1
|
Williamson MH, Clements WK. WNT16 primer. Differentiation 2025; 142:100833. [PMID: 39730242 PMCID: PMC12045490 DOI: 10.1016/j.diff.2024.100833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Affiliation(s)
- McLean H Williamson
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Wilson K Clements
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
2
|
Ren H, Ou Q, Pu Q, Lou Y, Yang X, Han Y, Liu S. Comprehensive Review on Bimolecular Fluorescence Complementation and Its Application in Deciphering Protein-Protein Interactions in Cell Signaling Pathways. Biomolecules 2024; 14:859. [PMID: 39062573 PMCID: PMC11274695 DOI: 10.3390/biom14070859] [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: 06/24/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Signaling pathways are responsible for transmitting information between cells and regulating cell growth, differentiation, and death. Proteins in cells form complexes by interacting with each other through specific structural domains, playing a crucial role in various biological functions and cell signaling pathways. Protein-protein interactions (PPIs) within cell signaling pathways are essential for signal transmission and regulation. The spatiotemporal features of PPIs in signaling pathways are crucial for comprehending the regulatory mechanisms of signal transduction. Bimolecular fluorescence complementation (BiFC) is one kind of imaging tool for the direct visualization of PPIs in living cells and has been widely utilized to uncover novel PPIs in various organisms. BiFC demonstrates significant potential for application in various areas of biological research, drug development, disease diagnosis and treatment, and other related fields. This review systematically summarizes and analyzes the technical advancement of BiFC and its utilization in elucidating PPIs within established cell signaling pathways, including TOR, PI3K/Akt, Wnt/β-catenin, NF-κB, and MAPK. Additionally, it explores the application of this technology in revealing PPIs within the plant hormone signaling pathways of ethylene, auxin, Gibberellin, and abscisic acid. Using BiFC in conjunction with CRISPR-Cas9, live-cell imaging, and ultra-high-resolution microscopy will enhance our comprehension of PPIs in cell signaling pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shiping Liu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (H.R.); (Q.O.); (Q.P.); (Y.L.); (X.Y.); (Y.H.)
| |
Collapse
|
3
|
Liu J, Murray JI. Mechanisms of lineage specification in Caenorhabditis elegans. Genetics 2023; 225:iyad174. [PMID: 37847877 PMCID: PMC11491538 DOI: 10.1093/genetics/iyad174] [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: 08/26/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
The studies of cell fate and lineage specification are fundamental to our understanding of the development of multicellular organisms. Caenorhabditis elegans has been one of the premiere systems for studying cell fate specification mechanisms at single cell resolution, due to its transparent nature, the invariant cell lineage, and fixed number of somatic cells. We discuss the general themes and regulatory mechanisms that have emerged from these studies, with a focus on somatic lineages and cell fates. We next review the key factors and pathways that regulate the specification of discrete cells and lineages during embryogenesis and postembryonic development; we focus on transcription factors and include numerous lineage diagrams that depict the expression of key factors that specify embryonic founder cells and postembryonic blast cells, and the diverse somatic cell fates they generate. We end by discussing some future perspectives in cell and lineage specification.
Collapse
Affiliation(s)
- Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - John Isaac Murray
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Konopelski Snavely SE, Srinivasan S, Dreyer CA, Tan J, Carraway KL, Ho HYH. Non-canonical WNT5A-ROR signaling: New perspectives on an ancient developmental pathway. Curr Top Dev Biol 2023; 153:195-227. [PMID: 36967195 PMCID: PMC11042798 DOI: 10.1016/bs.ctdb.2023.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Deciphering non-canonical WNT signaling has proven to be both fascinating and challenging. Discovered almost 30 years ago, non-canonical WNT ligands signal independently of the transcriptional co-activator β-catenin to regulate a wide range of morphogenetic processes during development. The molecular and cellular mechanisms that underlie non-canonical WNT function, however, remain nebulous. Recent results from various model systems have converged to define a core non-canonical WNT pathway consisting of the prototypic non-canonical WNT ligand, WNT5A, the receptor tyrosine kinase ROR, the seven transmembrane receptor Frizzled and the cytoplasmic scaffold protein Dishevelled. Importantly, mutations in each of these signaling components cause Robinow syndrome, a congenital disorder characterized by profound tissue morphogenetic abnormalities. Moreover, dysregulation of the pathway has also been linked to cancer metastasis. As new knowledge concerning the WNT5A-ROR pathway continues to grow, modeling these mutations will likely provide crucial insights into both the physiological regulation of the pathway and the etiology of WNT5A-ROR-driven diseases.
Collapse
Affiliation(s)
- Sara E Konopelski Snavely
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States
| | - Srisathya Srinivasan
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States
| | - Courtney A Dreyer
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, School of Medicine, Sacramento, CA, United States
| | - Jia Tan
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, School of Medicine, Sacramento, CA, United States
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States.
| |
Collapse
|
5
|
Zhang Q, Hrach H, Mangone M, Reiner DJ. Identifying the Caenorhabditis elegans vulval transcriptome. G3 (BETHESDA, MD.) 2022; 12:jkac091. [PMID: 35551383 PMCID: PMC9157107 DOI: 10.1093/g3journal/jkac091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022]
Abstract
Development of the Caenorhabditis elegans vulva is a classic model of organogenesis. This system, which starts with 6 equipotent cells, encompasses diverse types of developmental event, including developmental competence, multiple signaling events to control precise and faithful patterning of three cell fates, execution and proliferation of specific cell lineages, and a series of sophisticated morphogenetic events. Early events have been subjected to extensive mutational and genetic investigations and later events to cell biological analyses. We infer the existence of dramatically changing profiles of gene expression that accompanies the observed changes in development. Yet, except from serendipitous discovery of several transcription factors expressed in dynamic patterns in vulval lineages, our knowledge of the transcriptomic landscape during vulval development is minimal. This study describes the composition of a vulva-specific transcriptome. We used tissue-specific harvesting of mRNAs via immunoprecipitation of epitope-tagged poly(A) binding protein, PAB-1, heterologously expressed by a promoter known to express GFP in vulval cells throughout their development. The identified transcriptome was small but tightly interconnected. From this data set, we identified several genes with identified functions in development of the vulva and validated more with promoter-GFP reporters of expression. For one target, lag-1, promoter-GFP expression was limited but a fluorescent tag of the endogenous protein revealed extensive expression. Thus, we have identified a transcriptome of C. elegans vulval lineages as a launching pad for exploration of functions of these genes in organogenesis.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| | - Heather Hrach
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85281, USA
- Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, Tempe, AZ 85281, USA
| | - Marco Mangone
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85281, USA
- Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, Tempe, AZ 85281, USA
| | - David J Reiner
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| |
Collapse
|
6
|
Ka C, Gautam S, Marshall SR, Tice LP, Martinez-Bartolome M, Fenner JL, Range RC. Receptor Tyrosine Kinases ror1/2 and ryk Are Co-expressed with Multiple Wnt Signaling Components During Early Development of Sea Urchin Embryos. THE BIOLOGICAL BULLETIN 2021; 241:140-157. [PMID: 34706206 PMCID: PMC11257382 DOI: 10.1086/715237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
AbstractA combination of receptors, co-receptors, and secreted Wnt modulators form protein complexes at the cell surface that activate one or more of the three different Wnt signaling pathways (Wnt/β-catenin, Wnt/JNK, and Wnt/Ca2+). Two or more of these pathways are often active in the same cellular territories, forming Wnt signaling networks; however, the molecular mechanisms necessary to integrate information from these pathways in these situations are unclear in any in vivo model system. Recent studies have implicated two Wnt binding receptor tyrosine kinases, receptor tyrosine kinase-like orphan receptor (Ror) and related-to-receptor tyrosine kinase (Ryk), in the regulation of canonical and non-canonical Wnt signaling pathways, depending on the context; however, the spatiotemporal expression of these genes in relation to Wnt signaling components has not been well characterized in most deuterostome model systems. Here we use a combination of phylogenetic and spatiotemporal gene expression analyses to characterize Ror and Ryk orthologs in sea urchin embryos. Our phylogenetic analysis indicates that both ror1/2 and ryk originated as single genes from the metazoan ancestor. Expression analyses indicate that ror1/2 and ryk are expressed in the same domains of many Wnt ligands and Frizzled receptors essential for the specification and patterning of germ layers along the early anterior-posterior axis. In addition, both genes are co-expressed with Wnt signaling components in the gut, ventral ectoderm, and anterior neuroectoderm territories later in development. Together, our results indicate that Ror and Ryk have a complex evolutionary history and that their spatiotemporal expression suggests that they could contribute to the complexity of Wnt signaling in early sea urchin embryogenesis.
