107 results on '"Moens CB"'
Search Results
2. Position-independent functional refinement within the vagus motor topographic map.
- Author
-
Kaneko T, Boulanger-Weill J, Isabella AJ, and Moens CB
- Subjects
- Animals, Synaptic Transmission physiology, Axons physiology, Zebrafish physiology, Motor Neurons physiology, Vagus Nerve physiology
- Abstract
Motor neurons in the central nervous system often lie in a continuous topographic map, where neurons that innervate different body parts are spatially intermingled. This is the case for the efferent neurons of the vagus nerve, which innervate diverse muscle and organ targets in the head and viscera for brain-body communication. It remains elusive how neighboring motor neurons with different fixed peripheral axon targets develop the separate somatodendritic (input) connectivity they need to generate spatially precise body control. Here, we show that vagus motor neurons in the zebrafish indeed generate spatially appropriate peripheral responses to focal sensory stimulation even when they are transplanted into ectopic positions within the topographic map, indicating that circuit refinement occurs after the establishment of coarse topography. Refinement depends on motor neuron synaptic transmission, suggesting that an experience-dependent periphery-to-brain feedback mechanism establishes specific input connectivity among intermingled motor populations., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Development and regeneration of the vagus nerve.
- Author
-
Isabella AJ and Moens CB
- Subjects
- Vagus Nerve, Nerve Regeneration, Neurons physiology, Axons physiology
- Abstract
The vagus nerve, with its myriad constituent axon branches and innervation targets, has long been a model of anatomical complexity in the nervous system. The branched architecture of the vagus nerve is now appreciated to be highly organized around the topographic and/or molecular identities of the neurons that innervate each target tissue. However, we are only just beginning to understand the developmental mechanisms by which heterogeneous vagus neuron identity is specified, patterned, and used to guide the axons of particular neurons to particular targets. Here, we summarize our current understanding of the complex topographic and molecular organization of the vagus nerve, the developmental basis of neuron specification and patterned axon guidance that supports this organization, and the regenerative mechanisms that promote, or inhibit, the restoration of vagus nerve organization after nerve damage. Finally, we highlight key unanswered questions in these areas and discuss potential strategies to address these questions., Competing Interests: Declaration of Competing Interest None., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. A single-cell time-lapse of mouse prenatal development from gastrula to birth.
- Author
-
Qiu C, Martin BK, Welsh IC, Daza RM, Le TM, Huang X, Nichols EK, Taylor ML, Fulton O, O'Day DR, Gomes AR, Ilcisin S, Srivatsan S, Deng X, Disteche CM, Noble WS, Hamazaki N, Moens CB, Kimelman D, Cao J, Schier AF, Spielmann M, Murray SA, Trapnell C, and Shendure J
- Subjects
- Animals, Female, Mice, Pregnancy, Cell Differentiation genetics, Gastrulation genetics, Kidney cytology, Kidney embryology, Mesoderm cytology, Mesoderm enzymology, Neurons cytology, Neurons metabolism, Retina cytology, Retina embryology, Somites cytology, Somites embryology, Time Factors, Transcription Factors genetics, Transcription, Genetic, Organ Specificity genetics, Animals, Newborn embryology, Animals, Newborn genetics, Embryo, Mammalian cytology, Embryo, Mammalian embryology, Embryonic Development genetics, Gastrula cytology, Gastrula embryology, Single-Cell Analysis, Time-Lapse Imaging
- Abstract
The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans
1,2 . Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
5. Embryo-scale reverse genetics at single-cell resolution.
- Author
-
Saunders LM, Srivatsan SR, Duran M, Dorrity MW, Ewing B, Linbo TH, Shendure J, Raible DW, Moens CB, Kimelman D, and Trapnell C
- Subjects
- Animals, Gene Expression Profiling, Gene Expression Regulation, Developmental, Transcriptome genetics, Mutation, Notochord cytology, Notochord embryology, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Reverse Genetics methods, Zebrafish embryology, Zebrafish genetics, Single-Cell Analysis methods
- Abstract
The maturation of single-cell transcriptomic technologies has facilitated the generation of comprehensive cellular atlases from whole embryos
1-4 . 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., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
6. A single-cell transcriptional timelapse of mouse embryonic development, from gastrula to pup.
- Author
-
Qiu C, Martin BK, Welsh IC, Daza RM, Le TM, Huang X, Nichols EK, Taylor ML, Fulton O, O'Day DR, Gomes AR, Ilcisin S, Srivatsan S, Deng X, Disteche CM, Noble WS, Hamazaki N, Moens CB, Kimelman D, Cao J, Schier AF, Spielmann M, Murray SA, Trapnell C, and Shendure J
- Abstract
The house mouse, Mus musculus , is an exceptional model system, combining genetic tractability with close homology to human biology. Gestation in mouse development lasts just under three weeks, a period during which its genome orchestrates the astonishing transformation of a single cell zygote into a free-living pup composed of >500 million cells. Towards a global framework for exploring mammalian development, we applied single cell combinatorial indexing (sci-*) to profile the transcriptional states of 12.4 million nuclei from 83 precisely staged embryos spanning late gastrulation (embryonic day 8 or E8) to birth (postnatal day 0 or P0), with 2-hr temporal resolution during somitogenesis, 6-hr resolution through to birth, and 20-min resolution during the immediate postpartum period. From these data (E8 to P0), we annotate dozens of trajectories and hundreds of cell types and perform deeper analyses of the unfolding of the posterior embryo during somitogenesis as well as the ontogenesis of the kidney, mesenchyme, retina, and early neurons. Finally, we leverage the depth and temporal resolution of these whole embryo snapshots, together with other published data, to construct and curate a rooted tree of cell type relationships that spans mouse development from zygote to pup. Throughout this tree, we systematically nominate sets of transcription factors (TFs) and other genes as candidate drivers of the in vivo differentiation of hundreds of mammalian cell types. Remarkably, the most dramatic shifts in transcriptional state are observed in a restricted set of cell types in the hours immediately following birth, and presumably underlie the massive changes in physiology that must accompany the successful transition of a placental mammal to extrauterine life., Competing Interests: Competing Financial Interests Statement J.S. is a scientific advisory board member, consultant and/or co-founder of Scale Biosciences, Prime Medicine, Cajal Neuroscience, Guardant Health, Maze Therapeutics, Camp4 Therapeutics, Phase Genomics, Adaptive Biotechnologies, Sixth Street Capital and Pacific Biosciences. C.T. is a co-founder of Scale Biosciences. All other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
7. mTOR-regulated mitochondrial metabolism limits mycobacterium-induced cytotoxicity.
- Author
-
Pagán AJ, Lee LJ, Edwards-Hicks J, Moens CB, Tobin DM, Busch-Nentwich EM, Pearce EL, and Ramakrishnan L
- Subjects
- Animals, TOR Serine-Threonine Kinases metabolism, Zebrafish, Mycobacterium marinum, Mycobacterium tuberculosis metabolism, Tuberculosis
- Abstract
Necrosis of macrophages in the granuloma, the hallmark immunological structure of tuberculosis, is a major pathogenic event that increases host susceptibility. Through a zebrafish forward genetic screen, we identified the mTOR kinase, a master regulator of metabolism, as an early host resistance factor in tuberculosis. We found that mTOR complex 1 protects macrophages from mycobacterium-induced death by enabling infection-induced increases in mitochondrial energy metabolism fueled by glycolysis. These metabolic adaptations are required to prevent mitochondrial damage and death caused by the secreted mycobacterial virulence determinant ESAT-6. Thus, the host can effectively counter this early critical mycobacterial virulence mechanism simply by regulating energy metabolism, thereby allowing pathogen-specific immune mechanisms time to develop. Our findings may explain why Mycobacterium tuberculosis, albeit humanity's most lethal pathogen, is successful in only a minority of infected individuals., Competing Interests: Declaration of interests L.R. and E.L.P. are advisory board members for Cell. E.L.P. is a scientific advisory board member of ImmunoMet and a founder of Rheos Medicines. For the purpose of open access, the authors have applied for a CC BY public copyright license to any Author Accepted Manuscript version arising from this submission. This work is licensed under a Creative Commons Attribution 4.0 International License., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. Systematic reconstruction of cellular trajectories across mouse embryogenesis.
- Author
-
Qiu C, Cao J, Martin BK, Li T, Welsh IC, Srivatsan S, Huang X, Calderon D, Noble WS, Disteche CM, Murray SA, Spielmann M, Moens CB, Trapnell C, and Shendure J
- Subjects
- Animals, Embryo, Mammalian, Gastrulation genetics, Mammals, Mice, Embryonic Development genetics, Organogenesis
- Abstract
Mammalian embryogenesis is characterized by rapid cellular proliferation and diversification. Within a few weeks, a single-cell zygote gives rise to millions of cells expressing a panoply of molecular programs. Although intensively studied, a comprehensive delineation of the major cellular trajectories that comprise mammalian development in vivo remains elusive. Here, we set out to integrate several single-cell RNA-sequencing (scRNA-seq) datasets that collectively span mouse gastrulation and organogenesis, supplemented with new profiling of ~150,000 nuclei from approximately embryonic day 8.5 (E8.5) embryos staged in one-somite increments. Overall, we define cell states at each of 19 successive stages spanning E3.5 to E13.5 and heuristically connect them to their pseudoancestors and pseudodescendants. Although constructed through automated procedures, the resulting directed acyclic graph (TOME (trajectories of mammalian embryogenesis)) is largely consistent with our contemporary understanding of mammalian development. We leverage TOME to systematically nominate transcription factors (TFs) as candidate regulators of each cell type's specification, as well as 'cell-type homologs' across vertebrate evolution., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
9. Met is required for oligodendrocyte progenitor cell migration in Danio rerio.
- Author
-
Ali MF, Latimer AJ, Wang Y, Hogenmiller L, Fontenas L, Isabella AJ, Moens CB, Yu G, and Kucenas S
- Subjects
- Animals, Cell Differentiation, Oligodendroglia, Signal Transduction, Spinal Cord, Zebrafish, Oligodendrocyte Precursor Cells
- Abstract
During vertebrate central nervous system development, most oligodendrocyte progenitor cells (OPCs) are specified in the ventral spinal cord and must migrate throughout the neural tube until they become evenly distributed, occupying non-overlapping domains. While this process of developmental OPC migration is well characterized, the nature of the molecular mediators that govern it remain largely unknown. Here, using zebrafish as a model, we demonstrate that Met signaling is required for initial developmental migration of OPCs, and, using cell-specific knock-down of Met signaling, show that Met acts cell-autonomously in OPCs. Taken together, these findings demonstrate in vivo, the role of Met signaling in OPC migration and provide new insight into how OPC migration is regulated during development., (© The Author(s) 2021. Published by Oxford University Press on behalf of Genetics Society of America.)
