Bio

Academic Appointments


Administrative Appointments


  • Senior Associate Dean for Graduate Education and Postdoctoral Affairs, Stanford University School of Medicine (2015 - Present)
  • Chair, Department of Developmental Biology, Stanford University (2012 - 2015)

Honors & Awards


  • Pew Scholars Award in the Biomedical Sciences, Pew Charitable Trusts (1998-2002)
  • Rita Allen Foundation Scholars Award, Rita Allen Foundation (2002-2004)
  • Fellow, American Association for the Advancement of Science (2014)
  • Catherine R. Kennedy and Daniel L. Grossman Fellow in Human Biology, Stanford University (2014-Present)
  • Award for Excellence in Faculty Advising in Human Biology, Stanford University (2017)

Professional Education


  • Ph.D., Stanford University, Biochemistry (1993)
  • B.S., University of Florida, Microbiology (1987)

Research & Scholarship

Current Research and Scholarly Interests


Our research focuses on the development and function of glial cells in the vertebrate nervous system. Glia are non-neuronal cells with many essential functions, ranging from forming the myelin sheath to defending the brain against infection.

One of our goals is to use genetic approaches in zebrafish to discover new genes with essential functions in the glial cells that form the myelin sheath, which allows for rapid axonal conduction in vertebrates. Disruption of myelin underlies important human diseases, including Multiple Sclerosis and peripheral neuropathies. The formation of myelin, which involves reciprocal signaling between neurons and glial cells, a dramatic morphological transformation of the glial cells, and organization of the axon into different specialized domains, is fascinating but nonetheless poorly understood.

In genetic screens, we have identified mutations in more than 15 different genes that have specific functions in the development of myelinated axons. Among these are a novel G-protein coupled receptor that instructs Schwann cells to make myelin in peripheral nerves, receptors that control migration of glial cells along growing axons, a kinesin motor protein that is essential for mRNA localization and normal membrane compaction in myelinating oligodendrocytes, and a transcription factor that regulates the migration of the cells that form myelin in the brain and spinal cord.

Another goal of our research is to identify new genes that regulate microglia, which are specialized macrophages that are dedicated to the immune defense of the brain. Microglia also have critical roles in regulating synaptic connectivity and engulfing dead neurons to maintain homeostasis in the brain. Microglial dysfunction has been implicated a wide array of disorders, including autism and Alzheimer disease.

Starting with screens for mutants with abnormal microglia, we have identified novel genes regulate microglial development and function. Examples include a NOD-like receptor that suppresses inappropriate inflammation, a phosphate exporter that functions specifically in microglia and other tissue macrophages, and a regulator of lysosomal action that allows microglia to digest material that they engulf.

These projects provide new insights into glial cell development and function, generate new animal models of human disease, define pathways that may be disrupted in disease, and may provide new avenues toward therapies for diseases of glia.

Teaching

2017-18 Courses


Stanford Advisees


Graduate and Fellowship Programs


Publications

All Publications


  • An Anti-inflammatory NOD-like Receptor Is Required for Microglia Development CELL REPORTS Shiau, C. E., Monk, K. R., Joo, W., Talbot, W. S. 2013; 5 (5): 1342-1352

    Abstract

    Microglia are phagocytic cells that form the basis of the brain's immune system. They derive from primitive macrophages that migrate into the brain during embryogenesis, but the genetic control of microglial development remains elusive. Starting with a genetic screen in zebrafish, we show that the noncanonical NOD-like receptor (NLR) nlrc3-like is essential for microglial formation. Although most NLRs trigger inflammatory signaling, nlrc3-like acts cell autonomously in microglia precursor cells to suppress unwarranted inflammation in the absence of overt immune challenge. In nlrc3-like mutants, primitive macrophages initiate a systemic inflammatory response with increased proinflammatory cytokines and actively aggregate instead of migrating into the brain to form microglia. NLRC3-like requires both its pyrin and NACHT domains, and it can bind the inflammasome component apoptosis-associated speck-like protein. Our studies suggest that NLRC3-like may regulate the inflammasome and other inflammatory pathways. Together, these results demonstrate that NLRC3-like prevents inappropriate macrophage activation, thereby allowing normal microglial development.

    View details for DOI 10.1016/j.celrep.2013.11.004

    View details for Web of Science ID 000328266400017

    View details for PubMedID 24316075

  • Gpr126 is essential for peripheral nerve development and myelination in mammals DEVELOPMENT Monk, K. R., Oshima, K., Joers, S., Heller, S., Talbot, W. S. 2011; 138 (13): 2673-2680

    Abstract

    In peripheral nerves, Schwann cells form the myelin sheath that insulates axons and allows rapid propagation of action potentials. Although a number of regulators of Schwann cell development are known, the signaling pathways that control myelination are incompletely understood. In this study, we show that Gpr126 is essential for myelination and other aspects of peripheral nerve development in mammals. A mutation in Gpr126 causes a severe congenital hypomyelinating peripheral neuropathy in mice, and expression of differentiated Schwann cell markers, including Pou3f1, Egr2, myelin protein zero and myelin basic protein, is reduced. Ultrastructural studies of Gpr126-/- mice showed that axonal sorting by Schwann cells is delayed, Remak bundles (non-myelinating Schwann cells associated with small caliber axons) are not observed, and Schwann cells are ultimately arrested at the promyelinating stage. Additionally, ectopic perineurial fibroblasts form aberrant fascicles throughout the endoneurium of the mutant sciatic nerve. This analysis shows that Gpr126 is required for Schwann cell myelination in mammals, and defines new roles for Gpr126 in axonal sorting, formation of mature non-myelinating Schwann cells and organization of the perineurium.

    View details for DOI 10.1242/dev.062224

    View details for Web of Science ID 000291348700005

    View details for PubMedID 21613327

  • A G Protein-Coupled Receptor Is Essential for Schwann Cells to Initiate Myelination SCIENCE Monk, K. R., Naylor, S. G., Glenn, T. D., Mercurio, S., Perlin, J. R., Dominguez, C., Moens, C. B., Talbot, W. S. 2009; 325 (5946): 1402-1405

    Abstract

    The myelin sheath allows axons to conduct action potentials rapidly in the vertebrate nervous system. Axonal signals activate expression of specific transcription factors, including Oct6 and Krox20, that initiate myelination in Schwann cells. Elevation of cyclic adenosine monophosphate (cAMP) can mimic axonal contact in vitro, but the mechanisms that regulate cAMP levels in vivo are unknown. Using mutational analysis in zebrafish, we found that the G protein-coupled receptor Gpr126 is required autonomously in Schwann cells for myelination. In gpr126 mutants, Schwann cells failed to express oct6 and krox20 and were arrested at the promyelinating stage. Elevation of cAMP in gpr126 mutants, but not krox20 mutants, could restore myelination. We propose that Gpr126 drives the differentiation of promyelinating Schwann cells by elevating cAMP levels, thereby triggering Oct6 expression and myelination.