Collapse
Affiliation(s)
- C Ka
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - S Gautam
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - SR Marshall
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762
| | - LP Tice
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762
| | | | - JL Fenner
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - RC Range
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| |
Collapse
|
7
|
Shin H, Braendle C, Monahan KB, Kaplan REW, Zand TP, Mote FS, Peters EC, Reiner DJ. Developmental fidelity is imposed by genetically separable RalGEF activities that mediate opposing signals. PLoS Genet 2019; 15:e1008056. [PMID: 31086367 PMCID: PMC6534338 DOI: 10.1371/journal.pgen.1008056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/24/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
The six C. elegans vulval precursor cells (VPCs) are induced to form the 3°-3°-2°-1°-2°-3° pattern of cell fates with high fidelity. In response to EGF signal, the LET-60/Ras-LIN-45/Raf-MEK-2/MEK-MPK-1/ERK canonical MAP kinase cascade is necessary to induce 1° fate and synthesis of DSL ligands for the lateral Notch signal. In turn, LIN-12/Notch receptor is necessary to induce neighboring cells to become 2°. We previously showed that, in response to graded EGF signal, the modulatory LET-60/Ras-RGL-1/RalGEF-RAL-1/Ral signal promotes 2° fate in support of LIN-12. In this study, we identify two key differences between RGL-1 and RAL-1. First, deletion of RGL-1 confers no overt developmental defects, while previous studies showed RAL-1 to be essential for viability and fertility. From this observation, we hypothesize that the essential functions of RAL-1 are independent of upstream activation. Second, RGL-1 plays opposing and genetically separable roles in VPC fate patterning. RGL-1 promotes 2° fate via canonical GEF-dependent activation of RAL-1. Conversely, RGL-1 promotes 1° fate via a non-canonical GEF-independent activity. Our genetic epistasis experiments are consistent with RGL-1 functioning in the modulatory 1°-promoting AGE-1/PI3-Kinase-PDK-1-AKT-1 cascade. Additionally, animals lacking RGL-1 experience 15-fold higher rates of VPC patterning errors compared to the wild type. Yet VPC patterning in RGL-1 deletion mutants is not more sensitive to environmental perturbations. We propose that RGL-1 functions to orchestrate opposing 1°- and 2°-promoting modulatory cascades to decrease developmental stochasticity. We speculate that such switches are broadly conserved but mostly masked by paralog redundancy or essential functions. Developmental signals are increasingly conceptualized in the context of networks rather than linear pathways. Patterning of C. elegans vulval fates is mostly governed by two major signaling cascades that operate antagonistically to induce two cell identities. An additional pair of minor cascades support each of the major cascades. All components in this system are conserved in mammalian oncogenic signaling networks. We find that RGL-1, a component of one of the minor cascades, performs two antagonistic functions. Its deletion appears to abolish both opposing modulatory signals, resulting in a 15-fold increase in the basal error rate in development of these cells. We hypothesize that the bifunctional RGL-1 protein defines a novel mechanism by which signaling networks are interwoven to mitigate developmental errors.
Collapse
Affiliation(s)
- Hanna Shin
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX, United States of America
| | | | - Kimberly B Monahan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America
| | - Rebecca E W Kaplan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America
| | - Tanya P Zand
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America.,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Francisca Sefakor Mote
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX, United States of America
| | - Eldon C Peters
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America
| | - David J Reiner
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX, United States of America.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America.,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, United States of America
| |
Collapse
|
8
|
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: 2.1] [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
|
9
|
Pani AM, Goldstein B. Direct visualization of a native Wnt in vivo reveals that a long-range Wnt gradient forms by extracellular dispersal. eLife 2018; 7:38325. [PMID: 30106379 PMCID: PMC6143344 DOI: 10.7554/elife.38325] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/13/2018] [Indexed: 12/24/2022] Open
Abstract
Wnts are evolutionarily conserved signaling proteins with essential roles in development and disease that have often been thought to spread between cells and signal at a distance. However, recent studies have challenged this model, and whether long-distance extracellular Wnt dispersal occurs and is biologically relevant is debated. Understanding fundamental aspects of Wnt dispersal has been limited by challenges with observing endogenous ligands in vivo, which has prevented directly testing hypotheses. Here, we have generated functional, fluorescently tagged alleles for a C. elegans Wnt homolog and for the first time visualized a native, long-range Wnt gradient in a living animal. Live imaging of Wnt along with source and responding cell membranes provided support for free, extracellular dispersal. By limiting Wnt transfer between cells, we confirmed that extracellular spreading shapes a long-range gradient and is critical for neuroblast migration. These results provide direct evidence that Wnts spread extracellularly to regulate aspects of long-range signaling.
Collapse
Affiliation(s)
- Ariel M Pani
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, United States
| |
Collapse
|
10
|
Mattes B, Dang Y, Greicius G, Kaufmann LT, Prunsche B, Rosenbauer J, Stegmaier J, Mikut R, Özbek S, Nienhaus GU, Schug A, Virshup DM, Scholpp S. Wnt/PCP controls spreading of Wnt/β-catenin signals by cytonemes in vertebrates. eLife 2018; 7:36953. [PMID: 30060804 PMCID: PMC6086664 DOI: 10.7554/elife.36953] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022] Open
Abstract
Signaling filopodia, termed cytonemes, are dynamic actin-based membrane structures that regulate the exchange of signaling molecules and their receptors within tissues. However, how cytoneme formation is regulated remains unclear. Here, we show that Wnt/planar cell polarity (PCP) autocrine signaling controls the emergence of cytonemes, and that cytonemes subsequently control paracrine Wnt/β-catenin signal activation. Upon binding of the Wnt family member Wnt8a, the receptor tyrosine kinase Ror2 becomes activated. Ror2/PCP signaling leads to the induction of cytonemes, which mediate the transport of Wnt8a to neighboring cells. In the Wnt-receiving cells, Wnt8a on cytonemes triggers Wnt/β-catenin-dependent gene transcription and proliferation. We show that cytoneme-based Wnt transport operates in diverse processes, including zebrafish development, murine intestinal crypt and human cancer organoids, demonstrating that Wnt transport by cytonemes and its control via the Ror2 pathway is highly conserved in vertebrates. Communication helps the cells that make up tissues and organs to work together as a team. One way that cells share information with each other as tissues grow and develop is by exchanging signaling proteins. These interact with receptors on the surface of other cells; this causes the cell to change how it behaves. The Wnt family of signaling proteins orchestrate organ development. Wnt proteins influence which types of cells develop, how fast they divide, and how and when they move. Relatively few cells, or small groups of cells, in developing tissues produce Wnt proteins, while larger groups nearby respond to the signals. We do not fully understand how Wnt proteins travel between cells, but recent work revealed an unexpected mechanism – cells seem to hand-deliver their messages. Finger-like structures called cytonemes grow out of the cell membrane and carry Wnt proteins to their destination. If the cytonemes do not form properly the target cells do not behave correctly, which can lead to severe tissue malformation. Mattes et al. have now investigated how cytonemes form using a combination of state-of-the-art genetic and high-resolution imaging techniques. In initial experiments involving zebrafish cells that were grown in the laboratory, Mattes et al. found that the Wnt proteins kick start their own transport; before they travel to their destination, they act on the cells that made them. A Wnt protein called Wnt8a activates the receptor Ror2 on the surface of the signal-producing cell. Ror2 then triggers signals inside the cell that begin the assembly of the cytonemes. The more Ror2 is activated, the more cytonemes the cell makes, and the more Wnt signals it can send out. This mechanism operates in various tissues: Ror2 also controls the cytoneme transport process in living zebrafish embryos, the mouse intestine and human stomach tumors. This knowledge will help researchers to develop new ways to control Wnt signaling, which could help to produce new treatments for diseases ranging from cancers (for example in the stomach and bowel) to degenerative diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Benjamin Mattes
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Yonglong Dang
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gediminas Greicius
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Benedikt Prunsche
- Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Jakob Rosenbauer
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany
| | - Johannes Stegmaier
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Suat Özbek
- Centre of Organismal Studies, University of Heidelberg, Karlsruhe, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Department of Physics, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Alexander Schug
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany.,Steinbuch Centre for Computing, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - David M Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
11
|
Roy JP, Halford MM, Stacker SA. The biochemistry, signalling and disease relevance of RYK and other WNT-binding receptor tyrosine kinases. Growth Factors 2018; 36:15-40. [PMID: 29806777 DOI: 10.1080/08977194.2018.1472089] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The receptor tyrosine kinases (RTKs) are a well-characterized family of growth factor receptors that have central roles in human disease and are frequently therapeutically targeted. The RYK, ROR, PTK7 and MuSK subfamilies make up an understudied subset of WNT-binding RTKs. Numerous developmental, stem cell and pathological roles of WNTs, in particular WNT5A, involve signalling via these WNT receptors. The WNT-binding RTKs have highly context-dependent signalling outputs and stimulate the β-catenin-dependent, planar cell polarity and/or WNT/Ca2+ pathways. RYK, ROR and PTK7 members have a pseudokinase domain in their intracellular regions. Alternative signalling mechanisms, including proteolytic cleavage and protein scaffolding functions, have been identified for these receptors. This review explores the structure, signalling, physiological and pathological roles of RYK, with particular attention paid to cancer and the possibility of therapeutically targeting RYK. The other WNT-binding RTKs are compared with RYK throughout to highlight the similarities and differences within this subset of WNT receptors.
