1
|
Stewart RA, Ding Z, Jeon US, Goodman LB, Tran JJ, Zientko JP, Sabu M, Cadigan KM. Wnt target gene activation requires β-catenin separation into biomolecular condensates. PLoS Biol 2024; 22:e3002368. [PMID: 39316611 PMCID: PMC11460698 DOI: 10.1371/journal.pbio.3002368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/08/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
The Wnt/β-catenin signaling pathway plays numerous essential roles in animal development and tissue/stem cell maintenance. The activation of genes regulated by Wnt/β-catenin signaling requires the nuclear accumulation of β-catenin, a transcriptional co-activator. β-catenin is recruited to many Wnt-regulated enhancers through direct binding to T-cell factor/lymphoid enhancer factor (TCF/LEF) family transcription factors. β-catenin has previously been reported to form phase-separated biomolecular condensates (BMCs), which was implicated as a component of β-catenin's mechanism of action. This function required aromatic amino acid residues in the intrinsically disordered regions (IDRs) at the N- and C-termini of the protein. In this report, we further explore a role for β-catenin BMCs in Wnt target gene regulation. We find that β-catenin BMCs are miscible with LEF1 BMCs in vitro and in cultured cells. We characterized a panel of β-catenin mutants with different combinations of aromatic residue mutations in human cell culture and Drosophila melanogaster. Our data support a model in which aromatic residues across both IDRs contribute to BMC formation and signaling activity. Although different Wnt targets have different sensitivities to loss of β-catenin's aromatic residues, the activation of every target examined was compromised by aromatic substitution. These mutants are not defective in nuclear import or co-immunoprecipitation with several β-catenin binding partners. In addition, residues in the N-terminal IDR with no previously known role in signaling are clearly required for the activation of various Wnt readouts. Consistent with this, deletion of the N-terminal IDR results in a loss of signaling activity, which can be rescued by the addition of heterologous IDRs enriched in aromatic residues. Overall, our work supports a model in which the ability of β-catenin to form biomolecular condensates in the nucleus is tightly linked to its function as a transcriptional co-regulator.
Collapse
Affiliation(s)
- Richard A. Stewart
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Zhihao Ding
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ung Seop Jeon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lauren B. Goodman
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jeannine J. Tran
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John P. Zientko
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Malavika Sabu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ken M. Cadigan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
2
|
Gao H, Li Y, Zhang X, Zhang H, Tian Y, Li B. Unraveling the G protein-coupled receptor superfamily in aphids: Contractions and duplications linked to phloem feeding. Gen Comp Endocrinol 2024; 347:114435. [PMID: 38135222 DOI: 10.1016/j.ygcen.2023.114435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
The G Protein-Coupled Receptor (GPCR) superfamily is the largest and most diverse transmembrane receptor family, playing crucial roles in regulating various physiological processes. As one of the most destructive pests, aphids have been subject to previous studies, which revealed fewer GPCR superfamily members in Acyrthosiphon pisum and Aphis gossypii and the loss of multiple neuropeptide GPCRs. To elucidate the contraction patterns and evolutionary features of the aphid GPCR superfamily, we identified 97, 105, and 95 GPCR genes in Rhopalosiphum maidis, A. pisum, and A. gossypii, respectively. Comparative analysis and phylogenetic investigations with other hemipteran insects revealed a contracted GPCR superfamily in aphids. This contraction mainly occurred in biogenic amine receptors, GABA-B-R, and fz families, and several neuropeptide receptors such as ACPR, CrzR, and PTHR were completely lost. This phenomenon may be related to the parasitic nature of aphids. Additionally, several GPCRs associated with aphid feeding and water balance underwent duplication, including Lkr, NPFR, CCHa1-R, and DH-R, Type A LGRs, but the SK/CCKLR that inhibits feeding was completely lost, indicating changes in feeding genes that underpin the aphid's prolonged phloem feeding behavior. Furthermore, we observed fine-tuning in opsins, with reduced long-wavelength opsins and additional duplications of short-wavelength opsin, likely associated with daytime activity. Lastly, we found variations in the number of mthl genes in aphids. In conclusion, our investigation sheds light on the GPCR superfamily in aphids, revealing its association with diet lifestyle and laying the foundation for understanding and developing control strategies for the aphid GPCR superfamily.
Collapse
Affiliation(s)
- Han Gao
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yanxiao Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Xianzhen Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Hui Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Ying Tian
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
3
|
Hanly JJ, Loh LS, Mazo-Vargas A, Rivera-Miranda TS, Livraghi L, Tendolkar A, Day CR, Liutikaite N, Earls EA, Corning OBWH, D'Souza N, Hermina-Perez JJ, Mehta C, Ainsworth JA, Rossi M, Papa R, McMillan WO, Perry MW, Martin A. Frizzled2 receives WntA signaling during butterfly wing pattern formation. Development 2023; 150:dev201868. [PMID: 37602496 PMCID: PMC10560568 DOI: 10.1242/dev.201868] [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: 04/12/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023]
Abstract
Butterfly color patterns provide visible and biodiverse phenotypic readouts of the patterning processes. Although the secreted ligand WntA has been shown to instruct the color pattern formation in butterflies, its mode of reception remains elusive. Butterfly genomes encode four homologs of the Frizzled-family of Wnt receptors. Here, we show that CRISPR mosaic knockouts of frizzled2 (fz2) phenocopy the color pattern effects of WntA loss of function in multiple nymphalids. Whereas WntA mosaic clones result in intermediate patterns of reduced size, fz2 clones are cell-autonomous, consistent with a morphogen function. Shifts in expression of WntA and fz2 in WntA crispant pupae show that they are under positive and negative feedback, respectively. Fz1 is required for Wnt-independent planar cell polarity in the wing epithelium. Fz3 and Fz4 show phenotypes consistent with Wnt competitive-antagonist functions in vein formation (Fz3 and Fz4), wing margin specification (Fz3), and color patterning in the Discalis and Marginal Band Systems (Fz4). Overall, these data show that the WntA/Frizzled2 morphogen-receptor pair forms a signaling axis that instructs butterfly color patterning and shed light on the functional diversity of insect Frizzled receptors.
Collapse
Affiliation(s)
- Joseph J. Hanly
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
- Smithsonian Tropical Research Institute, Gamboa 0843-03092, Panama
| | - Ling S. Loh
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - Anyi Mazo-Vargas
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | | | - Luca Livraghi
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
- Smithsonian Tropical Research Institute, Gamboa 0843-03092, Panama
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Amruta Tendolkar
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - Christopher R. Day
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27708, USA
| | - Neringa Liutikaite
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - Emily A. Earls
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - Olaf B. W. H. Corning
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - Natalie D'Souza
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - José J. Hermina-Perez
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - Caroline Mehta
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
| | - Julia A. Ainsworth
- Department of Cell and Developmental Biology, UC San Diego, La Jolla, CA, USA
| | - Matteo Rossi
- Division of Evolutionary Biology, Ludwig Maximilian University, Munich 80539, Germany
| | - Riccardo Papa
- Department of Biology, University of Puerto Rico at Río Piedras, San Juan 00931, Puerto Rico
- Molecular Sciences and Research Center, University of Puerto Rico, San Juan 00931, Puerto Rico
- Dipartimento di Scienze Chimiche della Vita e della Sostenibilità Ambientale, Università di Parma, Parma 43121, Italy
| | - W. Owen McMillan
- Smithsonian Tropical Research Institute, Gamboa 0843-03092, Panama
| | - Michael W. Perry
- Department of Cell and Developmental Biology, UC San Diego, La Jolla, CA, USA
| | - Arnaud Martin
- Department of Biological Sciences, The George Washington University, Washington, DC 20052, USA
- Smithsonian Tropical Research Institute, Gamboa 0843-03092, Panama
| |
Collapse
|
4
|
Nguyen CT, Nguyen VM, Jeong S. Regulation of Off-track bidirectional signaling by Semaphorin-1a and Wnt signaling in the Drosophila motor axon guidance. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 150:103857. [PMID: 36244650 DOI: 10.1016/j.ibmb.2022.103857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/23/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Off-track receptor tyrosine kinase (OTK) has been shown to play an important role in the Drosophila motor axon pathfinding. The results of biochemical and genetic interactions previously suggested that OTK acts as a component of Semaphorin-1a/Plexin A (Sema-1a/PlexA) signaling during embryonic motor axon guidance and further showed that OTK binds to Wnt family members Wnt2 and Wnt4 and their common receptor Frizzled (Fz). However, the molecular mechanisms underlying the motor axon guidance function of OTK remain elusive. Here, we conclude that OTK mediates the forward and reverse signaling required for intersegmental nerve b (ISNb) motor axon pathfinding and we also demonstrate that the loss of two copies of Sema-1a synergistically enhances the bypass phenotype observed in otk mutants. Furthermore, the amorphic wnt2 mutation resulted in increased premature branching phenotypes, and the loss of fz function caused a frequent inability of ISNb motor axons to defasciculate at specific choice points. Consistent with a previous study, wnt4 mutant axons were often defective in recognizing target muscles. Interestingly, the bypass phenotype of otk mutants was robustly suppressed by loss of function mutations in wnt2, wnt4, or fz. In contrast, total ISNb defects of otk were increased by the loss-of-function alleles in wnt2 and wnt4, but not fz. These findings indicate that OTK may participate in the crosstalk between the Sema-1a/PlexA and Wnt signaling pathways, thereby contributing to ISNb motor axon pathfinding and target recognition.
