highly versatile amoeboid migration phenotype.


Subject(s)
Cell Movement , Embryo, Nonmammalian/cytology , Gastrula/cytology , Stem Cells/cytology , Zebrafish/embryology , Animals , Cell Adhesion , Cell Polarity
7.
Cell ; 160(1-2): 241-52, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25594182

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) can reconstitute and sustain the entire blood system. We generated a highly specific transgenic reporter of HSPCs in zebrafish. This allowed us to perform high-resolution live imaging on endogenous HSPCs not currently possible in mammalian bone marrow. Using this system, we have uncovered distinct interactions between single HSPCs and their niche. When an HSPC arrives in the perivascular niche, a group of endothelial cells remodel to form a surrounding pocket. This structure appears conserved in mouse fetal liver. Correlative light and electron microscopy revealed that endothelial cells surround a single HSPC attached to a single mesenchymal stromal cell. Live imaging showed that mesenchymal stromal cells anchor HSPCs and orient their divisions. A chemical genetic screen found that the compound lycorine promotes HSPC-niche interactions during development and ultimately expands the stem cell pool into adulthood. Our studies provide evidence for dynamic niche interactions upon stem cell colonization. PAPERFLICK:


Subject(s)
Endothelium/physiology , Hematopoietic Stem Cells/cytology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Division , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/physiology , Endothelium/cytology , Hematopoietic Stem Cells/physiology , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Stem Cell Niche , Stromal Cells/cytology , Stromal Cells/metabolism , Zebrafish/physiology
8.
Nature ; 625(7993): 126-133, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38123680

ABSTRACT

Chemical signalling is the primary means by which cells communicate in the embryo. The underlying principle refers to a group of ligand-producing cells and a group of cells that respond to this signal because they express the appropriate receptors1,2. In the zebrafish embryo, Wnt5b binds to the receptor Ror2 to trigger the Wnt-planar cell polarity (PCP) signalling pathway to regulate tissue polarity and cell migration3,4. However, it remains unclear how this lipophilic ligand is transported from the source cells through the aqueous extracellular space to the target tissue. In this study, we provide evidence that Wnt5b, together with Ror2, is loaded on long protrusions called cytonemes. Our data further suggest that the active Wnt5b-Ror2 complexes form in the producing cell and are handed over from these cytonemes to the receiving cell. Then, the receiving cell has the capacity to initiate Wnt-PCP signalling, irrespective of its functional Ror2 receptor status. On the tissue level, we further show that cytoneme-dependent spreading of active Wnt5b-Ror2 affects convergence and extension in the zebrafish gastrula. We suggest that cytoneme-mediated transfer of ligand-receptor complexes is a vital mechanism for paracrine signalling. This may prompt a reevaluation of the conventional concept of characterizing responsive and non-responsive tissues solely on the basis of the expression of receptors.


Subject(s)
Pseudopodia , Receptor Tyrosine Kinase-like Orphan Receptors , Wnt Proteins , Zebrafish , Animals , Gastrula/cytology , Gastrula/embryology , Gastrula/metabolism , Ligands , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Zebrafish/embryology , Zebrafish/metabolism , Cell Polarity , Cell Movement , Pseudopodia/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Paracrine Communication
9.
Nature ; 628(8007): 408-415, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38480883

ABSTRACT

During development, inflammation or tissue injury, macrophages may successively engulf and process multiple apoptotic corpses via efferocytosis to achieve tissue homeostasis1. How macrophages may rapidly adapt their transcription to achieve continuous corpse uptake is incompletely understood. Transcriptional pause/release is an evolutionarily conserved mechanism, in which RNA polymerase (Pol) II initiates transcription for 20-60 nucleotides, is paused for minutes to hours and is then released to make full-length mRNA2. Here we show that macrophages, within minutes of corpse encounter, use transcriptional pause/release to unleash a rapid transcriptional response. For human and mouse macrophages, the Pol II pause/release was required for continuous efferocytosis in vitro and in vivo. Interestingly, blocking Pol II pause/release did not impede Fc receptor-mediated phagocytosis, yeast uptake or bacterial phagocytosis. Integration of data from three genomic approaches-precision nuclear run-on sequencing, RNA sequencing, and assay for transposase-accessible chromatin using sequencing (ATAC-seq)-on efferocytic macrophages at different time points revealed that Pol II pause/release controls expression of select transcription factors and downstream target genes. Mechanistic studies on transcription factor EGR3, prominently regulated by pause/release, uncovered EGR3-related reprogramming of other macrophage genes involved in cytoskeleton and corpse processing. Using lysosomal probes and a new genetic fluorescent reporter, we identify a role for pause/release in phagosome acidification during efferocytosis. Furthermore, microglia from egr3-deficient zebrafish embryos displayed reduced phagocytosis of apoptotic neurons and fewer maturing phagosomes, supporting defective corpse processing. Collectively, these data indicate that macrophages use Pol II pause/release as a mechanism to rapidly alter their transcriptional programs for efficient processing of the ingested apoptotic corpses and for successive efferocytosis.