Collapse
Affiliation(s)
- James P Roy
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
- b Sir Peter MacCallum Department of Oncology , The University of Melbourne , Parkville , Australia
| | - Michael M Halford
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Steven A Stacker
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
- b Sir Peter MacCallum Department of Oncology , The University of Melbourne , Parkville , Australia
| |
Collapse
|
12
|
Wang J, Ding M. Robo and Ror function in a common receptor complex to regulate Wnt-mediated neurite outgrowth in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2018; 115:E2254-E2263. [PMID: 29463707 PMCID: PMC5877952 DOI: 10.1073/pnas.1717468115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Growing axons are exposed to various guidance cues en route to their targets, but the mechanisms that govern the response of growth cones to combinations of signals remain largely elusive. Here, we found that the sole Robo receptor, SAX-3, in Caenorhabditis elegans functions as a coreceptor for Wnt/CWN-2 molecules. SAX-3 binds to Wnt/CWN-2 and facilitates the membrane recruitment of CWN-2. SAX-3 forms a complex with the Ror/CAM-1 receptor and its downstream effector Dsh/DSH-1, promoting signal transduction from Wnt to Dsh. sax-3 functions in Wnt-responsive cells and the SAX-3 receptor is restricted to the side of the cell from which the neurite is extended. DSH-1 has a similar asymmetric distribution, which is disrupted by sax-3 mutation. Taking these results together, we propose that Robo receptor can function as a Wnt coreceptor to regulate Wnt-mediated biological processes in vivo.
Collapse
Affiliation(s)
- Jiaming Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101 Beijing, China
- Biological Science Department, University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Mei Ding
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101 Beijing, China;
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
13
|
Heppert JK, Pani AM, Roberts AM, Dickinson DJ, Goldstein B. A CRISPR Tagging-Based Screen Reveals Localized Players in Wnt-Directed Asymmetric Cell Division. Genetics 2018; 208:1147-1164. [PMID: 29348144 PMCID: PMC5844328 DOI: 10.1534/genetics.117.300487] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/08/2018] [Indexed: 11/18/2022] Open
Abstract
Oriented cell divisions are critical to establish and maintain cell fates and tissue organization. Diverse extracellular and intracellular cues have been shown to provide spatial information for mitotic spindle positioning; however, the molecular mechanisms by which extracellular signals communicate with cells to direct mitotic spindle positioning are largely unknown. In animal cells, oriented cell divisions are often achieved by the localization of force-generating motor protein complexes to discrete cortical domains. Disrupting either these force-generating complexes or proteins that globally affect microtubule stability results in defects in mitotic positioning, irrespective of whether these proteins function as spatial cues for spindle orientation. This poses a challenge to traditional genetic dissection of this process. Therefore, as an alternative strategy to identify key proteins that act downstream of intercellular signaling, we screened the localization of many candidate proteins by inserting fluorescent tags directly into endogenous gene loci, without overexpressing the proteins. We tagged 23 candidate proteins in Caenorhabditis elegans and examined each protein's localization in a well-characterized, oriented cell division in the four-cell-stage embryo. We used cell manipulations and genetic experiments to determine which cells harbor key localized proteins and which signals direct these localizations in vivo We found that Dishevelled and adenomatous polyposis coli homologs are polarized during this oriented cell division in response to a Wnt signal, but two proteins typically associated with mitotic spindle positioning, homologs of NuMA and Dynein, were not detectably polarized. These results suggest an unexpected mechanism for mitotic spindle positioning in this system, they pinpoint key proteins of interest, and they highlight the utility of a screening approach based on analyzing the localization of endogenously tagged proteins.
Collapse
Affiliation(s)
- Jennifer K Heppert
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Ariel M Pani
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Allyson M Roberts
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Daniel J Dickinson
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina 27599
| |
Collapse
|
14
|
Regulation of Axon Guidance by the Wnt Receptor Ror/CAM-1 in the PVT Guidepost Cell in Caenorhabditis elegans. Genetics 2017; 207:1533-1545. [PMID: 28993416 DOI: 10.1534/genetics.117.300375] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 09/27/2017] [Indexed: 01/24/2023] Open
Abstract
The Caenorhabditis elegans ventral nerve cord (VNC) consists of two asymmetric bundles of neurons and axons that are separated by the midline. How the axons are guided to stay on the correct sides of the midline remains poorly understood. Here we provide evidence that the conserved Wnt signaling pathway along with the Netrin and Robo pathways constitute a combinatorial code for midline guidance of PVP and PVQ axons that extend into the VNC. Combined loss of the Wnts CWN-1, CWN-2, and EGL-20 or loss of the Wnt receptor CAM-1 caused >70% of PVP and PVQ axons to inappropriately cross over from the left side to the right side. Loss of the Frizzled receptor LIN-17 or the planar cell polarity (PCP) protein VANG-1 also caused cross over defects that did not enhance those in the cam-1 mutant, indicating that the proteins function together in midline guidance. Strong cam-1 expression can be detected in the PVQs and the guidepost cell PVT that is located on the midline. However, only when cam-1 is expressed in PVT are the crossover defects of PVP and PVQ rescued, showing that CAM-1 functions nonautonomously in PVT to prevent axons from crossing the midline.
Collapse
|
15
|
Florica RO, Hipolito V, Bautista S, Anvari H, Rapp C, El-Rass S, Asgharian A, Antonescu CN, Killeen MT. The ENU-3 protein family members function in the Wnt pathway parallel to UNC-6/Netrin to promote motor neuron axon outgrowth in C. elegans. Dev Biol 2017; 430:249-261. [PMID: 28694018 DOI: 10.1016/j.ydbio.2017.06.036] [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: 03/30/2017] [Revised: 06/26/2017] [Accepted: 06/30/2017] [Indexed: 10/19/2022]
Abstract
The axons of the DA and DB classes of motor neurons fail to reach the dorsal cord in the absence of the guidance cue UNC-6/Netrin or its receptor UNC-5 in C. elegans. However, the axonal processes usually exit their cell bodies in the ventral cord in the absence of both molecules. Strains lacking functional versions of UNC-6 or UNC-5 have a low level of DA and DB motor neuron axon outgrowth defects. We found that mutations in the genes for all six of the ENU-3 proteins function to enhance the outgrowth defects of the DA and DB axons in strains lacking either UNC-6 or UNC-5. A mutation in the gene for the MIG-14/Wntless protein also enhances defects in a strain lacking either UNC-5 or UNC-6, suggesting that the ENU-3 and Wnt pathways function parallel to the Netrin pathway in directing motor neuron axon outgrowth. Our evidence suggests that the ENU-3 proteins are novel members of the Wnt pathway in nematodes. Five of the six members of the ENU-3 family are predicted to be single-pass trans-membrane proteins. The expression pattern of ENU-3.1 was consistent with plasma membrane localization. One family member, ENU-3.6, lacks the predicted signal peptide and the membrane-spanning domain. In HeLa cells ENU-3.6 had a cytoplasmic localization and caused actin dependent processes to appear. We conclude that the ENU-3 family proteins function in a pathway parallel to the UNC-6/Netrin pathway for motor neuron axon outgrowth, most likely in the Wnt pathway.
Collapse
Affiliation(s)
- Roxana Oriana Florica
- Graduate Program in Molecular Science Program, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3; Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Victoria Hipolito
- Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Stephen Bautista
- Graduate Program in Molecular Science Program, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3; Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Homa Anvari
- Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Chloe Rapp
- Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Suzan El-Rass
- Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Alimohammad Asgharian
- Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Costin N Antonescu
- Graduate Program in Molecular Science Program, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3; Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3
| | - Marie T Killeen
- Graduate Program in Molecular Science Program, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3; Dept. of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto, Ont., Canada M5B 2K3.
| |
Collapse
|
16
|
Regulation of WNT Signaling at the Neuromuscular Junction by the Immunoglobulin Superfamily Protein RIG-3 in Caenorhabditis elegans. Genetics 2017; 206:1521-1534. [PMID: 28515212 PMCID: PMC5500148 DOI: 10.1534/genetics.116.195297] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 05/11/2017] [Indexed: 12/17/2022] Open
Abstract
Perturbations in synaptic function could affect the normal behavior of an animal, making it important to understand the regulatory mechanisms of synaptic signaling. Previous work has shown that in Caenorhabditis elegans an immunoglobulin superfamily protein, RIG-3, functions in presynaptic neurons to maintain normal acetylcholine receptor levels at the neuromuscular junction (NMJ). In this study, we elucidate the molecular and functional mechanism of RIG-3. We demonstrate by genetic and BiFC (Bi-molecular Fluorescence Complementation) assays that presynaptic RIG-3 functions by directly interacting with the immunoglobulin domain of the nonconventional Wnt receptor, ROR receptor tyrosine kinase (RTK), CAM-1, which functions in postsynaptic body-wall muscles. This interaction in turn inhibits Wnt/LIN-44 signaling through the ROR/CAM-1 receptor, and allows for maintenance of normal acetylcholine receptor, AChR/ACR-16, levels at the neuromuscular synapse. Further, this work reveals that RIG-3 and ROR/CAM-1 function through the β-catenin/HMP-2 at the NMJ. Taken together, our results demonstrate that RIG-3 functions as an inhibitory molecule of the Wnt/LIN-44 signaling pathway through the RTK, CAM-1.
