1
|
Bell I, Khan H, Stutt N, Horn M, Hydzik T, Lum W, Rea V, Clapham E, Hoeg L, Van Raay TJ. Nkd1 functions downstream of Axin2 to attenuate Wnt signaling. Mol Biol Cell 2024; 35:ar93. [PMID: 38656801 PMCID: PMC11244159 DOI: 10.1091/mbc.e24-02-0059-t] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/10/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
Wnt signaling is a crucial developmental pathway involved in early development as well as stem-cell maintenance in adults and its misregulation leads to numerous diseases. Thus, understanding the regulation of this pathway becomes vitally important. Axin2 and Nkd1 are widely utilized negative feedback regulators in Wnt signaling where Axin2 functions to destabilize cytoplasmic β-catenin, and Nkd1 functions to inhibit the nuclear localization of β-catenin. Here, we set out to further understand how Axin2 and Nkd1 regulate Wnt signaling by creating axin2gh1/gh1, nkd1gh2/gh2 single mutants and axin2gh1/gh1;nkd1gh2/gh2 double mutant zebrafish using sgRNA/Cas9. All three Wnt regulator mutants were viable and had impaired heart looping, neuromast migration defects, and behavior abnormalities in common, but there were no signs of synergy in the axin2gh1/gh1;nkd1gh2/gh2 double mutants. Further, Wnt target gene expression by qRT-PCR and RNA-seq, and protein expression by mass spectrometry demonstrated that the double axin2gh1/gh1;nkd1gh2/gh2 mutant resembled the nkd1gh2/gh2 phenotype demonstrating that Nkd1 functions downstream of Axin2. In support of this, the data further demonstrates that Axin2 uniquely alters the properties of β-catenin-dependent transcription having novel readouts of Wnt activity compared with nkd1gh2/gh2 or the axin2gh1/gh1;nkd1gh2/gh2 double mutant. We also investigated the sensitivity of the Wnt regulator mutants to exacerbated Wnt signaling, where the single mutants displayed characteristic heightened Wnt sensitivity, resulting in an eyeless phenotype. Surprisingly, this phenotype was rescued in the double mutant, where we speculate that cross-talk between Wnt/β-catenin and Wnt/Planar Cell Polarity pathways could lead to altered Wnt signaling in some scenarios. Collectively, the data emphasizes both the commonality and the complexity in the feedback regulation of Wnt signaling.
Collapse
Affiliation(s)
- Ian Bell
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| | - Haider Khan
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| | - Nathan Stutt
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Matthew Horn
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| | - Teesha Hydzik
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| | - Whitney Lum
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| | - Victoria Rea
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| | - Emma Clapham
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| | - Lisa Hoeg
- Department of Bioinformatics, University of Guelph, Guelph, Ontario, N1G 2W1 Canada
| | - Terence J. Van Raay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, N1G 2W1 Ontario, Canada
| |
Collapse
|
2
|
Bátora D, Fischer J, Kaderli RM, Varga M, Lochner M, Gertsch J. Silicon-Rhodamine Functionalized Evocalcet Probes Potently and Selectively Label Calcium Sensing Receptors In Vitro, In Vivo, and Ex Vivo. ACS Pharmacol Transl Sci 2024; 7:1557-1570. [PMID: 38751613 PMCID: PMC11091967 DOI: 10.1021/acsptsci.4c00096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024]
Abstract
The calcium sensing receptor (CaSR) is a ubiquitously expressed G-protein coupled receptor (GPCR) that regulates extracellular calcium signals via the parathyroid glands. CaSR has recently also been implicated in noncalcitropic pathophysiologies like asthma, gut inflammation, and cancer. To date, molecular tools that enable the bioimaging of CaSR in tissues are lacking. Based on in silico analyses of available structure-activity relationship data on CaSR ligands, we designed and prepared silicon-rhodamine (SiR) conjugates of the clinically approved drug evocalcet. The new probes EvoSiR4 and EvoSiR6, with differing linker lengths at the evocalcet carboxyl end, both showed a 6-fold and 3-fold increase in potency toward CaSR (EC50 < 45 nM) compared to evocalcet and the evocalcet-linker conjugate, respectively, in an FLIPR-based cellular functional assay. The specificity of the EvoSiR probes toward CaSR binding and the impact of albumin was evaluated in live cell experiments. Both probes showed strong albumin binding, which facilitated the clearance of nonspecific binding interactions. Accordingly, in zebrafish embryos, EvoSiR4 specifically labeled the high CaSR expressing neuromasts of the lateral line in vivo. EvoSiR4 was also assessed in human parathyroid tissues ex vivo, showing a specific absolute CaSR-associated fluorescence compared to that of parathyroid autofluorescence. In summary, functionalization of evocalcet by SiR led to the preparation of potent and specific fluorescent CaSR probes. EvoSiR4 is a versatile small-molecular probe that can be employed in CaSR-related biomedical analyses where antibodies are not applicable.
Collapse
Affiliation(s)
- Daniel Bátora
- Institute
of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland
- Graduate
School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Jérôme
P. Fischer
- Institute
of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland
| | - Reto M. Kaderli
- Department
of Visceral Surgery and Medicine, Inselspital,
Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Máté Varga
- Department
of Genetics, ELTE Eötvös Loránd
University, 1117 Budapest, Hungary
| | - Martin Lochner
- Institute
of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland
| | - Jürg Gertsch
- Institute
of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
3
|
Powell GT, Faro A, Zhao Y, Stickney H, Novellasdemunt L, Henriques P, Gestri G, White ER, Ren J, Lu W, Young RM, Hawkins TA, Cavodeassi F, Schwarz Q, Dreosti E, Raible DW, Li VSW, Wright GJ, Jones EY, Wilson SW. Cachd1 interacts with Wnt receptors and regulates neuronal asymmetry in the zebrafish brain. Science 2024; 384:573-579. [PMID: 38696577 PMCID: PMC7615972 DOI: 10.1126/science.ade6970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/27/2024] [Indexed: 05/04/2024]
Abstract
Neurons on the left and right sides of the nervous system often show asymmetric properties, but how such differences arise is poorly understood. Genetic screening in zebrafish revealed that loss of function of the transmembrane protein Cachd1 resulted in right-sided habenula neurons adopting left-sided identity. Cachd1 is expressed in neuronal progenitors, functions downstream of asymmetric environmental signals, and influences timing of the normally asymmetric patterns of neurogenesis. Biochemical and structural analyses demonstrated that Cachd1 can bind simultaneously to Lrp6 and Frizzled family Wnt co-receptors. Consistent with this, lrp6 mutant zebrafish lose asymmetry in the habenulae, and epistasis experiments support a role for Cachd1 in modulating Wnt pathway activity in the brain. These studies identify Cachd1 as a conserved Wnt receptor-interacting protein that regulates lateralized neuronal identity in the zebrafish brain.
Collapse
Affiliation(s)
- Gareth T. Powell
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
| | - Ana Faro
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Yuguang Zhao
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Heather Stickney
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
- Ambry Genetics; Aliso Viejo, CA 92656, USA
| | - Laura Novellasdemunt
- The Francis Crick Institute; London, NW1 1AT, UK
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology; 08028, Barcelona, Spain
| | - Pedro Henriques
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Gaia Gestri
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | | | - Jingshan Ren
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Weixian Lu
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Rodrigo M. Young
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor; Camino La Piramide 5750, 8580745, Santiago, Chile
| | - Thomas A. Hawkins
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Florencia Cavodeassi
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- St. George’s, University of London; London, SW17 0RE, UK
| | - Quenten Schwarz
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
| | - Elena Dreosti
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - David W. Raible
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
| | | | - Gavin J. Wright
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York; York, YO10 5DD, UK
| | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Stephen W. Wilson
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| |
Collapse
|
4
|
Aguiar TFM, Rivas MP, de Andrade Silva EM, Pires SF, Dangoni GD, Macedo TC, Defelicibus A, Barros BDDF, Novak E, Cristofani LM, Odone V, Cypriano M, de Toledo SRC, da Cunha IW, da Costa CML, Carraro DM, Tojal I, de Oliveira Mendes TA, Krepischi ACV. First Transcriptome Analysis of Hepatoblastoma in Brazil: Unraveling the Pivotal Role of Noncoding RNAs and Metabolic Pathways. Biochem Genet 2024:10.1007/s10528-024-10764-y. [PMID: 38649558 DOI: 10.1007/s10528-024-10764-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/27/2024] [Indexed: 04/25/2024]
Abstract
Hepatoblastoma stands as the most prevalent liver cancer in the pediatric population. Characterized by a low mutational burden, chromosomal and epigenetic alterations are key drivers of its tumorigenesis. Transcriptome analysis is a powerful tool for unraveling the molecular intricacies of hepatoblastoma, shedding light on the effects of genetic and epigenetic changes on gene expression. In this study conducted in Brazilian patients, an in-depth whole transcriptome analysis was performed on 14 primary hepatoblastomas, compared to control liver tissues. The analysis unveiled 1,492 differentially expressed genes (1,031 upregulated and 461 downregulated), including 920 protein-coding genes (62%). Upregulated biological processes were linked to cell differentiation, signaling, morphogenesis, and development, involving known hepatoblastoma-associated genes (DLK1, MEG3, HDAC2, TET1, HMGA2, DKK1, DKK4), alongside with novel findings (GYNG4, CDH3, and TNFRSF19). Downregulated processes predominantly centered around oxidation and metabolism, affecting amines, nicotinamides, and lipids, featuring novel discoveries like the repression of SYT7, TTC36, THRSP, CCND1, GCK and CAMK2B. Two genes, which displayed a concordant pattern of DNA methylation alteration in their promoter regions and dysregulation in the transcriptome, were further validated by RT-qPCR: the upregulated TNFRSF19, a key gene in the embryonic development, and the repressed THRSP, connected to lipid metabolism. Furthermore, based on protein-protein interaction analysis, we identified genes holding central positions in the network, such as HDAC2, CCND1, GCK, and CAMK2B, among others, that emerged as prime candidates warranting functional validation in future studies. Notably, a significant dysregulation of non-coding RNAs (ncRNAs), predominantly upregulated transcripts, was observed, with 42% of the top 50 highly expressed genes being ncRNAs. An integrative miRNA-mRNA analysis revealed crucial biological processes associated with metabolism, oxidation reactions of lipids and carbohydrates, and methylation-dependent chromatin silencing. In particular, four upregulated miRNAs (miR-186, miR-214, miR-377, and miR-494) played a pivotal role in the network, potentially targeting multiple protein-coding transcripts, including CCND1 and CAMK2B. In summary, our transcriptome analysis highlighted disrupted embryonic development as well as metabolic pathways, particularly those involving lipids, emphasizing the emerging role of ncRNAs as epigenetic regulators in hepatoblastomas. These findings provide insights into the complexity of the hepatoblastoma transcriptome and identify potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Talita Ferreira Marques Aguiar
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, Human Genome and Stem-Cell Research Center, University of São Paulo, São Paulo, Brazil
- Columbia University Irving Medical Center, New York, NY, USA
| | - Maria Prates Rivas
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, Human Genome and Stem-Cell Research Center, University of São Paulo, São Paulo, Brazil
| | - Edson Mario de Andrade Silva
- Department of Biochemistry and Molecular Biology, Federal University of Viçosa, Minas Gerais, Brazil
- Horticultural Sciences Department, University of Florida, Gainesville, USA
| | - Sara Ferreira Pires
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, Human Genome and Stem-Cell Research Center, University of São Paulo, São Paulo, Brazil
| | - Gustavo Dib Dangoni
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, Human Genome and Stem-Cell Research Center, University of São Paulo, São Paulo, Brazil
| | - Taiany Curdulino Macedo
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, Human Genome and Stem-Cell Research Center, University of São Paulo, São Paulo, Brazil
| | | | | | - Estela Novak
- Pediatric Cancer Institute (ITACI) at the Pediatric Department, São Paulo University Medical School, São Paulo, Brazil
| | - Lilian Maria Cristofani
- Pediatric Cancer Institute (ITACI) at the Pediatric Department, São Paulo University Medical School, São Paulo, Brazil
| | - Vicente Odone
- Pediatric Cancer Institute (ITACI) at the Pediatric Department, São Paulo University Medical School, São Paulo, Brazil
| | - Monica Cypriano
- Department of Pediatrics, Adolescent and Child With Cancer Support Group (GRAACC), Federal University of São Paulo, São Paulo, Brazil
| | - Silvia Regina Caminada de Toledo
- Department of Pediatrics, Adolescent and Child With Cancer Support Group (GRAACC), Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Dirce Maria Carraro
- International Center for Research, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Israel Tojal
- International Center for Research, A. C. Camargo Cancer Center, São Paulo, Brazil
| | | | - Ana Cristina Victorino Krepischi
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, Human Genome and Stem-Cell Research Center, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
5
|
Rea V, Bell I, Ball T, Van Raay T. Gut-derived metabolites influence neurodevelopmental gene expression and Wnt signaling events in a germ-free zebrafish model. MICROBIOME 2022; 10:132. [PMID: 35996200 PMCID: PMC9396910 DOI: 10.1186/s40168-022-01302-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Small molecule metabolites produced by the microbiome are known to be neuroactive and are capable of directly impacting the brain and central nervous system, yet there is little data on the contribution of these metabolites to the earliest stages of neural development and neural gene expression. Here, we explore the impact of deriving zebrafish embryos in the absence of microbes on early neural development as well as investigate whether any potential changes can be rescued with treatment of metabolites derived from the zebrafish gut microbiota. RESULTS Overall, we did not observe any gross morphological changes between treatments but did observe a significant decrease in neural gene expression in embryos raised germ-free, which was rescued with the addition of zebrafish metabolites. Specifically, we identified 354 genes significantly downregulated in germ-free embryos compared to conventionally raised embryos via RNA-Seq analysis. Of these, 42 were rescued with a single treatment of zebrafish gut-derived metabolites to germ-free embryos. Gene ontology analysis revealed that these genes are involved in prominent neurodevelopmental pathways including transcriptional regulation and Wnt signaling. Consistent with the ontology analysis, we found alterations in the development of Wnt dependent events which was rescued in the germ-free embryos treated with metabolites. CONCLUSIONS These findings demonstrate that gut-derived metabolites are in part responsible for regulating critical signaling pathways in the brain, especially during neural development. Video abstract.