Collapse
Affiliation(s)
- Chinh Thanh Nguyen
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Van Minh Nguyen
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Sangyun Jeong
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
5
|
Ko EA, Zhou T. GPCR genes as a predictor of glioma severity and clinical outcome. J Int Med Res 2022; 50:3000605221113911. [PMID: 35903880 PMCID: PMC9340954 DOI: 10.1177/03000605221113911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To undertake a comprehensive analysis of the differential expression of the G protein-coupled receptor (GPCR) genes in order to construct a GPCR gene signature for human glioma prognosis. METHODS This current study investigated several glioma transcriptomic datasets and identified the GPCR genes potentially associated with glioma severity. RESULTS A gene signature comprising 13 GPCR genes (nine upregulated and four downregulated genes in high-grade glioma) was developed. The predictive power of the 13-gene signature was tested in two validation cohorts and a strong positive correlation (Spearman's rank correlation test: ρ = 0.649 for the Validation1 cohort; ρ = 0.693 for the Validation2 cohort) was observed between the glioma grade and 13-gene based severity score in both cohorts. The 13-gene signature was also predictive of glioma prognosis based on Kaplan-Meier survival curve analyses and Cox proportional hazard regression analysis in four cohorts of patients with glioma. CONCLUSIONS Knowledge of GPCR gene expression in glioma may help researchers gain a better understanding of the pathogenesis of high-grade glioma. Further studies are needed to validate the association between these GPCR genes and glioma pathogenesis.
Collapse
Affiliation(s)
- Eun-A Ko
- Department of Physiology, School of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
6
|
Mehta S, Hingole S, Chaudhary V. The Emerging Mechanisms of Wnt Secretion and Signaling in Development. Front Cell Dev Biol 2021; 9:714746. [PMID: 34485301 PMCID: PMC8415634 DOI: 10.3389/fcell.2021.714746] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/20/2021] [Indexed: 12/22/2022] Open
Abstract
Wnts are highly-conserved lipid-modified secreted proteins that activate multiple signaling pathways. These pathways regulate crucial processes during various stages of development and maintain tissue homeostasis in adults. One of the most fascinating aspects of Wnt protein is that despite being hydrophobic, they are known to travel several cell distances in the extracellular space. Research on Wnts in the past four decades has identified several factors and uncovered mechanisms regulating their expression, secretion, and mode of extracellular travel. More recently, analyses on the importance of Wnt protein gradients in the growth and patterning of developing tissues have recognized the complex interplay of signaling mechanisms that help in maintaining tissue homeostasis. This review aims to present an overview of the evidence for the various modes of Wnt protein secretion and signaling and discuss mechanisms providing precision and robustness to the developing tissues.
Collapse
Affiliation(s)
| | | | - Varun Chaudhary
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| |
Collapse
|
7
|
Sonavane PR, Willert K. Controlling Wnt Signaling Specificity and Implications for Targeting WNTs Pharmacologically. Handb Exp Pharmacol 2021; 269:3-28. [PMID: 34463853 DOI: 10.1007/164_2021_529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Wnt signaling is critical for proper development of the embryo and for tissue homeostasis in the adult. Activation of this signaling cascade is initiated by binding of the secreted Wnts to their receptors. With the mammalian genome encoding multiple Wnts and Wnt receptors, a longstanding question in the field has been how Wnt-receptor specificities are achieved. Emerging from these studies is a picture of exquisite control over Wnt protein production, secretion, distribution, and receptor interactions, culminating in activation of downstream signaling cascades that control a myriad of biological processes. Here we discuss mechanisms by which Wnt protein activities are tuned and illustrate how the multiple layers of regulation can be leveraged for therapeutic interventions in disease.
Collapse
Affiliation(s)
- Pooja R Sonavane
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Karl Willert
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Chaudhary V, Hingole S, Frei J, Port F, Strutt D, Boutros M. Robust Wnt signaling is maintained by a Wg protein gradient and Fz2 receptor activity in the developing Drosophila wing. Development 2019; 146:dev174789. [PMID: 31399474 PMCID: PMC6703709 DOI: 10.1242/dev.174789] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 07/10/2019] [Indexed: 12/19/2022]
Abstract
Wnts are secreted proteins that regulate cell fate during development of all metazoans. Wnt proteins were proposed to spread over several cells to activate signaling directly at a distance. In the Drosophila wing epithelium, an extracellular gradient of the Wnt1 homolog Wingless (Wg) was observed extending over several cells away from producing cells. Surprisingly, however, it was also shown that a membrane-tethered Neurotactin-Wg fusion protein (NRT-Wg) can largely replace endogenous Wg, leading to proper patterning of the wing. Therefore, the functional range of Wg and whether Wg spreading is required for correct tissue patterning remains controversial. Here, by capturing secreted Wg on cells away from the source, we show that Wg acts over a distance of up to 11 cell diameters to induce signaling. Furthermore, cells located outside the reach of extracellular Wg depend on the Frizzled2 receptor to maintain signaling. Frizzled2 expression is increased in the absence of Wg secretion and is required to maintain signaling and cell survival in NRT-wg wing discs. Together, these results provide insight into the mechanisms by which robust Wnt signaling is achieved in proliferating tissues.
Collapse
Affiliation(s)
- Varun Chaudhary
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
- Department of Biological Science, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, India
| | - Swapnil Hingole
- Department of Biological Science, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, India
| | - Jana Frei
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Fillip Port
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - David Strutt
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
9
|
Cho C, Wang Y, Smallwood PM, Williams J, Nathans J. Molecular determinants in Frizzled, Reck, and Wnt7a for ligand-specific signaling in neurovascular development. eLife 2019; 8:47300. [PMID: 31225798 PMCID: PMC6588345 DOI: 10.7554/elife.47300] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/07/2019] [Indexed: 12/28/2022] Open
Abstract
The molecular basis of Wnt-Frizzled specificity is a central question in developmental biology. Reck, a multi-domain and multi-functional glycosylphosphatidylinositol-anchored protein, specifically enhances beta-catenin signaling by Wnt7a and Wnt7b in cooperation with the 7-transmembrane protein Gpr124. Among amino acids that distinguish Wnt7a and Wnt7b from other Wnts, two clusters are essential for signaling in a Reck- and Gpr124-dependent manner. Both clusters are far from the site of Frizzled binding: one resides at the amino terminus and the second resides in a protruding loop. Within Reck, the fourth of five tandem repeats of an unusual domain with six-cysteines (the CC domain) is essential for Wnt7a stimulation: substitutions P256A and W261A in CC4 eliminate this activity without changing protein abundance or surface localization. Mouse embryos carrying ReckP256A,W261A have severe defects in forebrain angiogenesis, providing the strongest evidence to date that Reck promotes CNS angiogenesis by specifically stimulating Wnt7a and Wnt7b signaling.
Collapse
Affiliation(s)
- Chris Cho
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Philip M Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - John Williams
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
10
|
Ripp C, Loth J, Petrova I, Linnemannstöns K, Ulepic M, Fradkin L, Noordermeer J, Wodarz A. Drosophila Ror is a nervous system-specific co-receptor for Wnt ligands. Biol Open 2018; 7:bio.033001. [PMID: 30341100 PMCID: PMC6262871 DOI: 10.1242/bio.033001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Wnt ligands are secreted glycoproteins that control many developmental processes and are crucial for homeostasis of numerous tissues in the adult organism. Signal transduction of Wnts involves the binding of Wnts to receptor complexes at the surface of target cells. These receptor complexes are commonly formed between a member of the Frizzled family of seven-pass transmembrane proteins and a co-receptor, which is usually a single-pass transmembrane protein. Among these co-receptors are several with structural homology to receptor tyrosine kinases, including Ror, PTK7, Ryk and MUSK. In vertebrates, Ror-2 and PTK7 are important regulators of planar cell polarity (PCP). By contrast, PCP phenotypes were not reported for mutations in off-track (otk) and off-track2 (otk2), encoding the Drosophila orthologs of PTK7. Here we show that Drosophila Ror is expressed in the nervous system and localizes to the plasma membrane of perikarya and neurites. A null allele of Ror is homozygous viable and fertile, does not display PCP phenotypes and interacts genetically with mutations in otk and otk2. We show that Ror binds specifically to Wingless (Wg), Wnt4 and Wnt5 and also to Frizzled2 (Fz2) and Otk. Our findings establish Drosophila Ror as a Wnt co-receptor expressed in the nervous system. Summary:Drosophila Ror is a Wnt co-receptor expressed in the nervous system. A Ror null mutant allele is viable and shows genetic interaction with mutations in off-track and off-track2.