Subject(s)
Efferocytosis , Macrophages , RNA Polymerase II , Transcription Elongation, Genetic , Animals , Humans , Male , Mice , Apoptosis , Cytoskeleton/metabolism , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Efferocytosis/genetics , Hydrogen-Ion Concentration , Macrophages/immunology , Macrophages/metabolism , Neurons/metabolism , Phagosomes/metabolism , RNA Polymerase II/metabolism , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/genetics , Time Factors
10.
Cell ; 159(2): 415-27, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303534

ABSTRACT

Epithelial cells acquire functionally important shapes (e.g., squamous, cuboidal, columnar) during development. Here, we combine theory, quantitative imaging, and perturbations to analyze how tissue geometry, cell divisions, and mechanics interact to shape the presumptive enveloping layer (pre-EVL) on the zebrafish embryonic surface. We find that, under geometrical constraints, pre-EVL flattening is regulated by surface cell number changes following differentially oriented cell divisions. The division pattern is, in turn, determined by the cell shape distribution, which forms under geometrical constraints by cell-cell mechanical coupling. An integrated mathematical model of this shape-division feedback loop recapitulates empirical observations. Surprisingly, the model predicts that cell shape is robust to changes of tissue surface area, cell volume, and cell number, which we confirm in vivo. Further simulations and perturbations suggest the parameter linking cell shape and division orientation contributes to epithelial diversity. Together, our work identifies an evolvable design logic that enables robust cell-level regulation of tissue-level development.


Subject(s)
Epithelial Cells/cytology , Models, Biological , Morphogenesis , Zebrafish/embryology , Animals , Biomechanical Phenomena , Cell Count , Cell Division , Cell Shape , Embryo, Nonmammalian/cytology
11.
Cell ; 159(3): 662-75, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417113

ABSTRACT

Advancing our understanding of embryonic development is heavily dependent on identification of novel pathways or regulators. Although genome-wide techniques such as RNA sequencing are ideally suited for discovering novel candidate genes, they are unable to yield spatially resolved information in embryos or tissues. Microscopy-based approaches, using in situ hybridization, for example, can provide spatial information about gene expression, but are limited to analyzing one or a few genes at a time. Here, we present a method where we combine traditional histological techniques with low-input RNA sequencing and mathematical image reconstruction to generate a high-resolution genome-wide 3D atlas of gene expression in the zebrafish embryo at three developmental stages. Importantly, our technique enables searching for genes that are expressed in specific spatial patterns without manual image annotation. We envision broad applicability of RNA tomography as an accurate and sensitive approach for spatially resolved transcriptomics in whole embryos and dissected organs.


Subject(s)
Embryo, Nonmammalian/metabolism , Gene Expression Profiling , Sequence Analysis, RNA , Tomography/methods , Zebrafish/embryology , Animals , Imaging, Three-Dimensional
12.
Cell ; 159(7): 1698-710, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25497548

ABSTRACT

Cells control dynamic transitions in transcript levels by regulating transcription, processing, and/or degradation through an integrated regulatory strategy. Here, we combine RNA metabolic labeling, rRNA-depleted RNA-seq, and DRiLL, a novel computational framework, to quantify the level; editing sites; and transcription, processing, and degradation rates of each transcript at a splice junction resolution during the LPS response of mouse dendritic cells. Four key regulatory strategies, dominated by RNA transcription changes, generate most temporal gene expression patterns. Noncanonical strategies that also employ dynamic posttranscriptional regulation control only a minority of genes, but provide unique signal processing features. We validate Tristetraprolin (TTP) as a major regulator of RNA degradation in one noncanonical strategy. Applying DRiLL to the regulation of noncoding RNAs and to zebrafish embryogenesis demonstrates its broad utility. Our study provides a new quantitative approach to discover transcriptional and posttranscriptional events that control dynamic changes in transcript levels using RNA sequencing data.