    View details for DOI 10.1126/science.1173474

    View details for Web of Science ID 000269699100041

    View details for PubMedID 19745155

  • The Rag-Ragulator Complex Regulates Lysosome Function and Phagocytic Flux in Microglia. Cell reports Shen, K., Sidik, H., Talbot, W. S. 2016; 14 (3): 547-559

    Abstract

    Microglia are resident macrophages of the CNS that are essential for phagocytosis of apoptotic neurons and weak synapses during development. We show that RagA and Lamtor4, two components of the Rag-Ragulator complex, are essential regulators of lysosomes in microglia. In zebrafish lacking RagA function, microglia exhibit an expanded lysosomal compartment, but they are unable to properly digest apoptotic neuronal debris. Previous biochemical studies have placed the Rag-Ragulator complex upstream of mTORC1 activation in response to cellular nutrient availability. Nonetheless, RagA and mTOR mutant zebrafish have distinct phenotypes, indicating that the Rag-Ragulator complex has functions independent of mTOR signaling. Our analysis reveals an essential role of the Rag-Ragulator complex in proper lysosome function and phagocytic flux in microglia.

    View details for DOI 10.1016/j.celrep.2015.12.055

    View details for PubMedID 26774477

  • Mutations of GPR126 Are Responsible for Severe Arthrogryposis Multiplex Congenita AMERICAN JOURNAL OF HUMAN GENETICS Ravenscroft, G., Nolent, F., Rajagopalan, S., Meireles, A. M., Paavola, K. J., Gaillard, D., Alanio, E., Buckland, M., Arbuckle, S., Krivanek, M., Maluenda, J., Pannell, S., Gooding, R., Ong, R. W., Allcock, R. J., Carvalho, E. D., Carvalho, M. D., Kok, F., Talbot, W. S., Melki, J., Laing, N. G. 2015; 96 (6): 955-961

    Abstract

    Arthrogryposis multiplex congenita is defined by the presence of contractures across two or more major joints and results from reduced or absent fetal movement. Here, we present three consanguineous families affected by lethal arthrogryposis multiplex congenita. By whole-exome or targeted exome sequencing, it was shown that the probands each harbored a different homozygous mutation (one missense, one nonsense, and one frameshift mutation) in GPR126. GPR126 encodes G-protein-coupled receptor 126, which has been shown to be essential for myelination of axons in the peripheral nervous system in fish and mice. A previous study reported that Gpr126(-/-) mice have a lethal arthrogryposis phenotype. We have shown that the peripheral nerves in affected individuals from one family lack myelin basic protein, suggesting that this disease in affected individuals is due to defective myelination of the peripheral axons during fetal development. Previous work has suggested that autoproteolytic cleavage is important for activating GPR126 signaling, and our biochemical assays indicated that the missense substitution (p.Val769Glu [c.2306T>A]) impairs autoproteolytic cleavage of GPR126. Our data indicate that GPR126 is critical for myelination of peripheral nerves in humans. This study adds to the literature implicating defective axoglial function as a key cause of severe arthrogryposis multiplex congenita and suggests that GPR126 mutations should be investigated in individuals affected by this disorder.

    View details for DOI 10.1016/j.ajhg.2015.04.014

    View details for Web of Science ID 000355931200009

    View details for PubMedID 26004201

    View details for PubMedCentralID PMC4457946

  • Glial Cell Development and Function in Zebrafish COLD SPRING HARBOR PERSPECTIVES IN BIOLOGY Lyons, D. A., Talbot, W. S. 2015; 7 (2)
  • Differential Requirement for irf8 in Formation of Embryonic and Adult Macrophages in Zebrafish. PloS one Shiau, C. E., Kaufman, Z., Meireles, A. M., Talbot, W. S. 2015; 10 (1)

    Abstract

    Interferon regulatory factor 8 (Irf8) is critical for mammalian macrophage development and innate immunity, but its role in teleost myelopoiesis remains incompletely understood. In particular, genetic tools to analyze the role of Irf8 in zebrafish macrophage development at larval and adult stages are lacking. We generated irf8 null mutants in zebrafish using TALEN-mediated targeting. Our analysis defines different requirements for irf8 at different stages. irf8 is required for formation of all macrophages during primitive and transient definitive hematopoiesis, but not during adult-phase definitive hematopoiesis starting at 5-6 days postfertilization. At early stages, irf8 mutants have excess neutrophils and excess cell death in pu.1-expressing myeloid cells. Macrophage fates were recovered in irf8 mutants after wildtype irf8 expression in neutrophil and macrophage lineages, suggesting that irf8 regulates macrophage specification and survival. In juvenile irf8 mutant fish, mature macrophages are present, but at numbers significantly reduced compared to wildtype, indicating an ongoing requirement for irf8 after embryogenesis. As development progresses, tissue macrophages become apparent in zebrafish irf8 mutants, with the possible exception of microglia. Our study defines distinct requirement for irf8 in myelopoiesis before and after transition to the adult hematopoietic system.

    View details for DOI 10.1371/journal.pone.0117513

    View details for PubMedID 25615614

    View details for PubMedCentralID PMC4304715

  • The Phosphate Exporter xpr1b Is Required for Differentiation of Tissue-Resident Macrophages CELL REPORTS Meireles, A. M., Shiau, C. E., Guenther, C. A., Sidik, H., Kingsley, D. M., Talbot, W. S. 2014; 8 (6): 1659-1667
  • Type IV collagen is an activating ligand for the adhesion G protein-coupled receptor GPR126. Science signaling Paavola, K. J., Sidik, H., Zuchero, J. B., Eckart, M., Talbot, W. S. 2014; 7 (338): ra76-?

    Abstract

    GPR126 is an orphan heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptor (GPCR) that is essential for the development of diverse organs. We found that type IV collagen, a major constituent of the basement membrane, binds to Gpr126 and activates its signaling function. Type IV collagen stimulated the production of cyclic adenosine monophosphate in rodent Schwann cells, which require Gpr126 activity to differentiate, and in human embryonic kidney (HEK) 293 cells expressing exogenous Gpr126. Type IV collagen specifically bound to the extracellular amino-terminal region of Gpr126 containing the CUB (complement, Uegf, Bmp1) and pentraxin domains. Gpr126 derivatives lacking the entire amino-terminal region were constitutively active, suggesting that this region inhibits signaling and that ligand binding relieves this inhibition to stimulate receptor activity. A new zebrafish mutation that truncates Gpr126 after the CUB and pentraxin domains disrupted development of peripheral nerves and the inner ear. Thus, our findings identify type IV collagen as an activating ligand for GPR126, define its mechanism of activation, and highlight a previously unrecognized signaling function of type IV collagen in basement membranes.

    View details for DOI 10.1126/scisignal.2005347

    View details for PubMedID 25118328

  • Glial cell development and function in zebrafish. Cold Spring Harbor perspectives in biology Lyons, D. A., Talbot, W. S. 2014; 7 (2)

    Abstract

    The zebrafish is a premier vertebrate model system that offers many experimental advantages for in vivo imaging and genetic studies. This review provides an overview of glial cell types in the central and peripheral nervous system of zebrafish. We highlight some recent work that exploited the strengths of the zebrafish system to increase the understanding of the role of Gpr126 in Schwann cell myelination and illuminate the mechanisms controlling oligodendrocyte development and myelination. We also summarize similarities and differences between zebrafish radial glia and mammalian astrocytes and consider the possibility that their distinct characteristics may represent extremes in a continuum of cell identity. Finally, we focus on the emergence of zebrafish as a model for elucidating the development and function of microglia. These recent studies have highlighted the power of the zebrafish system for analyzing important aspects of glial development and function.