- Published
- 2021
- Full Text
- View/download PDF
10. Intrinsic positional memory guides target-specific axon regeneration in the zebrafish vagus nerve.
- Author
-
Isabella AJ, Stonick JA, Dubrulle J, and Moens CB
- Subjects
- Animals, Animals, Genetically Modified physiology, Gene Expression Regulation, Developmental physiology, Neurons physiology, Peripheral Nerve Injuries physiopathology, Axons physiology, Nerve Regeneration physiology, Vagus Nerve physiology, Zebrafish physiology
- Abstract
Regeneration after peripheral nerve damage requires that axons re-grow to the correct target tissues in a process called target-specific regeneration. Although much is known about the mechanisms that promote axon re-growth, re-growing axons often fail to reach the correct targets, resulting in impaired nerve function. We know very little about how axons achieve target-specific regeneration, particularly in branched nerves that require distinct targeting decisions at branch points. The zebrafish vagus motor nerve is a branched nerve with a well-defined topographic organization. Here, we track regeneration of individual vagus axons after whole-nerve laser severing and find a robust capacity for target-specific, functional re-growth. We then develop a new single-cell chimera injury model for precise manipulation of axon-environment interactions and find that (1) the guidance mechanism used during regeneration is distinct from the nerve's developmental guidance mechanism, (2) target selection is specified by neurons' intrinsic memory of their position within the brain, and (3) targeting to a branch requires its pre-existing innervation. This work establishes the zebrafish vagus nerve as a tractable regeneration model and reveals the mechanistic basis of target-specific regeneration., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2021. Published by The Company of Biologists Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
11. The field of neurogenetics: where it stands and where it is going.
- Author
-
Isabella AJ, Leyva-Díaz E, Kaneko T, Gratz SJ, Moens CB, Hobert O, O'Connor-Giles K, Thakur R, and Sun H
- Subjects
- Animals, Genetic Techniques, History, 20th Century, History, 21st Century, Neurosciences methods, Genetics history, Neurosciences history
- Published
- 2021
- Full Text
- View/download PDF
12. Retinoic Acid Organizes the Zebrafish Vagus Motor Topographic Map via Spatiotemporal Coordination of Hgf/Met Signaling.
- Author
-
Isabella AJ, Barsh GR, Stonick JA, Dubrulle J, and Moens CB
- Subjects
- Animals, Branchial Region drug effects, Branchial Region physiology, Hepatocyte Growth Factor genetics, Keratolytic Agents pharmacology, Proto-Oncogene Proteins c-met genetics, Signal Transduction, Spatio-Temporal Analysis, Vagus Nerve drug effects, Zebrafish Proteins genetics, Gene Expression Regulation, Developmental drug effects, Hepatocyte Growth Factor metabolism, Proto-Oncogene Proteins c-met metabolism, Tretinoin pharmacology, Vagus Nerve physiology, Zebrafish physiology, Zebrafish Proteins metabolism
- Abstract
Information flow through neural circuits often requires their organization into topographic maps in which the positions of cell bodies and synaptic targets correspond. To understand how topographic map development is controlled, we examine the mechanism underlying targeting of vagus motor axons to the pharyngeal arches in zebrafish. We reveal that retinoic acid organizes topography by specifying anterior-posterior identity in vagus motor neurons. We then show that chemoattractant signaling between Hgf and Met is required for vagus innervation of the pharyngeal arches. Finally, we find that retinoic acid controls the spatiotemporal dynamics of Hgf/Met signaling to coordinate axon targeting with the developmental progression of the pharyngeal arches and show that experimentally altering the timing of Hgf/Met signaling is sufficient to redirect axon targeting and disrupt the topographic map. These findings establish a mechanism of topographic map development in which the regulation of chemoattractant signaling in space and time guides axon targeting., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
13. Microtubules are required for the maintenance of planar cell polarity in monociliated floorplate cells.
- Author
-
Mathewson AW, Berman DG, and Moens CB
- Subjects
- Animals, Cilia genetics, Membrane Proteins genetics, Microtubules genetics, Protein Transport, Receptors, Neurotransmitter genetics, Zebrafish genetics, Zebrafish Proteins genetics, Cell Polarity, Cilia metabolism, Membrane Proteins metabolism, Microtubules metabolism, Receptors, Neurotransmitter metabolism, Zebrafish embryology, Zebrafish Proteins metabolism
- Abstract
The asymmetric localization of planar cell polarity (PCP) proteins is essential for the establishment of many planar polarized cellular processes, but the mechanisms that maintain these asymmetric distributions remain poorly understood. A body of evidence has tied oriented subapical microtubules (MTs) to the establishment of PCP protein polarity, yet recent studies have suggested that the MT cytoskeleton is later dispensable for the maintenance of this asymmetry. As MTs underlie the vesicular trafficking of membrane-bound proteins within cells, the requirement for MTs in the maintenance of PCP merited further investigation. We investigated the complex interactions between PCP proteins and the MT cytoskeleton in the polarized context of the floorplate of the zebrafish neural tube. We demonstrated that the progressive posterior polarization of the primary cilia of floorplate cells requires not only Vangl2 but also Fzd3a. We determined that GFP-Vangl2 asymmetrically localizes to anterior membranes whereas Fzd3a-GFP does not polarize on anterior or posterior membranes but maintains a cytosolic enrichment at the base of the primary cilium. Vesicular Fzd3a-GFP is rapidly trafficked along MTs primarily toward the apical membrane during a period of PCP maintenance, whereas vesicular GFP-Vangl2 is less frequently observed. Nocodazole-induced loss of MT polymerization disrupts basal body positioning as well as GFP-Vangl2 localization and reduces cytosolic Fzd3a-GFP movements. Removal of nocodazole after MT disruption restores MT polymerization but does not restore basal body polarity. Interestingly, GFP-Vangl2 repolarizes to anterior membranes and vesicular Fzd3a-GFP dynamics recover after multiple hours of recovery, even in the context of unpolarized basal bodies. Together our findings challenge previous work by revealing an ongoing role for MT-dependent transport of PCP proteins in maintaining both cellular and PCP protein asymmetry during development., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
14. The generation of granule cells during the development and evolution of the cerebellum.
- Author
-
Iulianella A, Wingate RJ, Moens CB, and Capaldo E
- Subjects
- Animals, Cell Differentiation, Cerebellum growth & development, Humans, Neocortex cytology, Rhombencephalon, Cerebellum cytology
- Abstract
The cerebellum coordinates vestibular input into the hindbrain to control balance and movement, and its anatomical complexity is increasingly viewed as a high-throughput processing center for sensory and cognitive functions. Cerebellum development however is relatively simple, and arises from a specialized structure in the anterior hindbrain called the rhombic lip, which along with the ventricular zone of the rostral-most dorsal hindbrain region, give rise to the distinct cell types that constitute the cerebellum. Granule cells, being the most numerous cell types, arise from the rhombic lip and form a dense and distinct layer of the cerebellar cortex. In this short review, we describe the various strategies used by amniotes and anamniotes to generate and diversify granule cell types during cerebellar development., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
15. Competition between TIAM1 and Membranes Balances Endophilin A3 Activity in Cancer Metastasis.
- Author
-
Poudel KR, Roh-Johnson M, Su A, Ho T, Mathsyaraja H, Anderson S, Grady WM, Moens CB, Conacci-Sorrell M, Eisenman RN, and Bai J
- Subjects
- Animals, Carcinogenesis pathology, Cell Line, Tumor, Cell Movement physiology, Cell Proliferation physiology, Cell Transformation, Neoplastic, Colonic Neoplasms pathology, Endocytosis physiology, GTP Phosphohydrolases metabolism, Guanine Nucleotide Exchange Factors metabolism, Humans, Mice, Neoplasm Metastasis, Signal Transduction, Zebrafish, rac1 GTP-Binding Protein metabolism, Acyltransferases metabolism, Colonic Neoplasms metabolism, T-Lymphoma Invasion and Metastasis-inducing Protein 1 metabolism
- Abstract
Normal cells acquire aggressive behavior by modifying signaling pathways. For instance, alteration of endocytosis profoundly impacts both proliferation and migration during tumorigenesis. Here we investigate the mechanisms that enable the endocytic machinery to coordinate these processes. We show that a membrane curvature-sensing protein, endophilin A3, promotes growth and migration of colon cancer cells through two competing mechanisms: an endocytosis pathway that is required for proliferation and a GTPase regulatory pathway that controls cell motility. EndoA3 stimulates cell migration by binding the Rac GEF TIAM1 leading to activation of small GTPases. Competing interactions of EndoA3 with membrane versus TIAM1 modulate hyperproliferative and metastatic phenotypes. Disruption of EndoA3-membrane interactions stimulates TIAM1 and small GTPases in vitro, and further promotes pro-metastatic phenotypes in vivo. Together, these results uncover a coupling mechanism, by which EndoA3 promotes growth and migration of colon cancers, by linking membrane dynamics to GTPase regulation., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