Subject(s)
Computer Simulation , Dendritic Cells/metabolism , Sequence Analysis, RNA/methods , Animals , Gene Expression Profiling/methods , Kinetics , Lipopolysaccharides/metabolism , Mice , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Untranslated/metabolism , Transcription, Genetic , Tristetraprolin/metabolism , Zebrafish/embryology
13.
Cell ; 159(3): 543-57, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417106

ABSTRACT

The transcriptional activation of one out of ?2800 olfactory receptor (OR) alleles is a poorly understood process. Here, we identify a plethora of putative OR enhancers and study their in vivo activity in olfactory neurons. Distinguished by an unusual epigenetic signature, candidate OR enhancers are characterized by extensive interchromosomal interactions associated with OR transcription and share a similar pattern of transcription factor footprints. In particular, we establish the role of the transcription factor Bptf as a facilitator of both enhancer interactions and OR transcription. Our observations agree with the model whereby OR transcription occurs in the context of multiple interacting enhancers. Disruption of these interchromosomal interactions results in weak and multigenic OR expression, suggesting that the rare coincidence of numerous enhancers over a stochastically chosen OR may account for the singularity and robustness in OR transcription.


Subject(s)
Enhancer Elements, Genetic , Receptors, Odorant/genetics , Transcriptional Activation , Animals , Animals, Genetically Modified , Antigens, Nuclear/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nucleoproteins/metabolism , Olfactory Receptor Neurons/metabolism , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/metabolism
14.
Nature ; 622(7981): 149-155, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37758945

ABSTRACT

A regular heartbeat is essential to vertebrate life. In the mature heart, this function is driven by an anatomically localized pacemaker. By contrast, pacemaking capability is broadly distributed in the early embryonic heart1-3, raising the question of how tissue-scale activity is first established and then maintained during embryonic development. The initial transition of the heart from silent to beating has never been characterized at the timescale of individual electrical events, and the structure in space and time of the early heartbeats remains poorly understood. Using all-optical electrophysiology, we captured the very first heartbeat of a zebrafish and analysed the development of cardiac excitability and conduction around this singular event. The first few beats appeared suddenly, had irregular interbeat intervals, propagated coherently across the primordial heart and emanated from loci that varied between animals and over time. The bioelectrical dynamics were well described by a noisy saddle-node on invariant circle bifurcation with action potential upstroke driven by CaV1.2. Our work shows how gradual and largely asynchronous development of single-cell bioelectrical properties produces a stereotyped and robust tissue-scale transition from quiescence to coordinated beating.


Subject(s)
Embryonic Development , Heart Rate , Heart , Zebrafish , Animals , Action Potentials , Heart/embryology , Heart/innervation , Heart/physiology , Heart Rate/physiology , Zebrafish/embryology , Zebrafish/physiology , Electrophysiology , Single-Cell Analysis
15.
Nature ; 623(7988): 782-791, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968389

ABSTRACT

The maturation of single-cell transcriptomic technologies has facilitated the generation of comprehensive cellular atlases from whole embryos1-4. A majority of these data, however, has been collected from wild-type embryos without an appreciation for the latent variation that is present in development. Here we present the 'zebrafish single-cell atlas of perturbed embryos': single-cell transcriptomic data from 1,812 individually resolved developing zebrafish embryos, encompassing 19 timepoints, 23 genetic perturbations and a total of 3.2 million cells. The high degree of replication in our study (eight or more embryos per condition) enables us to estimate the variance in cell type abundance organism-wide and to detect perturbation-dependent deviance in cell type composition relative to wild-type embryos. Our approach is sensitive to rare cell types, resolving developmental trajectories and genetic dependencies in the cranial ganglia neurons, a cell population that comprises less than 1% of the embryo. Additionally, time-series profiling of individual mutants identified a group of brachyury-independent cells with strikingly similar transcriptomes to notochord sheath cells, leading to new hypotheses about early origins of the skull. We anticipate that standardized collection of high-resolution, organism-scale single-cell data from large numbers of individual embryos will enable mapping of the genetic dependencies of zebrafish cell types, while also addressing longstanding challenges in developmental genetics, including the cellular and transcriptional plasticity underlying phenotypic diversity across individuals.