Collapse
Affiliation(s)
- Victoria Rea
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Ian Bell
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Taylor Ball
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Terence Van Raay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada.
| |
Collapse
|
6
|
Single-cell transcriptome analysis reveals three sequential phases of gene expression during zebrafish sensory hair cell regeneration. Dev Cell 2022; 57:799-819.e6. [PMID: 35316618 PMCID: PMC9188816 DOI: 10.1016/j.devcel.2022.03.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/19/2021] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
Loss of sensory hair cells (HCs) in the mammalian inner ear leads to permanent hearing and vestibular defects, whereas loss of HCs in zebrafish results in their regeneration. We used single-cell RNA sequencing (scRNA-seq) to characterize the transcriptional dynamics of HC regeneration in zebrafish at unprecedented spatiotemporal resolution. We uncovered three sequentially activated modules: first, an injury/inflammatory response and downregulation of progenitor cell maintenance genes within minutes after HC loss; second, the transient activation of regeneration-specific genes; and third, a robust re-activation of developmental gene programs, including HC specification, cell-cycle activation, ribosome biogenesis, and a metabolic switch to oxidative phosphorylation. The results are relevant not only for our understanding of HC regeneration and how we might be able to trigger it in mammals but also for regenerative processes in general. The data are searchable and publicly accessible via a web-based interface.
Collapse
|
7
|
Santra P, Amack JD. Loss of vacuolar-type H+-ATPase induces caspase-independent necrosis-like death of hair cells in zebrafish neuromasts. Dis Model Mech 2021; 14:dmm048997. [PMID: 34296747 PMCID: PMC8319552 DOI: 10.1242/dmm.048997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/15/2021] [Indexed: 01/24/2023] Open
Abstract
The vacuolar-type H+-ATPase (V-ATPase) is a multi-subunit proton pump that regulates cellular pH. V-ATPase activity modulates several cellular processes, but cell-type-specific functions remain poorly understood. Patients with mutations in specific V-ATPase subunits can develop sensorineural deafness, but the underlying mechanisms are unclear. Here, we show that V-ATPase mutations disrupt the formation of zebrafish neuromasts, which serve as a model to investigate hearing loss. V-ATPase mutant neuromasts are small and contain pyknotic nuclei that denote dying cells. Molecular markers and live imaging show that loss of V-ATPase induces mechanosensory hair cells in neuromasts, but not neighboring support cells, to undergo caspase-independent necrosis-like cell death. This is the first demonstration that loss of V-ATPase can lead to necrosis-like cell death in a specific cell type in vivo. Mechanistically, loss of V-ATPase reduces mitochondrial membrane potential in hair cells. Modulating the mitochondrial permeability transition pore, which regulates mitochondrial membrane potential, improves hair cell survival. These results have implications for understanding the causes of sensorineural deafness, and more broadly, reveal functions for V-ATPase in promoting survival of a specific cell type in vivo.
Collapse
Affiliation(s)
- Peu Santra
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY 13244, USA
| |
Collapse
|
8
|
Dries R, Lange A, Heiny S, Berghaus KI, Bastmeyer M, Bentrop J. Cell Proliferation and Collective Cell Migration During Zebrafish Lateral Line System Development Are Regulated by Ncam/Fgf-Receptor Interactions. Front Cell Dev Biol 2021; 8:591011. [PMID: 33520983 PMCID: PMC7841142 DOI: 10.3389/fcell.2020.591011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/24/2020] [Indexed: 11/27/2022] Open
Abstract
The posterior lateral line system (pLLS) of aquatic animals comprises small clustered mechanosensory organs along the side of the animal. They develop from proneuromasts, which are deposited from a migratory primordium on its way to the tip of the tail. We here show, that the Neural Cell Adhesion Molecule Ncam1b is an integral part of the pathways initiating and regulating the development of the pLLS in zebrafish. We find that morpholino-knockdowns of ncam1b (i) reduce cell proliferation within the primordium, (ii) reduce the expression of Fgf target gene erm, (iii) severely affect proneuromast formation, and (iv) affect primordium migration. Ncam1b directly interacts with Fgf receptor Fgfr1a, and a knockdown of fgfr1a causes similar phenotypic changes as observed in ncam1b-morphants. We conclude that Ncam1b is involved in activating proliferation by triggering the expression of erm. In addition, we demonstrate that Ncam1b is required for the expression of chemokine receptor Cxcr7b, which is crucial for directed primordial migration. Finally, we show that the knockdown of ncam1b destabilizes proneuromasts, suggesting a further function of Ncam1b in strengthening the cohesion of proneuromast cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Joachim Bentrop
- Zoological Institute, Cell- and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
9
|
Marsay KS, Greaves S, Mahabaleshwar H, Ho CM, Roehl H, Monk PN, Carney TJ, Partridge LJ. Tetraspanin Cd9b and Cxcl12a/Cxcr4b have a synergistic effect on the control of collective cell migration. PLoS One 2021; 16:e0260372. [PMID: 34847198 PMCID: PMC8631670 DOI: 10.1371/journal.pone.0260372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/08/2021] [Indexed: 11/30/2022] Open
Abstract
Collective cell migration is essential for embryonic development and homeostatic processes. During zebrafish development, the posterior lateral line primordium (pLLP) navigates along the embryo flank by collective cell migration. The chemokine receptors, Cxcr4b and Cxcr7b, as well as their cognate ligand, Cxcl12a, are essential for this process. We corroborate that knockdown of the zebrafish cd9 tetraspanin orthologue, cd9b, results in mild pLL abnormalities. Through generation of CRISPR and TALEN mutants, we show that cd9a and cd9b function partially redundantly in pLLP migration, which is delayed in the cd9b single and cd9a; cd9b double mutants. This delay led to a transient reduction in neuromast numbers. Loss of both Cd9a and Cd9b sensitized embryos to reduced Cxcr4b and Cxcl12a levels. Together these results provide evidence that Cd9 modulates collective cell migration of the pLLP during zebrafish development. One interpretation of these observations is that Cd9 contributes to more effective chemokine signalling.
Collapse
Affiliation(s)
- Katherine S. Marsay
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Sarah Greaves
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Harsha Mahabaleshwar
- Lee Kong Chian School of Medicine, Experimental Medicine Building, Yunnan Garden Campus, Nanyang Technological University, Singapore, Singapore
| | - Charmaine Min Ho
- Lee Kong Chian School of Medicine, Experimental Medicine Building, Yunnan Garden Campus, Nanyang Technological University, Singapore, Singapore
| | - Henry Roehl
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| | - Peter N. Monk
- Department of Infection, Immunity and Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Tom J. Carney
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Lee Kong Chian School of Medicine, Experimental Medicine Building, Yunnan Garden Campus, Nanyang Technological University, Singapore, Singapore
| | - Lynda J. Partridge
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
10
|
Dalle Nogare D, Chitnis AB. NetLogo agent-based models as tools for understanding the self-organization of cell fate, morphogenesis and collective migration of the zebrafish posterior Lateral Line primordium. Semin Cell Dev Biol 2019; 100:186-198. [PMID: 31901312 DOI: 10.1016/j.semcdb.2019.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 01/25/2023]
Abstract
Interactions between primordium cells and their environment determines the self-organization of the zebrafish posterior Lateral Line primordium as it migrates under the skin from the ear to the tip of the tail forming and depositing neuromasts to spearhead formation of the posterior Lateral Line sensory system. In this review we describe how the NetLogo agent-based programming environment has been used in our lab to visualize and explore how self-generated chemokine gradients determine collective migration, how the dynamics of Wnt signaling can be used to predict patterns of neuromast deposition, and how previously defined interactions between Wnt and Fgf signaling systems have the potential to determine the periodic formation of center-biased Fgf signaling centers in the wake of a shrinking Wnt system. We also describe how NetLogo was used as a database for storing and visualizing the results of in toto lineage analysis of all cells in the migrating primordium. Together, the models illustrate how this programming environment can be used in diverse ways to integrate what has been learnt from biological experiments about the nature of interactions between cells and their environment, and explore how these interactions could potentially determine emergent patterns of cell fate specification, morphogenesis and collective migration of the zebrafish posterior Lateral Line primordium.
Collapse
Affiliation(s)
- Damian Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD USA
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD USA.
| |
Collapse
|
11
|
Urasaki A, Morishita S, Naka K, Uozumi M, Abe K, Huang L, Watase E, Nakagawa O, Kawakami K, Matsui T, Bessho Y, Inagaki N. Shootins mediate collective cell migration and organogenesis of the zebrafish posterior lateral line system. Sci Rep 2019; 9:12156. [PMID: 31434971 PMCID: PMC6704158 DOI: 10.1038/s41598-019-48585-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/07/2019] [Indexed: 01/23/2023] Open
Abstract
The zebrafish sensory posterior lateral line is an excellent model system to study collective cell migration and organogenesis. Shootin1 is a cytoplasmic protein involved in neuronal polarization and axon guidance. Previous studies have shown that shootin1 couples actin filament retrograde flow with extracellular adhesive substrates at the leading edge of axonal growth cones, thereby producing mechanical force for the migration and guidance of axonal growth cones. However, the functions of shootin in peripheral cells remain unknown. Here we identified two novel shootin family members, shootin2 and shootin3. In zebrafish, shootin1 and shootin3 are expressed in the posterior lateral line primordium (PLLP) and neuromasts during embryonic development. A shootin1 mutant displayed a reduced speed of PLLP migration, while shootin1;shootin3 double mutation inhibited cell proliferation in the PLLP. Furthermore, our results suggest that shootin1 and shootin3 positively regulate the number of neuromasts and the number of cells in deposited neuromasts. Our study demonstrates that shootins mediate collective cell migration of the posterior lateral line primordium and formation of neuromasts in zebrafish.