16. Multiple zebrafish atoh1 genes specify a diversity of neuronal types in the zebrafish cerebellum.
- Author
-
Kidwell CU, Su CY, Hibi M, and Moens CB
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Differentiation genetics, Cell Movement genetics, Cerebellum metabolism, Fluorescent Antibody Technique, Gene Expression Regulation, Developmental, In Situ Hybridization, Neurons metabolism, Signal Transduction, Zebrafish embryology, Zebrafish genetics, Zebrafish Proteins metabolism, Basic Helix-Loop-Helix Transcription Factors metabolism, Cerebellum embryology, Neurogenesis genetics, Zebrafish Proteins genetics
- Abstract
A single Atoh1 basic-helix-loop-helix transcription factor specifies multiple neuron types in the mammalian cerebellum and anterior hindbrain. The zebrafish genome encodes three paralagous atoh1 genes whose functions in cerebellum and anterior hindbrain development we explore here. With use of a transgenic reporter, we report that zebrafish atoh1c-expressing cells are organized in two distinct domains that are separated both by space and developmental time. An early isthmic expression domain gives rise to an extracerebellar population in rhombomere 1 and an upper rhombic lip domain gives rise to granule cell progenitors that migrate to populate all four granule cell territories of the fish cerebellum. Using genetic mutants we find that of the three zebrafish atoh1 paralogs, atoh1c and atoh1a are required for the full complement of granule neurons. Surprisingly, the two genes are expressed in non-overlapping granule cell progenitor populations, indicating that fish use duplicate atoh1 genes to generate granule cell diversity that is not detected in mammals. Finally, live imaging of granule cell migration in wildtype and atoh1c mutant embryos reveals that while atoh1c is not required for granule cell specification per se, it is required for granule cells to delaminate and migrate away from the rhombic lip., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. Vagus Motor Neuron Topographic Map Determined by Parallel Mechanisms of hox5 Expression and Time of Axon Initiation.
- Author
-
Barsh GR, Isabella AJ, and Moens CB
- Subjects
- Animals, Animals, Genetically Modified embryology, Axons physiology, Embryo, Nonmammalian embryology, Gene Expression Regulation, Developmental genetics, Genes, Homeobox genetics, Motor Neurons physiology, Rhombencephalon embryology, Zebrafish embryology
- Abstract
Many networks throughout the nervous system are organized into topographic maps, where the positions of neuron cell bodies in the projecting field correspond with the positions of their axons in the target field. Previous studies of topographic map development show evidence for spatial patterning mechanisms, in which molecular determinants expressed across the projecting and target fields are matched directly in a point-to-point mapping process. Here, we describe a novel temporal mechanism of topographic map formation that depends on spatially regulated differences in the timing of axon outgrowth and functions in parallel with spatial point-to-point mapping mechanisms. We focus on the vagus motor neurons, which are topographically arranged in both mammals and fish. We show that cell position along the anterior-posterior axis of hindbrain rhombomere 8 determines expression of hox5 genes, which are expressed in posterior, but not anterior, vagus motor neurons. Using live imaging and transplantation in zebrafish embryos, we additionally reveal that axon initiation is delayed in posterior vagus motor neurons independent of neuron birth time. We show that hox5 expression directs topographic mapping without affecting time of axon outgrowth and that time of axon outgrowth directs topographic mapping without affecting hox5 expression. The vagus motor neuron topographic map is therefore determined by two mechanisms that act in parallel: a hox5-dependent spatial mechanism akin to classic mechanisms of topographic map formation and a novel axon outgrowth-dependent temporal mechanism in which time of axon formation is spatially regulated to direct axon targeting., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
18. Rpgrip1 is required for rod outer segment development and ciliary protein trafficking in zebrafish.
- Author
-
Raghupathy RK, Zhang X, Liu F, Alhasani RH, Biswas L, Akhtar S, Pan L, Moens CB, Li W, Liu M, Kennedy BN, and Shu X
- Subjects
- Animals, Codon, Nonsense, Protein Transport, Retina metabolism, Retina pathology, Retinal Degeneration pathology, Rhodopsin metabolism, Zebrafish growth & development, Zebrafish Proteins genetics, rab GTP-Binding Proteins metabolism, Cilia metabolism, Rod Cell Outer Segment metabolism, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
Mutations in the RPGR-interacting protein 1 (RPGRIP1) gene cause recessive Leber congenital amaurosis (LCA), juvenile retinitis pigmentosa (RP) and cone-rod dystrophy. RPGRIP1 interacts with other retinal disease-causing proteins and has been proposed to have a role in ciliary protein transport; however, its function remains elusive. Here, we describe a new zebrafish model carrying a nonsense mutation in the rpgrip1 gene. Rpgrip1homozygous mutants do not form rod outer segments and display mislocalization of rhodopsin, suggesting a role for RPGRIP1 in rhodopsin-bearing vesicle trafficking. Furthermore, Rab8, the key regulator of rhodopsin ciliary trafficking, was mislocalized in photoreceptor cells of rpgrip1 mutants. The degeneration of rod cells is early onset, followed by the death of cone cells. These phenotypes are similar to that observed in LCA and juvenile RP patients. Our data indicate RPGRIP1 is necessary for rod outer segment development through regulating ciliary protein trafficking. The rpgrip1 mutant zebrafish may provide a platform for developing therapeutic treatments for RP patients.
- Published
- 2017
- Full Text
- View/download PDF
19. Macrophage-Dependent Cytoplasmic Transfer during Melanoma Invasion In Vivo.
- Author
-
Roh-Johnson M, Shah AN, Stonick JA, Poudel KR, Kargl J, Yang GH, di Martino J, Hernandez RE, Gast CE, Zarour LR, Antoku S, Houghton AM, Bravo-Cordero JJ, Wong MH, Condeelis J, and Moens CB
- Subjects
- Animals, Cell Line, Tumor, Cell Movement physiology, Cytoplasm metabolism, Mice, Neoplasm Invasiveness, Zebrafish, Cell Communication physiology, Macrophages pathology, Melanoma pathology, Tumor Microenvironment physiology
- Abstract
Interactions between tumor cells and tumor-associated macrophages play critical roles in the initiation of tumor cell motility. To capture the cellular interactions of the tumor microenvironment with high-resolution imaging, we directly visualized tumor cells and their interactions with macrophages in zebrafish. Live imaging in zebrafish revealed that macrophages are dynamic, yet maintain sustained contact with tumor cells. In addition, the recruitment of macrophages to tumor cells promotes tumor cell dissemination. Using a Cre/LoxP strategy, we found that macrophages transfer cytoplasm to tumor cells in zebrafish and mouse models. Remarkably, macrophage cytoplasmic transfer correlated with melanoma cell dissemination. We further found that macrophages transfer cytoplasm to tumor cells upon cell contact in vitro. Thus, we present a model in which macrophage/tumor cell contact allows for the transfer of cytoplasmic molecules from macrophages to tumor cells corresponding to increased tumor cell motility and dissemination., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
20. Guidelines for morpholino use in zebrafish.
- Author
-
Stainier DYR, Raz E, Lawson ND, Ekker SC, Burdine RD, Eisen JS, Ingham PW, Schulte-Merker S, Yelon D, Weinstein BM, Mullins MC, Wilson SW, Ramakrishnan L, Amacher SL, Neuhauss SCF, Meng A, Mochizuki N, Panula P, and Moens CB
- Subjects
- Animals, Female, Male, Morpholinos adverse effects, Genetic Techniques standards, Morpholinos genetics, Zebrafish genetics
- Published
- 2017
- Full Text
- View/download PDF
21. A genetic basis for molecular asymmetry at vertebrate electrical synapses.
- Author
-
Miller AC, Whitebirch AC, Shah AN, Marsden KC, Granato M, O'Brien J, and Moens CB
- Subjects
- Animals, Zebrafish, Connexins genetics, Connexins metabolism, Electrical Synapses, Gap Junctions physiology, Neurons physiology
- Abstract
Neural network function is based upon the patterns and types of connections made between neurons. Neuronal synapses are adhesions specialized for communication and they come in two types, chemical and electrical. Communication at chemical synapses occurs via neurotransmitter release whereas electrical synapses utilize gap junctions for direct ionic and metabolic coupling. Electrical synapses are often viewed as symmetrical structures, with the same components making both sides of the gap junction. By contrast, we show that a broad set of electrical synapses in zebrafish, Danio rerio , require two gap-junction-forming Connexins for formation and function. We find that one Connexin functions presynaptically while the other functions postsynaptically in forming the channels. We also show that these synapses are required for the speed and coordination of escape responses. Our data identify a genetic basis for molecular asymmetry at vertebrate electrical synapses and show they are required for appropriate behavioral performance.