Collapse
|
17
|
Remédio L, Gribble KD, Lee JK, Kim N, Hallock PT, Delestrée N, Mentis GZ, Froemke RC, Granato M, Burden SJ. Diverging roles for Lrp4 and Wnt signaling in neuromuscular synapse development during evolution. Genes Dev 2017; 30:1058-69. [PMID: 27151977 PMCID: PMC4863737 DOI: 10.1101/gad.279745.116] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/31/2016] [Indexed: 11/25/2022]
Abstract
In this study, Remédio et al. use mice and zebrafish to show that muscle prepatterning in mammals and zebrafish is established by different mechanisms. Their findings demonstrate that Agrin/Lrp4/MuSK signaling plays an essential role in neuromuscular synapse formation in both fish and mammals, whereas Wnt signaling is dispensable. Motor axons approach muscles that are prepatterned in the prospective synaptic region. In mice, prepatterning of acetylcholine receptors requires Lrp4, a LDLR family member, and MuSK, a receptor tyrosine kinase. Lrp4 can bind and stimulate MuSK, strongly suggesting that association between Lrp4 and MuSK, independent of additional ligands, initiates prepatterning in mice. In zebrafish, Wnts, which bind the Frizzled (Fz)-like domain in MuSK, are required for prepatterning, suggesting that Wnts may contribute to prepatterning and neuromuscular development in mammals. We show that prepatterning in mice requires Lrp4 but not the MuSK Fz-like domain. In contrast, prepatterning in zebrafish requires the MuSK Fz-like domain but not Lrp4. Despite these differences, neuromuscular synapse formation in zebrafish and mice share similar mechanisms, requiring Lrp4, MuSK, and neuronal Agrin but not the MuSK Fz-like domain or Wnt production from muscle. Our findings demonstrate that evolutionary divergent mechanisms establish muscle prepatterning in zebrafish and mice.
Collapse
Affiliation(s)
- Leonor Remédio
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University Medical School, New York, New York 10016, USA
| | - Katherine D Gribble
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Jennifer K Lee
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University Medical School, New York, New York 10016, USA
| | - Natalie Kim
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University Medical School, New York, New York 10016, USA
| | - Peter T Hallock
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University Medical School, New York, New York 10016, USA
| | - Nicolas Delestrée
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, New York 10032, USA; Department of Neurology, Columbia University, New York, New York 10032, USA
| | - George Z Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, New York 10032, USA; Department of Neurology, Columbia University, New York, New York 10032, USA
| | - Robert C Froemke
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University Medical School, New York, New York 10016, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Steven J Burden
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University Medical School, New York, New York 10016, USA
| |
Collapse
|
18
|
Craft TR, Forrester WC. The Caenorhabditis elegans matrix non-peptidase MNP-1 is required for neuronal cell migration and interacts with the Ror receptor tyrosine kinase CAM-1. Dev Biol 2017; 424:18-27. [PMID: 28238735 DOI: 10.1016/j.ydbio.2017.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 10/20/2022]
Abstract
Directed cell migration is critical for metazoan development. During Caenorhabditis elegans development many neuronal, muscle and other cell types migrate. Multiple classes of proteins have been implicated in cell migration including secreted guidance cues, receptors for guidance cues and intracellular proteins that respond to cues to polarize cells and produce the forces that move them. In addition, cell surface and secreted proteases have been identified that may clear the migratory route and process guidance cues. We report here that mnp-1 is required for neuronal cell and growth cone migrations. MNP-1 is expressed by migrating cells and functions cell autonomously for cell migrations. We also find a genetic interaction between mnp-1 and cam-1, which encodes a Ror receptor tyrosine kinase required for some of the same cell migrations.
Collapse
Affiliation(s)
- Teresa R Craft
- Medical Sciences Program, Indiana University, Bloomington, IN 47405, United States
| | - Wayne C Forrester
- Medical Sciences Program, Indiana University, Bloomington, IN 47405, United States.
| |
Collapse
|
19
|
Abstract
ROR-family receptor tyrosine kinases form a small subfamily of receptor tyrosine kinases (RTKs), characterized by a conserved, unique domain architecture. ROR RTKs are evolutionary conserved throughout the animal kingdom and act as alternative receptors and coreceptors of WNT ligands. The intracellular signaling cascades activated downstream of ROR receptors are diverse, including but not limited to ROR-Frizzled-mediated activation of planar cell polarity signaling, RTK-like signaling, and antagonistic regulation of WNT/β-Catenin signaling. In line with their diverse repertoire of signaling functions, ROR receptors are involved in the regulation of multiple processes in embryonic development such as development of the axial and paraxial mesoderm, the nervous system and the neural crest, the axial and appendicular skeleton, and the kidney. In humans, mutations in the ROR2 gene cause two distinct developmental syndromes, recessive Robinow syndrome (RRS; MIM 268310) and dominant brachydactyly type B1 (BDB1; MIM 113000). In Robinow syndrome patients and animal models, the development of multiple organs is affected, whereas BDB1 results only in shortening of the distal phalanges of fingers and toes, reflecting the diversity of functions and signaling activities of ROR-family RTKs. In this chapter, we give an overview on ROR receptor structure and function. We discuss their signaling functions and role in vertebrate embryonic development with a focus on those developmental processes that are affected by mutations in the ROR2 gene in human patients.
Collapse
|
20
|
Schille C, Bayerlová M, Bleckmann A, Schambony A. Ror2 signaling is required for local upregulation of GDF6 and activation of BMP signaling at the neural plate border. Development 2016; 143:3182-94. [DOI: 10.1242/dev.135426] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022]
Abstract
The receptor tyrosine kinase Ror2 is a major Wnt receptor that activates β-catenin-independent signaling and plays a conserved role in the regulation of convergent extension movements and planar cell polarity in vertebrates. Mutations in the ROR2 gene cause recessive Robinow syndrome in humans, a short-limbed dwarfism associated with craniofacial malformations. Here, we show that Ror2 is required for local upregulation of gdf6 at the neural plate border in Xenopus embryos. Ror2 morphant embryos fail to upregulate neural plate border genes and show defects in the induction of neural crest cell fate. These embryos lack the spatially restricted activation of BMP signaling at the neural plate border at early neurula stages, which is required for neural crest induction. Ror2-dependent planar cell polarity signaling is required in the dorsolateral marginal zone during gastrulation indirectly to upregulate the BMP ligand Gdf6 at the neural plate border and Gdf6 is sufficient to rescue neural plate border specification in Ror2 morphant embryos. Thereby, Ror2 links Wnt/planar cell polarity signaling to BMP signaling in neural plate border specification and neural crest induction.
Collapse
Affiliation(s)
- Carolin Schille
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen 91058, Germany
| | - Michaela Bayerlová
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen 37073, Germany
| | - Annalen Bleckmann
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen 37073, Germany
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, Göttingen 37099, Germany
| | - Alexandra Schambony
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen 91058, Germany
| |
Collapse
|
21
|
Flibotte S, Kim BR, Van de Laar E, Brown L, Moghal N. The SWI/SNF chromatin remodeling complex exerts both negative and positive control over LET-23/EGFR-dependent vulval induction in Caenorhabditis elegans. Dev Biol 2016; 415:46-63. [PMID: 27207389 DOI: 10.1016/j.ydbio.2016.05.009] [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: 10/14/2015] [Revised: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 11/19/2022]
Abstract
Signaling by the epidermal growth factor receptor (EGFR) generates diverse developmental patterns. This requires precise control over the location and intensity of signaling. Elucidation of these regulatory mechanisms is important for understanding development and disease pathogenesis. In Caenorhabditis elegans, LIN-3/EGF induces vulval formation in the mid-body, which requires LET-23/EGFR activation only in P6.p, the vulval progenitor nearest the LIN-3 source. To identify mechanisms regulating this signaling pattern, we screened for mutations that cooperate with a let-23 gain-of-function allele to cause ectopic vulval induction. Here, we describe a dominant gain-of-function mutation in swsn-4, a component of SWI/SNF chromatin remodeling complexes. Loss-of-function mutations in multiple SWI/SNF components reveal that weak reduction in SWI/SNF activity causes ectopic vulval induction, while stronger reduction prevents adoption of vulval fates, a phenomenon also observed with increasing loss of LET-23 activity. High levels of LET-23 expression in P6.p are thought to locally sequester LIN-3, thereby preventing ectopic vulval induction, with slight reductions in its expression interfering with LIN-3 sequestration, but not vulval fate signaling. We find that SWI/SNF positively regulates LET-23 expression in P6.p descendants, providing an explanation for the similarities between let-23 and SWI/SNF mutant phenotypes. However, SWI/SNF regulation of LET-23 expression is cell-specific, with SWI/SNF repressing its expression in the ALA neuron. The swsn-4 gain-of-function mutation affects the PTH domain, and provides the first evidence that its auto-inhibitory function in yeast Sth1p is conserved in metazoan chromatin remodelers. Finally, our work supports broad use of SWI/SNF in regulating EGFR signaling during development, and suggests that dominant SWI/SNF mutations in certain human congenital anomaly syndromes may be gain-of-functions.
Collapse
Affiliation(s)
- Stephane Flibotte
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z4.
| | - Bo Ram Kim
- Princess Margaret Cancer Centre/University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 1L7.
| | - Emily Van de Laar
- Princess Margaret Cancer Centre/University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 1L7.
| | - Louise Brown
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5.
| | - Nadeem Moghal
- Princess Margaret Cancer Centre/University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 1L7.
| |
Collapse
|
22
|
Abstract
Wnt proteins are conserved signalling molecules that have an essential role in regulating diverse processes during embryogenesis and adult tissue homoeostasis. Wnts are post-translationally modified by palmitoylation, which is essential for Wnt secretion and function. Intriguingly, the crystal structure of XWnt8 in complex with the extracellular domain of the Frizzled 8 cysteine-rich domain (Fzd8-CRD) revealed that Wnts use the fatty acid as a 'hotspot' residue to engage its receptor, which is a unique mode of receptor-ligand recognition. In addition, there are several lines of evidence suggesting that Wnts engage several signalling modulators and alternative receptors by means of fatty acids as a critical contact residue. In the present article, we review our current understanding of Wnt acylation and its functional role in Wnt signalling regulation.