Collapse
Affiliation(s)
- Akihiro Urasaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.,Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Seiya Morishita
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Kosuke Naka
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Minato Uozumi
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Kouki Abe
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Liguo Huang
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Emiko Watase
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka, 411-8540, Japan
| | - Takaaki Matsui
- Laboratory of Gene Regulation Research, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Yasumasa Bessho
- Laboratory of Gene Regulation Research, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
12
|
Tang D, He Y, Li W, Li H. Wnt/β-catenin interacts with the FGF pathway to promote proliferation and regenerative cell proliferation in the zebrafish lateral line neuromast. Exp Mol Med 2019; 51:1-16. [PMID: 31123246 PMCID: PMC6533250 DOI: 10.1038/s12276-019-0247-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 01/13/2023] Open
Abstract
Wnt and FGF are highly conserved signaling pathways found in various organs and have been identified as important regulators of auditory organ development. In this study, we used the zebrafish lateral line system to study the cooperative roles of the Wnt and FGF pathways in regulating progenitor cell proliferation and regenerative cell proliferation. We found that activation of Wnt signaling induced cell proliferation and increased the number of hair cells in both developing and regenerating neuromasts. We further demonstrated that FGF signaling was critically involved in Wnt-regulated proliferation, and inhibition of FGF abolished the Wnt stimulation-mediated effects on cell proliferation, while activating FGF signaling with basic fibroblast growth factor (bFGF) led to a partial rescue of the proliferative failure and hair cell defects in the absence of Wnt activity. Whole-mount in situ hybridization analysis showed that the expression of several FGF pathway genes, including pea3 and fgfr1, was increased in neuromasts after treatment with the Wnt pathway inducer BIO. Interestingly, when SU5402 was used to inhibit FGF signaling, neuromast cells expressed much lower levels of the FGF receptor gene, fgfr1, but produced increased levels of Wnt target genes, including ctnnb1, ctnnb2, and tcf7l2, while bFGF treatment produced no alterations in the expression of those genes, suggesting that fgfr1 might restrict Wnt signaling in neuromasts during proliferation. In summary, our analysis demonstrates that both the Wnt and FGF pathways are tightly integrated to modulate the proliferation of progenitor cells during early neuromast development and regenerative cell proliferation after neomycin-induced injury in the zebrafish neuromast. Studying sensory organs on the skin of zebrafish is revealing details of molecular signaling pathways that may be relevant to our own sensory systems, especially the hair cells of the ear. These cells have fine hair-like structures that move in response to sound waves and help generate electrical signals to the brain that result in perception of sound. Huawei Li and colleagues at Fudan University, Shanghai, China, studied the roles of two well-known cellular signaling pathways in regulating the proliferation of similar sensory hair cells in zebrafish, a commonly used model organism. These pathways involve cell surface proteins that interact with small extracellular molecules to stimulate molecular changes within cells. Learning how the pathways control hair cell generation and multiplication may assist modification of similar systems in humans to study and treat hearing loss.
Collapse
Affiliation(s)
- Dongmei Tang
- ENT institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China
| | - Yingzi He
- ENT institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China
| | - Wenyan Li
- ENT institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China
| | - Huawei Li
- ENT institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China. .,Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, China. .,Shanghai Engineering Research Centre of Cochlear Implant, Shanghai, 200031, China. .,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Olson HM, Nechiporuk AV. Using Zebrafish to Study Collective Cell Migration in Development and Disease. Front Cell Dev Biol 2018; 6:83. [PMID: 30175096 PMCID: PMC6107837 DOI: 10.3389/fcell.2018.00083] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022] Open
Abstract
Cellular migration is necessary for proper embryonic development as well as maintenance of adult health. Cells can migrate individually or in groups in a process known as collective cell migration. Collectively migrating cohorts maintain cell-cell contacts, group polarization, and exhibit coordinated behavior. This mode of migration is important during numerous developmental processes including tracheal branching, blood vessel sprouting, neural crest cell migration and others. In the adult, collective cell migration is important for proper wound healing and is often misappropriated during cancer cell invasion. A variety of genetic model systems are used to examine and define the cellular and molecular mechanisms behind collective cell migration including border cell migration and tracheal branching in Drosophila melanogaster, neural crest cell migration in chick and Xenopus embryos, and posterior lateral line primordium (pLLP) migration in zebrafish. The pLLP is a group of about 100 cells that begins migrating around 22 hours post-fertilization along the lateral aspect of the trunk of the developing embryo. During migration, clusters of cells are deposited from the trailing end of the pLLP; these ultimately differentiate into mechanosensory organs of the lateral line system. As zebrafish embryos are transparent during early development and the pLLP migrates close to the surface of the skin, this system can be easily visualized and manipulated in vivo. These advantages together with the amenity to advance genetic methods make the zebrafish pLLP one of the premier model systems for studying collective cell migration. This review will describe the cellular behaviors and signaling mechanisms of the pLLP and compare the pLLP to collective cell migration in other popular model systems. In addition, we will examine how this type of migration is hijacked by collectively invading cancer cells.
Collapse
Affiliation(s)
- Hannah M Olson
- Department Cell, Developmental & Cancer Biology, The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States.,Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, United States
| | - Alex V Nechiporuk
- Department Cell, Developmental & Cancer Biology, The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
14
|
Neelathi UM, Dalle Nogare D, Chitnis AB. Cxcl12a induces snail1b expression to initiate collective migration and sequential Fgf-dependent neuromast formation in the zebrafish posterior lateral line primordium. Development 2018; 145:dev162453. [PMID: 29945870 PMCID: PMC6078336 DOI: 10.1242/dev.162453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 06/13/2018] [Indexed: 12/18/2022]
Abstract
The zebrafish posterior lateral line primordium migrates along a path defined by the chemokine Cxcl12a, periodically depositing neuromasts, to pioneer formation of the zebrafish posterior lateral line system. snail1b, known for its role in promoting cell migration, is expressed in leading cells of the primordium in response to Cxcl12a, whereas its expression in trailing cells is inhibited by Fgf signaling. snail1b knockdown delays initiation of primordium migration. This delay is associated with aberrant expansion of epithelial cell adhesion molecule (epcam) and reduction of cadherin 2 expression in the leading part of the primordium. Co-injection of snail1b morpholino with snail1b mRNA prevents the initial delay in migration and restores normal expression of epcam and cadherin 2 The delay in initiating primordium migration in snail1b morphants is accompanied by a delay in sequential formation of trailing Fgf signaling centers and associated protoneuromasts. This delay is not specifically associated with knockdown of snail1b but also with other manipulations that delay migration of the primordium. These observations reveal an unexpected link between the initiation of collective migration and sequential formation of protoneuromasts in the primordium.
Collapse
Affiliation(s)
- Uma M Neelathi
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Damian Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Guided genetic screen to identify genes essential in the regeneration of hair cells and other tissues. NPJ Regen Med 2018; 3:11. [PMID: 29872546 PMCID: PMC5986822 DOI: 10.1038/s41536-018-0050-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 04/18/2018] [Accepted: 05/08/2018] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine holds great promise for both degenerative diseases and traumatic tissue injury which represent significant challenges to the health care system. Hearing loss, which affects hundreds of millions of people worldwide, is caused primarily by a permanent loss of the mechanosensory receptors of the inner ear known as hair cells. This failure to regenerate hair cells after loss is limited to mammals, while all other non-mammalian vertebrates tested were able to completely regenerate these mechanosensory receptors after injury. To understand the mechanism of hair cell regeneration and its association with regeneration of other tissues, we performed a guided mutagenesis screen using zebrafish lateral line hair cells as a screening platform to identify genes that are essential for hair cell regeneration, and further investigated how genes essential for hair cell regeneration were involved in the regeneration of other tissues. We created genetic mutations either by retroviral insertion or CRISPR/Cas9 approaches, and developed a high-throughput screening pipeline for analyzing hair cell development and regeneration. We screened 254 gene mutations and identified 7 genes specifically affecting hair cell regeneration. These hair cell regeneration genes fell into distinct and somewhat surprising functional categories. By examining the regeneration of caudal fin and liver, we found these hair cell regeneration genes often also affected other types of tissue regeneration. Therefore, our results demonstrate guided screening is an effective approach to discover regeneration candidates, and hair cell regeneration is associated with other tissue regeneration. A study on zebrafish has genetically screened 254 genes and identified 7 genes implicated in the development and regeneration of hair cells and other tissues. Humans and other mammals cannot regrow hair cells—inner-ear sensory receptors that enable hearing—whereas non-mammalian vertebrates, including zebrafish, can regrow these following injury. Researchers from the United States, led by the National Institutes of Health’s Shawn Burgess, screened adult zebrafish for genes active during the regeneration of inner-ear epithelium. The researchers then produced zebrafish without these genes to study their functions. The studies tested 254 genes known to respond during regeneration, and identified seven specifically impacting regeneration. Most of these seven genes also functioned in liver and fin tissue regeneration. Understanding the mechanisms of these genes may enable future research into regenerative therapies in humans.
Collapse
|
16
|
Pfeiffer J, Tarbashevich K, Bandemer J, Palm T, Raz E. Rapid progression through the cell cycle ensures efficient migration of primordial germ cells - The role of Hsp90. Dev Biol 2018; 436:84-93. [PMID: 29477339 DOI: 10.1016/j.ydbio.2018.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/30/2017] [Accepted: 02/21/2018] [Indexed: 01/21/2023]
Abstract
Zebrafish primordial germ cells (PGCs) constitute a useful in vivo model to study cell migration and to elucidate the role of specific proteins in this process. Here we report on the role of the heat shock protein Hsp90aa1.2, a protein whose RNA level is elevated in the PGCs during their migration. Reducing Hsp90aa1.2 activity slows down the progression through the cell cycle and leads to defects in the control over the MTOC number in the migrating cells. These defects result in a slower migration rate and compromise the arrival of PGCs at their target, the region where the gonad develops. Our results emphasize the importance of ensuring rapid progression through the cell cycle during single-cell migration and highlight the role of heat shock proteins in the process.