Subject(s)
Embryo, Mammalian , Reverse Genetics , Single-Cell Analysis , Zebrafish , Animals , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Reverse Genetics/methods , Transcriptome/genetics , Zebrafish/embryology , Zebrafish/genetics , Mutation , Single-Cell Analysis/methods , Notochord/cytology , Notochord/embryology
16.
Nature ; 620(7974): 615-624, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37558872

ABSTRACT

The concomitant occurrence of tissue growth and organization is a hallmark of organismal development1-3. This often means that proliferating and differentiating cells are found at the same time in a continuously changing tissue environment. How cells adapt to architectural changes to prevent spatial interference remains unclear. Here, to understand how cell movements that are key for growth and organization are orchestrated, we study the emergence of photoreceptor neurons that occur during the peak of retinal growth, using zebrafish, human tissue and human organoids. Quantitative imaging reveals that successful retinal morphogenesis depends on the active bidirectional translocation of photoreceptors, leading to a transient transfer of the entire cell population away from the apical proliferative zone. This pattern of migration is driven by cytoskeletal machineries that differ depending on the direction: microtubules are exclusively required for basal translocation, whereas actomyosin is involved in apical movement. Blocking the basal translocation of photoreceptors induces apical congestion, which hampers the apical divisions of progenitor cells and leads to secondary defects in lamination. Thus, photoreceptor migration is crucial to prevent competition for space, and to allow concurrent tissue growth and lamination. This shows that neuronal migration, in addition to its canonical role in cell positioning4, can be involved in coordinating morphogenesis.


Subject(s)
Cell Movement , Morphogenesis , Photoreceptor Cells , Retina , Animals , Humans , Actomyosin/metabolism , Cell Competition , Cell Differentiation , Cell Movement/physiology , Cell Proliferation , Microtubules/metabolism , Morphogenesis/physiology , Organoids/cytology , Organoids/embryology , Photoreceptor Cells/cytology , Photoreceptor Cells/physiology , Retina/cytology , Retina/embryology , Zebrafish/embryology
17.
Nature ; 613(7945): 712-720, 2023 01.
Article in English | MEDLINE | ID: mdl-36653451

ABSTRACT

Ribosomes are produced in large quantities during oogenesis and are stored in the egg. However, the egg and early embryo are translationally repressed1-4. Here, using mass spectrometry and cryo-electron microscopy analyses of ribosomes isolated from zebrafish (Danio rerio) and Xenopus laevis eggs and embryos, we provide molecular evidence that ribosomes transition from a dormant state to an active state during the first hours of embryogenesis. Dormant ribosomes are associated with four conserved factors that form two modules, consisting of Habp4-eEF2 and death associated protein 1b (Dap1b) or Dap in complex with eIF5a. Both modules occupy functionally important sites and act together to stabilize ribosomes and repress translation. Dap1b (also known as Dapl1 in mammals) is a newly discovered translational inhibitor that stably inserts into the polypeptide exit tunnel. Addition of recombinant zebrafish Dap1b protein is sufficient to block translation and reconstitute the dormant egg ribosome state in a mammalian translation extract in vitro. Thus, a developmentally programmed, conserved ribosome state has a key role in ribosome storage and translational repression in the egg.


Subject(s)
Conserved Sequence , Evolution, Molecular , Ovum , Protein Biosynthesis , Ribosomes , Xenopus Proteins , Zebrafish Proteins , Animals , Cryoelectron Microscopy/methods , Peptides/metabolism , Ribosomes/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Mass Spectrometry , Xenopus laevis/embryology , Ovum/metabolism , Embryonic Structures , Embryonic Development , Female , Eukaryotic Translation Initiation Factor 5A
18.
Nature ; 618(7965): 543-549, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225983