Collapse
|
23
|
The ROR2 tyrosine kinase receptor regulates dendritic spine morphogenesis in hippocampal neurons. Mol Cell Neurosci 2015; 67:22-30. [DOI: 10.1016/j.mcn.2015.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 05/03/2015] [Accepted: 05/19/2015] [Indexed: 11/19/2022] Open
|
24
|
Gorrepati L, Krause MW, Chen W, Brodigan TM, Correa-Mendez M, Eisenmann DM. Identification of Wnt Pathway Target Genes Regulating the Division and Differentiation of Larval Seam Cells and Vulval Precursor Cells in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2015; 5:1551-66. [PMID: 26048561 PMCID: PMC4528312 DOI: 10.1534/g3.115.017715] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/18/2015] [Indexed: 12/29/2022]
Abstract
The evolutionarily conserved Wnt/β-catenin signaling pathway plays a fundamental role during metazoan development, regulating numerous processes including cell fate specification, cell migration, and stem cell renewal. Wnt ligand binding leads to stabilization of the transcriptional effector β-catenin and upregulation of target gene expression to mediate a cellular response. During larval development of the nematode Caenorhabditis elegans, Wnt/β-catenin pathways act in fate specification of two hypodermal cell types, the ventral vulval precursor cells (VPCs) and the lateral seam cells. Because little is known about targets of the Wnt signaling pathways acting during larval VPC and seam cell differentiation, we sought to identify genes regulated by Wnt signaling in these two hypodermal cell types. We conditionally activated Wnt signaling in larval animals and performed cell type-specific "mRNA tagging" to enrich for VPC and seam cell-specific mRNAs, and then used microarray analysis to examine gene expression compared to control animals. Two hundred thirty-nine genes activated in response to Wnt signaling were identified, and we characterized 50 genes further. The majority of these genes are expressed in seam and/or vulval lineages during normal development, and reduction of function for nine genes caused defects in the proper division, fate specification, fate execution, or differentiation of seam cells and vulval cells. Therefore, the combination of these techniques was successful at identifying potential cell type-specific Wnt pathway target genes from a small number of cells and at increasing our knowledge of the specification and behavior of these C. elegans larval hypodermal cells.
Collapse
Affiliation(s)
- Lakshmi Gorrepati
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | | | - Weiping Chen
- Intramural Research Program, NIDDK, Bethesda, Maryland 20814
| | | | - Margarita Correa-Mendez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | - David M Eisenmann
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| |
Collapse
|
25
|
Chien SCJ, Gurling M, Kim C, Craft T, Forrester W, Garriga G. Autonomous and nonautonomous regulation of Wnt-mediated neuronal polarity by the C. elegans Ror kinase CAM-1. Dev Biol 2015; 404:55-65. [PMID: 25917219 DOI: 10.1016/j.ydbio.2015.04.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 04/16/2015] [Accepted: 04/19/2015] [Indexed: 11/28/2022]
Abstract
Wnts are a conserved family of secreted glycoproteins that regulate various developmental processes in metazoans. Three of the five Caenorhabditis elegans Wnts, CWN-1, CWN-2 and EGL-20, and the sole Wnt receptor of the Ror kinase family, CAM-1, are known to regulate the anterior polarization of the mechanosensory neuron ALM. Here we show that CAM-1 and the Frizzled receptor MOM-5 act in parallel pathways to control ALM polarity. We also show that CAM-1 has two functions in this process: an autonomous signaling function that promotes anterior polarization and a nonautonomous Wnt-antagonistic function that inhibits anterior polarization. These antagonistic activities can account for the weak ALM phenotypes displayed by cam-1 mutants. Our observations suggest that CAM-1 could function as a Wnt receptor in many developmental processes, but the analysis of cam-1 mutants may fail to reveal CAM-1's role as a receptor in these processes because of its Wnt-antagonistic activity. In this model, loss of CAM-1 results in increased levels of Wnts that act through other Wnt receptors, masking CAM-1's autonomous role as a Wnt receptor.
Collapse
Affiliation(s)
- Shih-Chieh Jason Chien
- Department of Molecular and Cell Biology, University of California, Berkelry, CA 94720, United States
| | - Mark Gurling
- Department of Molecular and Cell Biology, University of California, Berkelry, CA 94720, United States
| | - Changsung Kim
- Department of Medical and Molecular Genetics, Indiana University Medical Sciences, Indiana University, Bloomington, IN 47405, United States
| | - Teresa Craft
- Department of Medical and Molecular Genetics, Indiana University Medical Sciences, Indiana University, Bloomington, IN 47405, United States
| | - Wayne Forrester
- Department of Medical and Molecular Genetics, Indiana University Medical Sciences, Indiana University, Bloomington, IN 47405, United States
| | - Gian Garriga
- Department of Molecular and Cell Biology, University of California, Berkelry, CA 94720, United States; Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, United States.
| |
Collapse
|
26
|
Roarty K, Shore AN, Creighton CJ, Rosen JM. Ror2 regulates branching, differentiation, and actin-cytoskeletal dynamics within the mammary epithelium. ACTA ACUST UNITED AC 2015; 208:351-66. [PMID: 25624393 PMCID: PMC4315251 DOI: 10.1083/jcb.201408058] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intricate cross-talk between classical and alternative Wnt signaling pathways includes an essential role for Ror2 in mammary epithelial development and differentiation. Wnt signaling encompasses β-catenin–dependent and –independent networks. How receptor context provides Wnt specificity in vivo to assimilate multiple concurrent Wnt inputs throughout development remains unclear. Here, we identified a refined expression pattern of Wnt/receptor combinations associated with the Wnt/β-catenin–independent pathway in mammary epithelial subpopulations. Moreover, we elucidated the function of the alternative Wnt receptor Ror2 in mammary development and provided evidence for coordination of this pathway with Wnt/β-catenin–dependent signaling in the mammary epithelium. Lentiviral short hairpin RNA (shRNA)-mediated depletion of Ror2 in vivo increased branching and altered the differentiation of the mammary epithelium. Microarray analyses identified distinct gene level alterations within the epithelial compartments in the absence of Ror2, with marked changes observed in genes associated with the actin cytoskeleton. Modeling of branching morphogenesis in vitro defined specific defects in cytoskeletal dynamics accompanied by Rho pathway alterations downstream of Ror2 loss. The current study presents a model of Wnt signaling coordination in vivo and assigns an important role for Ror2 in mammary development.
Collapse
Affiliation(s)
- Kevin Roarty
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Amy N Shore
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Chad J Creighton
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
27
|
Ackley BD. Wnt-signaling and planar cell polarity genes regulate axon guidance along the anteroposterior axis in C. elegans. Dev Neurobiol 2014; 74:781-96. [PMID: 24214205 PMCID: PMC4167394 DOI: 10.1002/dneu.22146] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 09/18/2013] [Accepted: 11/02/2013] [Indexed: 11/10/2022]
Abstract
During the development of the nervous system, neurons encounter signals that inform their outgrowth and polarization. Understanding how these signals combinatorially function to pattern the nervous system is of considerable interest to developmental neurobiologists. The Wnt ligands and their receptors have been well characterized in polarizing cells during asymmetric cell division. The planar cell polarity (PCP) pathway is also critical for cell polarization in the plane of an epithelium. The core set of PCP genes include members of the conserved Wnt-signaling pathway, such as Frizzled and Disheveled, but also the cadherin-domain protein Flamingo. In Drosophila, the Fat and Dachsous cadherins also function in PCP, but in parallel to the core PCP components. C. elegans also have two Fat-like and one Dachsous-like cadherins, at least one of which, cdh-4, contributes to neural development. In C. elegans Wnt ligands and the conserved PCP genes have been shown to regulate a number of different events, including embryonic cell polarity, vulval morphogenesis, and cell migration. As is also observed in vertebrates, the Wnt and PCP genes appear to function to primarily provide information about the anterior to posterior axis of development. Here, we review the recent work describing how mutations in the Wnt and core PCP genes affect axon guidance and synaptogenesis in C. elegans.
Collapse
Affiliation(s)
- Brian D Ackley
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, 66045
| |
Collapse
|
28
|
Poh WC, Shen Y, Inoue T. Function of the Ryk intracellular domain in C. elegans vulval development. Dev Dyn 2014; 243:1074-85. [PMID: 24975394 DOI: 10.1002/dvdy.24159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 05/05/2014] [Accepted: 06/01/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Ryk is a subfamily of receptor tyrosine kinases, which along with Frizzled and Ror, function as Wnt receptors. Vertebrate Ryk intracellular domain (ICD) is released from the cell membrane by a proteolytic cleavage in the transmembrane region and localizes to the nucleus. In C. elegans, Ryk is encoded by the lin-18 gene and regulates the polarity of the P7.p vulval cell. RESULTS Based on Western blots, we were unable to detect the presence of the cleaved LIN-18 ICD fragment. Functional assays found that LIN-18 intracellular domain is not absolutely required for LIN-18 function, consistent with previous results. However, overexpression of the LIN-18 intracellular domain fragment (LIN-18ICD) weakly enhanced the phenotype of lin-18 loss-of-function mutants. Furthermore, this activity was specific to the serine-rich juxtamembrane region. We also found that the nuclear localization of LIN-18ICD fragment can be regulated by Wnt pathway components including CAM-1/Ror, and by PAR-5/14-3-3. CONCLUSIONS Release of LIN-18ICD by cleavage at the membrane is not the main mechanism of LIN-18 signaling in vulval cells. However, our results suggest that LIN-18 intracellular domain interacts with Wnt pathway components and a 14-3-3 protein and likely plays a minor role in LIN-18 signaling.