Collapse
Affiliation(s)
- Jana Pfeiffer
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Katsiaryna Tarbashevich
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Jan Bandemer
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Thomas Palm
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany.
| |
Collapse
|
17
|
Dalle Nogare D, Chitnis AB. A framework for understanding morphogenesis and migration of the zebrafish posterior Lateral Line primordium. Mech Dev 2017; 148:69-78. [PMID: 28460893 PMCID: PMC10993927 DOI: 10.1016/j.mod.2017.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 12/31/2022]
Abstract
A description of zebrafish posterior Lateral Line (pLL) primordium development at single cell resolution together with the dynamics of Wnt, FGF, Notch and chemokine signaling in this system has allowed us to develop a framework to understand the self-organization of cell fate, morphogenesis and migration during its early development. The pLL primordium migrates under the skin, from near the ear to the tip of the tail, periodically depositing neuromasts. Nascent neuromasts, or protoneuromasts, form sequentially within the migrating primordium, mature, and are deposited from its trailing end. Initially broad Wnt signaling inhibits protoneuromast formation. However, protoneuromasts form sequentially in response to FGF signaling, starting from the trailing end, in the wake of a progressively shrinking Wnt system. While proliferation adds to the number of cells, the migrating primordium progressively shrinks as its trailing cells stop moving and are deposited. As it shrinks, the length of the migrating primordium correlates with the length of the leading Wnt system. Based on these observations we show how measuring the rate at which the Wnt system shrinks, the proliferation rate, the initial size of the primordium, its speed, and a few additional parameters allows us to predict the pattern of neuromast formation and deposition by the migrating primordium in both wild-type and mutant contexts. While the mechanism that links the length of the leading Wnt system to that of the primordium remains unclear, we discuss how it might be determined by access to factors produced in the leading Wnt active zone that are required for collective migration of trailing cells. We conclude by reviewing how FGFs, produced in response to Wnt signaling in leading cells, help determine collective migration of trailing cells, while a polarized response to a self-generated chemokine gradient serves as an efficient mechanism to steer primordium migration along its relatively long journey.
Collapse
Affiliation(s)
- Damian Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA.
| |
Collapse
|
18
|
Hübner K, Grassme KS, Rao J, Wenke NK, Zimmer CL, Korte L, Müller K, Sumanas S, Greber B, Herzog W. Wnt signaling positively regulates endothelial cell fate specification in the Fli1a-positive progenitor population via Lef1. Dev Biol 2017; 430:142-155. [DOI: 10.1016/j.ydbio.2017.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 01/06/2023]
|
19
|
He Y, Lu X, Qian F, Liu D, Chai R, Li H. Insm1a Is Required for Zebrafish Posterior Lateral Line Development. Front Mol Neurosci 2017; 10:241. [PMID: 28824372 PMCID: PMC5539400 DOI: 10.3389/fnmol.2017.00241] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/17/2017] [Indexed: 12/23/2022] Open
Abstract
Insulinoma-associated 1 (Insm1), a zinc-finger transcription factor, is widely expressed in the developing nervous system and plays important roles in cell cycle progression and cell fate specification. However, the functions of Insm1 in the embryonic development of the sensory system and its underlying molecular mechanisms remain largely unexplored. Here, through whole-mount in situ hybridization, we found that the zebrafish insm1a gene was expressed in the posterior lateral line (pLL) system, including both the migrating pLL primordium and the deposited neuromast cells. In order to decipher the specific roles of insm1a in zebrafish pLL development, we inhibited insm1a expression by using a morpholino knockdown strategy. The insm1a morphants exhibited primordium migration defects that resulted in reduced numbers of neuromasts. The inactivation of insm1a reduced the numbers of hair cells in neuromasts, and this defect could be a secondary consequence of disrupting rosette formation in the pLL primordium. Additionally, we showed that insm1a knockdown decreased the proliferation of pLL primordium cells, which likely contributed to these pLL defects. Furthermore, we showed that loss of insm1a resulted in elevated Wnt/β-catenin signaling and downregulation of Fgf target genes in the primordium. Insm1a knockdown also perturbed the expression patterns of chemokine signaling genes. Taken together, this study reveals a pivotal role for Insm1a in regulating pLL development during zebrafish embryogenesis.
Collapse
Affiliation(s)
- Yingzi He
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of NHFPCShanghai, China
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Fuping Qian
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Dong Liu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China.,Research Institute of OtolaryngologyNanjing, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of NHFPCShanghai, China.,Institutes of Biomedical Sciences, Fudan UniversityShanghai, China.,Shanghai Engineering Research Centre of Cochlear ImplantShanghai, China.,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan UniversityShanghai, China
| |
Collapse
|
20
|
Knutsdottir H, Zmurchok C, Bhaskar D, Palsson E, Dalle Nogare D, Chitnis AB, Edelstein-Keshet L. Polarization and migration in the zebrafish posterior lateral line system. PLoS Comput Biol 2017; 13:e1005451. [PMID: 28369079 PMCID: PMC5393887 DOI: 10.1371/journal.pcbi.1005451] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 04/17/2017] [Accepted: 03/13/2017] [Indexed: 12/19/2022] Open
Abstract
Collective cell migration plays an important role in development. Here, we study the posterior lateral line primordium (PLLP) a group of about 100 cells, destined to form sensory structures, that migrates from head to tail in the zebrafish embryo. We model mutually inhibitory FGF-Wnt signalling network in the PLLP and link tissue subdivision (Wnt receptor and FGF receptor activity domains) to receptor-ligand parameters. We then use a 3D cell-based simulation with realistic cell-cell adhesion, interaction forces, and chemotaxis. Our model is able to reproduce experimentally observed motility with leading cells migrating up a gradient of CXCL12a, and trailing (FGF receptor active) cells moving actively by chemotaxis towards FGF ligand secreted by the leading cells. The 3D simulation framework, combined with experiments, allows an investigation of the role of cell division, chemotaxis, adhesion, and other parameters on the shape and speed of the PLLP. The 3D model demonstrates reasonable behaviour of control as well as mutant phenotypes.
Collapse
Affiliation(s)
- Hildur Knutsdottir
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| | - Cole Zmurchok
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dhananjay Bhaskar
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eirikur Palsson
- Department of Biology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Damian Dalle Nogare
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Ajay B. Chitnis
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Leah Edelstein-Keshet
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
In toto imaging of the migrating Zebrafish lateral line primordium at single cell resolution. Dev Biol 2017; 422:14-23. [DOI: 10.1016/j.ydbio.2016.12.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/09/2016] [Accepted: 12/09/2016] [Indexed: 12/21/2022]
|
22
|
Mulvaney JF, Thompkins C, Noda T, Nishimura K, Sun WW, Lin SY, Coffin A, Dabdoub A. Kremen1 regulates mechanosensory hair cell development in the mammalian cochlea and the zebrafish lateral line. Sci Rep 2016; 6:31668. [PMID: 27550540 PMCID: PMC4994024 DOI: 10.1038/srep31668] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/21/2016] [Indexed: 02/06/2023] Open
Abstract
Here we present spatio-temporal localization of Kremen1, a transmembrane receptor, in the mammalian cochlea, and investigate its role in the formation of sensory organs in mammal and fish model organisms. We show that Kremen1 is expressed in prosensory cells during cochlear development and in supporting cells of the adult mouse cochlea. Based on this expression pattern, we investigated whether Kremen1 functions to modulate cell fate decisions in the prosensory domain of the developing cochlea. We used gain and loss-of-function experiments to show that Kremen1 is sufficient to bias cells towards supporting cell fate, and is implicated in suppression of hair cell formation. In addition to our findings in the mouse cochlea, we examined the effects of over expression and loss of Kremen1 in the zebrafish lateral line. In agreement with our mouse data, we show that over expression of Kremen1 has a negative effect on the number of mechanosensory cells that form in the zebrafish neuromasts, and that fish lacking Kremen1 protein develop more hair cells per neuromast compared to wild type fish. Collectively, these data support an inhibitory role for Kremen1 in hair cell fate specification.
Collapse
Affiliation(s)
- Joanna F Mulvaney
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON, M4N 3M5, Canada
| | - Cathrine Thompkins
- College of Arts and Sciences and Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, USA
| | - Teppei Noda
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON, M4N 3M5, Canada
| | - Koji Nishimura
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON, M4N 3M5, Canada
| | - Willy W Sun
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON, M4N 3M5, Canada
| | - Shuh-Yow Lin
- Department of Surgery, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Allison Coffin
- College of Arts and Sciences and Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, USA
| | - Alain Dabdoub
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON, M4N 3M5, Canada.,Department of Otolaryngology - Head and Neck Surgery, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
23
|
Venero Galanternik M, Nikaido M, Yu Z, McKinney SA, Piotrowski T. Localized Gene Induction by Infrared-Mediated Heat Shock. Zebrafish 2016; 13:537-540. [PMID: 27057799 DOI: 10.1089/zeb.2015.1161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Genetic manipulations are a vital instrument for the study of embryonic development where to understand how genes work, it is necessary to provoke a loss or gain of function of a particular gene in a spatial and temporal manner. In the zebrafish embryo, the Hsp70 promoter is the most commonly used tool to induce a transient global gene expression of a desired gene, in a temporal manner. However, Hsp70-driven global gene induction presents caveats when studying gene function in a tissue of interest as gene induction in the whole embryo can lead to cell-autonomous and non-cell-autonomous phenotypes. In the current article, we describe an innovative and cost effective protocol to activate Hsp70-dependent expression in a small subset of cells in the zebrafish embryo, by utilizing a localized infrared (IR) laser. Our IR laser set up can be incorporated to any microscope platform without the requirement for expensive equipment. Furthermore, our protocol allows for controlled localized induction of specific proteins under the control of the hsp70 promoter in small subsets of cells. We use the migrating zebrafish sensory lateral line primordium as a model, because of its relative simplicity and experimental accessibility; however, this technique can be applied to any tissue in the zebrafish embryo.
Collapse
Affiliation(s)
- Marina Venero Galanternik
- 1 Stowers Institute for Medical Research , Kansas City, Missouri.,2 Department of Neurobiology and Anatomy, University of Utah , Salt Lake City, Utah
| | - Masataka Nikaido
- 1 Stowers Institute for Medical Research , Kansas City, Missouri
| | - Zulin Yu
- 1 Stowers Institute for Medical Research , Kansas City, Missouri
| | - Sean A McKinney
- 1 Stowers Institute for Medical Research , Kansas City, Missouri
| | - Tatjana Piotrowski
- 1 Stowers Institute for Medical Research , Kansas City, Missouri.,2 Department of Neurobiology and Anatomy, University of Utah , Salt Lake City, Utah
| |
Collapse
|
24
|
Venero Galanternik M, Navajas Acedo J, Romero-Carvajal A, Piotrowski T. Imaging collective cell migration and hair cell regeneration in the sensory lateral line. Methods Cell Biol 2016; 134:211-56. [PMID: 27312495 DOI: 10.1016/bs.mcb.2016.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The accessibility of the lateral line system and its amenability to long-term in vivo imaging transformed the developing lateral line into a powerful model system to study fundamental morphogenetic events, such as guided migration, proliferation, cell shape changes, organ formation, organ deposition, cell specification and differentiation. In addition, the lateral line is not only amenable to live imaging during migration stages but also during postembryonic events such as sensory organ tissue homeostasis and regeneration. The robust regenerative capabilities of the mature, mechanosensory lateral line hair cells, which are homologous to inner ear hair cells and the ease with which they can be imaged, have brought zebrafish into the spotlight as a model to develop tools to treat human deafness. In this chapter, we describe protocols for long-term in vivo confocal imaging of the developing and regenerating lateral line.