Collapse
Affiliation(s)
- Caroline Ripp
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Julia Loth
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Iveta Petrova
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Karen Linnemannstöns
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Monique Ulepic
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Kerpener Str. 62, 50937 Köln, Germany
| | - Lee Fradkin
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2300RC Leiden, The Netherlands.,Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, LRB 760, Worcester, MA 01605, USA
| | - Jasprien Noordermeer
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Andreas Wodarz
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany .,Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Kerpener Str. 62, 50937 Köln, Germany.,Cluster of Excellence - Cellular stress response in aging-associated diseases (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| |
Collapse
|
11
|
Wingless Signaling: A Genetic Journey from Morphogenesis to Metastasis. Genetics 2018; 208:1311-1336. [PMID: 29618590 DOI: 10.1534/genetics.117.300157] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
Abstract
This FlyBook chapter summarizes the history and the current state of our understanding of the Wingless signaling pathway. Wingless, the fly homolog of the mammalian Wnt oncoproteins, plays a central role in pattern generation during development. Much of what we know about the pathway was learned from genetic and molecular experiments in Drosophila melanogaster, and the core pathway works the same way in vertebrates. Like most growth factor pathways, extracellular Wingless/Wnt binds to a cell surface complex to transduce signal across the plasma membrane, triggering a series of intracellular events that lead to transcriptional changes in the nucleus. Unlike most growth factor pathways, the intracellular events regulate the protein stability of a key effector molecule, in this case Armadillo/β-catenin. A number of mysteries remain about how the "destruction complex" destabilizes β-catenin and how this process is inactivated by the ligand-bound receptor complex, so this review of the field can only serve as a snapshot of the work in progress.
Collapse
|
12
|
Xun Q, Bi C, Cui X, Wu H, Wang M, Liao Y, Wang R, Xie H, Shen Z, Fang M. MagT1 is essential for Drosophila development through the shaping of Wingless and Decapentaplegic signaling pathways. Biochem Biophys Res Commun 2018; 503:1148-1153. [PMID: 29959918 DOI: 10.1016/j.bbrc.2018.06.133] [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: 06/08/2018] [Accepted: 06/23/2018] [Indexed: 12/01/2022]
Abstract
Magnesium transporter subtype 1 (MagT1) is a magnesium membrane transporter with channel like properties. We have previously identified MagT1 (CG7830) in Drosophila genome and characterized its protein product by electrophysiological means. Here, we report the generation of fly MagT1 mutants and show that MagT1 is essential for early embryonic development. In wings and primordial wings, by clonal analysis and RNAi knock down of MagT1, we have found that loss of MagT1 results in enhanced/ectopic Wingless (Wg, a fly Wnt) signaling and disrupted Decapentaplegic (Dpp) signaling, indicating the crucial role of MagT1 for fly development at later stages. Finally, we demonstrate directly that magnesium transportations are proportional with the MagT1 expressional levels in Drosophila S2 cells. Taken together, these findings may suggest that MagT1 is a major magnesium transporter/channel profoundly involved in fly development by affecting developmental signaling pathways, such as Wg and Dpp signaling.
Collapse
Affiliation(s)
- Qingying Xun
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China; Southeast University Medical School, Nanjing, 210009, China
| | - Caili Bi
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Xiaoying Cui
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Hongyan Wu
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Mingying Wang
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Yanlin Liao
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Rui Wang
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Hao Xie
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Zhijun Shen
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China
| | - Ming Fang
- Institute of Life Sciences, MOE Key Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
13
|
Grainger S, Willert K. Mechanisms of Wnt signaling and control. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10:e1422. [PMID: 29600540 PMCID: PMC6165711 DOI: 10.1002/wsbm.1422] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/14/2018] [Accepted: 02/22/2018] [Indexed: 01/17/2023]
Abstract
The Wnt signaling pathway is a highly conserved system that regulates complex biological processes across all metazoan species. At the cellular level, secreted Wnt proteins serve to break symmetry and provide cells with positional information that is critical to the patterning of the entire body plan. At the organismal level, Wnt signals are employed to orchestrate fundamental developmental processes, including the specification of the anterior-posterior body axis, induction of the primitive streak and ensuing gastrulation movements, and the generation of cell and tissue diversity. Wnt functions extend into adulthood where they regulate stem cell behavior, tissue homeostasis, and damage repair. Disruption of Wnt signaling activity during embryonic development or in adults results in a spectrum of abnormalities and diseases, including cancer. The molecular mechanisms that underlie the myriad of Wnt-regulated biological effects have been the subject of intense research for over three decades. This review is intended to summarize our current understanding of how Wnt signals are generated and interpreted. This article is categorized under: Biological Mechanisms > Cell Signaling Developmental Biology > Stem Cell Biology and Regeneration.
Collapse
Affiliation(s)
- Stephanie Grainger
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla California
| | - Karl Willert
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla California
| |
Collapse
|
14
|
Identification of Proteins Required for Precise Positioning of Apc2 in Dendrites. G3-GENES GENOMES GENETICS 2018; 8:1841-1853. [PMID: 29602811 PMCID: PMC5940173 DOI: 10.1534/g3.118.200205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In Drosophila neurons, uniform minus-end-out polarity in dendrites is maintained in part by kinesin-2-mediated steering of growing microtubules at branch points. Apc links the kinesin motor to growing microtubule plus ends and Apc2 recruits Apc to branch points where it functions. Because Apc2 acts to concentrate other steering proteins to branch points, we wished to understand how Apc2 is targeted. From an initial broad candidate RNAi screen, we found Miro (a mitochondrial transport protein), Ank2, Axin, spastin and Rac1 were required to position Apc2-GFP at dendrite branch points. YFP-Ank2-L8, Axin-GFP and mitochondria also localized to branch points suggesting the screen identified relevant proteins. By performing secondary screens, we found that energy production by mitochondria was key for Apc2-GFP positioning and spastin acted upstream of mitochondria. Ank2 seems to act independently from other players, except its membrane partner, Neuroglian (Nrg). Rac1 likely acts through Arp2/3 to generate branched actin to help recruit Apc2-GFP. Axin can function in a variety of wnt signaling pathways, one of which includes heterotrimeric G proteins and Frizzleds. Knockdown of Gαs, Gαo, Fz and Fz2, reduced targeting of Apc2 and Axin to branch points. Overall our data suggest that mitochondrial energy production, Nrg/Ank2, branched actin generated by Arp2/3 and Fz/G proteins/Axin function as four modules that control localization of the microtubule regulator Apc2 to its site of action in dendrite branch points.
Collapse
|
15
|
Ahmad SM. Conserved signaling mechanisms in Drosophila heart development. Dev Dyn 2017; 246:641-656. [PMID: 28598558 PMCID: PMC11546222 DOI: 10.1002/dvdy.24530] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/06/2017] [Accepted: 05/08/2017] [Indexed: 12/24/2022] Open
Abstract
Signal transduction through multiple distinct pathways regulates and orchestrates the numerous biological processes comprising heart development. This review outlines the roles of the FGFR, EGFR, Wnt, BMP, Notch, Hedgehog, Slit/Robo, and other signaling pathways during four sequential phases of Drosophila cardiogenesis-mesoderm migration, cardiac mesoderm establishment, differentiation of the cardiac mesoderm into distinct cardiac cell types, and morphogenesis of the heart and its lumen based on the proper positioning and cell shape changes of these differentiated cardiac cells-and illustrates how these same cardiogenic roles are conserved in vertebrates. Mechanisms bringing about the regulation and combinatorial integration of these diverse signaling pathways in Drosophila are also described. This synopsis of our present state of knowledge of conserved signaling pathways in Drosophila cardiogenesis and the means by which it was acquired should facilitate our understanding of and investigations into related processes in vertebrates. Developmental Dynamics 246:641-656, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shaad M. Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
16
|
Endocytosis of Wingless via a dynamin-independent pathway is necessary for signaling in Drosophila wing discs. Proc Natl Acad Sci U S A 2016; 113:E6993-E7002. [PMID: 27791132 DOI: 10.1073/pnas.1610565113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Endocytosis of ligand-receptor complexes regulates signal transduction during development. In particular, clathrin and dynamin-dependent endocytosis has been well studied in the context of patterning of the Drosophila wing disc, wherein apically secreted Wingless (Wg) encounters its receptor, DFrizzled2 (DFz2), resulting in a distinctive dorso-ventral pattern of signaling outputs. Here, we directly track the endocytosis of Wg and DFz2 in the wing disc and demonstrate that Wg is endocytosed from the apical surface devoid of DFz2 via a dynamin-independent CLIC/GEEC pathway, regulated by Arf1, Garz, and class I PI3K. Subsequently, Wg containing CLIC/GEEC endosomes fuse with DFz2-containing vesicles derived from the clathrin and dynamin-dependent endocytic pathway, which results in a low pH-dependent transfer of Wg to DFz2 within the merged and acidified endosome to initiate Wg signaling. The employment of two distinct endocytic pathways exemplifies a mechanism wherein cells in tissues leverage multiple endocytic pathways to spatially regulate signaling.