- Published
- 2017
- Full Text
- View/download PDF
22. Planar cell polarity in moving cells: think globally, act locally.
- Author
-
Davey CF and Moens CB
- Subjects
- Animals, Body Patterning physiology, Cellular Microenvironment physiology, Humans, Signal Transduction, Cell Movement physiology, Cell Polarity physiology
- Abstract
The planar cell polarity (PCP) pathway is best known for its role in polarizing epithelial cells within the plane of a tissue but it also plays a role in a range of cell migration events during development. The mechanism by which the PCP pathway polarizes stationary epithelial cells is well characterized, but how PCP signaling functions to regulate more dynamic cell behaviors during directed cell migration is much less understood. Here, we review recent discoveries regarding the localization of PCP proteins in migrating cells and their impact on the cell biology of collective and individual cell migratory behaviors., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
23. Regulation of Vegf signaling by natural and synthetic ligands.
- Author
-
Rossi A, Gauvrit S, Marass M, Pan L, Moens CB, and Stainier DYR
- Subjects
- Aging pathology, Animals, Arteries growth & development, Arteries pathology, Cell Differentiation, Genes, Dominant, Ligands, Mutation genetics, Neovascularization, Physiologic, Protein Engineering, RNA, Messenger genetics, RNA, Messenger metabolism, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor Receptor-1 metabolism, Zebrafish genetics, Signal Transduction, Vascular Endothelial Growth Factor A metabolism, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
The mechanisms that allow cells to bypass anti-vascular endothelial growth factor A (VEGFA) therapy remain poorly understood. Here we use zebrafish to investigate this question and first show that vegfaa mutants display a severe vascular phenotype that can surprisingly be rescued to viability by vegfaa messenger RNA injections at the 1-cell stage. Using vegfaa mutants as an in vivo test tube, we found that zebrafish Vegfbb, Vegfd, and Pgfb can also rescue these animals to viability. Taking advantage of a new vegfr1 tyrosine kinase-deficient mutant, we determined that Pgfb rescues vegfaa mutants via Vegfr1. Altogether, these data reveal potential resistance routes against current anti-VEGFA therapies. In order to circumvent this resistance, we engineered and validated new dominant negative Vegfa molecules that by trapping Vegf family members can block vascular development. Thus, our results show that Vegfbb, Vegfd, and Pgfb can sustain vascular development in the absence of VegfA, and our newly engineered Vegf molecules expand the toolbox for basic research and antiangiogenic therapy., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
24. MYC-nick promotes cell migration by inducing fascin expression and Cdc42 activation.
- Author
-
Anderson S, Poudel KR, Roh-Johnson M, Brabletz T, Yu M, Borenstein-Auerbach N, Grady WN, Bai J, Moens CB, Eisenman RN, and Conacci-Sorrell M
- Subjects
- Animals, Cell Line, Tumor, Cell Movement genetics, Colorectal Neoplasms pathology, F-Box-WD Repeat-Containing Protein 7 genetics, Gene Expression Regulation, Neoplastic, Humans, Mice, Neoplasm Metastasis, Signal Transduction, Stomach Neoplasms pathology, Transcriptional Activation genetics, Zebrafish, Carrier Proteins genetics, Colorectal Neoplasms genetics, Microfilament Proteins genetics, Proto-Oncogene Proteins c-myc genetics, Stomach Neoplasms genetics, cdc42 GTP-Binding Protein genetics
- Abstract
MYC-nick is a cytoplasmic, transcriptionally inactive member of the MYC oncoprotein family, generated by a proteolytic cleavage of full-length MYC. MYC-nick promotes migration and survival of cells in response to chemotherapeutic agents or withdrawal of glucose. Here we report that MYC-nick is abundant in colonic and intestinal tumors derived from mouse models with mutations in the Wnt, TGF-β, and PI3K pathways. Moreover, MYC-nick is elevated in colon cancer cells deleted for FBWX7, which encodes the major E3 ligase of full-length MYC frequently mutated in colorectal cancers. MYC-nick promotes the migration of colon cancer cells assayed in 3D cultures or grown as xenografts in a zebrafish metastasis model. MYC-nick accelerates migration by activating the Rho GTPase Cdc42 and inducing fascin expression. MYC-nick, fascin, and Cdc42 are frequently up-regulated in cells present at the invasive front of human colorectal tumors, suggesting a coordinated role for these proteins in tumor migration., Competing Interests: The authors declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
25. Cilia-Associated Genes Play Differing Roles in Aminoglycoside-Induced Hair Cell Death in Zebrafish.
- Author
-
Stawicki TM, Hernandez L, Esterberg R, Linbo T, Owens KN, Shah AN, Thapa N, Roberts B, Moens CB, Rubel EW, and Raible DW
- Subjects
- Animals, Cell Death, Cilia metabolism, Cilia ultrastructure, Cytoplasmic Dyneins genetics, Cytoplasmic Dyneins metabolism, Gene Expression, Hair Cells, Auditory cytology, Hair Cells, Auditory metabolism, Mechanotransduction, Cellular, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism, Zebrafish, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Aminoglycosides toxicity, Cilia drug effects, Drug Tolerance genetics, Hair Cells, Auditory drug effects, Mutation
- Abstract
Hair cells possess a single primary cilium, called the kinocilium, early in development. While the kinocilium is lost in auditory hair cells of most species it is maintained in vestibular hair cells. It has generally been believed that the primary role of the kinocilium and cilia-associated genes in hair cells is in the establishment of the polarity of actin-based stereocilia, the hair cell mechanotransduction apparatus. Through genetic screening and testing of candidate genes in zebrafish (Danio rerio) we have found that mutations in multiple cilia genes implicated in intraflagellar transport (dync2h1, wdr35, ift88, and traf3ip), and the ciliary transition zone (cc2d2a, mks1, and cep290) lead to resistance to aminoglycoside-induced hair cell death. These genes appear to have differing roles in hair cells, as mutations in intraflagellar transport genes, but not transition zone genes, lead to defects in kinocilia formation and processes dependent upon hair cell mechanotransduction activity. These mutants highlight a novel role of cilia-associated genes in hair cells, and provide powerful tools for further study., (Copyright © 2016 Stawicki et al.)
- Published
- 2016
- Full Text
- View/download PDF
26. Dachsous1b cadherin regulates actin and microtubule cytoskeleton during early zebrafish embryogenesis.
- Author
-
Li-Villarreal N, Forbes MM, Loza AJ, Chen J, Ma T, Helde K, Moens CB, Shin J, Sawada A, Hindes AE, Dubrulle J, Schier AF, Longmore GD, Marlow FL, and Solnica-Krezel L
- Published
- 2016
- Full Text
- View/download PDF
27. Rapid Reverse Genetic Screening Using CRISPR in Zebrafish.
- Author
-
Shah AN, Davey CF, Whitebirch AC, Miller AC, and Moens CB
- Subjects
- Animals, CRISPR-Cas Systems, Genetic Testing, Clustered Regularly Interspaced Short Palindromic Repeats, Zebrafish genetics
- Published
- 2016
- Full Text
- View/download PDF
28. Lysosomal Disorders Drive Susceptibility to Tuberculosis by Compromising Macrophage Migration.
- Author
-
Berg RD, Levitte S, O'Sullivan MP, O'Leary SM, Cambier CJ, Cameron J, Takaki KK, Moens CB, Tobin DM, Keane J, and Ramakrishnan L
- Subjects
- Animals, Granuloma metabolism, Macrophages cytology, Macrophages, Alveolar immunology, Mycobacterium marinum, Pulmonary Alveoli immunology, Smoking, Transcription Factors genetics, Transcription Factors metabolism, Transport Vesicles metabolism, Tuberculosis immunology, Tuberculosis pathology, Zebrafish, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Disease Susceptibility, Lysosomes metabolism, Macrophages immunology, Macrophages pathology, Mycobacterium Infections immunology, Mycobacterium Infections pathology
- Abstract
A zebrafish genetic screen for determinants of susceptibility to Mycobacterium marinum identified a hypersusceptible mutant deficient in lysosomal cysteine cathepsins that manifests hallmarks of human lysosomal storage diseases. Under homeostatic conditions, mutant macrophages accumulate undigested lysosomal material, which disrupts endocytic recycling and impairs their migration to, and thus engulfment of, dying cells. This causes a buildup of unengulfed cell debris. During mycobacterial infection, macrophages with lysosomal storage cannot migrate toward infected macrophages undergoing apoptosis in the tuberculous granuloma. The unengulfed apoptotic macrophages undergo secondary necrosis, causing granuloma breakdown and increased mycobacterial growth. Macrophage lysosomal storage similarly impairs migration to newly infecting mycobacteria. This phenotype is recapitulated in human smokers, who are at increased risk for tuberculosis. A majority of their alveolar macrophages exhibit lysosomal accumulations of tobacco smoke particulates and do not migrate to Mycobacterium tuberculosis. The incapacitation of highly microbicidal first-responding macrophages may contribute to smokers' susceptibility to tuberculosis., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
29. Approaching Perfection: New Developments in Zebrafish Genome Engineering.
- Author
-
Shah AN and Moens CB
- Subjects
- Animals, Genetic Engineering methods, Zebrafish genetics
- Abstract
Programmable nucleases have revolutionized zebrafish genetics by enabling targeted genome modifications. In this issue of Developmental Cell, Hoshijima et al. (2016) take genome modification in the zebrafish to the next level, demonstrating the efficient use of homologous recombination to make genetic tools for a range of applications., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
30. PCP Signaling between Migrating Neurons and their Planar-Polarized Neuroepithelial Environment Controls Filopodial Dynamics and Directional Migration.