    View details for DOI 10.1101/cshperspect.a020586

    View details for PubMedID 25395296

  • Signals regulating myelination in peripheral nerves and the Schwann cell response to injury. Current opinion in neurobiology Glenn, T. D., Talbot, W. S. 2013; 23 (6): 1041-1048

    Abstract

    In peripheral nerves, Schwann cells form myelin, which facilitates the rapid conduction of action potentials along axons in the vertebrate nervous system. Myelinating Schwann cells are derived from neural crest progenitors in a step-wise process that is regulated by extracellular signals and transcription factors. In addition to forming the myelin sheath, Schwann cells orchestrate much of the regenerative response that occurs after injury to peripheral nerves. In response to injury, myelinating Schwann cells dedifferentiate into repair cells that are essential for axonal regeneration, and then redifferentiate into myelinating Schwann cells to restore nerve function. Although this remarkable plasticity has long been recognized, many questions remain unanswered regarding the signaling pathways regulating both myelination and the Schwann cell response to injury.

    View details for DOI 10.1016/j.conb.2013.06.010

    View details for PubMedID 23896313

  • Analysis of Gpr126 function defines distinct mechanisms controlling the initiation and maturation of myelin DEVELOPMENT Glenn, T. D., Talbot, W. S. 2013; 140 (15): 3167-3175

    Abstract

    In peripheral nerves, Schwann cells form the myelin sheath, which allows the efficient propagation of action potentials along axons. The transcription factor Krox20 regulates the initiation of myelination in Schwann cells and is also required to maintain mature myelin. The adhesion G protein-coupled receptor (GPCR) Gpr126 is essential for Schwann cells to initiate myelination, but previous studies have not addressed the role of Gpr126 signaling in myelin maturation and maintenance. Through analysis of Gpr126 in zebrafish, we define two distinct mechanisms controlling the initiation and maturation of myelin. We show that gpr126 mutant Schwann cells elaborate mature myelin sheaths and maintain krox20 expression for months, provided that the early signaling defect is bypassed by transient elevation of cAMP. At the onset of myelination, Gpr126 and protein kinase A (PKA) function as a switch that allows Schwann cells to initiate krox20 expression and myelination. After myelination is initiated, krox20 expression is maintained and myelin maturation proceeds independently of Gpr126 signaling. Transgenic analysis indicates that the Krox20 cis-regulatory myelinating Schwann cell element (MSE) becomes active at the onset of myelination and that this activity is dependent on Gpr126 signaling. Activity of the MSE declines after initiation, suggesting that other elements are responsible for maintaining krox20 expression in mature nerves. We also show that elevated cAMP does not initiate myelination in the absence of functional Neuregulin 1 (Nrg1) signaling. These results indicate that the mechanisms regulating the initiation of myelination are distinct from those mediating the maturation and maintenance of myelin.

    View details for DOI 10.1242/dev.093401

    View details for Web of Science ID 000321864900010

    View details for PubMedID 23804499

  • Mutation of sec63 in zebrafish causes defects in myelinated axons and liver pathology DISEASE MODELS & MECHANISMS Monk, K. R., Voas, M. G., Franzini-Armstrong, C., Hakkinen, I. S., Talbot, W. S. 2013; 6 (1): 135-145

    Abstract

    Mutations in SEC63 cause polycystic liver disease in humans. Sec63 is a member of the endoplasmic reticulum (ER) translocon machinery, although it is unclear how mutations in SEC63 lead to liver cyst formation in humans. Here, we report the identification and characterization of a zebrafish sec63 mutant, which was discovered in a screen for mutations that affect the development of myelinated axons. Accordingly, we show that disruption of sec63 in zebrafish leads to abnormalities in myelinating glia in both the central and peripheral nervous systems. In the vertebrate nervous system, segments of myelin are separated by the nodes of Ranvier, which are unmyelinated regions of axonal membrane containing a high density of voltage-gated sodium channels. We show that sec63 mutants have morphologically abnormal and reduced numbers of clusters of voltage-gated sodium channels in the spinal cord and along peripheral nerves. Additionally, we observed reduced myelination in both the central and peripheral nervous systems, as well as swollen ER in myelinating glia. Markers of ER stress are upregulated in sec63 mutants. Finally, we show that sec63 mutants develop liver pathology. As in glia, the primary defect, detectable at 5 dpf, is fragmentation and swelling of the ER, indicative of accumulation of proteins in the lumen. At 8 dpf, ER swelling is severe; other pathological features include disrupted bile canaliculi, altered cytoplasmic matrix and accumulation of large lysosomes. Together, our analyses of sec63 mutant zebrafish highlight the possible role of ER stress in polycystic liver disease and suggest that these mutants will serve as a model for understanding the pathophysiology of this disease and other abnormalities involving ER stress.

    View details for DOI 10.1242/dmm.009217

    View details for Web of Science ID 000314865700015

    View details for PubMedID 22864019

  • Scube/You activity mediates release of dually lipid-modified Hedgehog signal in soluble form GENES & DEVELOPMENT Creanga, A., Glenn, T. D., Mann, R. K., Saunders, A. M., Talbot, W. S., Beachy, P. A. 2012; 26 (12): 1312-1325

    Abstract

    Owing to their covalent modification by cholesterol and palmitate, Hedgehog (Hh) signaling proteins are localized predominantly to the plasma membrane of expressing cells. Yet Hh proteins are also capable of mobilizing to and eliciting direct responses from distant cells. The zebrafish you gene, identified genetically >15 years ago, was more recently shown to encode a secreted glycoprotein that acts cell-nonautonomously in the Hh signaling pathway by an unknown mechanism. We investigated the function of the protein encoded by murine Scube2, an ortholog of you, and found that it mediates release in soluble form of the mature, cholesterol- and palmitate-modified Sonic hedgehog protein signal (ShhNp) when added to cultured cells or purified detergent-resistant membrane microdomains containing ShhNp. The efficiency of Scube2-mediated release of ShhNp is enhanced by the palmitate adduct of ShhNp and by coexpression in ShhNp-producing cells of mDispatchedA (mDispA), a transporter-like protein with a previously defined role in the release of lipid-modified Hh signals. The structural determinants of Scube2 required for its activity in cultured cell assays match those required for rescue of you mutant zebrafish embryos, and we thus conclude that the role of Scube/You proteins in Hh signaling in vivo is to facilitate the release and mobilization of Hh proteins for distant action.