Collapse
|
17
|
Gammons MV, Renko M, Johnson CM, Rutherford TJ, Bienz M. Wnt Signalosome Assembly by DEP Domain Swapping of Dishevelled. Mol Cell 2016; 64:92-104. [PMID: 27692984 PMCID: PMC5065529 DOI: 10.1016/j.molcel.2016.08.026] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/15/2016] [Accepted: 08/23/2016] [Indexed: 01/17/2023]
Abstract
Extracellular signals are often transduced by dynamic signaling complexes ("signalosomes") assembled by oligomerizing hub proteins following their recruitment to signal-activated transmembrane receptors. A paradigm is the Wnt signalosome, which is assembled by Dishevelled via reversible head-to-tail polymerization by its DIX domain. Its activity causes stabilization of β-catenin, a Wnt effector with pivotal roles in animal development and cancer. How Wnt triggers signalosome assembly is unknown. Here, we use structural analysis, as well as biophysical and cell-based assays, to show that the DEP domain of Dishevelled undergoes a conformational switch, from monomeric to swapped dimer, to trigger DIX-dependent polymerization and signaling to β-catenin. This occurs in two steps: binding of monomeric DEP to Frizzled followed by DEP domain swapping triggered by its high local concentration upon Wnt-induced recruitment into clathrin-coated pits. DEP domain swapping confers directional bias on signaling, and the dimerization provides cross-linking between Dishevelled polymers, illustrating a key principle underlying signalosome formation.
Collapse
Affiliation(s)
- Melissa V Gammons
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - Miha Renko
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Christopher M Johnson
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Trevor J Rutherford
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Mariann Bienz
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
18
|
Mottier-Pavie VI, Palacios V, Eliazer S, Scoggin S, Buszczak M. The Wnt pathway limits BMP signaling outside of the germline stem cell niche in Drosophila ovaries. Dev Biol 2016; 417:50-62. [PMID: 27364467 DOI: 10.1016/j.ydbio.2016.06.038] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/14/2016] [Accepted: 06/26/2016] [Indexed: 01/07/2023]
Abstract
The mechanisms that modulate and limit the signaling output of adult stem cell niches remain poorly understood. To gain further insights into how these microenvironments are regulated in vivo, we performed a candidate gene screen designed to identify factors that restrict BMP signal production to the cap cells that comprise the germline stem cell (GSC) niche of Drosophila ovaries. Through these efforts, we found that disruption of Wnt4 and components of the canonical Wnt pathway results in a complex germ cell phenotype marked by an expansion of GSC-like cells, pre-cystoblasts and cystoblasts in young females. This phenotype correlates with an increase of decapentaplegic (dpp) mRNA levels within escort cells and varying levels of BMP responsiveness in the germline. Further genetic experiments show that Wnt4, which exhibits graded expression in somatic cells of germaria, activates the Wnt pathway in posteriorly positioned escort cells. The activation of the Wnt pathway appears to be limited by the BMP pathway itself, as loss of Mad in escort cells results in the expansion of Wnt pathway activation. Wnt pathway activity changes within germaria during the course of aging, coincident with changes in dpp production. These data suggest that mutual antagonism between the BMP and Wnt pathways in somatic cells helps to regulate germ cell differentiation.
Collapse
Affiliation(s)
- Violaine I Mottier-Pavie
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Victor Palacios
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Susan Eliazer
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Shane Scoggin
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA.
| |
Collapse
|
19
|
Abstract
Frizzled proteins are the principal receptors for the Wnt family of ligands. They mediate canonical Wnt signaling together with Lrp5 and Lrp6 coreceptors. In conjunction with Celsr, Vangl, and a small number of additional membrane and membrane-associated proteins, they also play a central role in tissue polarity/planar cell polarity (PCP) signaling. Targeted mutations in 9 of the 10 mammalian Frizzled genes have revealed their roles in an extraordinarily diverse set of developmental and homeostatic processes, including morphogenetic movements responsible for palate, ventricular septum, ocular furrow, and neural tube closure; survival of thalamic neurons; bone formation; central nervous system (CNS) angiogenesis and blood-brain barrier formation and maintenance; and a wide variety of processes that orient subcellular, cellular, and multicellular structures relative to the body axes. The last group likely reflects the mammalian equivalent of tissue polarity/PCP signaling, as defined in Drosophila, and it includes CNS axon guidance, hair follicle and tongue papilla orientation, and inner ear sensory hair bundle orientation. Frizzled receptors are ubiquitous among multicellular animals and, with other signaling molecules, they very likely evolved to permit the development of the complex tissue architectures that provide multicellular animals with their enormous selective advantage.
Collapse
Affiliation(s)
- Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Chang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
20
|
Ahmad SM, Bhattacharyya P, Jeffries N, Gisselbrecht SS, Michelson AM. Two Forkhead transcription factors regulate cardiac progenitor specification by controlling the expression of receptors of the fibroblast growth factor and Wnt signaling pathways. Development 2015; 143:306-17. [PMID: 26657774 PMCID: PMC4725337 DOI: 10.1242/dev.122952] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 11/26/2015] [Indexed: 11/20/2022]
Abstract
Cardiogenesis involves the coordinated regulation of multiple biological processes by a finite set of transcription factors (TFs). Here, we show that the Forkhead TFs Checkpoint suppressor homologue (CHES-1-like) and Jumeau (Jumu), which govern cardiac progenitor cell divisions by regulating Polo kinase activity, play an additional, mutually redundant role in specifying the cardiac mesoderm (CM) as eliminating the functions of both Forkhead genes in the same Drosophila embryo results in defective hearts with missing hemisegments. This process is mediated by the Forkhead TFs regulating the fibroblast growth factor receptor Heartless (Htl) and the Wnt receptor Frizzled (Fz): CHES-1-like and jumu exhibit synergistic genetic interactions with htl and fz in CM specification, thereby implying that they function through the same genetic pathways, and transcriptionally activate the expression of both receptor-encoding genes. Furthermore, ectopic overexpression of either htl or fz in the mesoderm partially rescues the defective CM specification phenotype in embryos lacking both Forkhead genes. Together, these data emphasize the functional redundancy that leads to robustness in the cardiac progenitor specification process, and illustrate the pleiotropic functions of Forkhead TFs in different aspects of cardiogenesis. Summary: Checkpoint suppressor homologue and Jumeau, which are known to govern cardiac progenitor cell divisions, play additional, mutually redundant roles in specifying cardiac mesoderm in Drosophila.
Collapse
Affiliation(s)
- Shaad M Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pritha Bhattacharyya
- Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neal Jeffries
- Office of Biostatistics Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen S Gisselbrecht
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alan M Michelson
- Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Muñoz-Descalzo S, Hadjantonakis AK, Arias AM. Wnt/ß-catenin signalling and the dynamics of fate decisions in early mouse embryos and embryonic stem (ES) cells. Semin Cell Dev Biol 2015; 47-48:101-9. [PMID: 26321498 DOI: 10.1016/j.semcdb.2015.08.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 12/22/2022]
Abstract
Wnt/ß-catenin signalling is a widespread cell signalling pathway with multiple roles during vertebrate development. In mouse embryonic stem (mES) cells, there is a dual role for ß-catenin: it promotes differentiation when activated as part of the Wnt/ß-catenin signalling pathway, and promotes stable pluripotency independently of signalling. Although mES cells resemble the preimplantation epiblast progenitors, the first requirement for Wnt/ß-catenin signalling during mouse development has been reported at implantation [1,2]. The relationship between ß-catenin and pluripotency and that of mES cells with epiblast progenitors suggests that ß-catenin might have a functional role during preimplantation development. Here we summarize the expression and function of Wnt/ß-catenin signalling elements during the early stages of mouse development and consider the reasons why the requirement in ES cells do not reflect the embryo.
Collapse
Affiliation(s)
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | |
Collapse
|
22
|
Abstract
The Wnt/Wingless (Wg) signaling cascade controls a number of biological processes in animal development and adult life; aberrant Wnt/Wg signaling can cause diseases. In the 1980s genes were discovered that encode core Wnt/Wg pathway components: their mutant phenotypes were similar and an outline of a signaling cascade emerged. Over the years our knowledge of this important signaling system increased and more components were uncovered that are instrumental for Wnt/Wg secretion and transduction. Here we provide an overview of these discoveries, the technologies involved, with a particular focus on the important role Drosophila screens played in this process.