Collapse
Affiliation(s)
- M Venero Galanternik
- Stowers Institute for Medical Research, Kansas City, MO, United States; University of Utah, Salt Lake City, UT, United States
| | - J Navajas Acedo
- Stowers Institute for Medical Research, Kansas City, MO, United States
| | - A Romero-Carvajal
- Stowers Institute for Medical Research, Kansas City, MO, United States; University of Utah, Salt Lake City, UT, United States
| | - T Piotrowski
- Stowers Institute for Medical Research, Kansas City, MO, United States; University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
25
|
Valdivia LE, Lamb DB, Horner W, Wierzbicki C, Tafessu A, Williams AM, Gestri G, Krasnow AM, Vleeshouwer-Neumann TS, Givens M, Young RM, Lawrence LM, Stickney HL, Hawkins TA, Schwarz QP, Cavodeassi F, Wilson SW, Cerveny KL. Antagonism between Gdf6a and retinoic acid pathways controls timing of retinal neurogenesis and growth of the eye in zebrafish. Development 2016; 143:1087-98. [PMID: 26893342 PMCID: PMC4852494 DOI: 10.1242/dev.130922] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/08/2016] [Indexed: 12/27/2022]
Abstract
Maintaining neurogenesis in growing tissues requires a tight balance between progenitor cell proliferation and differentiation. In the zebrafish retina, neuronal differentiation proceeds in two stages with embryonic retinal progenitor cells (RPCs) of the central retina accounting for the first rounds of differentiation, and stem cells from the ciliary marginal zone (CMZ) being responsible for late neurogenesis and growth of the eye. In this study, we analyse two mutants with small eyes that display defects during both early and late phases of retinal neurogenesis. These mutants carry lesions in gdf6a, a gene encoding a BMP family member previously implicated in dorsoventral patterning of the eye. We show that gdf6a mutant eyes exhibit expanded retinoic acid (RA) signalling and demonstrate that exogenous activation of this pathway in wild-type eyes inhibits retinal growth, generating small eyes with a reduced CMZ and fewer proliferating progenitors, similar to gdf6a mutants. We provide evidence that RA regulates the timing of RPC differentiation by promoting cell cycle exit. Furthermore, reducing RA signalling in gdf6a mutants re-establishes appropriate timing of embryonic retinal neurogenesis and restores putative stem and progenitor cell populations in the CMZ. Together, our results support a model in which dorsally expressed gdf6a limits RA pathway activity to control the transition from proliferation to differentiation in the growing eye. Summary: In the vertebrate eye, dorsally expressed Gdf6a limits RA pathway activity to control the transition from proliferation to differentiation, thereby regulating eye size.
Collapse
Affiliation(s)
- Leonardo E Valdivia
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Dayna B Lamb
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Wilson Horner
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Claudia Wierzbicki
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Amanuel Tafessu
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Audrey M Williams
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Gaia Gestri
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Anna M Krasnow
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | | | - McKenzie Givens
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Rodrigo M Young
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Lisa M Lawrence
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Heather L Stickney
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Thomas A Hawkins
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Quenten P Schwarz
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Florencia Cavodeassi
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Kara L Cerveny
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| |
Collapse
|
26
|
Zhao Z, Ma X, Sung D, Li M, Kosti A, Lin G, Chen Y, Pertsemlidis A, Hsiao TH, Du L. microRNA-449a functions as a tumor suppressor in neuroblastoma through inducing cell differentiation and cell cycle arrest. RNA Biol 2016; 12:538-54. [PMID: 25760387 DOI: 10.1080/15476286.2015.1023495] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
microRNA-449a (miR-449a) has been identified to function as a tumor suppressor in several types of cancers. However, the role of miR-449a in neuroblastoma has not been intensively investigated. We recently found that the overexpression of miR-449a significantly induces neuroblastoma cell differentiation, suggesting its potential tumor suppressor function in neuroblastoma. In this study, we further investigated the mechanisms underlying the tumor suppressive function of miR-449a in neuroblastoma. We observed that miR-449a inhibits neuroblastoma cell survival and growth through 2 mechanisms--inducing cell differentiation and cell cycle arrest. Our comprehensive investigations on the dissection of the target genes of miR-449a revealed that 3 novel targets- MFAP4, PKP4 and TSEN15 -play important roles in mediating its differentiation-inducing function. In addition, we further found that its function in inducing cell cycle arrest involves down-regulating its direct targets CDK6 and LEF1. To determine the clinical significance of the miR-449a-mediated tumor suppressive mechanism, we examined the correlation between the expression of these 5 target genes in neuroblastoma tumor specimens and the survival of neuroblastoma patients. Remarkably, we noted that high tumor expression levels of all the 3 miR-449a target genes involved in regulating cell differentiation, but not the target genes involved in regulating cell cycle, are significantly correlated with poor survival of neuroblastoma patients. These results suggest the critical role of the differentiation-inducing function of miR-449a in determining neuroblastoma progression. Overall, our study provides the first comprehensive characterization of the tumor-suppressive function of miR-449a in neuroblastoma, and reveals the potential clinical significance of the miR-449a-mediated tumor suppressive pathway in neuroblastoma prognosis.
Collapse
Affiliation(s)
- Zhenze Zhao
- a Greehey Children's Cancer Research Institute; The University of Texas Health Science Center at San Antonio ; San Antonio , TX USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Solute Carrier Family 26 Member a2 (slc26a2) Regulates Otic Development and Hair Cell Survival in Zebrafish. PLoS One 2015; 10:e0136832. [PMID: 26375458 PMCID: PMC4573323 DOI: 10.1371/journal.pone.0136832] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/10/2015] [Indexed: 12/16/2022] Open
Abstract
Hearing loss is one of the most prevalent human birth defects. Genetic factors contribute to the pathogenesis of deafness. It is estimated that one-third of deafness genes have already been identified. The current work is an attempt to find novel genes relevant to hearing loss using guilt-by-profiling and guilt-by-association bioinformatics analyses of approximately 80 known non-syndromic hereditary hearing loss (NSHL) genes. Among the 300 newly identified candidate deafness genes, slc26a2 were selected for functional studies in zebrafish. The slc26a2 gene was knocked down using an antisense morpholino (MO), and significant defects were observed in otolith patterns, semicircular canal morphology, and lateral neuromast distributions in morphants. Loss-of-function defects are caused primarily by apoptosis, and morphants are insensitive to sound stimulation and imbalanced swimming behaviours. Morphant defects were found to be partially rescued by co-injection of human SLC26A2 mRNA. All the results suggest that bioinformatics is capable of predicting new deafness genes and this showed slc26a2 is to be a critical otic gene whose dysfunction may induce hearing impairment.
Collapse
|
28
|
Agarwala S, Duquesne S, Liu K, Boehm A, Grimm L, Link S, König S, Eimer S, Ronneberger O, Lecaudey V. Amotl2a interacts with the Hippo effector Yap1 and the Wnt/β-catenin effector Lef1 to control tissue size in zebrafish. eLife 2015; 4:e08201. [PMID: 26335201 PMCID: PMC4596637 DOI: 10.7554/elife.08201] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 09/02/2015] [Indexed: 12/17/2022] Open
Abstract
During development, proliferation must be tightly controlled for organs to reach their appropriate size. While the Hippo signaling pathway plays a major role in organ growth control, how it senses and responds to increased cell density is still unclear. In this study, we use the zebrafish lateral line primordium (LLP), a group of migrating epithelial cells that form sensory organs, to understand how tissue growth is controlled during organ formation. Loss of the cell junction-associated Motin protein Amotl2a leads to overproliferation and bigger LLP, affecting the final pattern of sensory organs. Amotl2a function in the LLP is mediated together by the Hippo pathway effector Yap1 and the Wnt/β-catenin effector Lef1. Our results implicate for the first time the Hippo pathway in size regulation in the LL system. We further provide evidence that the Hippo/Motin interaction is essential to limit tissue size during development. DOI:http://dx.doi.org/10.7554/eLife.08201.001 How do organs and tissues know when to stop growing? A cell communication pathway known as Hippo signaling plays a central role as it can tell cells to stop dividing. It is activated when cells in developing tissues come into contact with each other and causes a protein called Yap1 to be modified, which prevents it from entering the cell nucleus to activate genes that are involved in cell division. In a zebrafish embryo, an organ called the lateral line forms from a cluster of cells that migrate along the embryo's length. At regular intervals, the cluster deposits small bunches of cells from its trailing end. The resulting loss of cells from the cluster is balanced by cell division at the front of the cluster, which is triggered by another signaling pathway called Wnt signaling. A protein of the ‘Motin’ family called Amotl2a is present in this migrating cluster. Motin proteins form junctions between cells and inhibit the activity of Yap1, but it is not known whether they are involved in regulating the size of organs. Here, Agarwala et al. used the lateral line as a model to study the control of organ size in zebrafish embryos. The experiments show that when Amotl2a is absent, the migrating cell cluster becomes larger, with the highest levels of cell division occurring at its trailing end. Yap1 and a protein involved in Wnt signaling called Lef1 are also present in the cluster and are required for it to be normal in size. In zebrafish that lack Amotl2a, the additional loss of Yap1 prevents this cluster from becoming too large. From these and other results, it appears that Amotl2a regulates the size of the lateral line cell cluster by restricting the ability of Yap1 and Lef1 to promote cell division. Agarwala et al.'s findings demonstrate a role for Amotl2a in controlling the size of organs. A future challenge is to understand the details of how it restricts the activities of Yap1 and Lef1. DOI:http://dx.doi.org/10.7554/eLife.08201.002
Collapse
Affiliation(s)
- Sobhika Agarwala
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,Developmental Biology, Institute for Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Sandra Duquesne
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,Developmental Biology, Institute for Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Kun Liu
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,Image Analysis Lab, Institute for Computer Science, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Anton Boehm
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,Image Analysis Lab, Institute for Computer Science, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Lin Grimm
- Developmental Biology, Institute for Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Sandra Link
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,Developmental Biology, Institute for Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Sabine König
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Stefan Eimer
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,ZBSA Center for Biological Systems Analysis, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Olaf Ronneberger
- BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.,Image Analysis Lab, Institute for Computer Science, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Virginie Lecaudey
- Developmental Biology, Institute for Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
29
|
Miesfeld JB, Gestri G, Clark BS, Flinn MA, Poole RJ, Bader JR, Besharse JC, Wilson SW, Link BA. Yap and Taz regulate retinal pigment epithelial cell fate. Development 2015; 142:3021-32. [PMID: 26209646 PMCID: PMC4582179 DOI: 10.1242/dev.119008] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 07/10/2015] [Indexed: 12/20/2022]
Abstract
The optic vesicle comprises a pool of bi-potential progenitor cells from which the retinal pigment epithelium (RPE) and neural retina fates segregate during ocular morphogenesis. Several transcription factors and signaling pathways have been shown to be important for RPE maintenance and differentiation, but an understanding of the initial fate specification and determination of this ocular cell type is lacking. We show that Yap/Taz-Tead activity is necessary and sufficient for optic vesicle progenitors to adopt RPE identity in zebrafish. A Tead-responsive transgene is expressed within the domain of the optic cup from which RPE arises, and Yap immunoreactivity localizes to the nuclei of prospective RPE cells. yap (yap1) mutants lack a subset of RPE cells and/or exhibit coloboma. Loss of RPE in yap mutants is exacerbated in combination with taz (wwtr1) mutant alleles such that, when Yap and Taz are both absent, optic vesicle progenitor cells completely lose their ability to form RPE. The mechanism of Yap-dependent RPE cell type determination is reliant on both nuclear localization of Yap and interaction with a Tead co-factor. In contrast to loss of Yap and Taz, overexpression of either protein within optic vesicle progenitors leads to ectopic pigmentation in a dosage-dependent manner. Overall, this study identifies Yap and Taz as key early regulators of RPE genesis and provides a mechanistic framework for understanding the congenital ocular defects of Sveinsson's chorioretinal atrophy and congenital retinal coloboma.