    View details for DOI 10.1101/gad.191866.112

    View details for Web of Science ID 000305485300006

    View details for PubMedID 22677548

  • Neuronal Neuregulin 1 type III directs Schwann cell migration DEVELOPMENT Perlin, J. R., Lush, M. E., Stephens, W. Z., Piotrowski, T., Talbot, W. S. 2011; 138 (21): 4639-4648

    Abstract

    During peripheral nerve development, each segment of a myelinated axon is matched with a single Schwann cell. Tight regulation of Schwann cell movement, proliferation and differentiation is essential to ensure that these glial cells properly associate with axons. ErbB receptors are required for Schwann cell migration, but the operative ligand and its mechanism of action have remained unknown. We demonstrate that zebrafish Neuregulin 1 (Nrg1) type III, which signals through ErbB receptors, controls Schwann cell migration in addition to its previously known roles in proliferation and myelination. Chimera analyses indicate that ErbB receptors are required in all migrating Schwann cells, and that Nrg1 type III is required in neurons for migration. Surprisingly, expression of the ligand in a few axons is sufficient to induce migration along a chimeric nerve constituted largely of nrg1 type III mutant axons. These studies also reveal a mechanism that allows Schwann cells to fasciculate axons regardless of nrg1 type III expression. Time-lapse imaging of transgenic embryos demonstrated that misexpression of human NRG1 type III results in ectopic Schwann cell migration, allowing them to aberrantly enter the central nervous system. These results demonstrate that Nrg1 type III is an essential signal that controls Schwann cell migration to ensure that these glia are present in the correct numbers and positions in developing nerves.

    View details for DOI 10.1242/dev.068072

    View details for Web of Science ID 000296060100008

    View details for PubMedID 21965611

  • ErbB Signaling Has a Role in Radial Sorting Independent of Schwann Cell Number GLIA Raphael, A. R., Lyons, D. A., Talbot, W. S. 2011; 59 (7): 1047-1055

    Abstract

    In the peripheral nervous system, Schwann cells make myelin, a specialized sheath that is essential for rapid axonal conduction of action potentials. Immature Schwann cells initially interact with many axons, but, through a process termed radial sorting, eventually interact with one segment of a single axon as promyelinating Schwann cells. Previous studies have identified genes that are required for Schwann cell process extension and proliferation during radial sorting. Previous analyses also show that ErbB signaling is required for Schwann cell proliferation, myelination, radial sorting, and the proper formation of unmyelinated Remak bundles. Because ErbB signaling and Schwann cell proliferation are both required during radial sorting, we sought to determine if the primary function of ErbB signaling in this process is to regulate Schwann cell proliferation or if ErbB signaling also controls other aspects of radial sorting. To address this question, we applied small molecule inhibitors in vivo in zebrafish to independently block ErbB signaling and proliferation. Ultrastructural analysis of treated animals revealed that both ErbB signaling and Schwann cell proliferation are required for radial sorting in vivo. ErbB signaling, however, is required for Schwann cell process extension, while Schwann cell proliferation is not. These results provide in vivo evidence that ErbB signaling plays a direct role in process extension during radial sorting, in addition to its role in regulating Schwann cell proliferation.

    View details for DOI 10.1002/glia.21175

    View details for Web of Science ID 000291434500004

    View details for PubMedID 21491500

  • Schwann cells reposition a peripheral nerve to isolate it from postembryonic remodeling of its targets DEVELOPMENT Raphael, A. R., Perlin, J. R., Talbot, W. S. 2010; 137 (21): 3643-3649

    Abstract

    Although much is known about the initial construction of the peripheral nervous system (PNS), less well understood are the processes that maintain the position and connections of nerves during postembryonic growth. Here, we show that the posterior lateral line nerve in zebrafish initially grows in the epidermis and then rapidly transitions across the epidermal basement membrane into the subepidermal space. Our experiments indicate that Schwann cells, which myelinate axons in the PNS, are required to reposition the nerve. In mutants lacking Schwann cells, the nerve is mislocalized and the axons remain in the epidermis. Transplanting wild-type Schwann cells into these mutants rescues the position of the nerve. Analysis of chimeric embryos suggests that the process of nerve relocalization involves two discrete steps - the degradation and recreation of the epidermal basement membrane. Although the outgrowth of axons is normal in mutants lacking Schwann cells, the nerve becomes severely disorganized at later stages. In wild-type embryos, exclusion of the nerve from the epidermis isolates axons from migration of their targets (sensory neuromasts) within the epidermis. Without Schwann cells, axons remain within the epidermis and are dragged along with the migrating neuromasts. Our analysis of the posterior lateral line system defines a new process in which Schwann cells relocate a nerve beneath the epidermal basement membrane to insulate axons from the postembryonic remodeling of their targets.

    View details for DOI 10.1242/dev.057521

    View details for Web of Science ID 000283669300012

    View details for PubMedID 20876648

  • Schwann Cells Inhibit Ectopic Clustering of Axonal Sodium Channels JOURNAL OF NEUROSCIENCE Voas, M. G., Glenn, T. D., Raphael, A. R., Talbot, W. S. 2009; 29 (46): 14408-14414

    Abstract

    The clustering of voltage-gated sodium channels at the axon initial segment (AIS) and nodes of Ranvier is essential for the initiation and propagation of action potentials in myelinated axons. Sodium channels localize to the AIS through an axon-intrinsic mechanism driven by ankyrin G, while clustering at the nodes requires cues from myelinating glia that interact with axonal neurofascin186 (Sherman et al., 2005; Dzhashiashvili et al., 2007; Yang et al., 2007). Here, we report that in zebrafish mutants lacking Schwann cells in peripheral nerves (erbb2, erbb3, and sox10/colorless), axons form numerous aberrant sodium channel clusters throughout their length. Morpholino knockdown of ankyrin G, but not neurofascin, reduces the number of sodium channel clusters in Schwann cell-deficient mutants, suggesting that these aberrant clusters form by an axon-intrinsic mechanism. We also find that gpr126 mutants, in which Schwann cells are arrested at the promyelinating stage (Monk et al., 2009), are deficient in the clustering of neurofascin at the nodes of Ranvier. When Schwann cell migration in gpr126 mutants is blocked, there is an increase in the number of neurofascin clusters in peripheral axons. Our results suggest that Schwann cells inhibit the ability of ankyrin G to cluster sodium channels at ectopic locations, restricting its activity to the AIS and nodes of Ranvier.

    View details for DOI 10.1523/JNEUROSCI.0841-09.2009

    View details for Web of Science ID 000271944500004

    View details for PubMedID 19923275

  • Genetic dissection of myellinated axons in zebrafish CURRENT OPINION IN NEUROBIOLOGY Monk, K. R., Talbot, W. S. 2009; 19 (5): 486-490

    Abstract

    In the vertebrate nervous system, the myelin sheath allows for rapid and efficient conduction of action potentials along axons. Despite the essential function of myelin, many questions remain unanswered about the mechanisms that govern the development of myelinated axons. The fundamental properties of myelin are widely shared among vertebrates, and the zebrafish has emerged as a powerful system to study myelination in vivo. This review will highlight recent advances from genetic screens in zebrafish, including the discovery of the role of kif1b in mRNA localization in myelinating oligodendrocytes.

    View details for DOI 10.1016/j.conb.2009.08.006

    View details for Web of Science ID 000272922900005

    View details for PubMedID 19740648

  • Kif1b is essential for mRNA localization in oligodendrocytes and development of myelinated axons NATURE GENETICS Lyons, D. A., Naylor, S. G., Scholze, A., Talbot, W. S. 2009; 41 (7): 854-U121

    Abstract

    The kinesin motor protein Kif1b has previously been implicated in the axonal transport of mitochondria and synaptic vesicles. More recently, KIF1B has been associated with susceptibility to multiple sclerosis (MS). Here we show that Kif1b is required for the localization of mbp (myelin basic protein) mRNA to processes of myelinating oligodendrocytes in zebrafish. We observe the ectopic appearance of myelin-like membrane in kif1b mutants, coincident with the ectopic localization of myelin proteins in kif1b mutant oligodendrocyte cell bodies. These observations suggest that oligodendrocytes localize certain mRNA molecules, namely those encoding small basic proteins such as MBP, to prevent aberrant effects of these proteins elsewhere in the cell. We also find that Kif1b is required for outgrowth of some of the longest axons in the peripheral and central nervous systems. Our data demonstrate previously unknown functions of kif1b in vivo and provide insights into its possible roles in MS.

    View details for DOI 10.1038/ng.376

    View details for Web of Science ID 000267786200020

    View details for PubMedID 19503091

  • Axonal domains: Role for paranodal junction in node of Ranvier assembly CURRENT BIOLOGY Lyons, D. A., Talbot, W. S. 2008; 18 (18): R876-R879

    Abstract

    A new study shows that communication between axons and glia at the paranodal junction can orchestrate the formation of the node of Ranvier.

    View details for DOI 10.1016/j.cub.2008.07.070

    View details for Web of Science ID 000259523600018

    View details for PubMedID 18812088

  • KBP is essential for axonal structure, outgrowth and maintenance in zebrafish, providing insight into the cellular basis of Goldberg-Shprintzen syndrome DEVELOPMENT Lyons, D. A., Naylor, S. G., Mercurio, S., Dominguez, C., Talbot, W. S. 2008; 135 (3): 599-608

    Abstract

    Mutations in Kif1-binding protein/KIAA1279 (KBP) cause the devastating neurological disorder Goldberg-Shprintzen syndrome (GSS) in humans. The cellular function of KBP and the basis of the symptoms of GSS, however, remain unclear. Here, we report the identification and characterization of a zebrafish kbp mutant. We show that kbp is required for axonal outgrowth and maintenance. In vivo time-lapse analysis of neuronal development shows that the speed of early axonal outgrowth is reduced in both the peripheral and central nervous systems in kbp mutants. Ultrastructural studies reveal that kbp mutants have disruption to axonal microtubules during outgrowth. These results together suggest that kbp is an important regulator of the microtubule dynamics that drive the forward propulsion of axons. At later stages, we observe that many affected axons degenerate. Ultrastructural analyses at these stages demonstrate mislocalization of axonal mitochondria and a reduction in axonal number in the peripheral, central and enteric nervous systems. We propose that kbp is an important regulator of axonal development and that axonal cytoskeletal defects underlie the nervous system defects in GSS.

    View details for DOI 10.1242/dev.012377

    View details for Web of Science ID 000252254900019

    View details for PubMedID 18192286

  • Zebrafish bmp4 functions during late gastrulation to specify ventroposterior cell fates DEVELOPMENTAL BIOLOGY Stickney, H. L., Imai, Y., Draper, B., Moens, C., Talbot, W. S. 2007; 310 (1): 71-84

    Abstract

    Bone morphogenetic proteins (BMPs) are key mediators of dorsoventral patterning in vertebrates and are required for the induction of ventral fates in fish and frogs. A widely accepted model of dorsoventral patterning postulates that a morphogenetic BMP activity gradient patterns cell fates along the dorsoventral axis. Recent work in zebrafish suggests that the role of BMP signaling changes over time, with BMPs required for global dorsoventral patterning during early gastrulation and for tail patterning during late gastrulation and early somitogenesis. Key questions remain about the late phase, including which BMP ligands are required and how the functions of BMPs differ during the early and late gastrula stages. In a screen for dominant enhancers of mutations in the homeobox genes vox and vent, which function in parallel to bmp signaling, we identified an insertion mutation in bmp4. We then performed a reverse genetic screen to isolate a null allele of bmp4. We report the characterization of these two alleles and demonstrate that BMP4 is required during the later phase of BMP signaling for the specification of ventroposterior cell fates. Our results indicate that different bmp genes are essential at different stages. In addition, we present genetic evidence supporting a role for a morphogenetic BMP gradient in establishing mesodermal fates during the later phase of BMP signaling.

    View details for DOI 10.1016/j.ydbio.2007.07.027

    View details for Web of Science ID 000250080000007

    View details for PubMedID 17727832

  • Signals on the move: chemokine receptors and organogenesis in zebrafish. Science's STKE : signal transduction knowledge environment Perlin, J. R., Talbot, W. S. 2007; 2007 (400): pe45-?

    Abstract

    The chemokine SDF1 (stromal cell-derived factor 1) directs cell migration in many different contexts, ranging from embryogenesis to inflammation. SDF1a is the guidance cue for the zebrafish lateral line primordium, a tissue that moves along the flank of the embryo and deposits cells that form mechanosensory organs. The SDF1a receptor CXCR4b acts in cells at the leading edge of the primordium to direct its migration. Two new studies show that a second SDF1 receptor, CXCR7, is required only in the trailing cells of the primordium, and they explore how these two receptors orchestrate migration of the primordium. CXCR4b and CXCR7 are expressed in complementary domains, possibly through mutual repression in which each receptor inhibits expression of the other. These studies illustrate how the entire primordium can respond to a single signal, yet generate cell type-specific responses by using different receptors.

    View details for PubMedID 17712137

  • Putting the glue in glia: Necls mediate Schwann cell-axon adhesion JOURNAL OF CELL BIOLOGY Perlin, J. R., Talbot, W. S. 2007; 178 (5): 721-723

    Abstract

    Interactions between Schwann cells and axons are critical for the development and function of myelinated axons. Two recent studies (see Maurel et al. on p. 861 of this issue; Spiegel et al., 2007) report that the nectin-like (Necl) proteins Necl-1 and -4 are internodal adhesion molecules that are critical for myelination. These studies suggest that Necl proteins mediate a specific interaction between Schwann cells and axons that allows proper communication of the signals that trigger myelination.