Collapse
Affiliation(s)
- Fabian Heinz Jenny
- a University of Zurich; Institute of Molecular Life Sciences ; Zurich , Switzerland
| | | |
Collapse
|
23
|
Bipartite recognition of DNA by TCF/Pangolin is remarkably flexible and contributes to transcriptional responsiveness and tissue specificity of wingless signaling. PLoS Genet 2014; 10:e1004591. [PMID: 25188465 PMCID: PMC4154663 DOI: 10.1371/journal.pgen.1004591] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 07/08/2014] [Indexed: 11/19/2022] Open
Abstract
The T-cell factor (TCF) family of transcription factors are major mediators of Wnt/β-catenin signaling in metazoans. All TCFs contain a High Mobility Group (HMG) domain that possesses specific DNA binding activity. In addition, many TCFs contain a second DNA binding domain, the C-clamp, which binds to DNA motifs referred to as Helper sites. While HMG and Helper sites are both important for the activation of several Wnt dependent cis-regulatory modules (W-CRMs), the rules of what constitutes a functional HMG-Helper site pair are unknown. In this report, we employed a combination of in vitro binding, reporter gene analysis and bioinformatics to address this question, using the Drosophila family member TCF/Pangolin (TCF/Pan) as a model. We found that while there were constraints for the orientation and spacing of HMG-Helper pairs, the presence of a Helper site near a HMG site in any orientation increased binding and transcriptional response, with some orientations displaying tissue-specific patterns. We found that altering an HMG-Helper site pair from a sub-optimal to optimal orientation/spacing dramatically increased the responsiveness of a W-CRM in several fly tissues. In addition, we used the knowledge gained to bioinformatically identify two novel W-CRMs, one that was activated by Wnt/β-catenin signaling in the prothoracic gland, a tissue not previously connected to this pathway. In sum, this work extends the importance of Helper sites in fly W-CRMs and suggests that the type of HMG-Helper pair is a major factor in setting the threshold for Wnt activation and tissue-responsiveness. Regulation of gene expression is controlled in large part by proteins known as transcription factors, which bind to specific DNA sequences in the genome. The DNA binding domains of transcription factors recognize short stretches (5–11 base pairs) of DNA with considerable sequence degeneracy. This means that a single DNA binding domain, on its own, cannot find its targets in the vast excess of genomic sequence. We are studying this question using TCF/Pangolin, a Drosophila transcription factor that mediates Wnt/β-catenin signaling, an important developmental cell-cell communication pathway. TCF/Pangolin contains two DNA binding domains that bind to a pair of DNA motifs known as HMG and Helper sites. We used a combination of biochemistry, genetics and bioinformatics to elucidate the spacing and orientation constraints of HMG-Helper site pairs. We found that HMG-Helper site spacing/orientation influenced the sensitivity of a target to Wnt signaling, as well as its tissue-responsiveness. We used this information to improve our ability to search the Drosophila genome for Wnt targets, one of which was activated by the pathway in the fly ring gland, the major endocrine organ in insects. Our work is relevant to related mammalian TCF family members, which are implicated in development, stem cell biology and the progression of cancer.
Collapse
|
24
|
Structure-function analysis of the C-clamp of TCF/Pangolin in Wnt/ß-catenin signaling. PLoS One 2014; 9:e86180. [PMID: 24465946 PMCID: PMC3896468 DOI: 10.1371/journal.pone.0086180] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/05/2013] [Indexed: 02/03/2023] Open
Abstract
The evolutionarily conserved Wnt/ß-catenin (Wnt/ß-cat) pathway plays an important role in animal development in metazoans. Many Wnt targets are regulated by members of the TCF/LEF1 (TCF) family of transcription factors. All TCFs contain a High Mobility Group (HMG) domain that bind specific DNA sequences. Invertebrate TCFs and some vertebrate TCF isoforms also contain another domain, called the C-clamp, which allows TCFs to recognize an additional DNA motif known as the Helper site. While the C-clamp has been shown to be important for regulating several Wnt reporter genes in cell culture, its physiological role in regulating Wnt targets is less clear. In addition, little is known about this domain, except that two of the four conserved cysteines are functionally important. Here, we carried out a systematic mutagenesis and functional analysis of the C-clamp from the Drosophila TCF/Pangolin (TCF/Pan) protein. We found that the C-clamp is a zinc-binding domain that is sufficient for binding to the Helper site. In addition to this DNA-binding activity, the C-clamp also inhibits the HMG domain from binding its cognate DNA site. Point mutations were identified that specifically affected DNA-binding or reduced the inhibitory effect. These mutants were characterized in TCF/Pan rescue assays. The specific DNA-binding activity of the C-clamp was essential for TCF/Pan function in cell culture and in patterning the embryonic epidermis of Drosophila, demonstrating the importance of this C-clamp activity in regulating Wnt target gene expression. In contrast, the inhibitory mutation had a subtle effect in cell culture and no effect on TCF/Pan activity in embryos. These results provide important information about the functional domains of the C-clamp, and highlight its importance for Wnt/ß-cat signaling in Drosophila.
Collapse
|
25
|
Bejsovec A. Wingless/Wnt signaling in Drosophila: the pattern and the pathway. Mol Reprod Dev 2013; 80:882-94. [PMID: 24038436 DOI: 10.1002/mrd.22228] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/07/2013] [Indexed: 01/09/2023]
Abstract
Wnt signaling generates pattern in all animal embryos, from flies and worms to humans, and promotes the undifferentiated, proliferative state critical for stem cells in adult tissues. Inappropriate Wnt pathway activation is the major cause of colorectal cancers, a leading cause of cancer death in humans. Although this pathway has been studied extensively for years, large gaps remain in our understanding of how it switches on and off, and how its activation changes cellular behaviors. Much of what is known about the pathway comes from genetic studies in Drosophila, where a single Wnt molecule, encoded by wingless (wg), directs an array of cell-fate decisions similar to those made by the combined activities of all 19 Wnt family members in vertebrates. Although Wg specifies fate in many tissues, including the brain, limbs, and major organs, the fly embryonic epidermis has proven to be a very powerful system for dissecting pathway activity. It is a simple, accessible tissue, with a pattern that is highly sensitive to small changes in Wg pathway activity. This review discusses what we have learned about Wnt signaling from studying mutations that disrupt epidermal pattern in the fly embryo, highlights recent advances and controversies in the field, and sets these issues in the context of questions that remain about how this essential signaling pathway functions.
Collapse
Affiliation(s)
- Amy Bejsovec
- Department of Biology, Duke University, Durham, North Carolina
| |
Collapse
|
26
|
Tan Y, Yu D, Busto GU, Wilson C, Davis RL. Wnt signaling is required for long-term memory formation. Cell Rep 2013; 4:1082-9. [PMID: 24035392 DOI: 10.1016/j.celrep.2013.08.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 07/24/2013] [Accepted: 08/02/2013] [Indexed: 12/13/2022] Open
Abstract
Wnt signaling regulates synaptic plasticity and neurogenesis in the adult nervous system, suggesting a potential role in behavioral processes. Here, we probed the requirement for Wnt signaling during olfactory memory formation in Drosophila using an inducible RNAi approach. Interfering with β-catenin expression in adult mushroom body neurons specifically impaired long-term memory (LTM) without altering short-term memory. The impairment was reversible, being rescued by expression of a wild-type β-catenin transgene, and correlated with disruption of a cellular LTM trace. Inhibition of wingless, a Wnt ligand, and arrow, a Wnt coreceptor, also impaired LTM. Wingless expression in wild-type flies was transiently elevated in the brain after LTM conditioning. Thus, inhibiting three key components of the Wnt signaling pathway in adult mushroom bodies impairs LTM, indicating that this pathway mechanistically underlies this specific form of memory.
Collapse
Affiliation(s)
- Ying Tan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
27
|
tPA regulates neurite outgrowth by phosphorylation of LRP5/6 in neural progenitor cells. Mol Neurobiol 2013; 49:199-215. [PMID: 23925701 DOI: 10.1007/s12035-013-8511-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/08/2013] [Indexed: 10/26/2022]
Abstract
Despite the important role of tissue plasminogen activator (tPA) as a neuromodulator in neurons, microglia, and astrocytes, its role in neural progenitor cell (NPC) development is not clear yet. We identified that tPA is highly expressed in NPCs compared with neurons. Inhibition of tPA activity or expression using tPA stop, PAI-1, or tPA siRNA inhibited neurite outgrowth from NPCs, while overexpression or addition of exogenous tPA increased neurite outgrowth. The expression of Wnt and β-catenin as well as phosphorylation of LRP5 and LRP6, which has been implicated in Wnt-β-catenin signaling, was rapidly increased after tPA treatment and was decreased by tPA siRNA transfection. Knockdown of β-catenin or LRP5/6 expression by siRNA prevented tPA-induced neurite extension. NPCs obtained from tPA KO mice showed impaired neurite outgrowth compared with WT NPCs. In ischemic rat brains, axon density was higher in the brains transplanted with WT NPCs than in those with tPA KO NPCs, suggesting increased axonal sprouting by NPC-derived tPA. tPA-mediated regulation of neuronal maturation in NPCs may play an important role during development and in regenerative conditions.