Collapse
Affiliation(s)
- Joel B Miesfeld
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Gaia Gestri
- Department of Cell and Developmental Biology, UCL, London WC1E 6BT, UK
| | - Brian S Clark
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael A Flinn
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Richard J Poole
- Department of Cell and Developmental Biology, UCL, London WC1E 6BT, UK
| | - Jason R Bader
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Joseph C Besharse
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, UCL, London WC1E 6BT, UK
| | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
30
|
Jansson L, Kim GS, Cheng AG. Making sense of Wnt signaling-linking hair cell regeneration to development. Front Cell Neurosci 2015; 9:66. [PMID: 25814927 PMCID: PMC4356074 DOI: 10.3389/fncel.2015.00066] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 02/12/2015] [Indexed: 01/10/2023] Open
Abstract
Wnt signaling is a highly conserved pathway crucial for development and homeostasis of multicellular organisms. Secreted Wnt ligands bind Frizzled receptors to regulate diverse processes such as axis patterning, cell division, and cell fate specification. They also serve to govern self-renewal of somatic stem cells in several adult tissues. The complexity of the pathway can be attributed to the myriad of Wnt and Frizzled combinations as well as its diverse context-dependent functions. In the developing mouse inner ear, Wnt signaling plays diverse roles, including specification of the otic placode and patterning of the otic vesicle. At later stages, its activity governs sensory hair cell specification, cell cycle regulation, and hair cell orientation. In regenerating sensory organs from non-mammalian species, Wnt signaling can also regulate the extent of proliferative hair cell regeneration. This review describes the current knowledge of the roles of Wnt signaling and Wnt-responsive cells in hair cell development and regeneration. We also discuss possible future directions and the potential application and limitation of Wnt signaling in augmenting hair cell regeneration.
Collapse
Affiliation(s)
- Lina Jansson
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University Stanford, CA, USA
| | - Grace S Kim
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University Stanford, CA, USA
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University Stanford, CA, USA
| |
Collapse
|
31
|
Xing C, Gong B, Xue Y, Han Y, Wang Y, Meng A, Jia S. TGFβ1a regulates zebrafish posterior lateral line formation via Smad5 mediated pathway. J Mol Cell Biol 2015; 7:48-61. [PMID: 25603803 DOI: 10.1093/jmcb/mjv004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The zebrafish sensory posterior lateral line (pLL) has become an attractive model for studying collective cell migration and cell morphogenesis. Recent studies have indicated that chemokine, Wnt/β-catenin, Fgf, and Delta-Notch signaling pathways participate in regulating pLL development. However, it remains unclear whether TGFβ signaling pathway is involved in pLL development. Here we report a critical role of TGFβ1 in regulating morphogenesis of the pLL primordium (pLLP). The tgfβ1a gene is abundantly expressed in the lateral line primordium. Knockdown or knockout of tgfβ1a leads to a reduction of neuromast number, an increase of inter-neuromast distance, and a reduced number of hair cells. The aberrant morphogenesis in embryos depleted of tgfβ1a correlates with the reduced expression of atoh1a, deltaA, and n-cadherin/cdh2, which are known important regulators of the pLLP morphogenesis. Like tgfβ1a depletion, knockdown of smad5 that expresses in the pLLP, affects pLLP development whereas overexpression of a constitutive active Smad5 isoform rescues the defects in embryos depleted of tgfβ1a, indicating that Smad5 mediates tgfβ1a function in pLLP development. Therefore, TGFβ/Smad5 signaling plays an important role in the zebrafish lateral line formation.
Collapse
Affiliation(s)
- Cencan Xing
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bo Gong
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yu Xue
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanchao Han
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yixia Wang
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Anming Meng
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shunji Jia
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
32
|
Itch is required for lateral line development in zebrafish. PLoS One 2014; 9:e111799. [PMID: 25369329 PMCID: PMC4219781 DOI: 10.1371/journal.pone.0111799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 10/03/2014] [Indexed: 11/20/2022] Open
Abstract
The zebrafish posterior lateral line is formed during early development by the deposition of neuromasts from a migrating primordium. The molecular mechanisms regulating the regional organization and migration of the primordium involve interactions between Fgf and Wnt/β-catenin signaling and the establishment of specific cxcr4b and cxcr7b cytokine receptor expression domains. Itch has been identified as a regulator in several different signaling pathways, including Wnt and Cxcr4 signaling. We identified two homologous itch genes in zebrafish, itcha and itchb, with generalized expression patterns. By reducing itchb expression in particular upon morpholino knockdown, we demonstrated the importance of Itch in regulating lateral line development by perturbing the patterns of cxcr4b and cxcr7b expression. Itch knockdown results in a failure to down-regulate Wnt signaling and overexpression of cxcr4b in the primordium, slowing migration of the posterior lateral line primordium and resulting in abnormal development of the lateral line.
Collapse
|
33
|
Thomas ED, Cruz IA, Hailey DW, Raible DW. There and back again: development and regeneration of the zebrafish lateral line system. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 4:1-16. [PMID: 25330982 DOI: 10.1002/wdev.160] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/10/2014] [Accepted: 08/24/2014] [Indexed: 12/23/2022]
Abstract
The zebrafish lateral line is a sensory system used to detect changes in water flow. It is comprised of clusters of mechanosensory hair cells called neuromasts. The lateral line is initially established by a migratory group of cells, called a primordium, that deposits neuromasts at stereotyped locations along the surface of the fish. Wnt, FGF, and Notch signaling are all important regulators of various aspects of lateral line development, from primordium migration to hair cell specification. As zebrafish age, the organization of the lateral line becomes more complex in order to accommodate the fish's increased size. This expansion is regulated by many of the same factors involved in the initial development. Furthermore, unlike mammalian hair cells, lateral line hair cells have the capacity to regenerate after damage. New hair cells arise from the proliferation and differentiation of surrounding support cells, and the molecular and cellular pathways regulating this are beginning to be elucidated. All in all, the zebrafish lateral line has proven to be an excellent model in which to study a diverse array of processes, including collective cell migration, cell polarity, cell fate, and regeneration.
Collapse
Affiliation(s)
- Eric D Thomas
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, WA, USA; Department of Biological Structure, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
34
|
McGraw HF, Culbertson MD, Nechiporuk AV. Kremen1 restricts Dkk activity during posterior lateral line development in zebrafish. Development 2014; 141:3212-21. [PMID: 25038040 DOI: 10.1242/dev.102541] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Canonical Wnt signaling plays crucial roles during development and disease. How Wnt signaling is modulated in different in vivo contexts is currently not well understood. Here, we investigate the modulation of Wnt signaling in the posterior lateral line primordium (pLLP), a cohort of ~100 cells that collectively migrate along the trunk of the zebrafish embryo. The pLLP comprises proliferative progenitor cells and organized epithelial cells that will form the mechanosensory organs of the posterior lateral line. Wnt signaling is active in the leading progenitor zone of the pLLP and restricted from the trailing zone through expression of the secreted Wnt inhibitors dkk1b and dkk2. We have identified a zebrafish strain, krm1(nl10), which carries a mutation in the kremen1 gene, a non-obligate co-receptor for the Dkk family of proteins. Previous studies have shown that Kremen1 inhibits Wnt signaling by facilitating internalization of the Kremen1-Dkk-Lrp5/6 complex. Surprisingly, we found that disruption of Kremen1 in the pLLP exhibited molecular and cellular phenotypes associated with a decrease rather than overactivation of Wnt signaling. Transplantation of wild-type cells into the mutant primordia failed to rescue the krm1(nl10) phenotype, thus revealing that the effects of Kremen1 loss are non-cell-autonomous. Finally, ectopic expression of Dkk1b-mTangerine protein revealed larger spread of the fusion protein in the mutant primordia compared with the wild type. Based on our data, we propose a novel mechanism in which Kremen1 modulates Wnt activity by restricting the range of secreted Dkk proteins during collective cell migration in the pLLP.
Collapse
Affiliation(s)
- Hillary F McGraw
- Oregon Health & Science University, Department of Cell and Developmental Biology, Portland, OR 97239, USA
| | - Maya D Culbertson
- Oregon Health & Science University, Department of Cell and Developmental Biology, Portland, OR 97239, USA
| | - Alex V Nechiporuk
- Oregon Health & Science University, Department of Cell and Developmental Biology, Portland, OR 97239, USA
| |
Collapse
|
35
|
Ariza-Cosano A, Bensimon-Brito A, Gómez-Skarmeta JL, Bessa J. sox21a directs lateral line patterning by modulating FGF signaling. Dev Neurobiol 2014; 75:80-92. [PMID: 25044975 DOI: 10.1002/dneu.22211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 07/09/2014] [Accepted: 07/09/2014] [Indexed: 01/19/2023]
Abstract
The development of organs composed by repeated functional units is, in many cases, accomplished by the transition of cells from a progenitor to a differentiation domain, triggering a reiterated developmental program. Yet, how these discrete fields are formed during development is still a largely unresolved question. The posterior lateral line (pLL), a sensory organ present in fish and amphibians, develops from a primordium that migrates along the flanks of the animal periodically depositing neuromasts, the pLL functional units. In zebrafish (Danio rerio), the developmental program of the pLL is triggered by the transit of progenitor cells from a Wnt to a Fgf signaling domain. It has been proposed that these two fields are defined by the antagonistic activity of these two signaling pathways, but how they are formed and maintained is still an open question in the development of the pLL. In this work, we show that sox21a, an HMG -box transcription factor, is expressed within the Fgf domain. We demonstrate that, while the Fgf signaling pathway do not control sox21a, knockdown of sox21a causes impairment of Fgf signaling, expansion of the Wnt signaling domain and disruption of neuromast development. These results suggest that sox21a is a key player in the pLL primordium patterning, fine-tuning the border of the Fgf and Wnt signaling domains.
Collapse
Affiliation(s)
- Ana Ariza-Cosano
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Ctra. Utrera Km 1, Seville, 41013, Spain
| | | | | | | |
Collapse
|
36
|
Steiner AB, Kim T, Cabot V, Hudspeth AJ. Dynamic gene expression by putative hair-cell progenitors during regeneration in the zebrafish lateral line. Proc Natl Acad Sci U S A 2014; 111:E1393-401. [PMID: 24706895 PMCID: PMC3986164 DOI: 10.1073/pnas.1318692111] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hearing loss is most commonly caused by the destruction of mechanosensory hair cells in the ear. This condition is usually permanent: Despite the presence of putative hair-cell progenitors in the cochlea, hair cells are not naturally replenished in adult mammals. Unlike those of the mammalian ear, the progenitor cells of nonmammalian vertebrates can regenerate hair cells throughout life. The basis of this difference remains largely unexplored but may lie in molecular dissimilarities that affect how progenitors respond to hair-cell death. To approach this issue, we analyzed gene expression in hair-cell progenitors of the lateral-line system. We developed a transgenic line of zebrafish that expresses a red fluorescent protein in the presumptive hair-cell progenitors known as mantle cells. Fluorescence-activated cell sorting from the skins of transgenic larvae, followed by microarray-based expression analysis, revealed a constellation of transcripts that are specifically enriched in these cells. Gene expression analysis after hair-cell ablation uncovered a cohort of genes that are differentially regulated early in regeneration, suggesting possible roles in the response of progenitors to hair-cell death. These results provide a resource for studying hair-cell regeneration and the biology of sensory progenitor cells.
Collapse
Affiliation(s)
- Aaron B Steiner
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY 10065
| | | | | | | |
Collapse
|
37
|
Lush ME, Piotrowski T. ErbB expressing Schwann cells control lateral line progenitor cells via non-cell-autonomous regulation of Wnt/β-catenin. eLife 2014; 3:e01832. [PMID: 24642408 PMCID: PMC3957165 DOI: 10.7554/elife.01832] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 02/13/2014] [Indexed: 12/31/2022] Open
Abstract
Proper orchestration of quiescence and activation of progenitor cells is crucial during embryonic development and adult homeostasis. We took advantage of the zebrafish sensory lateral line to define niche-progenitor interactions to understand how integration of diverse signaling pathways spatially and temporally regulates the coordination of these processes. Our previous studies demonstrated that Schwann cells play a crucial role in negatively regulating lateral line progenitor proliferation. Here we demonstrate that ErbB/Neuregulin signaling is not only required for Schwann cell migration but that it plays a continued role in postmigratory Schwann cells. ErbB expressing Schwann cells inhibit lateral line progenitor proliferation and differentiation through non-cell-autonomous inhibition of Wnt/β-catenin signaling. Subsequent activation of Fgf signaling controls sensory organ differentiation, but not progenitor proliferation. In addition to the lateral line, these findings have important implications for understanding how niche-progenitor cells segregate interactions during development, and how they may go wrong in disease states. DOI: http://dx.doi.org/10.7554/eLife.01832.001.