ABSTRACT

The development of paired appendages was a key innovation during evolution and facilitated the aquatic to terrestrial transition of vertebrates. Largely derived from the lateral plate mesoderm (LPM), one hypothesis for the evolution of paired fins invokes derivation from unpaired median fins via a pair of lateral fin folds located between pectoral and pelvic fin territories1. Whilst unpaired and paired fins exhibit similar structural and molecular characteristics, no definitive evidence exists for paired lateral fin folds in larvae or adults of any extant or extinct species. As unpaired fin core components are regarded as exclusively derived from paraxial mesoderm, any transition presumes both co-option of a fin developmental programme to the LPM and bilateral duplication2. Here, we identify that the larval zebrafish unpaired pre-anal fin fold (PAFF) is derived from the LPM and thus may represent a developmental intermediate between median and paired fins. We trace the contribution of LPM to the PAFF in both cyclostomes and gnathostomes, supporting the notion that this is an ancient trait of vertebrates. Finally, we observe that the PAFF can be bifurcated by increasing bone morphogenetic protein signalling, generating LPM-derived paired fin folds. Our work provides evidence that lateral fin folds may have existed as embryonic anlage for elaboration to paired fins.


Subject(s)
Animal Fins , Biological Evolution , Mesoderm , Zebrafish , Animals , Animal Fins/anatomy & histology , Animal Fins/embryology , Animal Fins/growth & development , Larva/anatomy & histology , Larva/growth & development , Mesoderm/anatomy & histology , Mesoderm/embryology , Mesoderm/growth & development , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish/growth & development , Bone Morphogenetic Proteins/metabolism
19.
Nature ; 614(7949): 742-751, 2023 02.
Article in English | MEDLINE | ID: mdl-36755098

ABSTRACT

Cell identity is governed by the complex regulation of gene expression, represented as gene-regulatory networks1. Here we use gene-regulatory networks inferred from single-cell multi-omics data to perform in silico transcription factor perturbations, simulating the consequent changes in cell identity using only unperturbed wild-type data. We apply this machine-learning-based approach, CellOracle, to well-established paradigms-mouse and human haematopoiesis, and zebrafish embryogenesis-and we correctly model reported changes in phenotype that occur as a result of transcription factor perturbation. Through systematic in silico transcription factor perturbation in the developing zebrafish, we simulate and experimentally validate a previously unreported phenotype that results from the loss of noto, an established notochord regulator. Furthermore, we identify an axial mesoderm regulator, lhx1a. Together, these results show that CellOracle can be used to analyse the regulation of cell identity by transcription factors, and can provide mechanistic insights into development and differentiation.


Subject(s)
Cell Differentiation , Computer Simulation , Gene Regulatory Networks , Transcription Factors , Animals , Humans , Mice , Cell Differentiation/genetics , Embryonic Development/genetics , Phenotype , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics , Mesoderm/enzymology , Mesoderm/metabolism , Hematopoiesis/genetics
20.
Nature ; 613(7942): 153-159, 2023 01.
Article in English | MEDLINE | ID: mdl-36517597

ABSTRACT

Sequential segmentation creates modular body plans of diverse metazoan embryos1-4. Somitogenesis establishes the segmental pattern of the vertebrate body axis. A molecular segmentation clock in the presomitic mesoderm sets the pace of somite formation4. However, how cells are primed to form a segment boundary at a specific location remains unclear. Here we developed precise reporters for the clock and double-phosphorylated Erk (ppErk) gradient in zebrafish. We show that the Her1-Her7 oscillator drives segmental commitment by periodically lowering ppErk, therefore projecting its oscillation onto the ppErk gradient. Pulsatile inhibition of the ppErk gradient can fully substitute for the role of the clock, and kinematic clock waves are dispensable for sequential segmentation. The clock functions upstream of ppErk, which in turn enables neighbouring cells to discretely establish somite boundaries in zebrafish5. Molecularly divergent clocks and morphogen gradients were identified in sequentially segmenting species3,4,6-8. Our findings imply that versatile clocks may establish sequential segmentation in diverse species provided that they inhibit gradients.


Subject(s)
Body Patterning , Extracellular Signal-Regulated MAP Kinases , Periodicity , Somites , Zebrafish Proteins , Zebrafish , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Somites/drug effects , Somites/embryology , Somites/enzymology , Somites/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism , Biological Clocks , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL