1
|
Wu J, Shao Y, Hua X, Wang Y, Wang D. Nanoplastic at environmentally relevant concentrations induces toxicity across multiple generations associated with inhibition in germline G protein-coupled receptor CED-1 in Caenorhabditis elegans. CHEMOSPHERE 2024; 364:143011. [PMID: 39098352 DOI: 10.1016/j.chemosphere.2024.143011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/17/2024] [Accepted: 08/02/2024] [Indexed: 08/06/2024]
Abstract
Nanoplastics at environmentally relevant concentrations (ERCs) could cause transgenerational toxicity on organisms. Caenorhabditis elegans is an important model for the study of transgenerational toxicology of pollutants. Nevertheless, the underlying mechanisms for the control of transgenerational nanoplastic toxicity by germline signals remain largely unclear. In C. elegans, exposure to 1-100 μg/L polystyrene nanoparticle (PS-NP) decreased expression of germline ced-1 encoding a G protein-coupled receptor at parental generation (P0-G). After PS-NP exposure at P0-G, transgenerational decrease in germline ced-1 expression could be detected. Meanwhile, the susceptibility to transgenerational PS-NP toxicity was observed in ced-1(RNAi) animals. After PS-NP exposure at P0-G, germline RNAi of ced-1 increased expressions of met-2 and set-6 encoding histone methylation transferases. The susceptibility of ced-1(RNAi) to transgenerational PS-NP toxicity could be inhibited by RNAi of met-2 and set-6. Moreover, in PS-NP exposed met-2(RNAi) and set-6(RNAi) nematodes, expressions of ins-39, wrt-3, and/or efn-3 encoding secreted ligands were decreased. Therefore, our results demonstrated that inhibition in germline CED-1 mediated the toxicity induction of nanoplastics at ERCs across multiple generations in nematodes.
Collapse
Affiliation(s)
- Jingwei Wu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China
| | - Yuting Shao
- School of Public Health, Southeast University, Nanjing, China
| | - Xin Hua
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China
| | - Yuxing Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China
| | - Dayong Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China; Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, Shenzhen, China.
| |
Collapse
|
2
|
Lyu H, Chamberlin HM. Functional distinction in oncogenic Ras variant activity in Caenorhabditis elegans. Dis Model Mech 2024; 17:dmm050577. [PMID: 38946472 PMCID: PMC11340813 DOI: 10.1242/dmm.050577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 06/21/2024] [Indexed: 07/02/2024] Open
Abstract
Ras genes are important oncogenes that are frequently mutated in cancer. Human oncogenic variants exhibit functional distinctions in terms of their representation in different cancer types, impact on cellular targets and sensitivity to pharmacological treatments. However, how these distinct variants influence and respond to the cellular networks in which they are embedded is poorly understood. To identify novel participants in the complex interplay between Ras genotype and cell interaction networks in vivo, we have developed and tested an experimental framework using a simple vulva-development assay in the nematode C. elegans. Using this system, we evaluated a set of Ras oncogenic substitution changes at G12, G13 and Q61. We found that these variants fall into distinct groups based on phenotypic differences, sensitivity to gene dosage and inhibition of the downstream kinase MEK and their response to genetic modulators that influence Ras activity in a non-autonomous manner. Together, our results demonstrated that oncogenic C. elegans Ras variants exhibit clear distinctions in how they interface with the vulva-development network and showed that extracellular modulators yield variant-restricted effects in vivo.
Collapse
Affiliation(s)
- Haimeng Lyu
- Department of Molecular Genetics, Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
| | - Helen M. Chamberlin
- Department of Molecular Genetics, Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Paolillo VK, Ochs ME, Lundquist EA. MAB-5/Hox regulates the Q neuroblast transcriptome, including cwn-1/Wnt, to mediate posterior migration in Caenorhabditis elegans. Genetics 2024; 227:iyae045. [PMID: 38652773 PMCID: PMC11151924 DOI: 10.1093/genetics/iyae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/14/2024] [Indexed: 04/25/2024] Open
Abstract
Neurogenesis involves the precisely coordinated action of genetic programs controlling large-scale neuronal fate specification down to terminal events of neuronal differentiation. The Q neuroblasts in Caenorhabditis elegans, QL on the left and QR on the right, divide, differentiate, and migrate in a similar pattern to produce three neurons each. However, QL on the left migrates posteriorly, and QR on the right migrates anteriorly. The MAB-5/Hox transcription factor is necessary and sufficient for posterior Q lineage migration and is normally expressed only in the QL lineage. To define genes controlled by MAB-5 in the Q cells, fluorescence-activated cell sorting was utilized to isolate populations of Q cells at a time in early L1 larvae when MAB-5 first becomes active. Sorted Q cells from wild-type, mab-5 loss-of-function (lof), and mab-5 gain-of-function (gof) mutants were subject to RNA-seq and differential expression analysis. Genes enriched in Q cells included those involved in cell division, DNA replication, and DNA repair, consist with the neuroblast stem cell identity of the Q cells at this stage. Genes affected by mab-5 included those involved in neurogenesis, neural development, and interaction with the extracellular matrix. cwn-1, which encodes a Wnt signaling molecule, showed a paired response to mab-5 in the Q cells: cwn-1 expression was reduced in mab-5(lof) and increased in mab-5(gof), suggesting that MAB-5 is required for cwn-1 expression in Q cells. MAB-5 is required to prevent anterior migration of the Q lineage while it transcriptionally reprograms the Q lineage for posterior migration. Functional genetic analysis revealed that CWN-1 is required downstream of MAB-5 to inhibit anterior migration of the QL lineage, likely in parallel to EGL-20/Wnt in a noncanonical Wnt pathway. In sum, work here describes a Q cell transcriptome, and a set of genes regulated by MAB-5 in the QL lineage. One of these genes, cwn-1, acts downstream of mab-5 in QL migration, indicating that this gene set includes other genes utilized by MAB-5 to facilitate posterior neuroblast migration.
Collapse
Affiliation(s)
- Vitoria K Paolillo
- Department of Molecular Biosciences, KU Center for Genomics, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Matthew E Ochs
- Department of Molecular Biosciences, KU Center for Genomics, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Erik A Lundquist
- Department of Molecular Biosciences, KU Center for Genomics, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| |
Collapse
|
4
|
Paolillo VK, Ochs ME, Lundquist EA. MAB-5/Hox regulates the Q neuroblast transcriptome, including cwn-1/Wnt, to mediate posterior migration in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566461. [PMID: 37986999 PMCID: PMC10659417 DOI: 10.1101/2023.11.09.566461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Neurogenesis involves the precisely-coordinated action of genetic programs controlling large-scale neuronal fate specification down to terminal events of neuronal differentiation. The Q neuroblasts in C. elegans, QL on the left and QR on the right, divide, differentiate, and migrate in a similar pattern to produce three neurons each. However, QL on the left migrates posteriorly, and QR on the right migrates anteriorly. The MAB-5/Hox transcription factor is necessary and sufficient for posterior Q lineage migration, and is normally expressed only in the QL lineage. To define genes controlled by MAB-5 in the Q cells, fluorescence-activated cell sorting was utilized to isolate populations of Q cells at a time in early L1 larvae when MAB-5 first becomes active. Sorted Q cells from wild-type, mab-5 loss-of-function (lof), and mab-5 gain-of-function (gof) mutants were subject to RNA-seq and differential expression analysis. Genes enriched in Q cells included those involved in cell division, DNA replication, and DNA repair, consist with the neuroblast stem cell identity of the Q cells at this stage. Genes affected by mab-5 included those involved in neurogenesis, neural development, and interaction with the extracellular matrix. cwn-1, which encodes a Wnt signaling molecule, showed a paired response to mab-5 in the Q cells: cwn-1 expression was reduced in mab-5(lof) and increased in mab-5(gof), suggesting that MAB-5 is required for cwn-1 expression in Q cells. MAB-5 is required to prevent anterior migration of the Q lineage while it transcriptionally reprograms the Q lineage for posterior migration. Functional genetic analysis revealed that CWN-1 is required downstream of MAB-5 to inhibit anterior migration of the QL lineage, likely in parallel to EGL-20/Wnt in a non-canonical Wnt pathway. In sum, work here describes a Q cell transcriptome, and a set of genes regulated by MAB-5 in the QL lineage. One of these genes, cwn-1, acts downstream of mab-5 in QL migration, indicating that this gene set includes other genes utilized by MAB-5 to facilitate posterior neuroblast migration.
Collapse
Affiliation(s)
- Vitoria K Paolillo
- KU Center for Genomics, Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045
| | - Matthew E Ochs
- KU Center for Genomics, Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045
| | - Erik A Lundquist
- KU Center for Genomics, Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045
| |
Collapse
|
5
|
Heinze SD, Berger S, Engleitner S, Daube M, Hajnal A. Prolonging somatic cell proliferation through constitutive hox gene expression in C. elegans. Nat Commun 2023; 14:6850. [PMID: 37891160 PMCID: PMC10611754 DOI: 10.1038/s41467-023-42644-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
hox genes encode a conserved family of homeodomain transcription factors that are essential to determine the identity of body segments during embryogenesis and maintain adult somatic stem cells competent to regenerate organs. In contrast to higher organisms, somatic cells in C. elegans irreversibly exit the cell cycle after completing their cell lineage and the adult soma cannot regenerate. Here, we show that hox gene expression levels in C. elegans determine the temporal competence of somatic cells to proliferate. Down-regulation of the central hox gene lin-39 in dividing vulval cells results in their premature cell cycle exit, whereas constitutive lin-39 expression causes precocious Pn.p cell and sex myoblast divisions and prolongs the proliferative phase of the vulval cells past their normal point of arrest. Furthermore, ectopic expression of hox genes in the quiescent anchor cell re-activates the cell cycle and induces proliferation until young adulthood. Thus, constitutive expression of a single hox transcription factor is sufficient to prolong somatic cell proliferation beyond the restriction imposed by the cell lineage. The down-regulation of hox gene expression in most somatic cells at the end of larval development may be one cause for the absence of cell proliferation in adult C. elegans.
Collapse
Affiliation(s)
- Svenia D Heinze
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, CH-8057, Zürich, Switzerland
| | - Simon Berger
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Institute for Chemical- and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093, Zürich, Switzerland
| | - Stefanie Engleitner
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, CH-8057, Zürich, Switzerland
| | - Michael Daube
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Alex Hajnal
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.
| |
Collapse
|
6
|
Ayoub M, David LM, Shklyar B, Hakim-Mishnaevski K, Kurant E. Drosophila FGFR/Htl signaling shapes embryonic glia to phagocytose apoptotic neurons. Cell Death Discov 2023; 9:90. [PMID: 36898998 PMCID: PMC10006210 DOI: 10.1038/s41420-023-01382-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Glial phagocytosis of apoptotic neurons is crucial for development and proper function of the central nervous system. Relying on transmembrane receptors located on their protrusions, phagocytic glia recognize and engulf apoptotic debris. Like vertebrate microglia, Drosophila phagocytic glial cells form an elaborate network in the developing brain to reach and remove apoptotic neurons. However, the mechanisms controlling creation of the branched morphology of these glial cells critical for their phagocytic ability remain unknown. Here, we demonstrate that during early embryogenesis, the Drosophila fibroblast growth factor receptor (FGFR) Heartless (Htl) and its ligand Pyramus are essential in glial cells for the formation of glial extensions, the presence of which strongly affects glial phagocytosis of apoptotic neurons during later stages of embryonic development. Reduction in Htl pathway activity results in shorter lengths and lower complexity of glial branches, thereby disrupting the glial network. Our work thus illuminates the important role Htl signaling plays in glial subcellular morphogenesis and in establishing glial phagocytic ability.
Collapse
Affiliation(s)
- Malak Ayoub
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 3498838, Haifa, Israel
| | - Li-Mor David
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 3498838, Haifa, Israel
| | - Boris Shklyar
- Bioimaging Unit, Faculty of Natural Sciences, University of Haifa, 3498838, Haifa, Israel
| | - Ketty Hakim-Mishnaevski
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 3498838, Haifa, Israel
| | - Estee Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 3498838, Haifa, Israel.
| |
Collapse
|
7
|
Wang R, Leite DJ, Karadas L, Schiffer PH, Pechmann M. FGF signalling is involved in cumulus migration in the common house spider Parasteatoda tepidariorum. Dev Biol 2023; 494:35-45. [PMID: 36470448 DOI: 10.1016/j.ydbio.2022.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Cell migration is a fundamental component during the development of most multicellular organisms. In the early spider embryo, the collective migration of signalling cells, known as the cumulus, is required to set the dorsoventral body axis. Here, we show that FGF signalling plays an important role during cumulus migration in the spider Parasteatoda tepidariorum. Spider embryos with reduced FGF signalling show reduced or absent cumulus migration and display dorsoventral patterning defects. Our study reveals that the transcription factor Ets4 regulates the expression of several FGF signalling components in the cumulus. In conjunction with a previous study, we show that the expression of fgf8 in the germ-disc is regulated via the Hedgehog signalling pathway. We also demonstrate that FGF signalling influences the BMP signalling pathway activity in the region around cumulus cells. Finally, we show that FGFR signalling might also influence cumulus migration in basally branching spiders and we propose that fgf8 might act as a chemo-attractant to guide cumulus cells towards the future dorsal pole of the spider embryo.
Collapse
Affiliation(s)
- Ruixun Wang
- Institute for Zoology/Developmental Biology, Biocenter, University of Cologne, Zuelpicher Str. 47b, 50674, Cologne, Germany
| | - Daniel J Leite
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK
| | - Linda Karadas
- Institute for Zoology/Developmental Biology, Biocenter, University of Cologne, Zuelpicher Str. 47b, 50674, Cologne, Germany
| | - Philipp H Schiffer
- Institute for Zoology/Developmental Biology, Biocenter, University of Cologne, Zuelpicher Str. 47b, 50674, Cologne, Germany
| | - Matthias Pechmann
- Institute for Zoology/Developmental Biology, Biocenter, University of Cologne, Zuelpicher Str. 47b, 50674, Cologne, Germany.
| |
Collapse
|
8
|
Fergin A, Boesch G, Greter NR, Berger S, Hajnal A. Tissue-specific inhibition of protein sumoylation uncovers diverse SUMO functions during C. elegans vulval development. PLoS Genet 2022; 18:e1009978. [PMID: 35666766 PMCID: PMC9203017 DOI: 10.1371/journal.pgen.1009978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/16/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022] Open
Abstract
The sumoylation (SUMO) pathway is involved in a variety of processes during C. elegans development, such as gonadal and vulval fate specification, cell cycle progression and maintenance of chromosome structure. The ubiquitous expression and pleiotropic effects have made it difficult to dissect the tissue-specific functions of the SUMO pathway and identify its target proteins. To overcome these challenges, we have established tools to block protein sumoylation and degrade sumoylated target proteins in a tissue-specific and temporally controlled manner. We employed the auxin-inducible protein degradation system (AID) to down-regulate the SUMO E3 ligase GEI-17 or the SUMO ortholog SMO-1, either in the vulval precursor cells (VPCs) or in the gonadal anchor cell (AC). Our results indicate that the SUMO pathway acts in multiple tissues to control different aspects of vulval development, such as AC positioning, basement membrane (BM) breaching, VPC fate specification and morphogenesis. Inhibition of protein sumoylation in the VPCs resulted in abnormal toroid formation and ectopic cell fusions during vulval morphogenesis. In particular, sumoylation of the ETS transcription factor LIN-1 at K169 is necessary for the proper contraction of the ventral vulA toroids. Thus, the SUMO pathway plays several distinct roles throughout vulval development. Many proteins are chemically modified after they have been synthesized. In particular, conjugation with the Small Ubiquitin-like Modifier (SUMO) regulates the functions and activities of a large number of proteins in animal and plant cells. Here, we have used the Nematode Caenorhabditis elegans to study the various effects of SUMO protein modification on organ development. By applying a tissue-specific protein degradation system, we could selectively block the SUMO pathway in different tissues of the animals. We focused on the development of the egg-laying organ as a model, and found that the SUMO pathway acts in multiple tissues to regulate distinct cellular functions. Finally, we show that SUMO modification of one transcription factor, called LIN-1, is necessary for the proper morphogenesis of the organ. Our results indicate that the manifold effects of the SUMO pathway can be attributed to the combined action of a distinct number of SUMO modified proteins acting in different cell types.
Collapse
Affiliation(s)
- Aleksandra Fergin
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, Zürich, Switzerland
| | - Gabriel Boesch
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, Zürich, Switzerland
| | - Nadja R. Greter
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, Zürich, Switzerland
| | - Simon Berger
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, Zürich, Switzerland
| | - Alex Hajnal
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
9
|
A DNA replication-independent function of pre-replication complex genes during cell invasion in C. elegans. PLoS Biol 2022; 20:e3001317. [PMID: 35192608 PMCID: PMC8863262 DOI: 10.1371/journal.pbio.3001317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/01/2021] [Indexed: 11/19/2022] Open
Abstract
Cell invasion is an initiating event during tumor cell metastasis and an essential process during development. A screen of C. elegans orthologs of genes overexpressed in invasive human melanoma cells has identified several components of the conserved DNA pre-replication complex (pre-RC) as positive regulators of anchor cell (AC) invasion. The pre-RC genes function cell-autonomously in the G1-arrested AC to promote invasion, independently of their role in licensing DNA replication origins in proliferating cells. While the helicase activity of the pre-RC is necessary for AC invasion, the downstream acting DNA replication initiation factors are not required. The pre-RC promotes the invasive fate by regulating the expression of extracellular matrix genes and components of the PI3K signaling pathway. Increasing PI3K pathway activity partially suppressed the AC invasion defects caused by pre-RC depletion, suggesting that the PI3K pathway is one critical pre-RC target. We propose that the pre-RC, or a part of it, acts in the postmitotic AC as a transcriptional regulator that facilitates the switch to an invasive phenotype.
Collapse
|
10
|
Nakamura A, Goto Y, Kondo Y, Aoki K. Shedding light on developmental ERK signaling with genetically encoded biosensors. Development 2021; 148:271153. [PMID: 34338283 DOI: 10.1242/dev.199767] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The extracellular signal-regulated kinase (ERK) pathway governs cell proliferation, differentiation and migration, and therefore plays key roles in various developmental and regenerative processes. Recent advances in genetically encoded fluorescent biosensors have unveiled hitherto unrecognized ERK activation dynamics in space and time and their functional importance mainly in cultured cells. However, ERK dynamics during embryonic development have still only been visualized in limited numbers of model organisms, and we are far from a sufficient understanding of the roles played by developmental ERK dynamics. In this Review, we first provide an overview of the biosensors used for visualization of ERK activity in live cells. Second, we highlight the applications of the biosensors to developmental studies of model organisms and discuss the current understanding of how ERK dynamics are encoded and decoded for cell fate decision-making.
Collapse
Affiliation(s)
- Akinobu Nakamura
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Yuhei Goto
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Yohei Kondo
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Kazuhiro Aoki
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.,IRCC International Research Collaboration Center, National Institutes of Natural Sciences, 4-3-13 Toranomon, Minato-ku, Tokyo 105-0001, Japan
| |
Collapse
|
11
|
Andrikou C, Hejnol A. FGF signaling acts on different levels of mesoderm development within Spiralia. Development 2021; 148:264929. [PMID: 33999997 PMCID: PMC8180254 DOI: 10.1242/dev.196089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/08/2021] [Indexed: 01/23/2023]
Abstract
FGF signaling is involved in mesoderm induction in members of deuterostomes (e.g. tunicates, hemichordates), but not in flies and nematodes, in which it has a role in mesoderm patterning and migration. However, we need comparable studies in other protostome taxa in order to decipher whether this mesoderm-inducing function of FGF extends beyond the lineage of deuterostomes. Here, we investigated the role of FGF signaling in mesoderm development in three species of lophophorates, a clade within the protostome group Spiralia. Our gene expression analyses show that the mesodermal molecular patterning is conserved between brachiopods and phoronids, but the spatial and temporal recruitment of transcription factors differs significantly. Moreover, the use of the inhibitor SU5402 demonstrates that FGF signaling is involved in different steps of mesoderm development, as well as in morphogenetic movements of gastrulation and axial elongation. Our findings suggest that the mesoderm-inducing role of FGF extends beyond the group of deuterostomes.
Collapse
Affiliation(s)
- Carmen Andrikou
- University of Bergen, Department of Biological Sciences, Thormøhlensgate 55, 5006 Bergen, Norway.,Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006 Bergen, Norway
| | - Andreas Hejnol
- University of Bergen, Department of Biological Sciences, Thormøhlensgate 55, 5006 Bergen, Norway.,Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006 Bergen, Norway
| |
Collapse
|
12
|
de la Cova CC, Townley R, Greenwald I. Negative feedback by conserved kinases patterns the degradation of Caenorhabditis elegans Raf in vulval fate patterning. Development 2020; 147:226094. [PMID: 33144396 DOI: 10.1242/dev.195941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/27/2020] [Indexed: 11/20/2022]
Abstract
Activation of a canonical EGFR-Ras-Raf-ERK cascade initiates patterning of multipotent vulval precursor cells (VPCs) of Caenorhabditis elegans We have previously shown that this pathway includes a negative-feedback component in which MPK-1/ERK activity targets the upstream kinase LIN-45/Raf for degradation by the SEL-10/FBXW7 E3 ubiquitin ligase. This regulation requires a Cdc4 phosphodegron (CPD) in LIN-45 that is conserved in BRAF. Here, we identify and characterize the minimal degron that encompasses the CPD and is sufficient for SEL-10-mediated, MPK-1-dependent protein degradation. A targeted screen of conserved protein kinase-encoding genes yielded gsk-3 (an ortholog of human GSK3B) and cdk-2 (a CDK2-related kinase) as required for LIN-45 degron-mediated turnover. Genetic analysis revealed that LIN-45 degradation is blocked at the second larval stage due to cell cycle quiescence, and that relief of this block during the third larval stage relies on activation of CDKs. Additionally, activation of MPK-1 provides spatial pattern to LIN-45 degradation but does not bypass the requirement for gsk-3 and cdk-2 This analysis supports a model whereby MPK-1/ERK, GSK-3/GSK3 and CDK-2/CDK2, along with SEL-10/FBXW7, constitute a regulatory network that exerts spatial and temporal control of LIN-45/Raf degradation during VPC patterning.
Collapse
Affiliation(s)
- Claire C de la Cova
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA.,Department of Biological Sciences, University of Wisconsin Milwaukee, Milwaukee, WI 53201, USA
| | - Robert Townley
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA.,Department of Biological Sciences, University of Wisconsin Milwaukee, Milwaukee, WI 53201, USA
| | - Iva Greenwald
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|
13
|
Cell Non-autonomous Function of daf-18/PTEN in the Somatic Gonad Coordinates Somatic Gonad and Germline Development in C. elegans Dauer Larvae. Curr Biol 2019; 29:1064-1072.e8. [PMID: 30827916 DOI: 10.1016/j.cub.2019.01.076] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 12/21/2022]
Abstract
C. elegans larvae integrate environmental information and developmental decisions [1-3]. In favorable conditions, worms develop rapidly and continuously through four larval stages into reproductive adulthood. However, if conditions are unfavorable through the second larval stage, worms enter dauer diapause, a state of global and reversible developmental arrest in which precursor cells remain quiescent and preserve developmental potential, anticipating developmental progression if conditions improve. Signaling from neurons, hypodermis, and intestine regulate the appearance and behavior of dauer larvae and many aspects of developmental arrest of the non-gonadal soma [1, 4, 5]. Here, we show that the decision of somatic gonad blast cells (SGBs) and germline stem cells (GSCs) to be quiescent or progress developmentally is regulated differently from the non-gonadal soma: daf-18/PTEN acts non-autonomously within the somatic gonad to maintain developmental quiescence of both SGBs and GSCs. Our analysis suggests that daf-18 acts in somatic gonad cells to produce a "pro-quiescence" signal (or signals) that acts inter se and between the somatic gonad and the germline. The inferred signal does not require DAF-2/insulin receptor or maintain quiescence of the nearby sex myoblasts, and developmental progression in daf-18(0) does not require dafachronic acids. Abrogating quiescence in dauer results in post-dauer sterility. Our results implicate the somatic gonad as an endocrine organ to synchronize somatic gonad and germline development during dauer diapause and recovery, and our finding that PTEN acts non-autonomously to control blast cell quiescence may be relevant to its function as a tumor suppressor in mammals and to combating parasitic nematodes.
Collapse
|
14
|
A Receptor Tyrosine Kinase Network Regulates Neuromuscular Function in Response to Oxidative Stress in Caenorhabditis elegans. Genetics 2019; 211:1283-1295. [PMID: 30782598 DOI: 10.1534/genetics.119.302026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/14/2019] [Indexed: 11/18/2022] Open
Abstract
The transcription factor Nrf2 plays a critical role in the organism-wide regulation of the antioxidant stress response. The Nrf2 homolog SKN-1 functions in the intestinal cells nonautonomously to negatively regulate neuromuscular junction (NMJ) function in Caenorhabditis elegans To identify additional molecules that mediate SKN-1 signaling to the NMJ, we performed a candidate screen for suppressors of aldicarb resistance caused by acute treatment with the SKN-1 activator arsenite. We identified two receptor tyrosine kinases, EGL-15 (fibroblast growth factor receptor, FGFR) and DAF-2 (insulin-like peptide receptor), that are required for NMJ regulation in response to stress. Through double-mutant analysis, we found that EGL-15 functions downstream of, or parallel to, SKN-1 and SPHK-1 (sphingosine kinase), and that the EGL-15 ligand EGL-17 FGF and canonical EGL-15 effectors are required for oxidative stress-mediated regulation of NMJ function. DAF-2 also functions downstream of or parallel to SKN-1 to regulate NMJ function. Through tissue-specific rescue experiments, we found that FGFR signaling functions primarily in the hypodermis, whereas insulin-like peptide receptor signaling is required in multiple tissues. Our results support the idea that the regulation of NMJ function by SKN-1 occurs via a complex organism-wide signaling network involving receptor tyrosine kinase signaling in multiple tissues.
Collapse
|
15
|
Shin H, Reiner DJ. The Signaling Network Controlling C. elegans Vulval Cell Fate Patterning. J Dev Biol 2018; 6:E30. [PMID: 30544993 PMCID: PMC6316802 DOI: 10.3390/jdb6040030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/17/2022] Open
Abstract
EGF, emitted by the Anchor Cell, patterns six equipotent C. elegans vulval precursor cells to assume a precise array of three cell fates with high fidelity. A group of core and modulatory signaling cascades forms a signaling network that demonstrates plasticity during the transition from naïve to terminally differentiated cells. In this review, we summarize the history of classical developmental manipulations and molecular genetics experiments that led to our understanding of the signals governing this process, and discuss principles of signal transduction and developmental biology that have emerged from these studies.
Collapse
Affiliation(s)
- Hanna Shin
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
| | - David J Reiner
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
- College of Medicine, Texas A & M University, Houston, TX 77030, USA.
| |
Collapse
|
16
|
A New Tool for Inducible Gene Expression in Caenorhabditis elegans. Genetics 2018; 211:419-430. [PMID: 30504365 DOI: 10.1534/genetics.118.301705] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Controlling protein activity and localization is a key tool in modern biology. Mammalian steroid receptor ligand-binding domain (LBD) fusions have been used in a range of organisms and cell types to inactivate proteins of interest until the cognate steroid ligand is applied. Here, we demonstrate that the glucocorticoid receptor LBD confers ligand-gated control of a heterologous gene expression system (Q system) and the DAF-16 transcription factor in Caenorhabditis elegans These experiments provide a powerful tool for temporal control of protein activity, and will bolster existing tools used to modulate gene expression and protein activity in this animal.
Collapse
|
17
|
Fan TP, Ting HC, Yu JK, Su YH. Reiterative use of FGF signaling in mesoderm development during embryogenesis and metamorphosis in the hemichordate Ptychodera flava. BMC Evol Biol 2018; 18:120. [PMID: 30075704 PMCID: PMC6091094 DOI: 10.1186/s12862-018-1235-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/26/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Mesoderm is generally considered to be a germ layer that is unique to Bilateria, and it develops into diverse tissues, including muscle, and in the case of vertebrates, the skeleton and notochord. Studies on various deuterostome animals have demonstrated that fibroblast growth factor (FGF) signaling is required for the formation of many mesodermal structures, such as vertebrate somites, from which muscles are differentiated, and muscles in sea urchin embryos, suggesting an ancient role of FGF signaling in muscle development. However, the formation of trunk muscles in invertebrate chordates is FGF-independent, leading to ambiguity about this ancient role in deuterostomes. To further understand the role of FGF signaling during deuterostome evolution, we investigated the development of mesodermal structures during embryogenesis and metamorphosis in Ptychodera flava, an indirect-developing hemichordate that has larval morphology similar to echinoderms and adult body features that are similar to chordates. RESULTS Here we show that genes encoding FGF ligands, FGF receptors and transcription factors that are known to be involved in mesoderm formation and myogenesis are expressed dynamically during embryogenesis and metamorphosis. FGF signaling at the early gastrula stage is required for the specification of the mesodermal cell fate in P. flava. The mesoderm cells are then differentiated stepwise into the hydroporic canal, the pharyngeal muscle and the muscle string; formation of the last two muscular structures are controlled by FGF signaling. Moreover, augmentation of FGF signaling during metamorphosis accelerated the process, facilitating the transformation from cilia-driven swimming larvae into muscle-driven worm-like juveniles. CONCLUSIONS Our data show that FGF signaling is required for mesoderm induction and myogenesis in the P. flava embryo, and it is reiteratively used for the morphological transition during metamorphosis. The dependence of muscle development on FGF signaling in both planktonic larvae and sand-burrowing worms supports its ancestral role in deuterostomes.
Collapse
Affiliation(s)
- Tzu-Pei Fan
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei, 11529, Taiwan.,Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Hsiu-Chi Ting
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan
| | - Jr-Kai Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan
| | - Yi-Hsien Su
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei, 11529, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan. .,Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan.
| |
Collapse
|
18
|
Barr MM, García LR, Portman DS. Sexual Dimorphism and Sex Differences in Caenorhabditis elegans Neuronal Development and Behavior. Genetics 2018; 208:909-935. [PMID: 29487147 PMCID: PMC5844341 DOI: 10.1534/genetics.117.300294] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/05/2018] [Indexed: 01/05/2023] Open
Abstract
As fundamental features of nearly all animal species, sexual dimorphisms and sex differences have particular relevance for the development and function of the nervous system. The unique advantages of the nematode Caenorhabditis elegans have allowed the neurobiology of sex to be studied at unprecedented scale, linking ultrastructure, molecular genetics, cell biology, development, neural circuit function, and behavior. Sex differences in the C. elegans nervous system encompass prominent anatomical dimorphisms as well as differences in physiology and connectivity. The influence of sex on behavior is just as diverse, with biological sex programming innate sex-specific behaviors and modifying many other aspects of neural circuit function. The study of these differences has provided important insights into mechanisms of neurogenesis, cell fate specification, and differentiation; synaptogenesis and connectivity; principles of circuit function, plasticity, and behavior; social communication; and many other areas of modern neurobiology.
Collapse
Affiliation(s)
- Maureen M Barr
- Department of Genetics, Rutgers University, Piscataway, New Jersey 08854-8082
| | - L Rene García
- Department of Biology, Texas A&M University, College Station, Texas 77843-3258
| | - Douglas S Portman
- Department of Biomedical Genetics, University of Rochester, New York 14642
- Department of Neuroscience, University of Rochester, New York 14642
- Department of Biology, University of Rochester, New York 14642
| |
Collapse
|
19
|
Sherwood DR, Plastino J. Invading, Leading and Navigating Cells in Caenorhabditis elegans: Insights into Cell Movement in Vivo. Genetics 2018; 208:53-78. [PMID: 29301948 PMCID: PMC5753875 DOI: 10.1534/genetics.117.300082] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022] Open
Abstract
Highly regulated cell migration events are crucial during animal tissue formation and the trafficking of cells to sites of infection and injury. Misregulation of cell movement underlies numerous human diseases, including cancer. Although originally studied primarily in two-dimensional in vitro assays, most cell migrations in vivo occur in complex three-dimensional tissue environments that are difficult to recapitulate in cell culture or ex vivo Further, it is now known that cells can mobilize a diverse repertoire of migration modes and subcellular structures to move through and around tissues. This review provides an overview of three distinct cellular movement events in Caenorhabditis elegans-cell invasion through basement membrane, leader cell migration during organ formation, and individual cell migration around tissues-which together illustrate powerful experimental models of diverse modes of movement in vivo We discuss new insights into migration that are emerging from these in vivo studies and important future directions toward understanding the remarkable and assorted ways that cells move in animals.
Collapse
Affiliation(s)
- David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Durham, North Carolina 27705
| | - Julie Plastino
- Institut Curie, PSL Research University, CNRS, UMR 168, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, UMR 168, F-75005 Paris, France
| |
Collapse
|
20
|
Yang Q, Roiz D, Mereu L, Daube M, Hajnal A. The Invading Anchor Cell Induces Lateral Membrane Constriction during Vulval Lumen Morphogenesis in C. elegans. Dev Cell 2017; 42:271-285.e3. [PMID: 28787593 DOI: 10.1016/j.devcel.2017.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 05/15/2017] [Accepted: 07/11/2017] [Indexed: 12/16/2022]
Abstract
During epithelial tube morphogenesis, linear arrays of cells are converted into tubular structures through actomyosin-generated intracellular forces that induce tissue invagination and lumen formation. We have investigated lumen morphogenesis in the C. elegans vulva. The first discernible event initiating lumen formation is the apical constriction of the two innermost primary cells (VulF). The VulF cells thereafter constrict their lateral membranes along the apicobasal axis to extend the lumen dorsally. Lateral, but not apical, VulF constriction requires the prior invasion of the anchor cell (AC). The invading AC extends actin-rich protrusions toward VulF, resulting in the formation of a direct AC-VulF interface. The recruitment of the F-BAR-domain protein TOCA-1 to the AC-VulF interface induces the accumulation of force-generating actomyosin, causing a switch from apical to lateral membrane constriction and the dorsal extension of the lumen. Invasive cells may induce shape changes in adjacent cells to penetrate their target tissues.
Collapse
Affiliation(s)
- Qiutan Yang
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Zurich PhD Program in Molecular Life Sciences, Uni ETH Zürich, 8057 Zurich, Switzerland
| | - Daniel Roiz
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Zurich PhD Program in Molecular Life Sciences, Uni ETH Zürich, 8057 Zurich, Switzerland
| | - Louisa Mereu
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Zurich PhD Program in Molecular Life Sciences, Uni ETH Zürich, 8057 Zurich, Switzerland
| | - Michael Daube
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Alex Hajnal
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
21
|
de la Cova C, Townley R, Regot S, Greenwald I. A Real-Time Biosensor for ERK Activity Reveals Signaling Dynamics during C. elegans Cell Fate Specification. Dev Cell 2017; 42:542-553.e4. [PMID: 28826819 PMCID: PMC5595649 DOI: 10.1016/j.devcel.2017.07.014] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/19/2017] [Accepted: 07/20/2017] [Indexed: 01/06/2023]
Abstract
Kinase translocation reporters (KTRs) are genetically encoded fluorescent activity sensors that convert kinase activity into a nucleocytoplasmic shuttling equilibrium for visualizing single-cell signaling dynamics. Here, we adapt the first-generation KTR for extracellular signal-regulated kinase (ERK) to allow easy implementation in vivo. This sensor, "ERK-nKTR," allows quantitative and qualitative assessment of ERK activity by analysis of individual nuclei and faithfully reports ERK activity during development and neural function in diverse cell contexts in Caenorhabditis elegans. Analysis of ERK activity over time in the vulval precursor cells, a well-characterized paradigm of epidermal growth factor receptor (EGFR)-Ras-ERK signaling, has identified dynamic features not evident from analysis of developmental endpoints alone, including pulsatile frequency-modulated signaling associated with proximity to the EGF source. The toolkit described here will facilitate studies of ERK signaling in other C. elegans contexts, and the design features will enable implementation of this technology in other multicellular organisms.
Collapse
Affiliation(s)
- Claire de la Cova
- Department of Biological Sciences, Columbia University, New York, NY, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Medical Center, New York, NY, USA
| | - Robert Townley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Sergi Regot
- Department of Molecular Biology & Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Iva Greenwald
- Department of Biological Sciences, Columbia University, New York, NY, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
22
|
Wani S, Kuroyanagi H. An emerging model organism Caenorhabditis elegans for alternative pre-mRNA processing in vivo. WILEY INTERDISCIPLINARY REVIEWS-RNA 2017; 8. [PMID: 28703462 DOI: 10.1002/wrna.1428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 05/02/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022]
Abstract
A nematode Caenorhabditis elegans is an intron-rich organism and up to 25% of its pre-mRNAs are estimated to be alternatively processed. Its compact genomic organization enables construction of fluorescence splicing reporters with intact genomic sequences and visualization of alternative processing patterns of interest in the transparent living animals with single-cell resolution. Genetic analysis with the reporter worms facilitated identification of trans-acting factors and cis-acting elements, which are highly conserved in mammals. Analysis of unspliced and partially spliced pre-mRNAs in vivo raised models for alternative splicing regulation relying on specific order of intron excision. RNA-seq analysis of splicing factor mutants and CLIP-seq analysis of the factors allow global search for target genes in the whole animal. An mRNA surveillance system is not essential for its viability or fertility, allowing analysis of unproductively spliced noncoding mRNAs. These features offer C. elegans as an ideal model organism for elucidating alternative pre-mRNA processing mechanisms in vivo. Examples of isoform-specific functions of alternatively processed genes are summarized. WIREs RNA 2017, 8:e1428. doi: 10.1002/wrna.1428 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Shotaro Wani
- Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidehito Kuroyanagi
- Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
23
|
Dawes AT, Wu D, Mahalak KK, Zitnik EM, Kravtsova N, Su H, Chamberlin HM. A computational model predicts genetic nodes that allow switching between species-specific responses in a conserved signaling network. Integr Biol (Camb) 2017; 9:156-166. [DOI: 10.1039/c6ib00238b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alterations to only specific parameters in a model including EGF, Wnt and Notch lead to cell behavior differences.
Collapse
Affiliation(s)
- Adriana T. Dawes
- Department of Mathematics
- Ohio State University
- Columbus
- USA
- Department of Molecular Genetics
| | - David Wu
- Department of Mathematics
- Ohio State University
- Columbus
- USA
| | - Karley K. Mahalak
- Department of Molecular Genetics
- Ohio State University
- Columbus
- USA
- Graduate Program in Molecular
| | - Edward M. Zitnik
- Department of Molecular Genetics
- Ohio State University
- Columbus
- USA
| | - Natalia Kravtsova
- Department of Mathematics
- Ohio State University
- Columbus
- USA
- Department of Statistics
| | - Haiwei Su
- Department of Mathematics
- Ohio State University
- Columbus
- USA
| | | |
Collapse
|
24
|
Lussi YC, Mariani L, Friis C, Peltonen J, Myers TR, Krag C, Wong G, Salcini AE. Impaired removal of H3K4 methylation affects cell fate determination and gene transcription. Development 2016; 143:3751-3762. [PMID: 27578789 DOI: 10.1242/dev.139139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/20/2016] [Indexed: 01/30/2023]
Abstract
Methylation of histone 3 lysine 4 (H3K4) is largely associated with promoters and enhancers of actively transcribed genes and is finely regulated during development by the action of histone methyltransferases and demethylases. H3K4me3 demethylases of the KDM5 family have been previously implicated in development, but how the regulation of H3K4me3 level controls developmental processes is not fully established. Here, we show that the H3K4 demethylase RBR-2, the unique member of the KDM5 family in C. elegans, acts cell-autonomously and in a catalytic-dependent manner to control vulva precursor cells fate acquisition, by promoting the LIN-12/Notch pathway. Using genome-wide approaches, we show that RBR-2 reduces the H3K4me3 level at transcription start sites (TSSs) and in regions upstream of the TSSs, and acts both as a transcription repressor and activator. Analysis of the lin-11 genetic locus, a direct RBR-2 target gene required for vulva precursor cell fate acquisition, shows that RBR-2 controls the epigenetic signature of the lin-11 vulva-specific enhancer and lin-11 expression, providing in vivo evidence that RBR-2 can positively regulate transcription and cell fate acquisition by controlling enhancer activity.
Collapse
Affiliation(s)
- Yvonne C Lussi
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK-2200, Denmark.,Centre for Epigenetics, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Luca Mariani
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK-2200, Denmark.,Centre for Epigenetics, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Carsten Friis
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK-2200, Denmark.,Centre for Epigenetics, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Juhani Peltonen
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, Kuopio 70211, Finland
| | - Toshia R Myers
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK-2200, Denmark.,Centre for Epigenetics, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Claudia Krag
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Garry Wong
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, Kuopio 70211, Finland
| | - Anna Elisabetta Salcini
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK-2200, Denmark .,Centre for Epigenetics, University of Copenhagen, Copenhagen DK-2200, Denmark
| |
Collapse
|
25
|
Rella L, Fernandes Póvoa EE, Korswagen HC. The Caenorhabditis elegans Q neuroblasts: A powerful system to study cell migration at single-cell resolution in vivo. Genesis 2016; 54:198-211. [PMID: 26934462 DOI: 10.1002/dvg.22931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/09/2016] [Accepted: 02/25/2016] [Indexed: 11/08/2022]
Abstract
During development, cell migration plays a central role in the formation of tissues and organs. Understanding the molecular mechanisms that drive and control these migrations is a key challenge in developmental biology that will provide important insights into disease processes, including cancer cell metastasis. In this article, we discuss the Caenorhabditis elegans Q neuroblasts and their descendants as a tool to study cell migration at single-cell resolution in vivo. The highly stereotypical migration of these cells provides a powerful system to study the dynamic cytoskeletal processes that drive migration as well as the evolutionarily conserved signaling pathways (including different Wnt signaling cascades) that guide the cells along their specific trajectories. Here, we provide an overview of what is currently known about Q neuroblast migration and highlight the live-cell imaging, genome editing, and quantitative gene expression techniques that have been developed to study this process.
Collapse
Affiliation(s)
- Lorenzo Rella
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| | - Euclides E Fernandes Póvoa
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| | - Hendrik C Korswagen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| |
Collapse
|
26
|
Nunes QM, Li Y, Sun C, Kinnunen TK, Fernig DG. Fibroblast growth factors as tissue repair and regeneration therapeutics. PeerJ 2016; 4:e1535. [PMID: 26793421 PMCID: PMC4715458 DOI: 10.7717/peerj.1535] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/05/2015] [Indexed: 12/25/2022] Open
Abstract
Cell communication is central to the integration of cell function required for the development and homeostasis of multicellular animals. Proteins are an important currency of cell communication, acting locally (auto-, juxta-, or paracrine) or systemically (endocrine). The fibroblast growth factor (FGF) family contributes to the regulation of virtually all aspects of development and organogenesis, and after birth to tissue maintenance, as well as particular aspects of organism physiology. In the West, oncology has been the focus of translation of FGF research, whereas in China and to an extent Japan a major focus has been to use FGFs in repair and regeneration settings. These differences have their roots in research history and aims. The Chinese drive into biotechnology and the delivery of engineered clinical grade FGFs by a major Chinese research group were important enablers in this respect. The Chinese language clinical literature is not widely accessible. To put this into context, we provide the essential molecular and functional background to the FGF communication system covering FGF ligands, the heparan sulfate and Klotho co-receptors and FGF receptor (FGFR) tyrosine kinases. We then summarise a selection of clinical reports that demonstrate the efficacy of engineered recombinant FGF ligands in treating a wide range of conditions that require tissue repair/regeneration. Alongside, the functional reasons why application of exogenous FGF ligands does not lead to cancers are described. Together, this highlights that the FGF ligands represent a major opportunity for clinical translation that has been largely overlooked in the West.
Collapse
Affiliation(s)
- Quentin M. Nunes
- Department of Molecular and Clinical Cancer Medicine, NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, United Kingdom
| | - Yong Li
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Changye Sun
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Tarja K. Kinnunen
- Department of Biology, School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
| | - David G. Fernig
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
27
|
Andrikou C, Pai CY, Su YH, Arnone MI. Logics and properties of a genetic regulatory program that drives embryonic muscle development in an echinoderm. eLife 2015. [PMID: 26218224 PMCID: PMC4549668 DOI: 10.7554/elife.07343] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Evolutionary origin of muscle is a central question when discussing mesoderm evolution. Developmental mechanisms underlying somatic muscle development have mostly been studied in vertebrates and fly where multiple signals and hierarchic genetic regulatory cascades selectively specify myoblasts from a pool of naive mesodermal progenitors. However, due to the increased organismic complexity and distant phylogenetic position of the two systems, a general mechanistic understanding of myogenesis is still lacking. In this study, we propose a gene regulatory network (GRN) model that promotes myogenesis in the sea urchin embryo, an early branching deuterostome. A fibroblast growth factor signaling and four Forkhead transcription factors consist the central part of our model and appear to orchestrate the myogenic process. The topological properties of the network reveal dense gene interwiring and a multilevel transcriptional regulation of conserved and novel myogenic genes. Finally, the comparison of the myogenic network architecture among different animal groups highlights the evolutionary plasticity of developmental GRNs. DOI:http://dx.doi.org/10.7554/eLife.07343.001 Muscles, bones, and blood vessels all develop from a tissue called the mesoderm, which forms early on in the development of an embryo. Networks of genes control which parts of the mesoderm transform into different cell types. The gene networks that control the development of muscle cells from the mesoderm have so far been investigated in flies and several species of animals with backbones. However, these species are complex, which makes it difficult to work out the general principles that control muscle cell development. Sea urchins are often studied in developmental biology as they have many of the same genes as more complex animals, but are much simpler and easier to study in the laboratory. Andrikou et al. therefore investigated the ‘gene regulatory network’ that controls muscle development in sea urchins. This revealed that proteins called Forkhead transcription factors and a process called FGF signaling are crucial for controlling muscle development in sea urchins. These are also important factors for developing muscles in other animals. Andrikou et al. then produced models that show the interactions between the genes that control muscle formation at three different stages of embryonic development. These models reveal several important features of the muscle development gene regulatory network. For example, the network is robust: if one gene fails, the network is connected in a way that allows it to still make muscle. This also allows the network to adapt and evolve without losing the ability to perform any of its existing roles. Comparing the gene regulatory network that controls muscle development in sea urchins with the networks found in other animals showed that many of the same genes are used across different species, but are connected into different network structures. Investigating the similarities and differences of the regulatory networks in different species could help us to understand how muscles have evolved and could ultimately lead to a better understanding of the causes of developmental diseases. DOI:http://dx.doi.org/10.7554/eLife.07343.002
Collapse
Affiliation(s)
- Carmen Andrikou
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Napoli, Italy
| | - Chih-Yu Pai
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Hsien Su
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Maria Ina Arnone
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Napoli, Italy
| |
Collapse
|
28
|
Mok DZL, Sternberg PW, Inoue T. Morphologically defined sub-stages of C. elegans vulval development in the fourth larval stage. BMC DEVELOPMENTAL BIOLOGY 2015; 15:26. [PMID: 26066484 PMCID: PMC4464634 DOI: 10.1186/s12861-015-0076-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/03/2015] [Indexed: 01/06/2023]
Abstract
Background During the fourth larval (L4) stage, vulval cells of C. elegans undergo extensive morphogenesis accompanied by changes in gene expression. This phase of vulval development, occurring after the well-studied induction of vulval cells, is not well understood but is potentially a useful context in which to study how a complex temporal sequence of events is regulated during development. However, a system for precisely describing different phases of vulval development in the L4 stage has been lacking. Results We defined ten sub-stages of L4 based on morphological criteria as observed using Nomarski microscopy (L4.0 ~ L4.9). Precise timing of each sub-stage at 20 °C was determined. We also re-examined the timing of expression for several gene expression markers, and correlated the sub-stages with the timing of other developmental events in the vulva and the uterus. Conclusions This scheme allows the developmental timing of an L4 individual to be determined at approximately one-hour resolution without the need to resort to time course experiments. These well-defined developmental stages will enable more precise description of gene expression and other developmental events.
Collapse
Affiliation(s)
- Darren Z L Mok
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore
| | - Paul W Sternberg
- HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Takao Inoue
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore. .,HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
29
|
Díaz-Balzac CA, Lázaro-Peña MI, Ramos-Ortiz GA, Bülow HE. The Adhesion Molecule KAL-1/anosmin-1 Regulates Neurite Branching through a SAX-7/L1CAM-EGL-15/FGFR Receptor Complex. Cell Rep 2015; 11:1377-84. [PMID: 26004184 PMCID: PMC4464948 DOI: 10.1016/j.celrep.2015.04.057] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/20/2015] [Accepted: 04/27/2015] [Indexed: 01/10/2023] Open
Abstract
Neurite branching is essential for correct assembly of neural circuits, yet it remains a poorly understood process. For example, the neural cell adhesion molecule KAL-1/anosmin-1, which is mutated in Kallmann syndrome, regulates neurite branching through mechanisms largely unknown. Here, we show that KAL-1/anosmin-1 mediates neurite branching as an autocrine co-factor with EGL-17/FGF through a receptor complex consisting of the conserved cell adhesion molecule SAX-7/L1CAM and the fibroblast growth factor receptor EGL-15/FGFR. This protein complex, which appears conserved in humans, requires the immunoglobulin (Ig) domains of SAX-7/L1CAM and the FN(III) domains of KAL-1/anosmin-1 for formation in vitro as well as function in vivo. The kinase domain of the EGL-15/FGFR is required for branching, and genetic evidence suggests that ras-mediated signaling downstream of EGL-15/FGFR is necessary to effect branching. Our studies establish a molecular pathway that regulates neurite branching during development of the nervous system.
Collapse
Affiliation(s)
- Carlos A Díaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - María I Lázaro-Peña
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gibram A Ramos-Ortiz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biology, University of Puerto Rico-Río Piedras, San Juan 00931, Puerto Rico
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
30
|
Sharanya D, Fillis CJ, Kim J, Zitnik EM, Ward KA, Gallagher ME, Chamberlin HM, Gupta BP. Mutations in Caenorhabditis briggsae identify new genes important for limiting the response to EGF signaling during vulval development. Evol Dev 2015; 17:34-48. [PMID: 25627712 DOI: 10.1111/ede.12105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Studies of vulval development in the nematode C. elegans have identified many genes that are involved in cell division and differentiation processes. Some of these encode components of conserved signal transduction pathways mediated by EGF, Notch, and Wnt. To understand how developmental mechanisms change during evolution, we are doing a comparative analysis of vulva formation in C. briggsae, a species that is closely related to C. elegans. Here, we report 14 mutations in 7 Multivulva (Muv) genes in C. briggsae that inhibit inappropriate division of vulval precursors. We have developed a new efficient and cost-effective gene mapping method to localize Muv mutations to small genetic intervals on chromosomes, thus facilitating cloning and functional studies. We demonstrate the utility of our method by determining molecular identities of three of the Muv genes that include orthologs of Cel-lin-1 (ETS) and Cel-lin-31 (Winged-Helix) of the EGF-Ras pathway and Cel-pry-1 (Axin), of the Wnt pathway. The remaining four genes reside in regions that lack orthologs of known C. elegans Muv genes. Inhibitor studies demonstrate that the Muv phenotype of all four new genes is dependent on the activity of the EGF pathway kinase, MEK. One of these, Cbr-lin(gu167), shows modest increase in the expression of Cbr-lin-3/EGF compared to wild type. These results argue that while Cbr-lin(gu167) may act upstream of Cbr-lin-3/EGF, the other three genes influence the EGF pathway downstream or in parallel to Cbr-lin-3. Overall, our findings demonstrate that the genetic program underlying a conserved developmental process includes both conserved and divergent functional contributions.
Collapse
Affiliation(s)
- Devika Sharanya
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Ramakrishnan K, Okkema PG. Regulation of C. elegans neuronal differentiation by the ZEB-family factor ZAG-1 and the NK-2 homeodomain factor CEH-28. PLoS One 2014; 9:e113893. [PMID: 25474681 PMCID: PMC4256384 DOI: 10.1371/journal.pone.0113893] [Citation(s) in RCA: 11] [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: 09/16/2014] [Accepted: 11/02/2014] [Indexed: 12/28/2022] Open
Abstract
The C. elegans pharyngeal neuron M4 is a multi-functional cell that acts as a cholinergic motor neuron to stimulate peristaltic pharyngeal muscle contraction and as a neuroendocrine cell secreting neuropeptides and growth factors to affect other cells both inside and outside the pharynx. The conserved transcription factors ZAG-1 and CEH-28 are co-expressed in M4 through most of development, and here we examine how these factors contribute to M4 differentiation. We find ZAG-1 functions upstream of CEH-28 in a branched pathway to activate expression of different sets of M4 differentiation markers. CEH-28 activates expression of the growth factor genes dbl-1 and egl-17, and the neuropeptide genes flp-5 and flp-2, while ZAG-1 activates expression of the serotonin receptor ser-7, as well as expression of ceh-28 and its downstream targets. Other markers of M4 differentiation are expressed normally in both zag-1 and ceh-28 mutants, including the neuropeptide gene flp-21 and the acetylcholine biosynthetic gene unc-17. Unlike ceh-28 mutants, zag-1 mutants completely lack peristaltic muscle contractions resulting from broader defects in M4 differentiation. Despite these defects, neither ZAG-1 nor CEH-28 are terminal selectors of the M4 phenotype, and we suggest they function in a hierarchy to regulate different aspects of M4 differentiation.
Collapse
Affiliation(s)
- Kalpana Ramakrishnan
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Peter G. Okkema
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
32
|
Steciuk M, Cheong M, Waite C, You YJ, Avery L. Regulation of synaptic transmission at the Caenorhabditis elegans M4 neuromuscular junction by an antagonistic relationship between two calcium channels. G3 (BETHESDA, MD.) 2014; 4:2535-43. [PMID: 25378475 PMCID: PMC4267947 DOI: 10.1534/g3.114.014308] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/02/2014] [Indexed: 11/28/2022]
Abstract
In wild-type Caenorhabditis elegans, the synapse from motor neuron M4 to pharyngeal terminal bulb (TB) muscles is silent, and the muscles are instead excited by gap junction connections from adjacent muscles. An eat-5 innexin mutant lacking this electrical connection has few TB contractions and is unable to grow well on certain foods. We showed previously that this defect can be overcome by activation of the M4 → TB synapse. To identify genes that negatively regulate synaptic transmission, we isolated new suppressors of eat-5. To our surprise, these suppressors included null mutations in NPQR-type calcium channel subunit genes unc-2 and unc-36. Our results are consistent with the hypothesis that Ca(2+) entry through the NPQR-type channel inhibits synaptic transmission by activating the calcium-activated K(+) channel SLO-1, thus antagonizing the EGL-19 L-type calcium channel.
Collapse
Affiliation(s)
- Mark Steciuk
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148
| | - Mi Cheong
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia 23298-0551
| | - Christopher Waite
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia 23298-0551
| | - Young-Jai You
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia 23298-0614
| | - Leon Avery
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia 23298-0551
| |
Collapse
|
33
|
Mentink RA, Middelkoop TC, Rella L, Ji N, Tang CY, Betist MC, van Oudenaarden A, Korswagen HC. Cell intrinsic modulation of Wnt signaling controls neuroblast migration in C. elegans. Dev Cell 2014; 31:188-201. [PMID: 25373777 DOI: 10.1016/j.devcel.2014.08.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 07/11/2014] [Accepted: 08/07/2014] [Indexed: 01/25/2023]
Abstract
Members of the Wnt family of secreted signaling proteins are key regulators of cell migration and axon guidance. In the nematode C. elegans, the migration of the QR neuroblast descendants requires multiple Wnt ligands and receptors. We found that the migration of the QR descendants is divided into three sequential phases that are each mediated by a distinct Wnt signaling mechanism. Importantly, the transition from the first to the second phase, which is the main determinant of the final position of the QR descendants along the anteroposterior body axis, is mediated through a cell-autonomous process in which the time-dependent expression of a Wnt receptor turns on the canonical Wnt/β-catenin signaling response that is required to terminate long-range anterior migration. Our results show that, in addition to direct guidance of cell migration by Wnt morphogenic gradients, cell migration can also be controlled indirectly through cell-intrinsic modulation of Wnt signaling responses.
Collapse
Affiliation(s)
- Remco A Mentink
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Teije C Middelkoop
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Lorenzo Rella
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Ni Ji
- Department of Physics and Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Chung Yin Tang
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Marco C Betist
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Alexander van Oudenaarden
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Department of Physics and Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Hendrik C Korswagen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands.
| |
Collapse
|
34
|
Kuwasako K, Takahashi M, Unzai S, Tsuda K, Yoshikawa S, He F, Kobayashi N, Güntert P, Shirouzu M, Ito T, Tanaka A, Yokoyama S, Hagiwara M, Kuroyanagi H, Muto Y. RBFOX and SUP-12 sandwich a G base to cooperatively regulate tissue-specific splicing. Nat Struct Mol Biol 2014; 21:778-86. [DOI: 10.1038/nsmb.2870] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/14/2014] [Indexed: 12/25/2022]
|
35
|
LaBonty M, Szmygiel C, Byrnes LE, Hughes S, Woollard A, Cram EJ. CACN-1/Cactin plays a role in Wnt signaling in C. elegans. PLoS One 2014; 9:e101945. [PMID: 24999833 PMCID: PMC4084952 DOI: 10.1371/journal.pone.0101945] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 06/13/2014] [Indexed: 11/19/2022] Open
Abstract
Wnt signaling is tightly regulated during animal development and controls cell proliferation and differentiation. In C. elegans, activation of Wnt signaling alters the activity of the TCF/LEF transcription factor, POP-1, through activation of the Wnt/β-catenin or Wnt/β-catenin asymmetry pathways. In this study, we have identified CACN-1 as a potential regulator of POP-1 in C. elegans larval development. CACN-1/Cactin is a well-conserved protein of unknown molecular function previously implicated in the regulation of several developmental signaling pathways. Here we have used activation of POPTOP, a POP-1-responsive reporter construct, as a proxy for Wnt signaling. POPTOP requires POP-1 and SYS-1/β-catenin for activation in L4 uterine cells. RNAi depletion experiments show that CACN-1 is needed to prevent excessive activation of POPTOP and for proper levels and/or localization of POP-1. Surprisingly, high POPTOP expression correlates with increased levels of POP-1 in uterine nuclei, suggesting POPTOP may not mirror endogenous gene expression in all respects. Genetic interaction studies suggest that CACN-1 may act partially through LIT-1/NLK to alter POP-1 localization and POPTOP activation. Additionally, CACN-1 is required for proper proliferation of larval seam cells. Depletion of CACN-1 results in a loss of POP-1 asymmetry and reduction of terminal seam cell number, suggesting an adoption of the anterior, differentiated fate by the posterior daughter cells. These findings suggest CACN-1/Cactin modulates Wnt signaling during larval development.
Collapse
Affiliation(s)
- Melissa LaBonty
- Department of Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Cleo Szmygiel
- Department of Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Lauren E. Byrnes
- Department of Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Samantha Hughes
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Alison Woollard
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Erin J. Cram
- Department of Biology, Northeastern University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
36
|
Identification of late larval stage developmental checkpoints in Caenorhabditis elegans regulated by insulin/IGF and steroid hormone signaling pathways. PLoS Genet 2014; 10:e1004426. [PMID: 24945623 PMCID: PMC4063711 DOI: 10.1371/journal.pgen.1004426] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/20/2014] [Indexed: 01/10/2023] Open
Abstract
Organisms in the wild develop with varying food availability. During periods of nutritional scarcity, development may slow or arrest until conditions improve. The ability to modulate developmental programs in response to poor nutritional conditions requires a means of sensing the changing nutritional environment and limiting tissue growth. The mechanisms by which organisms accomplish this adaptation are not well understood. We sought to study this question by examining the effects of nutrient deprivation on Caenorhabditis elegans development during the late larval stages, L3 and L4, a period of extensive tissue growth and morphogenesis. By removing animals from food at different times, we show here that specific checkpoints exist in the early L3 and early L4 stages that systemically arrest the development of diverse tissues and cellular processes. These checkpoints occur once in each larval stage after molting and prior to initiation of the subsequent molting cycle. DAF-2, the insulin/insulin-like growth factor receptor, regulates passage through the L3 and L4 checkpoints in response to nutrition. The FOXO transcription factor DAF-16, a major target of insulin-like signaling, functions cell-nonautonomously in the hypodermis (skin) to arrest developmental upon nutrient removal. The effects of DAF-16 on progression through the L3 and L4 stages are mediated by DAF-9, a cytochrome P450 ortholog involved in the production of C. elegans steroid hormones. Our results identify a novel mode of C. elegans growth in which development progresses from one checkpoint to the next. At each checkpoint, nutritional conditions determine whether animals remain arrested or continue development to the next checkpoint. Organisms in the wild often face long periods in which food is scarce. This may occur due to seasonal effects, loss of territory, or changes in predator-to-prey ratio. During periods of scarcity, organisms undergo adaptations to conserve resources and prolong survival. When nutrient deprivation occurs during development, physical growth and maturation to adulthood is delayed. These effects are also observed in malnourished individuals, who are smaller and reach puberty at later ages. Developmental arrest in response to nutrient scarcity requires a means of sensing changing nutrient conditions and coordinating an organism-wide response. How this occurs is not well understood. We assessed the developmental response to nutrient withdrawal in the nematode worm Caenorhabditis elegans. By removing food in the late larval stages, a period of extensive tissue formation, we have uncovered previously unknown checkpoints that occur at precise times in development. Diverse tissues and cellular processes arrest at the checkpoints. Insulin-like signaling and steroid hormone signaling regulate tissue arrest following nutrient withdrawal. These pathways are conserved in mammals and are linked to growth processes and diseases. Given that the pathways that respond to nutrition are conserved in animals, it is possible that similar checkpoints may also be important in human development.
Collapse
|
37
|
Ramakrishnan K, Ray P, Okkema PG. CEH-28 activates dbl-1 expression and TGF-β signaling in the C. elegans M4 neuron. Dev Biol 2014; 390:149-59. [PMID: 24690231 PMCID: PMC4023489 DOI: 10.1016/j.ydbio.2014.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/22/2014] [Accepted: 03/24/2014] [Indexed: 02/07/2023]
Abstract
M4 is a multifunctional neuron in the Caenorhabditis elegans pharynx that can both stimulate peristaltic contractions of the muscles in the pharyngeal isthmus and function systemically to regulate an enhanced sensory response under hypoxic conditions. Here we identify a third function for M4 that depends on activation of the TGF-β family gene dbl-1 by the homeodomain transcription factor CEH-28. dbl-1 is expressed in M4 and a subset of other neurons, and we show CEH-28 specifically activates dbl-1 expression in M4. Characterization of the dbl-1 promoter indicates that CEH-28 targets an M4-specific enhancer within the dbl-1 promoter region, while expression in other neurons is mediated by separate regulatory sequences. Unlike ceh-28 mutants, dbl-1 mutants do not exhibit M4 synaptic and signaling defects. Instead, both ceh-28 and dbl-1 mutants exhibit morphological defects in the g1 gland cells located adjacent to M4 in the pharynx, and these defects can be partially rescued by M4-specific expression of dbl-1 in these mutants. Identical gland cell defects are observed in sma-6 and daf-4 mutants defective in the receptor for DBL-1, but they are not observed in sma-2 and sma-3 mutants lacking the R-Smads functioning downstream of this receptor. Together these results identify a novel neuroendocrine function for M4 and provide evidence for an R-Smad-independent mechanism for DBL-1 signaling in C. elegans.
Collapse
Affiliation(s)
- Kalpana Ramakrishnan
- Department of Biological Sciences, University of Illinois at Chicago, 900 S. Ashland Avenue (MC567), Chicago, IL 60607, USA
| | - Paramita Ray
- Department of Biological Sciences, University of Illinois at Chicago, 900 S. Ashland Avenue (MC567), Chicago, IL 60607, USA
| | - Peter G Okkema
- Department of Biological Sciences, University of Illinois at Chicago, 900 S. Ashland Avenue (MC567), Chicago, IL 60607, USA.
| |
Collapse
|
38
|
An in vivo EGF receptor localization screen in C. elegans Identifies the Ezrin homolog ERM-1 as a temporal regulator of signaling. PLoS Genet 2014; 10:e1004341. [PMID: 24785082 PMCID: PMC4006739 DOI: 10.1371/journal.pgen.1004341] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/16/2014] [Indexed: 11/19/2022] Open
Abstract
The subcellular localization of the epidermal growth factor receptor (EGFR) in polarized epithelial cells profoundly affects the activity of the intracellular signaling pathways activated after EGF ligand binding. Therefore, changes in EGFR localization and signaling are implicated in various human diseases, including different types of cancer. We have performed the first in vivo EGFR localization screen in an animal model by observing the expression of the EGFR ortholog LET-23 in the vulval epithelium of live C. elegans larvae. After systematically testing all genes known to produce an aberrant vulval phenotype, we have identified 81 genes regulating various aspects of EGFR localization and expression. In particular, we have found that ERM-1, the sole C. elegans Ezrin/Radixin/Moesin homolog, regulates EGFR localization and signaling in the vulval cells. ERM-1 interacts with the EGFR at the basolateral plasma membrane in a complex distinct from the previously identified LIN-2/LIN-7/LIN-10 receptor localization complex. We propose that ERM-1 binds to and sequesters basolateral LET-23 EGFR in an actin-rich inactive membrane compartment to restrict receptor mobility and signaling. In this manner, ERM-1 prevents the immediate activation of the entire pool of LET-23 EGFR and permits the generation of a long-lasting inductive signal. The regulation of receptor localization thus serves to fine-tune the temporal activation of intracellular signaling pathways. Abnormal signaling by the epidermal growth factor receptor (EGFR) contributes to the development of various human diseases, including different cancer types. One important mechanism that controls intracellular signal transduction is by regulation of the subcellular receptor localization in the signal-receiving cell. We are investigating the regulation of the EGFR homolog LET-23 in the Nematode C. elegans by observing the localization of the EGFR in the epithelial cells of live animals. This approach has allowed us to study the dynamics of receptor trafficking in cells embedded in their natural environment and receiving physiological concentrations of various extracellular signals. In a systematic RNA interference screen, we have identified 81 genes controlling EGFR localization and signaling in different subcellular compartments. One new regulator of EGFR signaling identified in this screen encodes the Ezrin Homolog ERM-1. We show genetic and biochemical evidence indicating that ERM-1 is part of a buffering mechanism to maintain a pool of immobile EGFR in the basolateral membrane compartment of the epithelial cells. This mechanism permits the generation of a long-lasting EGFR signal during multiple rounds of cell divisions. The control of receptor localization is thus necessary for the precise temporal regulation of signal transduction during animal development.
Collapse
|
39
|
Minor PJ, He TF, Sohn CH, Asthagiri AR, Sternberg PW. FGF signaling regulates Wnt ligand expression to control vulval cell lineage polarity in C. elegans. Development 2013; 140:3882-91. [PMID: 23946444 DOI: 10.1242/dev.095687] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The interpretation of extracellular cues leading to the polarization of intracellular components and asymmetric cell divisions is a fundamental part of metazoan organogenesis. The Caenorhabditis elegans vulva, with its invariant cell lineage and interaction of multiple cell signaling pathways, provides an excellent model for the study of cell polarity within an organized epithelial tissue. Here, we show that the fibroblast growth factor (FGF) pathway acts in concert with the Frizzled homolog LIN-17 to influence the localization of SYS-1, a component of the Wnt/β-catenin asymmetry pathway, indirectly through the regulation of cwn-1. The source of the FGF ligand is the primary vulval precursor cell (VPC) P6.p, which controls the orientation of the neighboring secondary VPC P7.p by signaling through the sex myoblasts (SMs), activating the FGF pathway. The Wnt CWN-1 is expressed in the posterior body wall muscle of the worm as well as in the SMs, making it the only Wnt expressed on the posterior and anterior sides of P7.p at the time of the polarity decision. Both sources of cwn-1 act instructively to influence P7.p polarity in the direction of the highest Wnt signal. Using single molecule fluorescence in situ hybridization, we show that the FGF pathway regulates the expression of cwn-1 in the SMs. These results demonstrate an interaction between FGF and Wnt in C. elegans development and vulval cell lineage polarity, and highlight the promiscuous nature of Wnts and the importance of Wnt gradient directionality within C. elegans.
Collapse
Affiliation(s)
- Paul J Minor
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | | | | |
Collapse
|
40
|
Masoudi N, Fancsalszky L, Pourkarimi E, Vellai T, Alexa A, Reményi A, Gartner A, Mehta A, Takács-Vellai K. The NM23-H1/H2 homolog NDK-1 is required for full activation of Ras signaling in C. elegans. Development 2013; 140:3486-95. [PMID: 23900546 PMCID: PMC3737725 DOI: 10.1242/dev.094011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2013] [Indexed: 11/21/2022]
Abstract
The group I members of the Nm23 (non-metastatic) gene family encode nucleoside diphosphate kinases (NDPKs) that have been implicated in the regulation of cell migration, proliferation and differentiation. Despite their developmental and medical significance, the molecular functions of these NDPKs remain ill defined. To minimize confounding effects of functional compensation between closely related Nm23 family members, we studied ndk-1, the sole Caenorhabditis elegans ortholog of group I NDPKs, and focused on its role in Ras/mitogen-activated protein kinase (MAPK)-mediated signaling events during development. ndk-1 inactivation leads to a protruding vulva phenotype and affects vulval cell fate specification through the Ras/MAPK cascade. ndk-1 mutant worms show severe reduction of activated, diphosphorylated MAPK in somatic tissues, indicative of compromised Ras/MAPK signaling. A genetic epistasis analysis using the vulval induction system revealed that NDK-1 acts downstream of LIN-45/Raf, but upstream of MPK-1/MAPK, at the level of the kinase suppressors of ras (KSR-1/2). KSR proteins act as scaffolds facilitating Ras signaling events by tethering signaling components, and we suggest that NDK-1 modulates KSR activity through direct physical interaction. Our study reveals that C. elegans NDK-1/Nm23 influences differentiation by enhancing the level of Ras/MAPK signaling. These results might help to better understand how dysregulated Nm23 in humans contributes to tumorigenesis.
Collapse
Affiliation(s)
- Neda Masoudi
- Department of Genetics, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary
- Wellcome Trust Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Luca Fancsalszky
- Department of Genetics, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary
| | - Ehsan Pourkarimi
- Department of Genetics, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary
- Wellcome Trust Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary
| | - Anita Alexa
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary
- Institute of Molecular Pharmacology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Pusztaszeri út 59-67, H-1025 Budapest, Hungary
| | - Attila Reményi
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary
- Institute of Molecular Pharmacology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Pusztaszeri út 59-67, H-1025 Budapest, Hungary
| | - Anton Gartner
- Wellcome Trust Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Anil Mehta
- Division of Medical Sciences, Centre for CVS and Lung Biology, Ninewells Hospital Medical School, Dundee DD1 9SY, UK
| | - Krisztina Takács-Vellai
- Department of Genetics, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117 Budapest, Hungary
| |
Collapse
|
41
|
Tamayo JV, Gujar M, Macdonald SJ, Lundquist EA. Functional transcriptomic analysis of the role of MAB-5/Hox in Q neuroblast migration in Caenorhabditis elegans. BMC Genomics 2013; 14:304. [PMID: 23642123 PMCID: PMC3651406 DOI: 10.1186/1471-2164-14-304] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 05/01/2013] [Indexed: 01/11/2023] Open
Abstract
Background Directed cell migration is a fundamental process in normal development and in tumor metastasis. In C. elegans the MAB-5/Hox transcription factor is a determinant of posterior migration of the Q neuroblast descendants. In this work, mab-5 transcriptional targets that control Q descendant migration are identified by comparing RNA-seq profiles in wild type and mab-5 mutant backgrounds. Results Transcriptome profiling is a widely-used and potent tool to identify genes involved in developmental and pathological processes, and is most informative when RNA can be isolated from individual cell or tissue types. Cell-specific RNA samples can be difficult to obtain from invertebrate model organisms such as Drosophila and C. elegans. Here we test the utility of combining a whole organism RNA-seq approach with mab-5 loss and gain-of-function mutants and functional validation using RNAi to identify genes regulated by MAB-5 to control Q descendant migration. We identified 22 genes whose expression was controlled by mab-5 and that controlled Q descendant migration. Genes regulated by mab-5 were enriched for secreted and transmembrane molecules involved in basement membrane interaction and modification, and some affected Q descendant migration. Conclusions Our results indicate that a whole-organism RNA-seq approach, when combined with mutant analysis and functional validation, can be a powerful method to identify genes involved in a specific developmental process, in this case Q descendant posterior migration. These genes could act either autonomously in the Q cells, or non-autonomously in other cells that express MAB-5. The identities of the genes regulated by MAB-5 indicate that MAB-5 acts by modifying interactions with the basement membrane, resulting in posterior versus anterior migration.
Collapse
Affiliation(s)
- Joel V Tamayo
- Department of Molecular Biosciences, Programs in Genetics and Molecular, Cellular, and Developmental Biology, The University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | | | | | | |
Collapse
|
42
|
Green SA, Norris RP, Terasaki M, Lowe CJ. FGF signaling induces mesoderm in the hemichordate Saccoglossus kowalevskii. Development 2013; 140:1024-33. [PMID: 23344709 DOI: 10.1242/dev.083790] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
FGFs act in vertebrate mesoderm induction and also play key roles in early mesoderm formation in ascidians and amphioxus. However, in sea urchins initial characterizations of FGF function do not support a role in early mesoderm induction, making the ancestral roles of FGF signaling and mechanisms of mesoderm specification in deuterostomes unclear. In order to better characterize the evolution of mesoderm formation, we have examined the role of FGF signaling during mesoderm development in Saccoglossus kowalevskii, an experimentally tractable representative of hemichordates. We report the expression of an FGF ligand, fgf8/17/18, in ectoderm overlying sites of mesoderm specification within the archenteron endomesoderm. Embryological experiments demonstrate that mesoderm induction in the archenteron requires contact with ectoderm, and loss-of-function experiments indicate that both FGF ligand and receptor are necessary for mesoderm specification. fgf8/17/18 gain-of-function experiments establish that FGF8/17/18 is sufficient to induce mesoderm in adjacent endomesoderm. These experiments suggest that FGF signaling is necessary from the earliest stages of mesoderm specification and is required for all mesoderm development. Furthermore, they suggest that the archenteron is competent to form mesoderm or endoderm, and that FGF signaling from the ectoderm defines the location and amount of mesoderm. When considered in a comparative context, these data support a phylogenetically broad requirement for FGF8/17/18 signaling in mesoderm specification and suggest that FGF signaling played an ancestral role in deuterostome mesoderm formation.
Collapse
Affiliation(s)
- Stephen A Green
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
43
|
Modzelewska K, Lauritzen A, Hasenoeder S, Brown L, Georgiou J, Moghal N. Neurons refine the Caenorhabditis elegans body plan by directing axial patterning by Wnts. PLoS Biol 2013; 11:e1001465. [PMID: 23319891 PMCID: PMC3539944 DOI: 10.1371/journal.pbio.1001465] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 11/16/2012] [Indexed: 12/14/2022] Open
Abstract
Metazoans display remarkable conservation of gene families, including growth factors, yet somehow these genes are used in different ways to generate tremendous morphological diversity. While variations in the magnitude and spatio-temporal aspects of signaling by a growth factor can generate different body patterns, how these signaling variations are organized and coordinated during development is unclear. Basic body plans are organized by the end of gastrulation and are refined as limbs, organs, and nervous systems co-develop. Despite their proximity to developing tissues, neurons are primarily thought to act after development, on behavior. Here, we show that in Caenorhabditis elegans, the axonal projections of neurons regulate tissue progenitor responses to Wnts so that certain organs develop with the correct morphology at the right axial positions. We find that foreshortening of the posteriorly directed axons of the two canal-associated neurons (CANs) disrupts mid-body vulval morphology, and produces ectopic vulval tissue in the posterior epidermis, in a Wnt-dependent manner. We also provide evidence that suggests that the posterior CAN axons modulate the location and strength of Wnt signaling along the anterior-posterior axis by employing a Ror family Wnt receptor to bind posteriorly derived Wnts, and hence, refine their distributions. Surprisingly, despite high levels of Ror expression in many other cells, these cells cannot substitute for the CAN axons in patterning the epidermis, nor can cells expressing a secreted Wnt inhibitor, SFRP-1. Thus, unmyelinated axon tracts are critical for patterning the C. elegans body. Our findings suggest that the evolution of neurons not only improved metazoans by increasing behavioral complexity, but also by expanding the diversity of developmental patterns generated by growth factors such as Wnts.
Collapse
Affiliation(s)
- Katarzyna Modzelewska
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Amara Lauritzen
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Stefan Hasenoeder
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Louise Brown
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - John Georgiou
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Nadeem Moghal
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
44
|
Goh KY, Ng NW, Hagen T, Inoue T. p21-activated kinase interacts with Wnt signaling to regulate tissue polarity and gene expression. Proc Natl Acad Sci U S A 2012; 109:15853-8. [PMID: 23019370 PMCID: PMC3465426 DOI: 10.1073/pnas.1120795109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Wnt signaling is mediated by three classes of receptors, Frizzled, Ryk, and Ror. In Caenorhabditis elegans, Wnt signaling regulates the anterior/posterior polarity of the P7.p vulval lineage, and mutations in lin-17/Frizzled cause loss or reversal of P7.p lineage polarity. We found that pak-1/Pak (p21-activated kinase), along with putative activators of Pak, nck-1/Nck, and ced-10/Rac, regulates P7.p polarity. Mutations in these genes suppress the polarity defect of lin-17 mutants. Furthermore, mutations in pak-1, nck-1, and ced-10 cause constitutive dauer formation at 27 °C, a phenotype also observed in egl-20/Wnt and cam-1/Ror mutants. In HEK293T cells, Pak1 can antagonize canonical Wnt signaling. Moreover, overexpression of Ror2 leads to phosphorylation of Pak1. Together, these results indicate that Pak interacts with Wnt signaling to regulate tissue polarity and gene expression.