Collapse
Affiliation(s)
- Woon Cheng Poh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
29
|
Kennedy LM, Grishok A. Neuronal migration is regulated by endogenous RNAi and chromatin-binding factor ZFP-1/AF10 in Caenorhabditis elegans. Genetics 2014; 197:207-20. [PMID: 24558261 PMCID: PMC4012481 DOI: 10.1534/genetics.114.162917] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/13/2014] [Indexed: 01/05/2023] Open
Abstract
Endogenous short RNAs and the conserved plant homeodomain (PHD) zinc-finger protein ZFP-1/AF10 regulate overlapping sets of genes in Caenorhabditis elegans, which suggests that they control common biological pathways. We have shown recently that the RNAi factor RDE-4 and ZFP-1 negatively modulate transcription of the insulin/PI3 signaling-dependent kinase PDK-1 to promote C. elegans fitness. Moreover, we have demonstrated that the insulin/IGF-1-PI3K-signaling pathway regulates the activity of the DAF-16/FOXO transcription factor in the hypodermis to nonautonomously promote the anterior migrations of the hermaphrodite-specific neurons (HSNs) during embryogenesis of C. elegans. In this study, we implicate the PHD-containing isoform of ZFP-1 and endogenous RNAi in the regulation of HSN migration. ZFP-1 affects HSN migration in part through its negative effect on pdk-1 transcription and modulation of downstream DAF-16 activity. We also identify a novel role for ZFP-1 and RNAi pathway components, including RDE-4, in the regulation of HSN migration in parallel with DAF-16. Therefore, the coordinated activities of DAF-16, ZFP-1, and endogenous RNAi contribute to gene regulation during development to ensure proper neuronal positioning.
Collapse
Affiliation(s)
- Lisa M. Kennedy
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032
| | - Alla Grishok
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032
| |
Collapse
|
30
|
Borcherding N, Kusner D, Liu GH, Zhang W. ROR1, an embryonic protein with an emerging role in cancer biology. Protein Cell 2014; 5:496-502. [PMID: 24752542 PMCID: PMC4085287 DOI: 10.1007/s13238-014-0059-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 03/27/2014] [Indexed: 11/23/2022] Open
Abstract
Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a member of the ROR family consisting of ROR1 and ROR2. RORs contain two distinct extracellular cysteine-rich domains and one transmembrane domain. Within the intracellular portion, ROR1 possesses a tyrosine kinase domain, two serine/threonine-rich domains and a proline-rich domain. RORs have been studied in the context of embryonic patterning and neurogenesis through a variety of homologs. These physiologic functions are dichotomous based on the requirement of the kinase domain. A growing literature has established ROR1 as a marker for cancer, such as in CLL and other blood malignancies. In addition, ROR1 is critically involved in progression of a number of blood and solid malignancies. ROR1 has been shown to inhibit apoptosis, potentiate EGFR signaling, and induce epithelial-mesenchymal transition (EMT). Importantly, ROR1 is only detectable in embryonic tissue and generally absent in adult tissue, making the protein an ideal drug target for cancer therapy.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Department of Pathology, College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | | | | | | |
Collapse
|
31
|
Green J, Nusse R, van Amerongen R. The role of Ryk and Ror receptor tyrosine kinases in Wnt signal transduction. Cold Spring Harb Perspect Biol 2014; 6:cshperspect.a009175. [PMID: 24370848 DOI: 10.1101/cshperspect.a009175] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Receptor tyrosine kinases of the Ryk and Ror families were initially classified as orphan receptors because their ligands were unknown. They are now known to contain functional extracellular Wnt-binding domains and are implicated in Wnt-signal transduction in multiple species. Although their signaling mechanisms still remain to be resolved in detail, both Ryk and Ror control important developmental processes in different tissues. However, whereas many other Wnt-signaling responses affect cell proliferation and differentiation, Ryk and Ror are mostly associated with controlling processes that rely on the polarized migration of cells. Here we discuss what is currently known about the involvement of this exciting class of receptors in development and disease.
Collapse
Affiliation(s)
- Jennifer Green
- Department of Developmental Biology and Howard Hughes Medical Institute, Stanford University, Stanford, California 94305
| | | | | |
Collapse
|
32
|
Schindler AJ, Sherwood DR. Morphogenesis of the caenorhabditis elegans vulva. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 2:75-95. [PMID: 23418408 DOI: 10.1002/wdev.87] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Understanding how cells move, change shape, and alter cellular behaviors to form organs, a process termed morphogenesis, is one of the great challenges of developmental biology. Formation of the Caenorhabditis elegans vulva is a powerful, simple, and experimentally accessible model for elucidating how morphogenetic processes produce an organ. In the first step of vulval development, three epithelial precursor cells divide and differentiate to generate 22 cells of 7 different vulval subtypes. The 22 vulval cells then rearrange from a linear array into a tube, with each of the seven cell types undergoing characteristic morphogenetic behaviors that construct the vulva. Vulval morphogenesis entails many of the same cellular activities that underlie organogenesis and tissue formation across species, including invagination, lumen formation, oriented cell divisions, cell–cell adhesion, cell migration, cell fusion, extracellular matrix remodeling, and cell invasion. Studies of vulval development have led to pioneering discoveries in a number of these processes and are beginning to bridge the gap between the pathways that specify cells and their connections to morphogenetic behaviors. The simplicity of the vulva and the experimental tools available in C. elegans will continue to make vulval morphogenesis a powerful paradigm to further our understanding of the largely mysterious mechanisms that build tissues and organs.
Collapse
|
33
|
Wnt signaling through the Ror receptor in the nervous system. Mol Neurobiol 2013; 49:303-15. [PMID: 23990374 DOI: 10.1007/s12035-013-8520-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/18/2013] [Indexed: 01/04/2023]
Abstract
The receptor tyrosine kinase-like orphan receptor (Ror) proteins are conserved tyrosine kinase receptors that play roles in a variety of cellular processes that pattern tissues and organs during vertebrate and invertebrate development. Ror signaling is required for skeleton and neuronal development and modulates cell migration, cell polarity, and convergent extension. Ror has also been implicated in two human skeletal disorders, brachydactyly type B and Robinow syndrome. Rors are widely expressed during metazoan development including domains in the nervous system. Here, we review recent progress in understanding the roles of the Ror receptors in neuronal migration, axonal pruning, axon guidance, and synaptic plasticity. The processes by which Ror signaling execute these diverse roles are still largely unknown, but they likely converge on cytoskeletal remodeling. In multiple species, Rors have been shown to act as Wnt receptors signaling via novel non-canonical Wnt pathways mediated in some tissues by the adapter protein disheveled and the non-receptor tyrosine kinase Src. Rors can either activate or repress Wnt target expression depending on the cellular context and can also modulate signal transduction by sequestering Wnt ligands away from their signaling receptors. Future challenges include the identification of signaling components of the Ror pathways and bettering our understanding of the roles of these pleiotropic receptors in patterning the nervous system.
Collapse
|
34
|
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: 1.3] [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
|
35
|
Abstract
Receptor Tyrosine Kinase (RTK)-Ras-Extracellular signal-regulated kinase (ERK) signaling pathways control many aspects of C. elegans development and behavior. Studies in C. elegans helped elucidate the basic framework of the RTK-Ras-ERK pathway and continue to provide insights into its complex regulation, its biological roles, how it elicits cell-type appropriate responses, and how it interacts with other signaling pathways to do so. C. elegans studies have also revealed biological contexts in which alternative RTK- or Ras-dependent pathways are used instead of the canonical pathway.
Collapse
Affiliation(s)
- Meera V Sundaram
- Dept. of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA.
| |
Collapse
|
36
|
Zhang J, Li X, Jevince AR, Guan L, Wang J, Hall DH, Huang X, Ding M. Neuronal target identification requires AHA-1-mediated fine-tuning of Wnt signaling in C. elegans. PLoS Genet 2013; 9:e1003618. [PMID: 23825972 PMCID: PMC3694823 DOI: 10.1371/journal.pgen.1003618] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 05/23/2013] [Indexed: 11/29/2022] Open
Abstract
Electrical synaptic transmission through gap junctions is a vital mode of intercellular communication in the nervous system. The mechanism by which reciprocal target cells find each other during the formation of gap junctions, however, is poorly understood. Here we show that gap junctions are formed between BDU interneurons and PLM mechanoreceptors in C. elegans and the connectivity of BDU with PLM is influenced by Wnt signaling. We further identified two PAS-bHLH family transcription factors, AHA-1 and AHR-1, which function cell-autonomously within BDU and PLM to facilitate the target identification process. aha-1 and ahr-1 act genetically upstream of cam-1. CAM-1, a membrane-bound receptor tyrosine kinase, is present on both BDU and PLM cells and likely serves as a Wnt antagonist. By binding to a cis-regulatory element in the cam-1 promoter, AHA-1 enhances cam-1 transcription. Our study reveals a Wnt-dependent fine-tuning mechanism that is crucial for mutual target cell identification during the formation of gap junction connections. The establishment of functional neuronal circuits requires that different neurons respond selectively to guidance molecules at particular times and in specific locations. In the target region, where cells connect, the same guidance molecules steer the growth of neurites from both the neuron and its target cell. The spatial, temporal, and cell-type-specific regulation of neuronal connection needs to be tightly regulated and precisely coordinated within the neuron and its target cell to achieve effective connection. In this study, we found that the precise connectivity of the BDU interneuron and the PLM mechanoreceptor in the nematode worm Caenorhabditis elegans is influenced by Wnt signaling. BDU-PLM contact also depends on the transcription factor AHA-1, which functions within both BDU and PLM cells to enhance transcription of the gene encoding the trans-membrane receptor CAM-1. CAM-1 is present on BDU and PLM and likely serves as a Wnt antagonist, thus linking transcriptional regulation by AHA-1 to modulation of Wnt signaling. Therefore, our study reveals a locally confined, cell type-specific and cell-autonomous mechanism that mediates mutual target identification.