- Author
-
Davey CF, Mathewson AW, and Moens CB
- Subjects
- Animals, Cytoskeleton genetics, Cytoskeleton metabolism, Mice, Motor Neurons metabolism, Neuroepithelial Cells metabolism, Pseudopodia metabolism, Rhombencephalon metabolism, Signal Transduction, Zebrafish, Cell Movement genetics, Cell Polarity genetics, Pseudopodia genetics, Rhombencephalon growth & development
- Abstract
The planar cell polarity (PCP) pathway is a cell-contact mediated mechanism for transmitting polarity information between neighboring cells. PCP "core components" (Vangl, Fz, Pk, Dsh, and Celsr) are essential for a number of cell migratory events including the posterior migration of facial branchiomotor neurons (FBMNs) in the plane of the hindbrain neuroepithelium in zebrafish and mice. While the mechanism by which PCP signaling polarizes static epithelial cells is well understood, how PCP signaling controls highly dynamic processes like neuronal migration remains an important outstanding question given that PCP components have been implicated in a range of directed cell movements, particularly during vertebrate development. Here, by systematically disrupting PCP signaling in a rhombomere-restricted manner we show that PCP signaling is required both within FBMNs and the hindbrain rhombomere 4 environment at the time when they initiate their migration. Correspondingly, we demonstrate planar polarized localization of PCP core components Vangl2 and Fzd3a in the hindbrain neuroepithelium, and transient localization of Vangl2 at the tips of retracting FBMN filopodia. Using high-resolution timelapse imaging of FBMNs in genetic chimeras we uncover opposing cell-autonomous and non-cell-autonomous functions for Fzd3a and Vangl2 in regulating FBMN protrusive activity. Within FBMNs, Fzd3a is required to stabilize filopodia while Vangl2 has an antagonistic, destabilizing role. However, in the migratory environment Fzd3a acts to destabilize FBMN filopodia while Vangl2 has a stabilizing role. Together, our findings suggest a model in which PCP signaling between the planar polarized neuroepithelial environment and FBMNs directs migration by the selective stabilization of FBMN filopodia.
- Published
- 2016
- Full Text
- View/download PDF
31. Targeted candidate gene screens using CRISPR/Cas9 technology.
- Author
-
Shah AN, Moens CB, and Miller AC
- Subjects
- Animals, Phenotype, Zebrafish genetics, Zebrafish growth & development, CRISPR-Cas Systems genetics, Genetic Association Studies methods, Genetic Engineering methods, RNA, Guide, CRISPR-Cas Systems genetics
- Abstract
In the postgenomic era, the ability to quickly, efficiently, and inexpensively assign function to the zebrafish proteome is critical. Clustered regularly interspaced short palindromic repeats (CRISPRs) have revolutionized the ability to perform reverse genetics because of its simplicity and broad applicability. The CRISPR system is composed of an engineered, gene-specific single guide RNA (sgRNA) and a Cas9 enzyme that causes double-stranded breaks in DNA at the targeted site. This simple, two-part system, when injected into one-cell stage zebrafish embryos, efficiently mutates target loci at a frequency such that injected embryos phenocopy known mutant phenotypes. This property allows for CRISPR-based F0 screening in zebrafish, which provides a means to screen through a large number of candidate genes for their role in a phenotype of interest. While there are important considerations for any successful genetic screen, CRISPR screening has significant benefits over conventional methods and can be accomplished in any lab with modest molecular biology experience., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
32. The Ciliopathy Protein CC2D2A Associates with NINL and Functions in RAB8-MICAL3-Regulated Vesicle Trafficking.
- Author
-
Bachmann-Gagescu R, Dona M, Hetterschijt L, Tonnaer E, Peters T, de Vrieze E, Mans DA, van Beersum SE, Phelps IG, Arts HH, Keunen JE, Ueffing M, Roepman R, Boldt K, Doherty D, Moens CB, Neuhauss SC, Kremer H, and van Wijk E
- Subjects
- Abnormalities, Multiple genetics, Abnormalities, Multiple metabolism, Abnormalities, Multiple pathology, Animals, Cerebellum metabolism, Cerebellum pathology, Cilia genetics, Cilia metabolism, Cilia pathology, Ciliary Motility Disorders metabolism, Ciliary Motility Disorders pathology, Cytoskeletal Proteins, Encephalocele metabolism, Encephalocele pathology, Eye Abnormalities genetics, Eye Abnormalities metabolism, Eye Abnormalities pathology, Gene Knockdown Techniques, Humans, Kidney Diseases, Cystic genetics, Kidney Diseases, Cystic metabolism, Kidney Diseases, Cystic pathology, Microtubule-Associated Proteins genetics, Mixed Function Oxygenases metabolism, Mutation, Nuclear Proteins genetics, Polycystic Kidney Diseases metabolism, Polycystic Kidney Diseases pathology, Protein Transport genetics, Proteins metabolism, Retina metabolism, Retina pathology, Retinitis Pigmentosa, Signal Transduction, Zebrafish, rab GTP-Binding Proteins metabolism, Cerebellum abnormalities, Ciliary Motility Disorders genetics, Encephalocele genetics, Microtubule-Associated Proteins metabolism, Mixed Function Oxygenases genetics, Nuclear Proteins metabolism, Polycystic Kidney Diseases genetics, Proteins genetics, Retina abnormalities, rab GTP-Binding Proteins genetics
- Abstract
Ciliopathies are a group of human disorders caused by dysfunction of primary cilia, ubiquitous microtubule-based organelles involved in transduction of extra-cellular signals to the cell. This function requires the concentration of receptors and channels in the ciliary membrane, which is achieved by complex trafficking mechanisms, in part controlled by the small GTPase RAB8, and by sorting at the transition zone located at the entrance of the ciliary compartment. Mutations in the transition zone gene CC2D2A cause the related Joubert and Meckel syndromes, two typical ciliopathies characterized by central nervous system malformations, and result in loss of ciliary localization of multiple proteins in various models. The precise mechanisms by which CC2D2A and other transition zone proteins control protein entrance into the cilium and how they are linked to vesicular trafficking of incoming cargo remain largely unknown. In this work, we identify the centrosomal protein NINL as a physical interaction partner of CC2D2A. NINL partially co-localizes with CC2D2A at the base of cilia and ninl knockdown in zebrafish leads to photoreceptor outer segment loss, mislocalization of opsins and vesicle accumulation, similar to cc2d2a-/- phenotypes. Moreover, partial ninl knockdown in cc2d2a-/- embryos enhances the retinal phenotype of the mutants, indicating a genetic interaction in vivo, for which an illustration is found in patients from a Joubert Syndrome cohort. Similar to zebrafish cc2d2a mutants, ninl morphants display altered Rab8a localization. Further exploration of the NINL-associated interactome identifies MICAL3, a protein known to interact with Rab8 and to play an important role in vesicle docking and fusion. Together, these data support a model where CC2D2A associates with NINL to provide a docking point for cilia-directed cargo vesicles, suggesting a mechanism by which transition zone proteins can control the protein content of the ciliary compartment.
- Published
- 2015
- Full Text
- View/download PDF
33. Dachsous1b cadherin regulates actin and microtubule cytoskeleton during early zebrafish embryogenesis.
- Author
-
Li-Villarreal N, Forbes MM, Loza AJ, Chen J, Ma T, Helde K, Moens CB, Shin J, Sawada A, Hindes AE, Dubrulle J, Schier AF, Longmore GD, Marlow FL, and Solnica-Krezel L
- Subjects
- Animals, Cadherins genetics, DNA Primers genetics, Exocytosis physiology, Female, Immunohistochemistry, In Situ Hybridization, Microscopy, Confocal, Optical Imaging, Ovary anatomy & histology, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Zebrafish Proteins genetics, Actins metabolism, Cadherins metabolism, Cytoskeleton physiology, Microtubules metabolism, Zebrafish embryology, Zebrafish Proteins metabolism
- Abstract
Dachsous (Dchs), an atypical cadherin, is an evolutionarily conserved regulator of planar cell polarity, tissue size and cell adhesion. In humans, DCHS1 mutations cause pleiotropic Van Maldergem syndrome. Here, we report that mutations in zebrafish dchs1b and dchs2 disrupt several aspects of embryogenesis, including gastrulation. Unexpectedly, maternal zygotic (MZ) dchs1b mutants show defects in the earliest developmental stage, egg activation, including abnormal cortical granule exocytosis (CGE), cytoplasmic segregation, cleavages and maternal mRNA translocation, in transcriptionally quiescent embryos. Later, MZdchs1b mutants exhibit altered dorsal organizer and mesendodermal gene expression, due to impaired dorsal determinant transport and Nodal signaling. Mechanistically, MZdchs1b phenotypes can be explained in part by defective actin or microtubule networks, which appear bundled in mutants. Accordingly, disruption of actin cytoskeleton in wild-type embryos phenocopied MZdchs1b mutant defects in cytoplasmic segregation and CGE, whereas interfering with microtubules in wild-type embryos impaired dorsal organizer and mesodermal gene expression without perceptible earlier phenotypes. Moreover, the bundled microtubule phenotype was partially rescued by expressing either full-length Dchs1b or its intracellular domain, suggesting that Dchs1b affects microtubules and some developmental processes independent of its known ligand Fat. Our results indicate novel roles for vertebrate Dchs in actin and microtubule cytoskeleton regulation in the unanticipated context of the single-celled embryo., (© 2015. Published by The Company of Biologists Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
34. Rapid reverse genetic screening using CRISPR in zebrafish.
- Author
-
Shah AN, Davey CF, Whitebirch AC, Miller AC, and Moens CB
- Subjects
- Animals, Gene Expression Regulation, Gene Expression Regulation, Developmental, Genetic Loci, Pigmentation genetics, Pigmentation physiology, RNA, Messenger genetics, RNA, Messenger metabolism, Retinal Pigment Epithelium embryology, Zebrafish embryology, Zebrafish Proteins genetics, CRISPR-Cas Systems, Embryo, Nonmammalian physiology, Genetic Testing methods, Zebrafish genetics
- Abstract
Identifying genes involved in biological processes is critical for understanding the molecular building blocks of life. We used engineered CRISPR (clustered regularly interspaced short palindromic repeats) to efficiently mutate specific loci in zebrafish (Danio rerio) and screen for genes involved in vertebrate biological processes. We found that increasing CRISPR efficiency by injecting optimized amounts of Cas9-encoding mRNA and multiplexing single guide RNAs (sgRNAs) allowed for phenocopy of known mutants across many phenotypes in embryos. We performed a proof-of-concept screen in which we used intersecting, multiplexed pool injections to examine 48 loci and identified two new genes involved in electrical-synapse formation. By deep sequencing target loci, we found that 90% of the genes were effectively screened. We conclude that CRISPR can be used as a powerful reverse genetic screening strategy in vivo in a vertebrate system.