Collapse
|
28
|
Structure-function dissection of the frizzled receptor in Drosophila melanogaster suggests different mechanisms of action in planar polarity and canonical Wnt signaling. Genetics 2012; 192:1295-313. [PMID: 23023003 PMCID: PMC3512140 DOI: 10.1534/genetics.112.144592] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Members of the Frizzled family of sevenpass transmembrane receptors signal via the canonical Wnt pathway and also via noncanonical pathways of which the best characterized is the planar polarity pathway. Activation of both canonical and planar polarity signaling requires interaction between Frizzled receptors and cytoplasmic proteins of the Dishevelled family; however, there has been some dispute regarding whether the Frizzled-Dishevelled interactions are the same in both cases. Studies looking at mutated forms of Dishevelled suggested that stable recruitment of Dishevelled to membranes by Frizzled was required only for planar polarity activity, implying that qualitatively different Frizzled-Dishevelled interactions underlie canonical signaling. Conversely, studies looking at the sequence requirements of Frizzled receptors in the fruit fly Drosophila melanogaster for canonical and planar polarity signaling have concluded that there is most likely a common mechanism of action. To understand better Frizzled receptor function, we have carried out a large-scale mutagenesis in Drosophila to isolate novel mutations in frizzled that affect planar polarity activity and have identified a group of missense mutations in cytosolic-facing regions of the Frizzled receptor that block Dishevelled recruitment. Interestingly, although some of these affect both planar polarity and canonical activity, as previously reported for similar lesions, we find a subset that affect only planar polarity activity. These results support the view that qualitatively different Frizzled-Dishevelled interactions underlie planar polarity and canonical Wnt signaling.
Collapse
|
29
|
Three decades of Wnts: a personal perspective on how a scientific field developed. EMBO J 2012; 31:2670-84. [PMID: 22617420 DOI: 10.1038/emboj.2012.146] [Citation(s) in RCA: 319] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 04/24/2012] [Indexed: 12/13/2022] Open
Abstract
Wnt genes and components of Wnt signalling pathways have been implicated in a wide spectrum of important biological phenomena, ranging from early organismal development to cell behaviours to several diseases, especially cancers. Emergence of the field of Wnt signalling can be largely traced back to the discovery of the first mammalian Wnt gene in 1982. In this essay, we mark the thirtieth anniversary of that discovery by describing some of the critical scientific developments that led to the flowering of this field of research.
Collapse
|
30
|
Kuroda J, Nakamura M, Yoshida M, Yamamoto H, Maeda T, Taniguchi K, Nakazawa N, Hatori R, Ishio A, Ozaki A, Shimaoka S, Ito T, Iida H, Okumura T, Maeda R, Matsuno K. Canonical Wnt signaling in the visceral muscle is required for left-right asymmetric development of the Drosophila midgut. Mech Dev 2011; 128:625-39. [PMID: 22198363 DOI: 10.1016/j.mod.2011.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/24/2011] [Accepted: 12/11/2011] [Indexed: 11/19/2022]
Abstract
Many animals develop left-right (LR) asymmetry in their internal organs. The mechanisms of LR asymmetric development are evolutionarily divergent, and are poorly understood in invertebrates. Therefore, we studied the genetic pathway of LR asymmetric development in Drosophila. Drosophila has several organs that show directional and stereotypic LR asymmetry, including the embryonic gut, which is the first organ to develop LR asymmetry during Drosophila development. In this study, we found that genes encoding components of the Wnt-signaling pathway are required for LR asymmetric development of the anterior part of the embryonic midgut (AMG). frizzled 2 (fz2) and Wnt4, which encode a receptor and ligand of Wnt signaling, respectively, were required for the LR asymmetric development of the AMG. arrow (arr), an ortholog of the mammalian gene encoding low-density lipoprotein receptor-related protein 5/6, which is a co-receptor of the Wnt-signaling pathway, was also essential for LR asymmetric development of the AMG. These results are the first demonstration that Wnt signaling contributes to LR asymmetric development in invertebrates, as it does in vertebrates. The AMG consists of visceral muscle and an epithelial tube. Our genetic analyses revealed that Wnt signaling in the visceral muscle but not the epithelium of the midgut is required for the AMG to develop its normal laterality. Furthermore, fz2 and Wnt4 were expressed in the visceral muscles of the midgut. Consistent with these results, we observed that the LR asymmetric rearrangement of the visceral muscle cells, the first visible asymmetry of the developing AMG, did not occur in embryos lacking Wnt4 expression. Our results also suggest that canonical Wnt/β-catenin signaling, but not non-canonical Wnt signaling, is responsible for the LR asymmetric development of the AMG. Canonical Wnt/β-catenin signaling is reported to have important roles in LR asymmetric development in zebrafish. Thus, the contribution of canonical Wnt/β-catenin signaling to LR asymmetric development may be an evolutionarily conserved feature between vertebrates and invertebrates.
Collapse
Affiliation(s)
- Junpei Kuroda
- Department of Biological Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Liu L, Rao JN, Zou T, Xiao L, Smith A, Zhuang R, Turner DJ, Wang JY. Activation of Wnt3a signaling stimulates intestinal epithelial repair by promoting c-Myc-regulated gene expression. Am J Physiol Cell Physiol 2011; 302:C277-85. [PMID: 21975427 DOI: 10.1152/ajpcell.00341.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In response to mucosal injury, epithelial cells modify the patterns of expressed genes to repair damaged tissue rapidly. Our previous studies have demonstrated that the transcription factor c-Myc is necessary for stimulation of epithelial cell renewal during mucosal healing, but the up-stream signaling initiating c-Myc gene expression after injury remains unknown. Wnts are cysteine-rich glycoproteins that act as short-range ligands to locally activate receptor-mediated signaling pathways and correlate with the increased expression of the c-Myc gene. The current study tested the hypothesis that Wnt3a signaling is implicated in intestinal epithelial repair after wounding by stimulating c-Myc expression. Elevated Wnt3a signaling in intestinal epithelial cells (IEC-6 line) by coculturing with stable Wnt3a-transfected fibroblasts or ectopic overexpression of the Wnt3a gene enhanced intestinal epithelial repair after wounding. This stimulatory effect on epithelial repair was prevented by silencing the Wnt coreceptor LRP6 or by c-Myc silencing. Activation of the Wnt3a signaling pathway increased β-catenin nuclear translocation by decreasing its phosphorylation and stimulated c-Myc expression during epithelial repair after wounding. In stable Wnt3a-transfected IEC-6 cells, increased levels of c-Myc were associated with an increase in expression of c-Myc-regulated genes cyclcin D1 and cyclin E, whereas c-Myc silencing inhibited expression of cyclin D1 and cyclin E and delayed epithelial repair. These results indicate that elevated Wnt3a signaling in intestinal epithelial cells after wounding stimulates epithelial repair by promoting c-Myc-regulated gene expression.
Collapse
Affiliation(s)
- Lan Liu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Yanfeng WA, Berhane H, Mola M, Singh J, Jenny A, Mlodzik M. Functional dissection of phosphorylation of Disheveled in Drosophila. Dev Biol 2011; 360:132-42. [PMID: 21963539 DOI: 10.1016/j.ydbio.2011.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 08/23/2011] [Accepted: 09/14/2011] [Indexed: 01/25/2023]
Abstract
Disheveled/Dsh proteins (Dvl in mammals) are core components of both Wnt/Wg-signaling pathways: canonical β-catenin signaling and Frizzled (Fz)-planar cell polarity (PCP) signaling. Although Dsh is a key cytoplasmic component of both Wnt/Fz-pathways, regulation of its signaling specificity is not well understood. Dsh is phosphorylated, but the functional significance of its phosphorylation remains unclear. We have systematically investigated the phosphorylation of Dsh by combining mass-spectrometry analyses, biochemical studies, and in vivo genetic methods in Drosophila. Our approaches identified multiple phospho-residues of Dsh in vivo. Our data define three novel and unexpected conclusions: (1) strikingly and in contrast to common assumptions, all conserved serines/threonines are non-essential for Dsh function in either pathway; (2) phosphorylation of conserved Tyrosine473 in the DEP domain is critical for PCP-signaling - Dsh(Y473F) behaves like a PCP-specific allele; and (3) defects associated with the PCP specific dsh(1) allele, Dsh(K417M), located within a putative Protein Kinase C consensus site, are likely due to a post-translational modification requirement of Lys417, rather than phosphorylation nearby. In summary, our combined data indicate that while many Ser/Thr and Tyr residues are indeed phosphorylated in vivo, strikingly most of these phosphorylation events are not critical for Dsh function with the exception of DshY473.