    View details for DOI 10.1083/jcb.200708019

    View details for Web of Science ID 000249240800002

    View details for PubMedID 17724116

  • alpha II-spectrin is essential for assembly of the nodes of Ranvier in myelinated axons CURRENT BIOLOGY Voas, M. G., Lyons, D. A., Naylor, S. G., Arana, N., Rasband, M. N., Talbot, W. S. 2007; 17 (6): 562-568

    Abstract

    Saltatory conduction in myelinated axons requires organization of the nodes of Ranvier, where voltage-gated sodium channels are prominently localized [1]. Previous results indicate that alphaII-spectrin, a component of the cortical cytoskeleton [2], is enriched at the paranodes [3, 4], which flank the node of Ranvier, but alphaII-spectrin's function has not been investigated. Starting with a genetic screen in zebrafish, we discovered in alphaII-spectrin (alphaII-spn) a mutation that disrupts nodal sodium-channel clusters in myelinated axons of the PNS and CNS. In alphaII-spn mutants, the nodal sodium-channel clusters are reduced in number and disrupted at early stages. Analysis of chimeric animals indicated that alphaII-spn functions autonomously in neurons. Ultrastructural studies show that myelin forms in the posterior lateral line nerve and in the ventral spinal cord in alphaII-spn mutants and that the node is abnormally long; these findings indicate that alphaII-spn is required for the assembly of a mature node of the correct length. We find that alphaII-spectrin is enriched in nodes and paranodes at early stages and that the nodal expression diminishes as nodes mature. Our results provide functional evidence that alphaII-spectrin in the axonal cytoskeleton is essential for stabilizing nascent sodium-channel clusters and assembling the mature node of Ranvier.

    View details for DOI 10.1016/j.cub.2007.01.071

    View details for Web of Science ID 000245225500031

    View details for PubMedID 17331725

  • A genetic screen identifies genes essential for development of myelinated axons in zebrafish DEVELOPMENTAL BIOLOGY Pogoda, H., Sternheim, N., Lyons, D. A., Diamond, B., Hawkins, T. A., Woods, I. G., Bhatt, D. H., Franzini-Armstrong, C., Dominguez, C., Arana, N., Jacobs, J., Nix, R., Fetcho, J. R., Talbot, W. S. 2006; 298 (1): 118-131

    Abstract

    The myelin sheath insulates axons in the vertebrate nervous system, allowing rapid propagation of action potentials via saltatory conduction. Specialized glial cells, termed Schwann cells in the PNS and oligodendrocytes in the CNS, wrap axons to form myelin, a compacted, multilayered sheath comprising specific proteins and lipids. Disruption of myelinated axons causes human diseases, including multiple sclerosis and Charcot-Marie-Tooth peripheral neuropathies. Despite the progress in identifying human disease genes and other mutations disrupting glial development and myelination, many important unanswered questions remain about the mechanisms that coordinate the development of myelinated axons. To address these questions, we began a genetic dissection of myelination in zebrafish. Here we report a genetic screen that identified 13 mutations, which define 10 genes, disrupting the development of myelinated axons. We present the initial characterization of seven of these mutations, defining six different genes, along with additional characterization of mutations that we have described previously. The different mutations affect the PNS, the CNS, or both, and phenotypic analyses indicate that the genes affect a wide range of steps in glial development, from fate specification through terminal differentiation. The analysis of these mutations will advance our understanding of myelination, and the mutants will serve as models of human diseases of myelin.

    View details for DOI 10.1016/j.ydbio.2006.06.021

    View details for Web of Science ID 000241071100011

    View details for PubMedID 16875686

  • nsf is essential for organization of myelinated axons in zebrafish CURRENT BIOLOGY Woods, I. G., Lyons, D. A., Voas, M. G., Pogoda, H. M., Talbot, W. S. 2006; 16 (7): 636-648

    Abstract

    Myelinated axons are essential for rapid conduction of action potentials in the vertebrate nervous system. Of particular importance are the nodes of Ranvier, sites of voltage-gated sodium channel clustering that allow action potentials to be propagated along myelinated axons by saltatory conduction. Despite their critical role in the function of myelinated axons, little is known about the mechanisms that organize the nodes of Ranvier.Starting with a forward genetic screen in zebrafish, we have identified an essential requirement for nsf (N-ethylmaleimide sensitive factor) in the organization of myelinated axons. Previous work has shown that NSF is essential for membrane fusion in eukaryotes and has a critical role in vesicle fusion at chemical synapses. Zebrafish nsf mutants are paralyzed and have impaired response to light, reflecting disrupted nsf function in synaptic transmission and neural activity. In addition, nsf mutants exhibit defects in Myelin basic protein expression and in localization of sodium channel proteins at nodes of Ranvier. Analysis of chimeric larvae indicates that nsf functions autonomously in neurons, such that sodium channel clusters are evident in wild-type neurons transplanted into the nsf mutant hosts. Through pharmacological analyses, we show that neural activity and function of chemical synapses are not required for sodium channel clustering and myelination in the larval nervous system.Zebrafish nsf mutants provide a novel vertebrate system to investigate Nsf function in vivo. Our results reveal a previously unknown role for nsf, independent of its function in synaptic vesicle fusion, in the formation of the nodes of Ranvier in the vertebrate nervous system.

    View details for DOI 10.1016/j.cub.2006.02.067

    View details for Web of Science ID 000236649400021

    View details for PubMedID 16581508

  • The zebrafish gene map defines ancestral vertebrate chromosomes GENOME RESEARCH Woods, I. G., Wilson, C., Friedlander, B., Chang, P., Reyes, D. K., Nix, R., Kelly, P. D., Chu, F., Postlethwait, J. H., Talbot, W. S. 2005; 15 (9): 1307-1314

    Abstract

    Genetic screens in zebrafish (Danio rerio) have identified mutations that define the roles of hundreds of essential vertebrate genes. Genetic maps can link mutant phenotype with gene sequence by providing candidate genes for mutations and polymorphic genetic markers useful in positional cloning projects. Here we report a zebrafish genetic map comprising 4073 polymorphic markers, with more than twice the number of coding sequences localized in previously reported zebrafish genetic maps. We use this map in comparative studies to identify numerous regions of synteny conserved among the genomes of zebrafish, Tetraodon, and human. In addition, we use our map to analyze gene duplication in the zebrafish and Tetraodon genomes. Current evidence suggests that a whole-genome duplication occurred in the teleost lineage after it split from the tetrapod lineage, and that only a subset of the duplicates have been retained in modern teleost genomes. It has been proposed that differential retention of duplicate genes may have facilitated the isolation of nascent species formed during the vast radiation of teleosts. We find that different duplicated genes have been retained in zebrafish and Tetraodon, although similar numbers of duplicates remain in both genomes. Finally, we use comparative mapping data to address the proposal that the common ancestor of vertebrates had a genome consisting of 12 chromosomes. In a three-way comparison between the genomes of zebrafish, Tetraodon, and human, our analysis delineates the gene content for 11 of these 12 proposed ancestral chromosomes.

    View details for DOI 10.1101/gr.4134305

    View details for Web of Science ID 000231720500015

    View details for PubMedID 16109975

  • erbb3 and erbb2 are essential for Schwann cell migration and myelination in zebrafish CURRENT BIOLOGY Lyons, D. A., Pogoda, H. M., Voas, M. G., Woods, I. G., Diamond, B., Nix, R., Arana, N., Jacobs, J., Talbot, W. S. 2005; 15 (6): 513-524

    Abstract

    Myelin is critical for efficient axonal conduction in the vertebrate nervous system. Neuregulin (Nrg) ligands and their ErbB receptors are required for the development of Schwann cells, the glial cells that form myelin in the peripheral nervous system. Previous studies have not determined whether Nrg-ErbB signaling is essential in vivo for Schwann cell fate specification, proliferation, survival, migration, or the onset of myelination.In genetic screens for mutants with disruptions in myelinated nerves, we identified mutations in erbb3 and erbb2, which together encode a heteromeric tyrosine kinase receptor for Neuregulin ligands. Phenotypic analysis shows that both genes are essential for development of Schwann cells. BrdU-incorporation studies and time-lapse analysis reveal that Schwann cell proliferation and migration, but not survival, are disrupted in erbb3 mutants. We show that Schwann cells can migrate in the absence of DNA replication. This uncoupling of proliferation and migration indicates that erbb gene function is required independently for these two processes. Pharmacological inhibition of ErbB signaling at different stages reveals a continuing requirement for ErbB function during migration and also provides evidence that ErbB signaling is required after migration for proliferation and the terminal differentiation of myelinating Schwann cells.These results provide in vivo evidence that Neuregulin-ErbB signaling is essential for directed Schwann cell migration and demonstrate that this pathway is also required for the onset of myelination in postmigratory Schwann cells.