- Published
- 2015
- Full Text
- View/download PDF
35. Rapid identification and recovery of ENU-induced mutations with next-generation sequencing and Paired-End Low-Error analysis.
- Author
-
Pan L, Shah AN, Phelps IG, Doherty D, Johnson EA, and Moens CB
- Subjects
- Animals, Codon, Nonsense drug effects, DNA analysis, DNA isolation & purification, Gene Library, Genomics standards, Male, Mutation drug effects, RNA Splice Sites genetics, Sequence Analysis, DNA, Spermatozoa metabolism, Zebrafish genetics, Zebrafish metabolism, Ethylnitrosourea toxicity, Genome drug effects, Genomics methods, High-Throughput Nucleotide Sequencing standards
- Abstract
Background: Targeting Induced Local Lesions IN Genomes (TILLING) is a reverse genetics approach to directly identify point mutations in specific genes of interest in genomic DNA from a large chemically mutagenized population. Classical TILLING processes, based on enzymatic detection of mutations in heteroduplex PCR amplicons, are slow and labor intensive., Results: Here we describe a new TILLING strategy in zebrafish using direct next generation sequencing (NGS) of 250 bp amplicons followed by Paired-End Low-Error (PELE) sequence analysis. By pooling a genomic DNA library made from over 9,000 N-ethyl-N-nitrosourea (ENU) mutagenized F1 fish into 32 equal pools of 288 fish, each with a unique Illumina barcode, we reduce the complexity of the template to a level at which we can detect mutations that occur in a single heterozygous fish in the entire library. MiSeq sequencing generates 250 base-pair overlapping paired-end reads, and PELE analysis aligns the overlapping sequences to each other and filters out any imperfect matches, thereby eliminating variants introduced during the sequencing process. We find that this filtering step reduces the number of false positive calls 50-fold without loss of true variant calls. After PELE we were able to validate 61.5% of the mutant calls that occurred at a frequency between 1 mutant call:100 wildtype calls and 1 mutant call:1000 wildtype calls in a pool of 288 fish. We then use high-resolution melt analysis to identify the single heterozygous mutation carrier in the 288-fish pool in which the mutation was identified., Conclusions: Using this NGS-TILLING protocol we validated 28 nonsense or splice site mutations in 20 genes, at a two-fold higher efficiency than using traditional Cel1 screening. We conclude that this approach significantly increases screening efficiency and accuracy at reduced cost and can be applied in a wide range of organisms.
- Published
- 2015
- Full Text
- View/download PDF
36. Neurobeachin is required postsynaptically for electrical and chemical synapse formation.
- Author
-
Miller AC, Voelker LH, Shah AN, and Moens CB
- Subjects
- Animals, Behavior, Animal physiology, Mutation, Zebrafish, Zebrafish Proteins, Nerve Tissue Proteins physiology, Nervous System growth & development, Neurons physiology, Synapses physiology
- Abstract
Background: Neural networks and their function are defined by synapses, which are adhesions specialized for intercellular communication that can be either chemical or electrical. At chemical synapses, transmission between neurons is mediated by neurotransmitters, whereas at electrical synapses, direct ionic and metabolic coupling occur via gap junctions between neurons. The molecular pathways required for electrical synaptogenesis are not well understood, and whether they share mechanisms of formation with chemical synapses is not clear., Results: Here, using a forward genetic screen in zebrafish, we find that the autism-associated gene neurobeachin (nbea), which encodes a BEACH-domain-containing protein implicated in endomembrane trafficking, is required for both electrical and chemical synapse formation. Additionally, we find that nbea is dispensable for axonal formation and early dendritic outgrowth but is required to maintain dendritic complexity. These synaptic and morphological defects correlate with deficiencies in behavioral performance. Using chimeric animals in which individually identifiable neurons are either mutant or wild-type, we find that Nbea is necessary and sufficient autonomously in the postsynaptic neuron for both synapse formation and dendritic arborization., Conclusions: Our data identify a surprising link between electrical and chemical synapse formation and show that Nbea acts as a critical regulator in the postsynaptic neuron for the coordination of dendritic morphology with synaptogenesis., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
37. Pard3 regulates contact between neural crest cells and the timing of Schwann cell differentiation but is not essential for neural crest migration or myelination.
- Author
-
Blasky AJ, Pan L, Moens CB, and Appel B
- Subjects
- Animals, Axons metabolism, Carrier Proteins genetics, Cell Polarity physiology, Gene Expression Regulation, Developmental physiology, Motor Neurons cytology, Motor Neurons metabolism, Neural Crest cytology, Schwann Cells cytology, Zebrafish genetics, Zebrafish Proteins genetics, Carrier Proteins biosynthesis, Cell Differentiation physiology, Cell Movement physiology, Neural Crest embryology, Schwann Cells metabolism, Zebrafish embryology, Zebrafish Proteins biosynthesis
- Abstract
Background: Schwann cells, which arise from the neural crest, are the myelinating glia of the peripheral nervous system. During development neural crest and their Schwann cell derivatives engage in a sequence of events that comprise delamination from the neuroepithelium, directed migration, axon ensheathment, and myelin membrane synthesis. At each step neural crest and Schwann cells are polarized, suggesting important roles for molecules that create cellular asymmetries. In this work we investigated the possibility that one polarity protein, Pard3, contributes to the polarized features of neural crest and Schwann cells that are associated with directed migration and myelination., Results: We analyzed mutant zebrafish embryos deficient for maternal and zygotic pard3 function. Time-lapse imaging revealed that neural crest delamination was normal but that migrating cells were disorganized with substantial amounts of overlapping membrane. Nevertheless, neural crest cells migrated to appropriate peripheral targets. Schwann cells wrapped motor axons and, although myelin gene expression was delayed, myelination proceeded to completion., Conclusions: Pard3 mediates contact inhibition between neural crest cells and promotes timely myelin gene expression but is not essential for neural crest migration or myelination., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
38. Distinct Notch signaling outputs pattern the developing arterial system.
- Author
-
Quillien A, Moore JC, Shin M, Siekmann AF, Smith T, Pan L, Moens CB, Parsons MJ, and Lawson ND
- Subjects
- Animals, Animals, Genetically Modified, Arteries cytology, Cell Differentiation genetics, Embryo, Nonmammalian, Endothelium, Vascular embryology, Morphogenesis genetics, Neovascularization, Physiologic genetics, Signal Transduction physiology, Veins embryology, Zebrafish embryology, Zebrafish genetics, Arteries embryology, Body Patterning genetics, Receptors, Notch physiology
- Abstract
Differentiation of arteries and veins is essential for the development of a functional circulatory system. In vertebrate embryos, genetic manipulation of Notch signaling has demonstrated the importance of this pathway in driving artery endothelial cell differentiation. However, when and where Notch activation occurs to affect endothelial cell fate is less clear. Using transgenic zebrafish bearing a Notch-responsive reporter, we demonstrate that Notch is activated in endothelial progenitors during vasculogenesis prior to blood vessel morphogenesis and is maintained in arterial endothelial cells throughout larval stages. Furthermore, we find that endothelial progenitors in which Notch is activated are committed to a dorsal aorta fate. Interestingly, some arterial endothelial cells subsequently downregulate Notch signaling and then contribute to veins during vascular remodeling. Lineage analysis, together with perturbation of both Notch receptor and ligand function, further suggests several distinct developmental windows in which Notch signaling acts to promote artery commitment and maintenance. Together, these findings demonstrate that Notch acts in distinct contexts to initiate and maintain artery identity during embryogenesis.
- Published
- 2014
- Full Text
- View/download PDF
39. Hoxb1b controls oriented cell division, cell shape and microtubule dynamics in neural tube morphogenesis.
- Author
-
Zigman M, Laumann-Lipp N, Titus T, Postlethwait J, and Moens CB
- Subjects
- Animals, Branchial Region embryology, Branchial Region metabolism, Cell Polarity, Epithelium embryology, Epithelium metabolism, Gene Expression Regulation, Developmental, Homeodomain Proteins genetics, Mitosis, Mutation genetics, Neural Tube metabolism, Rhombencephalon cytology, Rhombencephalon embryology, Zebrafish metabolism, Cell Division, Cell Shape, Homeodomain Proteins metabolism, Microtubules metabolism, Morphogenesis, Neural Tube cytology, Zebrafish embryology
- Abstract
Hox genes are classically ascribed to function in patterning the anterior-posterior axis of bilaterian animals; however, their role in directing molecular mechanisms underlying morphogenesis at the cellular level remains largely unstudied. We unveil a non-classical role for the zebrafish hoxb1b gene, which shares ancestral functions with mammalian Hoxa1, in controlling progenitor cell shape and oriented cell division during zebrafish anterior hindbrain neural tube morphogenesis. This is likely distinct from its role in cell fate acquisition and segment boundary formation. We show that, without affecting major components of apico-basal or planar cell polarity, Hoxb1b regulates mitotic spindle rotation during the oriented neural keel symmetric mitoses that are required for normal neural tube lumen formation in the zebrafish. This function correlates with a non-cell-autonomous requirement for Hoxb1b in regulating microtubule plus-end dynamics in progenitor cells in interphase. We propose that Hox genes can influence global tissue morphogenesis by control of microtubule dynamics in individual cells in vivo.