Collapse
Affiliation(s)
- Wang A Yanfeng
- Dept. of Developmental & Regenerative Biology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
33
|
Song R, Yosypiv IV. (Pro)renin Receptor in Kidney Development and Disease. Int J Nephrol 2011; 2011:247048. [PMID: 21755055 PMCID: PMC3132641 DOI: 10.4061/2011/247048] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 03/20/2011] [Accepted: 04/20/2011] [Indexed: 12/24/2022] Open
Abstract
The renin-angiotensin system (RAS), a key regulator of the blood pressure and fluid/electrolyte homeostasis, also plays a critical role in kidney development. All the components of the RAS are expressed in the developing metanephros. Moreover, mutations in the genes encoding components of the RAS in mice or humans are associated with a broad spectrum of congenital anomalies of the kidney and urinary tract (CAKUT). These forms of CAKUT include renal papillary hypoplasia, hydronephrosis, duplicated collecting system, renal tubular dysgenesis, renal vascular abnormalities, and aberrant glomerulogenesis. Emerging evidence indicates that (pro)renin receptor (PRR), a novel component of the RAS, is essential for proper kidney development and that aberrant PRR signaling is causally linked to cardiovascular and renal disease. This paper describes the role of the RAS in kidney development and highlights emerging insights into the cellular and molecular mechanisms by which the PRR may regulate this critical morphogenetic process.
Collapse
Affiliation(s)
- Renfang Song
- Section of Pediatric Nephrology, Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | |
Collapse
|
34
|
Yellow submarine of the Wnt/Frizzled signaling: submerging from the G protein harbor to the targets. Biochem Pharmacol 2011; 82:1311-9. [PMID: 21689640 DOI: 10.1016/j.bcp.2011.06.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 05/30/2011] [Accepted: 06/02/2011] [Indexed: 10/18/2022]
Abstract
The Wnt/Frizzled signaling pathway plays multiple functions in animal development and, when deregulated, in human disease. The G-protein coupled receptor (GPCR) Frizzled and its cognate heterotrimeric Gi/o proteins initiate the intracellular signaling cascades resulting in cell fate determination and polarization. In this review, we summarize the knowledge on the ligand recognition, biochemistry, modifications and interacting partners of the Frizzled proteins viewed as GPCRs. We also discuss the effectors of the heterotrimeric Go protein in Frizzled signaling. One group of these effectors is represented by small GTPases of the Rab family, which amplify the initial Wnt/Frizzled signal. Another effector is the negative regulator of Wnt signaling Axin, which becomes deactivated in response to Go action. The discovery of the GPCR properties of Frizzled receptors not only provides mechanistic understanding to their signaling pathways, but also paves new avenues for the drug discovery efforts.
Collapse
|
35
|
Sugimura R, Li L. Noncanonical Wnt signaling in vertebrate development, stem cells, and diseases. ACTA ACUST UNITED AC 2011; 90:243-56. [PMID: 21181886 DOI: 10.1002/bdrc.20195] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Wnt signaling regulates many aspects of vertebrate development and adult stem cells. Deregulation of Wnt signaling causes development defect and cancer. The signaling is categorized in two pathways: canonical and noncanonical. Both pathways are initiated by Wnt ligands and Frizzled receptors. Canonical pathway leads to β-catenin:T-cell factor/lymphoid enhancer factor-mediated gene expression, which regulates proliferation and differentiation of cells. Noncanonical Wnt signaling is mediated by intracellular calcium ion and JNK. This signaling leads to NFAT, a key transcriptional factor regulating gene expression. In addition, β-catenin:T-cell factor/lymphoid enhancer factor-mediated gene expression is downregulated by CaMKII-TAK1-NLK. Cellular polarity and motility are the main outcomes of the signaling. During development, noncanonical Wnt signaling is required for tissue formation. Recent studies have shown that atypical cadherin Flamingo contributes to noncanonical Wnt signaling by directing the migration of cells. Also, noncanonical Wnt signaling is required for maintenance of adult stem cells. In the field of cancer research, noncanonical Wnt signaling has been considered a tumor suppressor; however, recent evidence has shown that the signaling also enhances cancer progression in the later stages of disease. In this review, we describe and discuss components of noncanonical Wnt signaling, diseases caused by deregulation of the signaling, regulation of adult stem cells by the signaling, and implications in cancer biology.
Collapse
Affiliation(s)
- Ryohichi Sugimura
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, Missouri 64110, USA
| | | |
Collapse
|
36
|
Strutt D, Vincent JP. Frizzled signaling: Gαo and Rab5 at the crossroads of the canonical and PCP pathways? Sci Signal 2010; 3:pe43. [PMID: 21081753 DOI: 10.1126/scisignal.3148pe43] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025]
Abstract
Endocytosis modulates the activities of members of a family of receptors known as Frizzled (Fz) proteins, which control canonical β-catenin signaling and the planar cell polarity pathway; however, what determines which of the two pathways is activated is unclear. Evidence now suggests that the guanosine triphosphatase Rab5, a key regulator of endocytosis, binds directly to Fz proteins and that subsequent trafficking steps determine the outcome of Fz signaling.
Collapse
Affiliation(s)
- David Strutt
- Medical Research Council (MRC) Centre for Developmental and Biomedical Genetics, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | | |
Collapse
|
37
|
Niehrs C, Shen J. Regulation of Lrp6 phosphorylation. Cell Mol Life Sci 2010; 67:2551-62. [PMID: 20229235 PMCID: PMC11115861 DOI: 10.1007/s00018-010-0329-3] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 02/08/2010] [Accepted: 02/19/2010] [Indexed: 12/14/2022]
Abstract
The Wnt/beta-catenin signaling pathway plays important roles in embryonic development and tissue homeostasis, and is implicated in human disease. Wnts transduce signals via transmembrane receptors of the Frizzled (Fzd/Fz) family and the low density lipoprotein receptor-related protein 5/6 (Lrp5/6). A key mechanism in their signal transduction is that Wnts induce Lrp6 signalosomes, which become phosphorylated at multiple conserved sites, notably at PPSPXS motifs. Lrp6 phosphorylation is crucial to beta-catenin stabilization and pathway activation by promoting Axin and Gsk3 recruitment to phosphorylated sites. Here, we summarize how proline-directed kinases (Gsk3, PKA, Pftk1, Grk5/6) and non-proline-directed kinases (CK1 family) act upon Lrp6, how the phosphorylation is regulated by ligand binding and mitosis, and how Lrp6 phosphorylation leads to beta-catenin stabilization.
Collapse
Affiliation(s)
- Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 581, Heidelberg, Germany.
| | | |
Collapse
|
38
|
Oh IH. Microenvironmental targeting of Wnt/β-catenin signals for hematopoietic stem cell regulation. Expert Opin Biol Ther 2010; 10:1315-29. [DOI: 10.1517/14712598.2010.504705] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
39
|
Sienknecht UJ, Fekete DM. Mapping of Wnt, frizzled, and Wnt inhibitor gene expression domains in the avian otic primordium. J Comp Neurol 2010; 517:751-64. [PMID: 19842206 DOI: 10.1002/cne.22169] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Wnt signaling activates at least three different pathways involved in development and disease. Interactions of secreted ligands and inhibitors with cell-surface receptors result in the activation or regulation of particular downstream intracellular cascades. During the developmental stages of otic vesicle closure and beginning morphogenesis, the forming inner ear transcribes a plethora of Wnt-related genes. We report expression of 23 genes out of 25 tested in situ hybridization probes on tissue serial sections. Sensory primordia and Frizzled gene expression share domains, with Fzd1 being a continuous marker. Prospective nonsensory domains express Wnts, whose transcripts mainly flank prosensory regions. Finally, Wnt inhibitor domains are superimposed over both prosensory and nonsensory otic regions. Three Wnt antagonists, Dkk1, SFRP2, and Frzb are prominent. Their gene expression patterns partly overlap and change over time, which adds to the diversity of molecular microenvironments. Strikingly, prosensory domains express Wnts transiently. This includes: 1) the prosensory otic region of high proliferation, neuroblast delamination, and programmed cell death at stage 20/21 (Wnt3, -5b, -7b, -8b, -9a, and -11); and 2) sensory primordia at stage 25 (Wnt7a and Wnt9a). In summary, robust Wnt-related gene expression shows both spatial and temporal tuning during inner ear development as the otic vesicle initiates morphogenesis and prosensory cell fate determination.
Collapse
Affiliation(s)
- Ulrike J Sienknecht
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA
| | | |
Collapse
|
40
|
Kim JA, Kang YJ, Park G, Kim M, Park YO, Kim H, Leem SH, Chu IS, Lee JS, Jho EH, Oh IH. Identification of a stroma-mediated Wnt/beta-catenin signal promoting self-renewal of hematopoietic stem cells in the stem cell niche. Stem Cells 2009; 27:1318-29. [PMID: 19489023 DOI: 10.1002/stem.52] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
With contrasting observations on the effects of beta-catenin on hematopoietic stem cells (HSCs), the precise role of Wnt/beta-catenin signals on HSC regulation remains unclear. Here, we show a distinct mode of Wnt/beta-catenin signal that can regulate HSCs in a stroma-dependent manner. Stabilization of beta-catenin in the bone marrow stromal cells promoted maintenance and self-renewal of HSCs in a contact-dependent manner, whereas direct stabilization in hematopoietic cells caused loss of HSCs. Interestingly, canonical Wnt receptors and beta-catenin accumulation were predominantly enriched in the stromal rather than the hematopoietic compartment of bone marrows. Moreover, the active form of beta-catenin accumulated selectively in the trabecular endosteum in "Wnt 3a-stimulated" or "irradiation-stressed," but not in "steady-state" marrows. Notably, notch ligands were induced in Wnt/beta-catenin activated bone marrow stroma and downstream notch signal activation was seen in the HSCs in contact with the activated stroma. Taken together, Wnt/beta-catenin activated stroma and their cross-talk with HSCs may function as a physiologically regulated microenvironmental cue for HSC self-renewal in the stem cell niche.