    View details for DOI 10.1016/j.cub.2005.02.030

    View details for Web of Science ID 000228208400019

    View details for PubMedID 15797019

  • The you gene encodes an EGF-CUB protein essential for hedgehog signaling in zebrafish PLOS BIOLOGY Woods, I. G., Talbot, W. S. 2005; 3 (3): 476-487

    Abstract

    Hedgehog signaling is required for many aspects of development in vertebrates and invertebrates. Misregulation of the Hedgehog pathway causes developmental abnormalities and has been implicated in certain types of cancer. Large-scale genetic screens in zebrafish have identified a group of mutations, termed you-class mutations, that share common defects in somite shape and in most cases disrupt Hedgehog signaling. These mutant embryos exhibit U-shaped somites characteristic of defects in slow muscle development. In addition, Hedgehog pathway mutations disrupt spinal cord patterning. We report the positional cloning of you, one of the original you-class mutations, and show that it is required for Hedgehog signaling in the development of slow muscle and in the specification of ventral fates in the spinal cord. The you gene encodes a novel protein with conserved EGF and CUB domains and a secretory pathway signal sequence. Epistasis experiments support an extracellular role for You upstream of the Hedgehog response mechanism. Analysis of chimeras indicates that you mutant cells can appropriately respond to Hedgehog signaling in a wild-type environment. Additional chimera analysis indicates that wild-type you gene function is not required in axial Hedgehog-producing cells, suggesting that You is essential for transport or stability of Hedgehog signals in the extracellular environment. Our positional cloning and functional studies demonstrate that You is a novel extracellular component of the Hedgehog pathway in vertebrates.

    View details for DOI 10.1371/journal.pbio.0030066

    View details for Web of Science ID 000227984000015

    View details for PubMedID 15660164

  • Molecular genetics of axis formation in zebrafish ANNUAL REVIEW OF GENETICS Schier, A. F., Talbot, W. S. 2005; 39: 561-613

    Abstract

    The basic vertebrate body plan of the zebrafish embryo is established in the first 10 hours of development. This period is characterized by the formation of the anterior-posterior and dorsal-ventral axes, the development of the three germ layers, the specification of organ progenitors, and the complex morphogenetic movements of cells. During the past 10 years a combination of genetic, embryological, and molecular analyses has provided detailed insights into the mechanisms underlying this process. Maternal determinants control the expression of transcription factors and the location of signaling centers that pattern the blastula and gastrula. Bmp, Nodal, FGF, canonical Wnt, and retinoic acid signals generate positional information that leads to the restricted expression of transcription factors that control cell type specification. Noncanonical Wnt signaling is required for the morphogenetic movements during gastrulation. We review how the coordinated interplay of these molecules determines the fate and movement of embryonic cells.

    View details for DOI 10.1146/annurev.genet.37.110801.143752

    View details for Web of Science ID 000234685200024

    View details for PubMedID 16285872

  • The role of the zebrafish nodal-related genes squint and cyclops in patterning of mesendoderm DEVELOPMENT Dougan, S. T., Warga, R. M., Kane, D. A., Schier, A. F., Talbot, W. S. 2003; 130 (9): 1837-1851

    Abstract

    Nodal signals, a subclass of the TGFbeta superfamily of secreted factors, induce formation of mesoderm and endoderm in vertebrate embryos. We have examined the possible dorsoventral and animal-vegetal patterning roles for Nodal signals by using mutations in two zebrafish nodal-related genes, squint and cyclops, to manipulate genetically the levels and timing of Nodal activity. squint mutants lack dorsal mesendodermal gene expression at the late blastula stage, and fate mapping and gene expression studies in sqt(-/-); cyc(+/+) and sqt(-/-); cyc(+/-) mutants show that some dorsal marginal cells inappropriately form hindbrain and spinal cord instead of dorsal mesendodermal derivatives. The effects on ventrolateral mesendoderm are less severe, although the endoderm is reduced and muscle precursors are located nearer to the margin than in wild type. Our results support a role for Nodal signals in patterning the mesendoderm along the animal-vegetal axis and indicate that dorsal and ventrolateral mesoderm require different levels of squint and cyclops function. Dorsal marginal cells were not transformed toward more lateral fates in either sqt(-/-); cyc(+/-) or sqt(-/-); cyc(+/+) embryos, arguing against a role for the graded action of Nodal signals in dorsoventral patterning of the mesendoderm. Differential regulation of the cyclops gene in these cells contributes to the different requirements for nodal-related gene function in these cells. Dorsal expression of cyclops requires Nodal-dependent autoregulation, whereas other factors induce cyclops expression in ventrolateral cells. In addition, the differential timing of dorsal mesendoderm induction in squint and cyclops mutants suggests that dorsal marginal cells can respond to Nodal signals at stages ranging from the mid-blastula through the mid-gastrula.

    View details for DOI 10.1242/dev.00400

    View details for Web of Science ID 000182811600011

    View details for PubMedID 12642489

  • fast1 is required for the development of dorsal axial structures in zebrafish CURRENT BIOLOGY Sirotkin, H. I., GATES, M. A., Kelly, P. D., Schier, A. F., Talbot, W. S. 2000; 10 (17): 1051-1054

    Abstract

    Nodal-related signals comprise a subclass of the transforming growth factor-beta (TGF-beta) superfamily and regulate key events in vertebrate embryogenesis, including mesoderm formation, establishment of left-right asymmetry and neural patterning [1-8]. Nodal ligands are thought to act with EGF-CFC protein co-factors to activate activin type I and II or related receptors, which phosphorylate Smad2 and trigger nuclear translocation of a Smad2/4 complex [8-12]. The winged-helix transcription factor forkhead activin signal transducer-1 (Fast-1) acts as a co-factor for Smad2 [12-20]. Xenopus Fast-1 is thought to function as a transcriptional effector of Nodal signals during mesoderm formation [17], but no mutations in the Fast-1 gene have been identified. We report the identification of the zebrafish fast1 gene and show that it is disrupted in schmalspur (sur) mutants, which have defects in the development of dorsal midline cell types and establishment of left-right asymmetry [21-25]. We find that prechordal plate and notochord are strongly reduced in maternal-zygotic sur mutants, whereas other mesendodermal structures are present - a less severe phenotype than that caused by complete loss of Nodal signaling. These results show that fast1 is required for development of dorsal axial structures and left-right asymmetry, and suggest that Nodal signals act through Fast1-dependent and independent pathways.