- Published
- 2014
- Full Text
- View/download PDF
40. Cerebellar development in the absence of Gbx function in zebrafish.
- Author
-
Su CY, Kemp HA, and Moens CB
- Subjects
- Alleles, Animals, Animals, Genetically Modified, Body Patterning, Cell Differentiation, Cerebellum metabolism, Epistasis, Genetic, Fibroblast Growth Factors metabolism, Genotype, Homeodomain Proteins genetics, Mice, Morphogenesis, Mutation, Neurons metabolism, Otx Transcription Factors metabolism, Phenotype, Signal Transduction, Zebrafish genetics, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Cerebellum embryology, Gene Expression Regulation, Developmental, Homeodomain Proteins physiology, Zebrafish embryology, Zebrafish Proteins physiology
- Abstract
The midbrain-hindbrain boundary (MHB) is a well-known organizing center during vertebrate brain development. The MHB forms at the expression boundary of Otx2 and Gbx2, mutually repressive homeodomain transcription factors expressed in the midbrain/forebrain and anterior hindbrain, respectively. The genetic hierarchy of gene expression at the MHB is complex, involving multiple positive and negative feedback loops that result in the establishment of non-overlapping domains of Wnt1 and Fgf8 on either side of the boundary and the consequent specification of the cerebellum. The cerebellum derives from the dorsal part of the anterior-most hindbrain segment, rhombomere 1 (r1), which undergoes a distinctive morphogenesis to give rise to the cerebellar primordium within which the various cerebellar neuron types are specified. Previous studies in the mouse have shown that Gbx2 is essential for cerebellar development. Using zebrafish mutants we show here that in the zebrafish gbx1 and gbx2 are required redundantly for morphogenesis of the cerebellar primordium and subsequent cerebellar differentiation, but that this requirement is alleviated by knocking down Otx. Expression of fgf8, wnt1 and the entire MHB genetic program is progressively lost in gbx1-;gbx2- double mutants but is rescued by Otx knock-down. This rescue of the MHB genetic program depends on rescued Fgf signaling, however the rescue of cerebellar primordium morphogenesis is independent of both Gbx and Fgf. Based on our findings we propose a revised model for the role of Gbx in cerebellar development., (© 2013 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
41. Role of mef2ca in developmental buffering of the zebrafish larval hyoid dermal skeleton.
- Author
-
DeLaurier A, Huycke TR, Nichols JT, Swartz ME, Larsen A, Walker C, Dowd J, Pan L, Moens CB, and Kimmel CB
- Subjects
- Animals, Genes, Homeobox, Zebrafish genetics, Zebrafish growth & development, Bone Development genetics, Larva growth & development, Zebrafish embryology, Zebrafish Proteins genetics
- Abstract
Phenotypic robustness requires a process of developmental buffering that is largely not understood, but which can be disrupted by mutations. Here we show that in mef2ca(b1086) loss of function mutant embryos and early larvae, development of craniofacial hyoid bones, the opercle (Op) and branchiostegal ray (BR), becomes remarkably unstable; the large magnitude of the instability serves as a positive attribute to learn about features of this developmental buffering. The OpBR mutant phenotype variably includes bone expansion and fusion, Op duplication, and BR homeosis. Formation of a novel bone strut, or a bone bridge connecting the Op and BR together occurs frequently. We find no evidence that the phenotypic stability in the wild type is provided by redundancy between mef2ca and its co-ortholog mef2cb, or that it is related to the selector (homeotic) gene function of mef2ca. Changes in dorsal-ventral patterning of the hyoid arch also might not contribute to phenotypic instability in mutants. However, subsequent development of the bone lineage itself, including osteoblast differentiation and morphogenetic outgrowth, shows marked variation. Hence, steps along the developmental trajectory appear differentially sensitive to the loss of buffering, providing focus for the future study., (© 2013 Published by Elsevier Inc.)
- Published
- 2014
- Full Text
- View/download PDF
42. Notch3 establishes brain vascular integrity by regulating pericyte number.
- Author
-
Wang Y, Pan L, Moens CB, and Appel B
- Subjects
- Animals, Animals, Genetically Modified, Blood-Brain Barrier growth & development, Blood-Brain Barrier metabolism, Brain blood supply, Cell Count, Cell Differentiation, Cell Proliferation, Cerebral Hemorrhage etiology, Cerebral Hemorrhage genetics, Cerebral Hemorrhage metabolism, Gene Expression Regulation, Developmental, Humans, Mutation, Receptor, Notch3, Receptor, Platelet-Derived Growth Factor beta genetics, Receptor, Platelet-Derived Growth Factor beta metabolism, Receptors, Notch deficiency, Receptors, Notch genetics, Signal Transduction, Zebrafish genetics, Zebrafish Proteins deficiency, Zebrafish Proteins genetics, Brain growth & development, Brain metabolism, Pericytes cytology, Pericytes metabolism, Receptors, Notch metabolism, Zebrafish growth & development, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
Brain pericytes are important regulators of brain vascular integrity, permeability and blood flow. Deficiencies of brain pericytes are associated with neonatal intracranial hemorrhage in human fetuses, as well as stroke and neurodegeneration in adults. Despite the important functions of brain pericytes, the mechanisms underlying their development are not well understood and little is known about how pericyte density is regulated across the brain. The Notch signaling pathway has been implicated in pericyte development, but its exact roles remain ill defined. Here, we report an investigation of the Notch3 receptor using zebrafish as a model system. We show that zebrafish brain pericytes express notch3 and that notch3 mutant zebrafish have a deficit of brain pericytes and impaired blood-brain barrier function. Conditional loss- and gain-of-function experiments provide evidence that Notch3 signaling positively regulates brain pericyte proliferation. These findings establish a new role for Notch signaling in brain vascular development whereby Notch3 signaling promotes expansion of the brain pericyte population.
- Published
- 2014
- Full Text
- View/download PDF
43. Retinal regeneration in adult zebrafish requires regulation of TGFβ signaling.
- Author
-
Lenkowski JR, Qin Z, Sifuentes CJ, Thummel R, Soto CM, Moens CB, and Raymond PA
- Subjects
- Animals, Animals, Genetically Modified, Cell Proliferation, Disease Models, Animal, Extracellular Matrix Proteins genetics, Extracellular Matrix Proteins metabolism, Eye Proteins genetics, Gliosis genetics, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Homeodomain Proteins genetics, Mutation genetics, Nerve Tissue Proteins genetics, Photic Stimulation adverse effects, Retina pathology, Retinal Degeneration etiology, Transforming Growth Factor beta genetics, Up-Regulation genetics, Zebrafish, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Homeobox Protein SIX3, Ependymoglial Cells metabolism, Nerve Regeneration physiology, Retinal Degeneration pathology, Signal Transduction physiology, Transforming Growth Factor beta metabolism
- Abstract
Müller glia are the resident radial glia in the vertebrate retina. The response of mammalian Müller glia to retinal damage often results in a glial scar and no functional replacement of lost neurons. Adult zebrafish Müller glia, in contrast, are considered tissue-specific stem cells that can self-renew and generate neurogenic progenitors to regenerate all retinal neurons after damage. Here, we demonstrate that regulation of TGFβ signaling by the corepressors Tgif1 and Six3b is critical for the proliferative response to photoreceptor destruction in the adult zebrafish retina. When function of these corepressors is disrupted, Müller glia and their progeny proliferate less, leading to a significant reduction in photoreceptor regeneration. Tgif1 expression and regulation of TGFβ signaling are implicated in the function of several types of stem cells, but this is the first demonstration that this regulatory network is necessary for regeneration of neurons., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
44. Notch3 signaling gates cell cycle entry and limits neural stem cell amplification in the adult pallium.
- Author
-
Alunni A, Krecsmarik M, Bosco A, Galant S, Pan L, Moens CB, and Bally-Cuif L
- Subjects
- Animals, Animals, Genetically Modified genetics, Animals, Genetically Modified metabolism, Brain cytology, Brain metabolism, Embryo, Nonmammalian cytology, Embryo, Nonmammalian metabolism, Gene Knockdown Techniques, Morpholinos, Neural Stem Cells cytology, Neuroglia metabolism, Neurons cytology, Neurons metabolism, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, Receptor, Notch3, Receptors, Notch genetics, Zebrafish genetics, Zebrafish metabolism, Zebrafish Proteins genetics, Cell Cycle, Cell Proliferation, Neural Stem Cells metabolism, Neuroglia cytology, Receptors, Notch metabolism, Signal Transduction, Zebrafish Proteins metabolism
- Abstract
Maintaining the homeostasis of germinal zones in adult organs is a fundamental but mechanistically poorly understood process. In particular, what controls stem cell activation remains unclear. We have previously shown that Notch signaling limits neural stem cell (NSC) proliferation in the adult zebrafish pallium. Combining pharmacological and genetic manipulations, we demonstrate here that long-term Notch invalidation primarily induces NSC amplification through their activation from quiescence and increased occurrence of symmetric divisions. Expression analyses, morpholino-mediated invalidation and the generation of a notch3-null mutant directly implicate Notch3 in these effects. By contrast, abrogation of notch1b function results in the generation of neurons at the expense of the activated NSC state. Together, our results support a differential involvement of Notch receptors along the successive steps of NSC recruitment. They implicate Notch3 at the top of this hierarchy to gate NSC activation and amplification, protecting the homeostasis of adult NSC reservoirs under physiological conditions.