Collapse
Affiliation(s)
- Mark E Lush
- Stowers Institute for Medical Research, Kansas City, United States
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, United States
| | - Tatjana Piotrowski
- Stowers Institute for Medical Research, Kansas City, United States
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, United States
| |
Collapse
|
38
|
Han HW, Chou CM, Chu CY, Cheng CH, Yang CH, Hung CC, Hwang PP, Lee SJ, Liao YF, Huang CJ. The Nogo-C2/Nogo receptor complex regulates the morphogenesis of zebrafish lateral line primordium through modulating the expression of dkk1b, a Wnt signal inhibitor. PLoS One 2014; 9:e86345. [PMID: 24466042 PMCID: PMC3897714 DOI: 10.1371/journal.pone.0086345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 12/06/2013] [Indexed: 12/19/2022] Open
Abstract
The fish lateral line (LL) is a mechanosensory system closely related to the hearing system of higher vertebrates, and it is composed of several neuromasts located on the surface of the fish. These neuromasts can detect changes in external water flow, to assist fish in maintaining a stationary position in a stream. In the present study, we identified a novel function of Nogo/Nogo receptor signaling in the formation of zebrafish neuromasts. Nogo signaling in zebrafish, like that in mammals, involves three ligands and four receptors, as well as three co-receptors (TROY, p75, and LINGO-1). We first demonstrated that Nogo-C2, NgRH1a, p75, and TROY are able to form a Nogo-C2 complex, and that disintegration of this complex causes defective neuromast formation in zebrafish. Time-lapse recording of the CldnB::lynEGFP transgenic line revealed that functional obstruction of the Nogo-C2 complex causes disordered morphogenesis, and reduces rosette formation in the posterior LL (PLL) primordium during migration. Consistent with these findings, hair-cell progenitors were lost from the PLL primordium in p75, TROY, and Nogo-C2/NgRH1a morphants. Notably, the expression levels of pea3, a downstream marker of Fgf signaling, and dkk1b, a Wnt signaling inhibitor, were both decreased in p75, TROY, and Nogo-C2/NgRH1a morphants; moreover, dkk1b mRNA injection could rescue the defects in neuromast formation resulting from knockdown of p75 or TROY. We thus suggest that a novel Nogo-C2 complex, consisting of Nogo-C2, NgRH1a, p75, and TROY, regulates Fgf signaling and dkk1b expression, thereby ensuring stable organization of the PLL primordium.
Collapse
Affiliation(s)
- Hao-Wei Han
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chih-Ming Chou
- Department of Biochemistry, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ying Chu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chia-Hsiung Cheng
- Department of Biochemistry, Taipei Medical University, Taipei, Taiwan
| | | | - Chin-Chun Hung
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Shyh-Jye Lee
- Institute of Zoology, National Taiwan University, Taipei, Taiwan
| | - Yung-Feng Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- * E-mail: (CJH); (YFL)
| | - Chang-Jen Huang
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- * E-mail: (CJH); (YFL)
| |
Collapse
|
39
|
He Y, Wu J, Mei H, Yu H, Sun S, Shou J, Li H. Histone deacetylase activity is required for embryonic posterior lateral line development. Cell Prolif 2013; 47:91-104. [PMID: 24267956 DOI: 10.1111/cpr.12081] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/21/2013] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES The posterior lateral line (PLL) system in zebrafish has recently become a model for investigating tissue morphogenesis. PLL primordium periodically deposits neuromasts as it migrates along the horizontal myoseptum from head to tail of the embryonic fish, and this migration requires activity of various molecular mechanisms. Histone deacetylases (HDACs) have been implicated in numerous biological processes of development, by regulating gene transcription, but their roles in regulating PLL during embryonic development have up to now remained unexplored. MATERIAL AND METHODS In this study, we used HDAC inhibitors to investigate the role of HDACs in early development of the zebrafish PLL sensory system. We further investigated development of the PLL by cell-specific immunostaining and in situ hybridization. RESULTS Our analysis showed that HDACs were involved in zebrafish PLL development as pharmacological inhibition of HDACs resulted in its defective formation. We observed that migration of PLL primordium was altered and accompanied by disrupted development of PLL neuromasts in HDAC inhibitor-treated embryos. In these, positions of PLL neuromasts were affected. In particular, the first PLL neuromast was displaced posteriorly in a treatment dose-dependent manner. Primordium cell proliferation was reduced upon HDAC inhibition. Finally, we showed that inhibition of HDAC function reduced numbers of hair cells in PLL neuromasts of HDAC inhibitor-treated embryos. CONCLUSION Here, we have revealed a novel role for HDACs in orchestrating PLL morphogenesis. Our results suggest that HDAC activity is necessary for control of cell proliferation and migration of PLL primordium and hair cell differentiation during early stages of PLL development in zebrafish.
Collapse
Affiliation(s)
- Y He
- Institutes of Biomedical Sciences of Fudan University, Shanghai, 200032, China; Department of Otolaryngology, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, 200031, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Jacques BE, Montgomery WH, Uribe PM, Yatteau A, Asuncion JD, Resendiz G, Matsui JI, Dabdoub A. The role of Wnt/β-catenin signaling in proliferation and regeneration of the developing basilar papilla and lateral line. Dev Neurobiol 2013; 74:438-56. [PMID: 24115534 DOI: 10.1002/dneu.22134] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/09/2013] [Accepted: 09/16/2013] [Indexed: 12/22/2022]
Abstract
Canonical Wnt/β-catenin signaling has been implicated in multiple developmental events including the regulation of proliferation, cell fate, and differentiation. In the inner ear, Wnt/β-catenin signaling is required from the earliest stages of otic placode specification through the formation of the mature cochlea. Within the avian inner ear, the basilar papilla (BP), many Wnt pathway components are expressed throughout development. Here, using reporter constructs for Wnt/β-catenin signaling, we show that this pathway is active throughout the BP (E6-E14) in both hair cells (HCs) and supporting cells. To characterize the role of Wnt/β-catenin activity in developing HCs, we performed gain- and loss-of-function experiments in vitro and in vivo in the chick BP and zebrafish lateral line systems, respectively. Pharmacological inhibition of Wnt signaling in the BP and lateral line neuromasts during the periods of proliferation and HC differentiation resulted in reduced proliferation and decreased HC formation. Conversely, pharmacological activation of this pathway significantly increased the number of HCs in the lateral line and BP. Results demonstrated that this increase was the result of up-regulated cell proliferation within the Sox2-positive cells of the prosensory domains. Furthermore, Wnt/β-catenin activation resulted in enhanced HC regeneration in the zebrafish lateral line following aminoglycoside-induced HC loss. Combined, our data suggest that Wnt/β-catenin signaling specifies the number of cells within the prosensory domain and subsequently the number of HCs. This ability to induce proliferation suggests that the modulation of Wnt/β-catenin signaling could play an important role in therapeutic HC regeneration.
Collapse
Affiliation(s)
- Bonnie E Jacques
- Department of Surgery/Otolaryngology, UCSD School of Medicine, La Jolla, CA, 92093
| | | | | | | | | | | | | | | |
Collapse
|
41
|
A Hox gene controls lateral line cell migration by regulating chemokine receptor expression downstream of Wnt signaling. Proc Natl Acad Sci U S A 2013; 110:16892-7. [PMID: 24082091 DOI: 10.1073/pnas.1306282110] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The posterior lateral line primordium in zebrafish provides an amenable model to study mechanisms of collective cell migration. The directed migration of the cell cluster along the path of Sdf1a chemokine requires two receptors, Cxcr4b and Cxcr7b, which are expressed in the leading and trailing part of the primordium, respectively. The polarized expression of receptors is regulated by Wnt signaling, but downstream players mediating this control remain to be found. Here, we show that the Hox homeobox gene Hoxb8a is a critical component that acts downstream of the Wnt pathway to coordinate the expression of both chemokine receptors. We find that Hoxb8a is expressed in the leading part of the primordium and is required for the correct speed and extent of migration. Hoxb8a expression is dependent upon Wnt activity and needed both for cxcr4b expression and to repress and thus restrict cxcr7b expression to the trailing zone of the primordium. In the absence of Wnt activity, overexpressed Hoxb8a is able to repress cxcr7b but not up-regulate cxcr4b expression. Together with results from expressing dominant activator and repressor constructs, these findings suggest that Hoxb8a is induced by and cooperates with Wnt signaling to up-regulate cxcr4b, and acts through multiple mechanisms to repress cxcr7b expression.
Collapse
|
42
|
Increased LEF1 expression and decreased Notch2 expression are strong predictors of poor outcomes in colorectal cancer patients. DISEASE MARKERS 2013; 35:395-405. [PMID: 24223455 PMCID: PMC3810108 DOI: 10.1155/2013/983981] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/14/2013] [Accepted: 08/05/2013] [Indexed: 01/27/2023]
Abstract
Background/Objective. We aimed to examine the expression of lymphoid enhancer factor 1 (LEF1) and Notch2 in colorectal cancer (CRC) and their association with clinicopathologic variables and CRC patients' prognosis. Methods. Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot analysis were performed to assess the expression of LEF1 and Notch2 in 184 patients with CRC. Results. We observed a strong negative correlation between LEF1 expression and Notch2 expression (P < 0.001). Both LEF1 mRNA and protein expression increased while the Notch2 mRNA and protein expression decreased in tumor specimens compared with the matched paratumorous normal tissue (P < 0.001). An increase in LEF1 protein expression was significantly associated with lymph node metastases, distant metastasis, advanced TNM (tumor-node-metastasis) stage, and shorter overall survival. A decrease in Notch2 protein expression was associated with poorly differentiated tumors, lymph node metastases, distant metastasis, advanced TNM stage, and shorter overall survival of patients. In the multivariate Cox regression analysis, the LEF1 protein expression (P < 0.001), Notch2 protein expression (P < 0.001), TNM stage (P < 0.001), and the combination of increased LEF1 protein coexpression and decreased Notch2 protein coexpression (P < 0.001) were found to be independent prognostic indicators in CRC. Conclusion. Our results suggest that increased LEF1 coexpression and decreased Notch2 coexpression represent a risk factor for poor overall survival of CRC patients.
Collapse
|
43
|
Wada H, Ghysen A, Asakawa K, Abe G, Ishitani T, Kawakami K. Wnt/Dkk negative feedback regulates sensory organ size in zebrafish. Curr Biol 2013; 23:1559-65. [PMID: 23891113 DOI: 10.1016/j.cub.2013.06.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 05/16/2013] [Accepted: 06/13/2013] [Indexed: 11/18/2022]
Abstract
Correct organ size must involve a balance between promotion and inhibition of cell proliferation. A mathematical model has been proposed in which an organ is assumed to produce its own growth activator as well as a growth inhibitor [1], but there is as yet no molecular evidence to support this model [2]. The mechanosensory organs of the fish lateral line system (neuromasts) are composed of a core of sensory hair cells surrounded by nonsensory support cells. Sensory cells are constantly replaced and are regenerated from surrounding nonsensory cells [3], while each organ retains the same size throughout life. Moreover, neuromasts also bud off new neuromasts, which stop growing when they reach the same size [4, 5]. Here, we show that the size of neuromasts is controlled by a balance between growth-promoting Wnt signaling activity in proliferation-competent cells and Wnt-inhibiting Dkk activity produced by differentiated sensory cells. This negative feedback loop from Dkk (secreted by differentiated cells) on Wnt-dependent cell proliferation (in surrounding cells) also acts during regeneration to achieve size constancy. This study establishes Wnt/Dkk as a novel mechanism to determine the final size of an organ.