Collapse
Affiliation(s)
- Jingyan Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xia Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Angela R. Jevince
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Liying Guan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Jiaming Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - David H. Hall
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- * E-mail: (XH); (MD)
| | - Mei Ding
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- * E-mail: (XH); (MD)
| |
Collapse
|
37
|
Abstract
Tyrosine phosphorylation plays a significant role in a wide range of cellular processes. The Drosophila genome encodes more than 20 receptor tyrosine kinases and extensive studies in the past 20 years have illustrated their diverse roles and complex signaling mechanisms. Although some receptor tyrosine kinases have highly specific functions, others strikingly are used in rather ubiquitous manners. Receptor tyrosine kinases regulate a broad expanse of processes, ranging from cell survival and proliferation to differentiation and patterning. Remarkably, different receptor tyrosine kinases share many of the same effectors and their hierarchical organization is retained in disparate biological contexts. In this comprehensive review, we summarize what is known regarding each receptor tyrosine kinase during Drosophila development. Astonishingly, very little is known for approximately half of all Drosophila receptor tyrosine kinases.
Collapse
Affiliation(s)
- Richelle Sopko
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
38
|
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: 1.3] [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
|
39
|
Abstract
30 years after the identification of WNTs, their signal transduction has become increasingly complex, with the discovery of more than 15 receptors and co-receptors in seven protein families. The recent discovery of three receptor classes for the R-spondin family of WNT agonists further adds to this complexity. What emerges is an intricate network of receptors that form higher-order ligand-receptor complexes routing downstream signalling. These are regulated both extracellularly by agonists such as R-spondin and intracellularly by post-translational modifications such as phosphorylation, proteolytic processing and endocytosis.
Collapse
Affiliation(s)
- Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, DKFZ, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
40
|
Abstract
In addition to activating β-catenin/TCF transcriptional complexes, Wnt proteins can elicit a variety of other responses. These are often lumped together under the denominator "alternative" or "non-canonical" Wnt signaling, but they likely comprise distinct signaling events. In this article, I discuss how the use of different ligand and receptor combinations is thought to give rise to these alternative Wnt-signaling responses. Although many of the biochemical details remain to be resolved, it is evident that alternative Wnt signaling plays important roles in regulating tissue morphogenesis during embryonic development.
Collapse
Affiliation(s)
- Renée van Amerongen
- Department of Developmental Biology, Stanford University, Stanford, California 94305, USA.
| |
Collapse
|
41
|
Bae YK, Sung JY, Kim YN, Kim S, Hong KM, Kim HT, Choi MS, Kwon JY, Shim J. An in vivo C. elegans model system for screening EGFR-inhibiting anti-cancer drugs. PLoS One 2012; 7:e42441. [PMID: 22957020 PMCID: PMC3434183 DOI: 10.1371/journal.pone.0042441] [Citation(s) in RCA: 20] [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: 03/20/2012] [Accepted: 07/09/2012] [Indexed: 11/20/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a well-established target for cancer treatment. EGFR tyrosine kinase (TK) inhibitors, such as gefinitib and erlotinib, have been developed as anti-cancer drugs. Although non-small cell lung carcinoma with an activating EGFR mutation, L858R, responds well to gefinitib and erlotinib, tumors with a doubly mutated EGFR, T790M-L858R, acquire resistance to these drugs. The C. elegans EGFR homolog LET-23 and its downstream signaling pathway have been studied extensively to provide insight into regulatory mechanisms conserved from C. elegans to humans. To develop an in vivo screening system for potential cancer drugs targeting specific EGFR mutants, we expressed three LET-23 chimeras in which the TK domain was replaced with either the human wild-type TK domain (LET-23::hEGFR-TK), a TK domain with the L858R mutation (LET-23::hEGFR-TK[L858R]), or a TK domain with the T790M-L858R mutations (LET-23::hEGFR-TK[T790M-L858R]) in C. elegans vulval cells using the let-23 promoter. The wild-type hEGFR-TK chimeric protein rescued the let-23 mutant phenotype, and the activating mutant hEGFR-TK chimeras induced a multivulva (Muv) phenotype in a wild-type C. elegans background. The anti-cancer drugs gefitinib and erlotinib suppressed the Muv phenotype in LET-23::hEGFR-TK[L858R]-expressing transgenic animals, but not in LET-23::hEGFR-TK[T790M-L858R] transgenic animals. As a pilot screen, 8,960 small chemicals were tested for Muv suppression, and AG1478 (an EGFR-TK inhibitor) and U0126 (a MEK inhibitor) were identified as potential inhibitors of EGFR-mediated biological function. In conclusion, transgenic C. elegans expressing chimeric LET-23::hEGFR-TK proteins are a model system that can be used in mutation-specific screens for new anti-cancer drugs.
Collapse
Affiliation(s)
- Young-Ki Bae
- Comparative Biomedicine Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Jee Young Sung
- Pediatric Oncology Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Yong-Nyun Kim
- Comparative Biomedicine Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Sunshin Kim
- New Experimental Therapeutics Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Kyeong Man Hong
- Cancer Cell and Molecular Biology Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Heung Tae Kim
- Center for Lung Cancer, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Min Sung Choi
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Jae Young Kwon
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Jaegal Shim
- Comparative Biomedicine Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
- * E-mail:
| |
Collapse
|
42
|
Schneider J, Skelton RL, Von Stetina SE, Middelkoop TC, van Oudenaarden A, Korswagen HC, Miller DM. UNC-4 antagonizes Wnt signaling to regulate synaptic choice in the C. elegans motor circuit. Development 2012; 139:2234-45. [PMID: 22619391 DOI: 10.1242/dev.075184] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Coordinated movement depends on the creation of synapses between specific neurons in the motor circuit. In C. elegans, this important decision is regulated by the UNC-4 homeodomain protein. unc-4 mutants are unable to execute backward locomotion because VA motor neurons are mis-wired with inputs normally reserved for their VB sisters. We have proposed that UNC-4 functions in VAs to block expression of VB genes. This model is substantiated by the finding that ectopic expression of the VB gene ceh-12 (encoding a homolog of the homeodomain protein HB9) in unc-4 mutants results in the mis-wiring of posterior VA motor neurons with VB-like connections. Here, we show that VA expression of CEH-12 depends on a nearby source of the Wnt protein EGL-20. Our results indicate that UNC-4 prevents VAs from responding to a local EGL-20 cue by disabling a canonical Wnt signaling cascade involving the Frizzled receptors MIG-1 and MOM-5. CEH-12 expression in VA motor neurons is also opposed by a separate pathway that includes the Wnt ligand LIN-44. This work has revealed a transcriptional mechanism for modulating the sensitivity of specific neurons to diffusible Wnt ligands and thereby defines distinct patterns of synaptic connectivity. The existence of comparable Wnt gradients in the vertebrate spinal cord could reflect similar roles for Wnt signaling in vertebrate motor circuit assembly.
Collapse
Affiliation(s)
- Judsen Schneider
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Jackson BM, Eisenmann DM. β-catenin-dependent Wnt signaling in C. elegans: teaching an old dog a new trick. Cold Spring Harb Perspect Biol 2012; 4:a007948. [PMID: 22745286 PMCID: PMC3405868 DOI: 10.1101/cshperspect.a007948] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Wnt signaling is an evolutionarily ancient pathway used to regulate many events during metazoan development. Genetic results from Caenorhabditis elegans more than a dozen years ago suggested that Wnt signaling in this nematode worm might be different than in vertebrates and Drosophila: the worm had a small number of Wnts, too many β-catenins, and some Wnt pathway components functioned in an opposite manner than in other species. Work over the ensuing years has clarified that C. elegans does possess a canonical Wnt/β-catenin signaling pathway similar to that in other metazoans, but that the majority of Wnt signaling in this species may proceed via a variant Wnt/β-catenin signaling pathway that uses some new components (mitogen-activated protein kinase signaling enzymes), and in which some conserved pathway components (β-catenin, T-cell factor [TCF]) are used in new and interesting ways. This review summarizes our current understanding of the canonical and novel TCF/β-catenin-dependent signaling pathways in C. elegans.