    View details for Web of Science ID 000089303900017

    View details for PubMedID 10996072

  • Bozozok and squint act in parallel to specify dorsal mesoderm and anterior neuroectoderm in zebrafish DEVELOPMENT Sirotkin, H. I., Dougan, S. T., Schier, A. F., Talbot, W. S. 2000; 127 (12): 2583-2592

    Abstract

    In vertebrate embryos, maternal (beta)-catenin protein activates the expression of zygotic genes that establish the dorsal axial structures. Among the zygotically acting genes with key roles in the specification of dorsal axial structures are the homeobox gene bozozok (boz) and the nodal-related (TGF-(beta) family) gene squint (sqt). Both genes are expressed in the dorsal yolk syncytial layer, a source of dorsal mesoderm inducing signals, and mutational analysis has indicated that boz and sqt are required for dorsal mesoderm development. Here we examine the regulatory interactions among boz, sqt and a second nodal-related gene, cyclops (cyc). Three lines of evidence indicate that boz and sqt act in parallel to specify dorsal mesoderm and anterior neuroectoderm. First, boz requires sqt function to induce high levels of ectopic dorsal mesoderm, consistent with sqt acting either downstream or in parallel to boz. Second, sqt mRNA is expressed in blastula stage boz mutants, indicating that boz is not essential for activation of sqt transcription, and conversely, boz mRNA is expressed in blastula stage sqt mutants. Third, boz;sqt double mutants have a much more severe phenotype than boz and sqt single mutants. Double mutants consistently lack the anterior neural tube and axial mesoderm, and ventral fates are markedly expanded. Expression of chordin and noggin1 is greatly reduced in boz;sqt mutants, indicating that the boz and sqt pathways have overlapping roles in activating secreted BMP antagonists. In striking contrast to boz;sqt double mutants, anterior neural fates are specified in boz;sqt;cyc triple mutants. This indicates that cyc represses anterior neural development, and that boz and sqt counteract this repressive function. Our results support a model in which boz and sqt act in parallel to induce dorsalizing BMP-antagonists and to counteract the repressive function of cyc in neural patterning.

    View details for Web of Science ID 000087948900007

    View details for PubMedID 10821757

  • Analysis of chromosomal rearrangements induced by postmeiotic mutagenesis with ethylnitrosourea in zebrafish GENETICS Imai, Y., Feldman, B., Schier, A. F., Talbot, W. S. 2000; 155 (1): 261-272

    Abstract

    Mutations identified in zebrafish genetic screens allow the dissection of a wide array of problems in vertebrate biology. Most screens have examined mutations induced by treatment of spermatogonial (premeiotic) cells with the chemical mutagen N-ethyl-N-nitrosourea (ENU). Treatment of postmeiotic gametes with ENU induces specific-locus mutations at a higher rate than premeiotic regimens, suggesting that postmeiotic mutagenesis protocols could be useful in some screening strategies. Whereas there is extensive evidence that ENU induces point mutations in premeiotic cells, the range of mutations induced in postmeiotic zebrafish germ cells has been less thoroughly characterized. Here we report the identification and analysis of five mutations induced by postmeiotic ENU treatment. One mutation, snh(st1), is a translocation involving linkage group (LG) 11 and LG 14. The other four mutations, oep(st2), kny(st3), Df(LG 13)(st4), and cyc(st5), are deletions, ranging in size from less than 3 cM to greater than 20 cM. These results show that germ cell stage is an important determinant of the type of mutations induced. The induction of chromosomal rearrangements may account for the elevated frequency of specific-locus mutations observed after treatment of postmeiotic gametes with ENU.

    View details for Web of Science ID 000086869200022

    View details for PubMedID 10790400

  • Genetic linkage mapping of zebrafish genes and ESTs GENOME RESEARCH Kelly, P. D., Chu, F., Woods, I. G., Ngo-Hazelett, P., Cardozo, T., Huang, H., Kimm, F., Liao, L. Y., Yan, Y. L., Zhou, Y. Y., Johnson, S. L., Abagyan, R., Schier, A. F., Postlethwait, J. H., Talbot, W. S. 2000; 10 (4): 558-567

    Abstract

    Genetic screens in zebrafish (Danio rerio) have isolated mutations in hundreds of genes essential for vertebrate development, physiology, and behavior. We have constructed a genetic linkage map that will facilitate the identification of candidate genes for these mutations and allow comparisons among the genomes of zebrafish and other vertebrates. On this map, we have localized 771 zebrafish genes and expressed sequence tags (ESTs) by scoring single-stranded conformational polymorphisms (SSCPs) in a meiotic mapping panel. Of these sequences, 642 represent previously unmapped genes and ESTs. The mapping panel was comprised of 42 homozygous diploid individuals produced by heat shock treatment of haploid embryos at the one-cell stage (HS diploids). This "doubled haploid" strategy combines the advantages of mapping in haploid and standard diploid systems, because heat shock diploid individuals have only one allele at each locus and can survive to adulthood, enabling a relatively large quantity of genomic DNA to be prepared from each individual in the mapping panel. To integrate this map with others, we also scored 593 previously mapped simple-sequence length polymorphisms (SSLPs) in the mapping panel. This map will accelerate the molecular analysis of zebrafish mutations and facilitate comparative analysis of vertebrate genomes.

    View details for Web of Science ID 000086744300021

    View details for PubMedID 10779498

    View details for PubMedCentralID PMC310859

  • Zebrafish organizer development and germ-layer formation require nodal-related signals NATURE Feldman, B., GATES, M. A., Egan, E. S., Dougan, S. T., Rennebeck, G., Sirotkin, H. I., Schier, A. F., Talbot, W. S. 1998; 395 (6698): 181-185

    Abstract

    The vertebrate body plan is established during gastrulation, when cells move inwards to form the mesodermal and endodermal germ layers. Signals from a region of dorsal mesoderm, which is termed the organizer, pattern the body axis by specifying the fates of neighbouring cells. The organizer is itself induced by earlier signals. Although members of the transforming growth factor-beta (TGF-beta) and Wnt families have been implicated in the formation of the organizer, no endogenous signalling molecule is known to be required for this process. Here we report that the zebrafish squint (sqt) and cyclops (cyc) genes have essential, although partly redundant, functions in organizer development and also in the formation of mesoderm and endoderm. We show that the sqt gene encodes a member of the TGF-beta superfamily that is related to mouse nodal. cyc encodes another nodal-related proteins, which is consistent with our genetic evidence that sqt and cyc have overlapping functions. The sqt gene is expressed in a dorsal region of the blastula that includes the extraembryonic yolk syncytial layer (YSL). The YSL has been implicated as a source of signals that induce organizer development and mesendoderm formation. Misexpression of sqt RNA within the embryo or specifically in the YSL induces expanded or ectopic dorsal mesoderm. These results establish an essential role for nodal-related signals in organizer development and mesendoderm formation.

    View details for Web of Science ID 000075829900044

    View details for PubMedID 9744277