Collapse
Affiliation(s)
- Hironori Wada
- PRESTO, Japan Science and Technology Agency (JST), Kawaguchi, Saitama 322-0012, Japan.
| | | | | | | | | | | |
Collapse
|
44
|
Head JR, Gacioch L, Pennisi M, Meyers JR. Activation of canonical Wnt/β-catenin signaling stimulates proliferation in neuromasts in the zebrafish posterior lateral line. Dev Dyn 2013; 242:832-46. [PMID: 23606225 DOI: 10.1002/dvdy.23973] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 03/22/2013] [Accepted: 03/23/2013] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The posterior lateral line in zebrafish develops from a migrating primordium that deposits clusters of cells that differentiate into neuromasts at regular intervals along the trunk. The deposition of these neuromasts is known to be coordinated by Wnt and FGF signals that control the proliferation, migration, and organization of the primordium. However, little is known about the control of proliferation in the neuromasts following their deposition. RESULTS We show that pharmacological activation of the Wnt/β-catenin signaling pathway with 1-azakenpaullone upregulates proliferation in neuromasts post-deposition. This results in increased size of the neuromasts and overproduction of sensory hair cells. We also show that activation of Wnt signaling returns already quiescent supporting cells to a proliferative state in mature neuromasts. Additionally, activation of Wnt signaling increases the number of supporting cells that return to the cell cycle in response to hair cell damage and the number of regenerated hair cells. Finally, we show that inhibition of Wnt signaling by overexpression of dkk1b suppresses proliferation during both differentiation and regeneration. CONCLUSIONS These data suggest that Wnt/β-catenin signaling is both necessary and sufficient for the control of proliferation of lateral line progenitors during development, ongoing growth of the neuromasts, and hair cell regeneration.
Collapse
Affiliation(s)
- Jeffery R Head
- Neuroscience Program, Colgate University, Hamilton, New York, USA
| | | | | | | |
Collapse
|
45
|
Moro E, Vettori A, Porazzi P, Schiavone M, Rampazzo E, Casari A, Ek O, Facchinello N, Astone M, Zancan I, Milanetto M, Tiso N, Argenton F. Generation and application of signaling pathway reporter lines in zebrafish. Mol Genet Genomics 2013; 288:231-42. [PMID: 23674148 PMCID: PMC3664755 DOI: 10.1007/s00438-013-0750-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/02/2013] [Indexed: 12/22/2022]
Abstract
In the last years, we have seen the emergence of different tools that have changed the face of biology from a simple modeling level to a more systematic science. The transparent zebrafish embryo is one of the living models in which, after germline transformation with reporter protein-coding genes, specific fluorescent cell populations can be followed at single-cell resolution. The genetically modified embryos, larvae and adults, resulting from the transformation, are individuals in which time lapse analysis, digital imaging quantification, FACS sorting and next-generation sequencing can be performed in specific times and tissues. These multifaceted genetic and cellular approaches have permitted to dissect molecular interactions at the subcellular, intercellular, tissue and whole-animal level, thus allowing integration of cellular and developmental genetics with molecular imaging in the resulting frame of modern biology. In this review, we describe a new step in the zebrafish road to system biology, based on the use of transgenic biosensor animals expressing fluorescent proteins under the control of signaling pathway-responsive cis-elements. In particular, we provide here the rationale and details of this powerful tool, trying to focus on its huge potentialities in basic and applied research, while also discussing limits and potential technological evolutions of this approach.
Collapse
Affiliation(s)
- Enrico Moro
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padua, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Matsuda M, Nogare DD, Somers K, Martin K, Wang C, Chitnis AB. Lef1 regulates Dusp6 to influence neuromast formation and spacing in the zebrafish posterior lateral line primordium. Development 2013; 140:2387-97. [PMID: 23637337 DOI: 10.1242/dev.091348] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The posterior lateral line primordium (PLLp) migrates caudally and periodically deposits neuromasts. Coupled, but mutually inhibitory, Wnt-FGF signaling systems regulate proto-neuromast formation in the PLLp: FGF ligands expressed in response to Wnt signaling activate FGF receptors and initiate proto-neuromast formation. FGF receptor signaling, in turn, inhibits Wnt signaling. However, mechanisms that determine periodic neuromast formation and deposition in the PLLp remain poorly understood. Previous studies showed that neuromasts are deposited closer together and the PLLp terminates prematurely in lef1-deficient zebrafish embryos. It was suggested that this results from reduced proliferation in the leading domain of the PLLp and/or premature incorporation of progenitors into proto-neuromasts. We found that rspo3 knockdown reduces proliferation in a manner similar to that seen in lef1 morphants. However, it does not cause closer neuromast deposition or premature termination of the PLLp, suggesting that such changes in lef1-deficient embryos are not linked to changes in proliferation. Instead, we suggest that they are related to the role of Lef1 in regulating the balance of Wnt and FGF functions in the PLLp. Lef1 determines expression of the FGF signaling inhibitor Dusp6 in leading cells and regulates incorporation of cells into neuromasts; reduction of Dusp6 in leading cells in lef1-deficient embryos allows new proto-neuromasts to form closer to the leading edge. This is associated with progressively slower PLLp migration, reduced spacing between deposited neuromasts and premature termination of the PLLp system.
Collapse
Affiliation(s)
- Miho Matsuda
- Program in Genomics of Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20852, USA
| | | | | | | | | | | |
Collapse
|
47
|
Innervation is required for sense organ development in the lateral line system of adult zebrafish. Proc Natl Acad Sci U S A 2013; 110:5659-64. [PMID: 23509277 DOI: 10.1073/pnas.1214004110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Superficial mechanosensory organs (neuromasts) distributed over the head and body of fishes and amphibians form the "lateral line" system. During zebrafish adulthood, each neuromast of the body (posterior lateral line system, or PLL) produces "accessory" neuromasts that remain tightly clustered, thereby increasing the total number of PLL neuromasts by a factor of more than 10. This expansion is achieved by a budding process and is accompanied by branches of the afferent nerve that innervates the founder neuromast. Here we show that innervation is essential for the budding process, in complete contrast with the development of the embryonic PLL, where innervation is entirely dispensable. To obtain insight into the molecular mechanisms that underlie the budding process, we focused on the terminal system that develops at the posterior tip of the body and on the caudal fin. In this subset of PLL neuromasts, bud neuromasts form in a reproducible sequence over a few days, much faster than for other PLL neuromasts. We show that wingless/int (Wnt) signaling takes place during, and is required for, the budding process. We also show that the Wnt activator R-spondin is expressed by the axons that innervate budding neuromasts. We propose that the axon triggers Wnt signaling, which itself is involved in the proliferative phase that leads to bud formation. Finally, we show that innervation is required not only for budding, but also for long-term maintenance of all PLL neuromasts.
Collapse
|
48
|
Wang X, Kopinke D, Lin J, McPherson AD, Duncan RN, Otsuna H, Moro E, Hoshijima K, Grunwald DJ, Argenton F, Chien CB, Murtaugh LC, Dorsky RI. Wnt signaling regulates postembryonic hypothalamic progenitor differentiation. Dev Cell 2013; 23:624-36. [PMID: 22975330 DOI: 10.1016/j.devcel.2012.07.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 05/15/2012] [Accepted: 07/17/2012] [Indexed: 12/21/2022]
Abstract
Previous studies have raised the possibility that Wnt signaling may regulate both neural progenitor maintenance and neuronal differentiation within a single population. Here we investigate the role of Wnt/β-catenin activity in the zebrafish hypothalamus and find that the pathway is first required for the proliferation of unspecified hypothalamic progenitors in the embryo. At later stages, including adulthood, sequential activation and inhibition of Wnt activity is required for the differentiation of neural progenitors and negatively regulates radial glia differentiation. The presence of Wnt activity is conserved in hypothalamic progenitors of the adult mouse, where it plays a conserved role in inhibiting the differentiation of radial glia. This study establishes the vertebrate hypothalamus as a model for Wnt-regulated postembryonic neural progenitor differentiation and defines specific roles for Wnt signaling in neurogenesis.
Collapse
Affiliation(s)
- Xu Wang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gaete M, Tucker AS. Organized emergence of multiple-generations of teeth in snakes is dysregulated by activation of Wnt/beta-catenin signalling. PLoS One 2013; 8:e74484. [PMID: 24019968 PMCID: PMC3760860 DOI: 10.1371/journal.pone.0074484] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 08/02/2013] [Indexed: 02/01/2023] Open
Abstract
In contrast to mammals, most reptiles constantly regenerate their teeth. In the snake, the epithelial dental lamina ends in a successional lamina, which proliferates and elongates forming multiple tooth generations, all linked by a permanent dental lamina. To investigate the mechanisms used to control the initiation of new tooth germs in an ordered sequential pattern we utilized the polyphodont (multiple-generation) corn snake (Pantherophis guttatus). We observed that the dental lamina expressed the transcription factor Sox2, a multipotent stem cell marker, whereas the successional lamina cells expressed the transcription factor Lef1, a Wnt/β-catenin pathway target gene. Activation of the Wnt/β-catenin pathway in culture increased the number of developing tooth germs, in comparison to control untreated cultures. These additional tooth germs budded off from ectopic positions along the dental lamina, rather than in an ordered sequence from the successional lamina. Wnt/β-catenin activation enhanced cell proliferation, particularly in normally non-odontogenic regions of the dental lamina, which widely expressed Lef1, restricting the Sox2 domain. This suggests an expansion of the successional lamina at the expense of the dental lamina. Activation of the Wnt/β-catenin pathway in cultured snake dental organs, therefore, led to changes in proliferation and to the molecular pattern of the dental lamina, resulting in loss of the organised emergence of tooth germs. These results suggest that epithelial compartments are critical for the arrangement of organs that develop in sequence, and highlight the role of Wnt/β-catenin signalling in such processes.
Collapse
Affiliation(s)
- Marcia Gaete
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, London, United Kingdom
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Abigail S. Tucker
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Breau MA, Wilson D, Wilkinson DG, Xu Q. Chemokine and Fgf signalling act as opposing guidance cues in formation of the lateral line primordium. Development 2012; 139:2246-53. [PMID: 22619392 DOI: 10.1242/dev.080275] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The directional migration of many cell populations occurs as a coherent group. An amenable model is provided by the posterior lateral line in zebrafish, which is formed by a cohesive primordium that migrates from head to tail and deposits future neuromasts at intervals. We found that prior to the onset of migration, the compact state of the primordium is not fully established, as isolated cells with lateral line identity are present caudal to the main primordium. These isolated cells are retained in position such that they fuse with the migrating primordium as it advances, and later contribute to the leading zone and terminal neuromasts. We found that the isolated lateral line cells are positioned by two antagonistic cues: Fgf signalling attracts them towards the primordium, which counteracts Sdf1α/Cxcr4b-mediated caudal attraction. These findings reveal a novel chemotactic role for Fgf signalling in which it enables the coalescence of the lateral line primordium from an initial fuzzy pattern into a compact group of migrating cells.
Collapse
Affiliation(s)
- Marie A Breau
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, UK
| | | | | | | |
Collapse
|