Collapse
Affiliation(s)
- Kah Yee Goh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Natalie Weili Ng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Thilo Hagen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Takao Inoue
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| |
Collapse
|
45
|
Skorobogata O, Rocheleau CE. RAB-7 antagonizes LET-23 EGFR signaling during vulva development in Caenorhabditis elegans. PLoS One 2012; 7:e36489. [PMID: 22558469 PMCID: PMC3340361 DOI: 10.1371/journal.pone.0036489] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 04/07/2012] [Indexed: 12/20/2022] Open
Abstract
The Rab7 GTPase regulates late endosome trafficking of the Epidermal Growth Factor Receptor (EGFR) to the lysosome for degradation. However, less is known about how Rab7 activity, functioning late in the endocytic pathway, affects EGFR signaling. Here we used Caenorhabditis elegans vulva cell fate induction, a paradigm for genetic analysis of EGFR/Receptor Tyrosine Kinase (RTK) signaling, to assess the genetic requirements for rab-7. Using a rab-7 deletion mutant, we demonstrate that rab-7 antagonizes LET-23 EGFR signaling to a similar extent, but in a distinct manner, as previously described negative regulators such as sli-1 c-Cbl. Epistasis analysis places rab-7 upstream of or in parallel to lin-3 EGF and let-23 EGFR. However, expression of gfp::rab-7 in the Vulva Presursor Cells (VPCs) is sufficient to rescue the rab-7(-) VPC induction phenotypes indicating that RAB-7 functions in the signal receiving cell. We show that components of the Endosomal Sorting Complex Required for Transport (ESCRT)-0, and -I, complexes, hgrs-1 Hrs, and vps-28, also antagonize signaling, suggesting that LET-23 EGFR likely transits through Multivesicular Bodies (MVBs) en route to the lysosome. Consistent with RAB-7 regulating LET-23 EGFR trafficking, rab-7 mutants have increased number of LET-23::GFP-positive endosomes. Our data imply that Rab7, by mediating EGFR trafficking and degradation, plays an important role in downregulation of EGFR signaling. Failure to downregulate EGFR signaling contributes to oncogenesis, and thus Rab7 could possess tumor suppressor activity in humans.
Collapse
Affiliation(s)
- Olga Skorobogata
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University and McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | - Christian E. Rocheleau
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University and McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| |
Collapse
|
46
|
Nusser-Stein S, Beyer A, Rimann I, Adamczyk M, Piterman N, Hajnal A, Fisher J. Cell-cycle regulation of NOTCH signaling during C. elegans vulval development. Mol Syst Biol 2012; 8:618. [PMID: 23047528 PMCID: PMC3501274 DOI: 10.1038/msb.2012.51] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 09/04/2012] [Indexed: 01/05/2023] Open
Abstract
C. elegans vulval development is one of the best-characterized systems to study cell fate specification during organogenesis. The detailed knowledge of the signaling pathways determining vulval precursor cell (VPC) fates permitted us to create a computational model based on the antagonistic interactions between the epidermal growth factor receptor (EGFR)/RAS/MAPK and the NOTCH pathways that specify the primary and secondary fates, respectively. A key notion of our model is called bounded asynchrony, which predicts that a limited degree of asynchrony in the progression of the VPCs is necessary to break their equivalence. While searching for a molecular mechanism underlying bounded asynchrony, we discovered that the termination of NOTCH signaling is tightly linked to cell-cycle progression. When single VPCs were arrested in the G1 phase, intracellular NOTCH failed to be degraded, resulting in a mixed primary/secondary cell fate. Moreover, the G1 cyclins CYD-1 and CYE-1 stabilize NOTCH, while the G2 cyclin CYB-3 promotes NOTCH degradation. Our findings reveal a synchronization mechanism that coordinates NOTCH signaling with cell-cycle progression and thus permits the formation of a stable cell fate pattern.
Collapse
Affiliation(s)
- Stefanie Nusser-Stein
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Sciences PhD program, Uni ETH Zürich, Switzerland
| | - Antje Beyer
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Ivo Rimann
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Magdalene Adamczyk
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Sciences PhD program, Uni ETH Zürich, Switzerland
| | - Nir Piterman
- Department of Computer Science, University of Leicester, Leicester, UK
| | - Alex Hajnal
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
47
|
Pellegrino MW, Farooqui S, Fröhli E, Rehrauer H, Kaeser-Pebernard S, Müller F, Gasser RB, Hajnal A. LIN-39 and the EGFR/RAS/MAPK pathway regulate C. elegans vulval morphogenesis via the VAB-23 zinc finger protein. Development 2011; 138:4649-60. [PMID: 21989912 DOI: 10.1242/dev.071951] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Morphogenesis represents a phase of development during which cell fates are executed. The conserved hox genes are key cell fate determinants during metazoan development, but their role in controlling organ morphogenesis is less understood. Here, we show that the C. elegans hox gene lin-39 regulates epidermal morphogenesis via its novel target, the essential zinc finger protein VAB-23. During the development of the vulva, the egg-laying organ of the hermaphrodite, the EGFR/RAS/MAPK signaling pathway activates, together with LIN-39 HOX, the expression of VAB-23 in the primary cell lineage to control the formation of the seven vulval toroids. VAB-23 regulates the formation of homotypic contacts between contralateral pairs of cells with the same sub-fates at the vulval midline by inducing smp-1 (semaphorin) transcription. In addition, VAB-23 prevents ectopic vulval cell fusions by negatively regulating expression of the fusogen eff-1. Thus, LIN-39 and the EGFR/RAS/MAPK signaling pathway, which specify cell fates earlier during vulval induction, continue to act during the subsequent phase of cell fate execution by regulating various aspects of epidermal morphogenesis. Vulval cell fate specification and execution are, therefore, tightly coupled processes.
Collapse
Affiliation(s)
- Mark W Pellegrino
- The University of Melbourne, Department of Veterinary Science, Werribee, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ulm EA, Sleiman SF, Chamberlin HM. Developmental functions for the Caenorhabditis elegans Sp protein SPTF-3. Mech Dev 2011; 128:428-41. [PMID: 21884786 DOI: 10.1016/j.mod.2011.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/12/2011] [Accepted: 08/14/2011] [Indexed: 10/17/2022]
Abstract
Sp factors are important for animal development and the transcriptional regulation of a wide variety of genes. How they influence the developmental decisions of individual cells within the organism, however, is poorly understood. To better understand the developmental functions for Sp transcription factors, we have characterized the functions of Caenorhabditis elegans SPTF-3 using RNAi knockdown and a non-null, hypomorphic mutant allele. We find that disruption of sptf-3 confers a variety of developmental defects, including defects in development of the egg-laying system, oocyte production, and embryonic morphogenesis. sptf-3 mutants exhibit defects in vulval lineage polarity, a phenotype previously only observed in mutants defective in Wnt signaling. We show that the embryonic function of sptf-3 is dependent on germline activity, arguing that the gene has an important maternal contribution to embryonic development. An evaluation of reporter gene expression suggests that SPTF-3 exhibits specificity, in that it can influence the expression of a given gene in some cells but not others, and that SPTF-3 participates in the maintenance of gene expression states in differentiated cells. We propose SPTF-3 provides a good model to study the in vivo functions for Sp transcription factors during animal development.
Collapse
Affiliation(s)
- Elizabeth A Ulm
- Department of Molecular Genetics, Ohio State University, 484 W.12th Ave., Columbus, OH 43210, USA.
| | | | | |
Collapse
|
49
|
Functional and spatial analysis of C. elegans SYG-1 and SYG-2, orthologs of the Neph/nephrin cell adhesion module directing selective synaptogenesis. PLoS One 2011; 6:e23598. [PMID: 21858180 PMCID: PMC3156230 DOI: 10.1371/journal.pone.0023598] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 07/20/2011] [Indexed: 01/25/2023] Open
Abstract
The assembly of specific synaptic connections represents a prime example of cellular recognition. Members of the Ig superfamily are among the most ancient proteins represented in the genomes of both mammalian and invertebrate organisms, where they constitute a trans-synaptic adhesion system. The correct connectivity patterns of the highly conserved immunoglobulin superfamily proteins nephrin and Neph1 are crucial for the assembly of functional neuronal circuits and the formation of the kidney slit diaphragm, a synapse-like structure forming the filtration barrier. Here, we utilize the nematode C. elegans model for studying the requirements of synaptic specificity mediated by nephrin-Neph proteins. In C. elegans, the nephrin/Neph1 orthologs SYG-2 and SYG-1 form intercellular contacts strictly in trans between epithelial guidepost cells and neurons specifying the localization of synapses. We demonstrate a functional conservation between mammalian nephrin and SYG-2. Expression of nephrin effectively compensated loss of syg-2 function in C. elegans and restored defective synaptic connectivity further establishing the C. elegans system as a valuable model for slit diaphragm proteins. Next, we investigated the effect of SYG-1 and SYG-2 trans homodimerization respectively. Strikingly, synapse assembly could be induced by homophilic SYG-1 but not SYG-2 binding indicating a critical role of SYG-1 intracellular signalling for morphogenetic events and pointing toward the dynamic and stochastic nature of extra- and intracellular nephrin-Neph interactions to generate reproducible patterns of synaptic connectivity.
Collapse
|
50
|
Mondal S, Ahlawat S, Rau K, Venkataraman V, Koushika SP. Imaging in vivo neuronal transport in genetic model organisms using microfluidic devices. Traffic 2011; 12:372-85. [PMID: 21199219 DOI: 10.1111/j.1600-0854.2010.01157.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Microfluidic devices have been developed for imaging behavior and various cellular processes in Caenorhabditis elegans, but not subcellular processes requiring high spatial resolution. In neurons, essential processes such as axonal, dendritic, intraflagellar and other long-distance transport can be studied by acquiring fast time-lapse images of green fluorescent protein (GFP)-tagged moving cargo. We have achieved two important goals in such in vivo studies namely, imaging several transport processes in unanesthetized intact animals and imaging very early developmental stages. We describe a microfluidic device for immobilizing C. elegans and Drosophila larvae that allows imaging without anesthetics or dissection. We observed that for certain neuronal cargoes in C. elegans, anesthetics have significant and sometimes unexpected effects on the flux. Further, imaging the transport of certain cargo in early developmental stages was possible only in the microfluidic device. Using our device we observed an increase in anterograde synaptic vesicle transport during development corresponding with synaptic growth. We also imaged Q neuroblast divisions and mitochondrial transport during early developmental stages of C. elegans and Drosophila, respectively. Our simple microfluidic device offers a useful means to image high-resolution subcellular processes in C. elegans and Drosophila and can be readily adapted to other transparent or translucent organisms.
Collapse
Affiliation(s)
- Sudip Mondal
- Neurobiology, NCBS-TIFR, Bellary Road, Bangalore 560065, India
| | | | | | | | | |
Collapse
|