- Published
- 2013
- Full Text
- View/download PDF
45. The first mecp2-null zebrafish model shows altered motor behaviors.
- Author
-
Pietri T, Roman AC, Guyon N, Romano SA, Washbourne P, Moens CB, de Polavieja GG, and Sumbre G
- Subjects
- Age Factors, Animals, Animals, Genetically Modified, Female, Humans, Male, Methyl-CpG-Binding Protein 2 genetics, Mutation genetics, Pregnancy, Zebrafish, Methyl-CpG-Binding Protein 2 deficiency, Methyl-CpG-Binding Protein 2 physiology, Models, Animal, Motor Activity physiology
- Abstract
Rett syndrome (RTT) is an X-linked neurodevelopmental disorder and one of the most common causes of mental retardation in affected girls. Other symptoms include a rapid regression of motor and cognitive skills after an apparently early normal development. Sporadic mutations in the transcription factor MECP2 has been shown to be present in more than 90% of the patients and several models of MeCP2-deficient mice have been created to understand the role of this gene. These models have pointed toward alterations in the maintenance of the central nervous system rather than its development, in line with the late onset of the disease in humans. However, the exact functions of MeCP2 remain difficult to delineate and the animal models have yielded contradictory results. Here, we present the first mecp2-null allele mutation zebrafish model. Surprisingly and in contrast to MeCP2-null mouse models, mecp2-null zebrafish are viable and fertile. They present nonetheless clear behavioral alterations during their early development, including spontaneous and sensory-evoked motor anomalies, as well as defective thigmotaxis.
- Published
- 2013
- Full Text
- View/download PDF
46. barx1 represses joints and promotes cartilage in the craniofacial skeleton.
- Author
-
Nichols JT, Pan L, Moens CB, and Kimmel CB
- Subjects
- Animals, Cartilage metabolism, Facial Bones embryology, Joints metabolism, Skull embryology, Transcription Factors genetics, Zebrafish Proteins genetics, Cartilage embryology, Facial Bones metabolism, Joints embryology, Skull metabolism, Transcription Factors metabolism, Zebrafish embryology, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
The evolution of joints, which afford skeletal mobility, was instrumental in vertebrate success. Here, we explore the molecular genetics and cell biology that govern jaw joint development. Genetic manipulation experiments in zebrafish demonstrate that functional loss, or gain, of the homeobox-containing gene barx1 produces gain, or loss, of joints, respectively. Ectopic joints in barx1 mutant animals are present in every pharyngeal segment, and are associated with disrupted attachment of bone, muscles and teeth. We find that ectopic joints develop at the expense of cartilage. Time-lapse experiments suggest that barx1 controls the skeletal precursor cell choice between differentiating into cartilage versus joint cells. We discovered that barx1 functions in this choice, in part, by regulating the transcription factor hand2. We further show that hand2 feeds back to negatively regulate barx1 expression. We consider the possibility that changes in barx1 function in early vertebrates were among the key innovations fostering the evolution of skeletal joints.
- Published
- 2013
- Full Text
- View/download PDF
47. Tardbpl splicing rescues motor neuron and axonal development in a mutant tardbp zebrafish.
- Author
-
Hewamadduma CA, Grierson AJ, Ma TP, Pan L, Moens CB, Ingham PW, Ramesh T, and Shaw PJ
- Subjects
- Amyotrophic Lateral Sclerosis embryology, Amyotrophic Lateral Sclerosis metabolism, Animals, DNA-Binding Proteins metabolism, Disease Models, Animal, Female, Gene Knockout Techniques, Humans, Male, Mutation, Zebrafish embryology, Zebrafish genetics, Zebrafish Proteins metabolism, Amyotrophic Lateral Sclerosis genetics, Axons metabolism, DNA-Binding Proteins genetics, Motor Neurons metabolism, RNA Splicing, Zebrafish metabolism, Zebrafish Proteins genetics
- Abstract
Mutations in the transactive response DNA binding protein-43 (TARDBP/TDP-43) gene, which regulates transcription and splicing, causes a familial form of amyotrophic lateral sclerosis (ALS). Here, we characterize and report the first tardbp mutation in zebrafish, which introduces a premature stop codon (Y220X), eliminating expression of the Tardbp protein. Another TARDBP ortholog, tardbpl, in zebrafish is shown to encode a Tardbp-like protein which is truncated compared with Tardbp itself and lacks part of the C-terminal glycine-rich domain (GRD). Here, we show that tardbp mutation leads to the generation of a novel tardbpl splice form (tardbpl-FL) capable of making a full-length Tardbp protein (Tardbpl-FL), which compensates for the loss of Tardbp. This finding provides a novel in vivo model to study TDP-43-mediated splicing regulation. Additionally, we show that elimination of both zebrafish TARDBP orthologs results in a severe motor phenotype with shortened motor axons, locomotion defects and death at around 10 days post fertilization. The Tardbp/Tardbpl knockout model generated in this study provides an excellent in vivo system to study the role of the functional loss of Tardbp and its involvement in ALS pathogenesis.
- Published
- 2013
- Full Text
- View/download PDF
48. RNA-seq-based mapping and candidate identification of mutations from forward genetic screens.
- Author
-
Miller AC, Obholzer ND, Shah AN, Megason SG, and Moens CB
- Subjects
- Animals, Computational Biology methods, Gene Expression Regulation, Genetic Linkage, Genome, High-Throughput Nucleotide Sequencing, Internet, Polymorphism, Single Nucleotide, RNA Splicing, Reproducibility of Results, Zebrafish genetics, Chromosome Mapping, Genetic Testing methods, Mutation, Sequence Analysis, RNA methods
- Abstract
Forward genetic screens have elucidated molecular pathways required for innumerable aspects of life; however, identifying the causal mutations from such screens has long been the bottleneck in the process, particularly in vertebrates. We have developed an RNA-seq-based approach that identifies both the region of the genome linked to a mutation and candidate lesions that may be causal for the phenotype of interest. We show that our method successfully identifies zebrafish mutations that cause nonsense or missense changes to codons, alter transcript splicing, or alter gene expression levels. Furthermore, we develop an easily accessible bioinformatics pipeline allowing for implementation of all steps of the method. Overall, we show that RNA-seq is a fast, reliable, and cost-effective method to map and identify mutations that will greatly facilitate the power of forward genetics in vertebrate models.
- Published
- 2013
- Full Text
- View/download PDF
49. Wnt-dependent epithelial transitions drive pharyngeal pouch formation.
- Author
-
Choe CP, Collazo A, Trinh le A, Pan L, Moens CB, and Crump JG
- Subjects
- Adherens Junctions metabolism, Animals, Embryonic Development, Epithelium growth & development, Gene Expression Regulation, Developmental, Mesoderm growth & development, Mesoderm metabolism, Pharynx growth & development, Pharynx metabolism, Signal Transduction, Wnt Signaling Pathway, rac1 GTP-Binding Protein genetics, rac1 GTP-Binding Protein metabolism, Body Patterning genetics, Body Patterning physiology, Wnt Proteins genetics, Wnt Proteins metabolism, Wnt4 Protein genetics, Wnt4 Protein metabolism, Zebrafish genetics, Zebrafish growth & development, Zebrafish Proteins genetics, Zebrafish Proteins metabolism
- Abstract
The pharyngeal pouches, which form by budding of the foregut endoderm, are essential for segmentation of the vertebrate face. To date, the cellular mechanism and segmental nature of such budding have remained elusive. Here, we find that Wnt11r and Wnt4a from the head mesoderm and ectoderm, respectively, play distinct roles in the segmental formation of pouches in zebrafish. Time-lapse microscopy, combined with mutant and tissue-specific transgenic experiments, reveal requirements of Wnt signaling in two phases of endodermal epithelial transitions. Initially, Wnt11r and Rac1 destabilize the endodermal epithelium to promote the lateral movement of pouch-forming cells. Next, Wnt4a and Cdc42 signaling induce the rearrangement of maturing pouch cells into bilayers through junctional localization of the Alcama immunoglobulin-domain protein, which functions to restabilize adherens junctions. We propose that this dynamic control of epithelial morphology by Wnt signaling may be a common theme for the budding of organ anlagen from the endoderm., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
50. The differentiation and movement of presomitic mesoderm progenitor cells are controlled by Mesogenin 1.
- Author
-
Fior R, Maxwell AA, Ma TP, Vezzaro A, Moens CB, Amacher SL, Lewis J, and Saúde L
- Subjects
- Animals, Animals, Genetically Modified, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Tracking, Embryonic Development genetics, Embryonic Stem Cells metabolism, Mesoderm cytology, Mesoderm metabolism, Somites embryology, Somites metabolism, T-Box Domain Proteins genetics, T-Box Domain Proteins metabolism, T-Box Domain Proteins physiology, Tail embryology, Torso embryology, Zebrafish embryology, Zebrafish genetics, Zebrafish metabolism, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Basic Helix-Loop-Helix Transcription Factors physiology, Cell Differentiation genetics, Cell Movement genetics, Embryonic Stem Cells physiology, Mesoderm embryology, Zebrafish Proteins physiology
- Abstract
Somites are formed from the presomitic mesoderm (PSM) and give rise to the axial skeleton and skeletal muscles. The PSM is dynamic; somites are generated at the anterior end, while the posterior end is continually renewed with new cells entering from the tailbud progenitor region. Which genes control the conversion of tailbud progenitors into PSM and how is this process coordinated with cell movement? Using loss- and gain-of-function experiments and heat-shock transgenics we show in zebrafish that the transcription factor Mesogenin 1 (Msgn1), acting with Spadetail (Spt), has a central role. Msgn1 allows progression of the PSM differentiation program by switching off the progenitor maintenance genes ntl, wnt3a, wnt8 and fgf8 in the future PSM cells as they exit from the tailbud, and subsequently induces expression of PSM markers such as tbx24. msgn1 is itself positively regulated by Ntl/Wnt/Fgf, creating a negative-feedback loop that might be crucial to regulate homeostasis of the progenitor population until somitogenesis ends. Msgn1 drives not only the changes in gene expression in the nascent PSM cells but also the movements by which they stream out of the tailbud into the PSM. Loss of Msgn1 reduces the flux of cells out of the tailbud, producing smaller somites and an enlarged tailbud, and, by delaying exhaustion of the progenitor population, results in supernumerary tail somites. Through its combined effects on gene expression and cell movement, Msgn1 (with Spt) plays a key role both in genesis of the paraxial mesoderm and in maintenance of the progenitor population from which it derives.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.