Collapse
Affiliation(s)
- Belinda M Jackson
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | | |
Collapse
|
44
|
van Amerongen R, Fuerer C, Mizutani M, Nusse R. Wnt5a can both activate and repress Wnt/β-catenin signaling during mouse embryonic development. Dev Biol 2012; 369:101-14. [PMID: 22771246 PMCID: PMC3435145 DOI: 10.1016/j.ydbio.2012.06.020] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/20/2012] [Accepted: 06/27/2012] [Indexed: 01/04/2023]
Abstract
Embryonic development is controlled by a small set of signal transduction pathways, with vastly different phenotypic outcomes depending on the time and place of their recruitment. How the same molecular machinery can elicit such specific and distinct responses, remains one of the outstanding questions in developmental biology. Part of the answer may lie in the high inherent genetic complexity of these signaling cascades, as observed for the Wnt-pathway. The mammalian genome encodes multiple Wnt proteins and receptors, each of which show dynamic and tightly controlled expression patterns in the embryo. Yet how these components interact in the context of the whole organism remains unknown. Here we report the generation of a novel, inducible transgenic mouse model that allows spatiotemporal control over the expression of Wnt5a, a protein implicated in many developmental processes and multiple Wnt-signaling responses. We show that ectopic Wnt5a expression from E10.5 onwards results in a variety of developmental defects, including loss of hair follicles and reduced bone formation in the skull. Moreover, we find that Wnt5a can have dual signaling activities during mouse embryonic development. Specifically, Wnt5a is capable of both inducing and repressing β-catenin/TCF signaling in vivo, depending on the time and site of expression and the receptors expressed by receiving cells. These experiments show for the first time that a single mammalian Wnt protein can have multiple signaling activities in vivo, thereby furthering our understanding of how signaling specificity is achieved in a complex developmental context.
Collapse
Affiliation(s)
- Renée van Amerongen
- Department of Developmental Biology and Howard Hughes Medical Institute, Lorry I. Lokey Stem Cell Research Building, Stanford University, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
45
|
Jensen M, Hoerndli FJ, Brockie PJ, Wang R, Johnson E, Maxfield D, Francis MM, Madsen DM, Maricq AV. Wnt signaling regulates acetylcholine receptor translocation and synaptic plasticity in the adult nervous system. Cell 2012; 149:173-87. [PMID: 22464329 DOI: 10.1016/j.cell.2011.12.038] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 11/08/2011] [Accepted: 12/30/2011] [Indexed: 12/20/2022]
Abstract
The adult nervous system is plastic, allowing us to learn, remember, and forget. Experience-dependent plasticity occurs at synapses--the specialized points of contact between neurons where signaling occurs. However, the mechanisms that regulate the strength of synaptic signaling are not well understood. Here, we define a Wnt-signaling pathway that modifies synaptic strength in the adult nervous system by regulating the translocation of one class of acetylcholine receptors (AChRs) to synapses. In Caenorhabditis elegans, we show that mutations in CWN-2 (Wnt ligand), LIN-17 (Frizzled), CAM-1 (Ror receptor tyrosine kinase), or the downstream effector DSH-1 (disheveled) result in similar subsynaptic accumulations of ACR-16/α7 AChRs, a consequent reduction in synaptic current, and predictable behavioral defects. Photoconversion experiments revealed defective translocation of ACR-16/α7 to synapses in Wnt-signaling mutants. Using optogenetic nerve stimulation, we demonstrate activity-dependent synaptic plasticity and its dependence on ACR-16/α7 translocation mediated by Wnt signaling via LIN-17/CAM-1 heteromeric receptors.
Collapse
Affiliation(s)
- Michael Jensen
- Department of Biology, University of Utah, Salt Lake City, UT 84112-0840, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Rebagay G, Yan S, Liu C, Cheung NK. ROR1 and ROR2 in Human Malignancies: Potentials for Targeted Therapy. Front Oncol 2012; 2:34. [PMID: 22655270 PMCID: PMC3356025 DOI: 10.3389/fonc.2012.00034] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 03/22/2012] [Indexed: 11/13/2022] Open
Abstract
Targeted therapies require cellular protein expression that meets specific requirements that will maximize effectiveness, minimize off-target toxicities, and provide an opportunity for a therapeutic effect. The receptor tyrosine kinase-like orphan receptors (ROR) are possible targets for therapy that may meet such requirements. RORs are transmembrane proteins that are part of the receptor tyrosine kinase (RTK) family. The RORs have been shown to play a role in tumor-like behavior, such as cell migration and cell invasiveness and are normally not expressed in normal adult tissue. As part of the large effort in target discovery, ROR proteins have recently been found to be expressed in human cancers. Their unique expression profiles may provide a novel class of therapeutic targets for small molecules against the kinase or for antibody-based therapies against these receptors. Being restricted on tumor cells and not on most normal tissues, RORs are excellent targets for the treatment of minimal residual disease, the final hurdle in the curative approach to many cancers, including solid tumors such as neuroblastoma. In this review, we summarize the biology of RORs as they relate to human cancer, and highlight the therapeutic approaches directed toward them.
Collapse
Affiliation(s)
- Guilly Rebagay
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center New York, NY, USA
| | | | | | | |
Collapse
|
47
|
Tyrosine phosphorylation-mediated signaling pathways in dictyostelium. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:894351. [PMID: 21776390 PMCID: PMC3135261 DOI: 10.1155/2011/894351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 02/21/2011] [Indexed: 01/21/2023]
Abstract
While studies on metazoan cell proliferation, cell differentiation, and cytokine signaling laid the foundation of the current paradigms of tyrosine kinase signaling, similar studies using lower eukaryotes have provided invaluable insight for the understanding of mammalian pathways, such as Wnt and STAT pathways. Dictyostelium is one of the leading lower eukaryotic model systems where stress-induced cellular responses, Wnt-like pathways, and STAT-mediated pathways are well investigated. These Dictyostelium pathways will be reviewed together with their mammalian counterparts to facilitate the comparative understanding of these variant and noncanonical pathways.
Collapse
|
48
|
Feike AC, Rachor K, Gentzel M, Schambony A. Wnt5a/Ror2-induced upregulation of xPAPC requires xShcA. Biochem Biophys Res Commun 2010; 400:500-6. [DOI: 10.1016/j.bbrc.2010.08.074] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 08/17/2010] [Indexed: 10/19/2022]
|
49
|
Song S, Zhang B, Sun H, Li X, Xiang Y, Liu Z, Huang X, Ding M. A Wnt-Frz/Ror-Dsh pathway regulates neurite outgrowth in Caenorhabditis elegans. PLoS Genet 2010; 6:e1001056. [PMID: 20711352 PMCID: PMC2920835 DOI: 10.1371/journal.pgen.1001056] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 07/08/2010] [Indexed: 11/18/2022] Open
Abstract
One of the challenges to understand the organization of the nervous system has been to determine how axon guidance molecules govern axon outgrowth. Through an unbiased genetic screen, we identified a conserved Wnt pathway which is crucial for anterior-posterior (A/P) outgrowth of neurites from RME head motor neurons in Caenorhabditis elegans. The pathway is composed of the Wnt ligand CWN-2, the Frizzled receptors CFZ-2 and MIG-1, the co-receptor CAM-1/Ror, and the downstream component Dishevelled/DSH-1. Among these, CWN-2 acts as a local attractive cue for neurite outgrowth, and its activity can be partially substituted with other Wnts, suggesting that spatial distribution plays a role in the functional specificity of Wnts. As a co-receptor, CAM-1 functions cell-autonomously in neurons and, together with CFZ-2 and MIG-1, transmits the Wnt signal to downstream effectors. Yeast two-hybrid screening identified DSH-1 as a binding partner for CAM-1, indicating that CAM-1 could facilitate CWN-2/Wnt signaling by its physical association with DSH-1. Our study reveals an important role of a Wnt-Frz/Ror-Dsh pathway in regulating neurite A/P outgrowth.
Collapse
Affiliation(s)
- Song Song
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Bo Zhang
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Hui Sun
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xia Li
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yanhui Xiang
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Zhonghua Liu
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xun Huang
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Mei Ding
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Receptor tyrosine kinase-like orphan receptor 2 (ROR2) and Indian hedgehog regulate digit outgrowth mediated by the phalanx-forming region. Proc Natl Acad Sci U S A 2010; 107:14211-6. [PMID: 20660756 DOI: 10.1073/pnas.1009314107] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Elongation of the digit rays resulting in the formation of a defined number of phalanges is a process poorly understood in mammals, whereas in the chicken distal mesenchymal bone morphogenetic protein (BMP) signaling in the so-called phalanx-forming region (PFR) or digit crescent (DC) seems to be involved. The human brachydactylies (BDs) are inheritable conditions characterized by variable degrees of digit shortening, thus providing an ideal model to analyze the development and elongation of phalanges. We used a mouse model for BDB1 (Ror2(W749X/W749X)) lacking middle phalanges and show that a signaling center corresponding to the chick PFR exists in the mouse, which is diminished in BDB1 mice. This resulted in a strongly impaired elongation of the digit condensations due to reduced chondrogenic commitment of undifferentiated distal mesenchymal cells. We further show that a similar BMP-based mechanism accounts for digit shortening in a mouse model for the closely related condition BDA1 (Ihh(E95K/E95K)), altogether indicating the functional significance of the PFR in mammals. Genetic interaction experiments as well as pathway analysis in BDB1 mice suggest that Indian hedgehog and WNT/beta-catenin signaling, which we show is inhibited by receptor tyrosine kinase-like orphan receptor 2 (ROR2) in distal limb mesenchyme, are acting upstream of BMP signaling in the PFR.
Collapse
|