Collapse
Affiliation(s)
- Jin-A Kim
- Catholic Cell Therapy Center and Department of Cellular Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sinenko SA, Mandal L, Martinez-Agosto JA, Banerjee U. Dual role of wingless signaling in stem-like hematopoietic precursor maintenance in Drosophila. Dev Cell 2009; 16:756-63. [PMID: 19460351 PMCID: PMC2718753 DOI: 10.1016/j.devcel.2009.03.003] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 02/24/2009] [Accepted: 03/03/2009] [Indexed: 12/11/2022]
Abstract
In Drosophila, blood development occurs in a specialized larval hematopoietic organ, the lymph gland (LG), within which stem-like hemocyte precursors or prohemocytes differentiate to multiple blood cell types. Here we show that components of the Wingless (Wg) signaling pathway are expressed in prohemocytes. Loss- and gain-of-function analysis indicates that canonical Wg signaling is required for maintenance of prohemocytes and negatively regulates their differentiation. Wg signals locally in a short-range fashion within different compartments of the LG. In addition, Wg signaling positively regulates the proliferation and maintenance of cells that function as a hematopoietic niche in Drosophila, the posterior signaling center (PSC), and in the proliferation of crystal cells. Our studies reveal a conserved function of Wg signaling in the maintenance of stem-like blood progenitors and reveal an involvement of this pathway in the regulation of hemocyte differentiation through its action in the hematopoietic niche.
Collapse
Affiliation(s)
- Sergey A. Sinenko
- Department of Molecular, Cell and Developmental Biology, Molecular Biology Institute, University of California, Los Angeles; Los Angeles, California 90095, USA
| | - Lolitika Mandal
- Department of Molecular, Cell and Developmental Biology, Molecular Biology Institute, University of California, Los Angeles; Los Angeles, California 90095, USA
| | - Julian A. Martinez-Agosto
- Department of Molecular, Cell and Developmental Biology, Molecular Biology Institute, University of California, Los Angeles; Los Angeles, California 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, California 90095, USA
- Mattel Childrens Hospital UCLA, University of California, Los Angeles; Los Angeles, California 90095, USA
| | - Utpal Banerjee
- Department of Molecular, Cell and Developmental Biology, Molecular Biology Institute, University of California, Los Angeles; Los Angeles, California 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, California 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles; Los Angeles, California 90095, USA
| |
Collapse
|
42
|
Singh AK, Gupta S, Jiang Y, Younus M, Ramzan M. In vitro Neurogenesis from Neural Progenitor Cells Isolated from the Hippocampus Region of the Brain of Adult Rats Exposed to Ethanol during Early Development through Their Alcohol-Drinking Mothers. Alcohol Alcohol 2009; 44:185-98. [DOI: 10.1093/alcalc/agn109] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
43
|
Lin G, Xu N, Xi R. Paracrine Wingless signalling controls self-renewal of Drosophila intestinal stem cells. Nature 2008; 455:1119-23. [PMID: 18806781 DOI: 10.1038/nature07329] [Citation(s) in RCA: 271] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2008] [Accepted: 08/08/2008] [Indexed: 01/05/2023]
Abstract
In the Drosophila midgut, multipotent intestinal stem cells (ISCs) that are scattered along the epithelial basement membrane maintain tissue homeostasis by their ability to steadily produce daughters that differentiate into either enterocytes or enteroendocrine cells, depending on the levels of Notch activity. However, the mechanisms controlling ISC self-renewal remain elusive. Here we show that a canonical Wnt signalling pathway controls ISC self-renewal. The ligand Wingless (Wg) is specifically expressed in the circular muscles next to ISCs, separated by a thin layer of basement membrane. Reduced function of wg causes ISC quiescence and differentiation, whereas wg overexpression produces excessive ISC-like cells that express high levels of the Notch ligand, Delta. Clonal analysis shows that the main downstream components of the Wg pathway, including Frizzled, Dishevelled and Armadillo, are autonomously required for ISC self-renewal. Furthermore, epistatic analysis suggests that Notch acts downstream of the Wg pathway and a hierarchy of Wg/Notch signalling pathways controls the balance between self-renewal and differentiation of ISCs. These data suggest that the underlying circular muscle constitutes the ISC niche, which produce Wg signals that act directly on ISCs to promote ISC self-renewal. This study demonstrates markedly conserved mechanisms regulating ISCs from Drosophila to mammals. The identification of the Drosophila ISC niche and the principal self-renewal signal will facilitate further understanding of intestinal homeostasis control and tumorigenesis.
Collapse
Affiliation(s)
- Guonan Lin
- Graduate program, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | | | | |
Collapse
|
44
|
Burns CJ, Zhang J, Brown EC, Van Bibber AM, Van Es J, Clevers H, Ishikawa TO, Taketo MM, Vetter ML, Fuhrmann S. Investigation of Frizzled-5 during embryonic neural development in mouse. Dev Dyn 2008; 237:1614-26. [PMID: 18489003 PMCID: PMC2562763 DOI: 10.1002/dvdy.21565] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Recent studies revealed that the Wnt receptor Frizzled-5 (Fzd5) is required for eye and retina development in zebrafish and Xenopus, however, its role during mammalian eye development is unknown. In the mouse embryo, Fzd5 is prominently expressed in the pituitary, distal optic vesicle, and optic stalk, then later in the progenitor zone of the developing retina. To elucidate the role of Fzd5 during eye development, we analyzed embryos with a germline disruption of the Fzd5 gene at E10.25, just before embryos die due to defects in yolk sac angiogenesis. We observed severe defects in optic cup morphogenesis and lens development. However, in embryos with conditional inactivation of Fzd5 using Six3-Cre, we observed no obvious early eye defects. Analysis of Axin2 mRNA expression and TCF/LEF-responsive reporter activation demonstrate that Fzd5 does not regulate the Wnt/beta-catenin pathway in the eye. Thus, the function of Fzd5 during eye development appears to be species-dependent.
Collapse
Affiliation(s)
- Carole J. Burns
- Department of Neurobiology and Anatomy, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Jianmin Zhang
- Department of Neurobiology and Anatomy, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Erinn C. Brown
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Alyssa M. Van Bibber
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Johan Van Es
- Netherlands Institute for Developmental Biology, Center for Biomedical Research, Hubrecht Institute, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Hans Clevers
- Netherlands Institute for Developmental Biology, Center for Biomedical Research, Hubrecht Institute, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Tomo-o Ishikawa
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - M. Mark Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Monica L. Vetter
- Department of Neurobiology and Anatomy, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Sabine Fuhrmann
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
45
|
Ma L, Wang HY. Mitogen-activated protein kinase p38 regulates the Wnt/cyclic GMP/Ca2+ non-canonical pathway. J Biol Chem 2007; 282:28980-28990. [PMID: 17684012 DOI: 10.1074/jbc.m702840200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The non-canonical Wnt/cyclic GMP/Ca(2+)/NF-AT pathway operates via Frizzled-2, a member of the superfamily of G protein-coupled receptors. In scanning for signaling events downstream of the Frizzled-2/G alpha t2/PDE6 triad activated in response to Wnt5a, we observed a strong activation of the mitogen-activated protein kinase p38 in mouse F9 teratocarcinoma embryonal cells. The activation of p38 is essential for NF-AT transcriptional activation mediated via Frizzled2. Wnt5a-stimulated p38 activation was rapid, sensitive to pertussis toxin, to siRNA against either G alpha t2 or p38 alpha, and to the p38 inhibitor SB203580. Real-time analysis of intracellular cyclic GMP using the Cygnet2 biosensor revealed p38 to act at the level of cyclic GMP, upstream of the mobilization of intracellular Ca(2+). Fluorescence resonance energy transfer (FRET) imaging reveals the changes in cyclic GMP in response to Wnt5a predominate about the cell membrane, and likewise sensitive to either siRNA targeting p38 or to treatment with SB203580. Dishevelled is not required for Wnt5a activation of p38; siRNAs targeting Dishevelleds and expression of the Dishevelled antagonist Dapper-1 do not suppress the p38 response to Wnt5a stimulation. These novel results are the first to detail a Dishevelled-independent Wnt response, demonstrating a critical role of the mitogen-activated protein kinase p38 in regulating the Wnt non-canonical pathway.
Collapse
Affiliation(s)
- Li Ma
- Departments of Physiology & Biophysics, Diabetes & Metabolic Diseases Research Center, School of Medicine, State University of New York/Stony Brook, Stony Brook, New York 11794-8661
| | - Hsien-Yu Wang
- Departments of Physiology & Biophysics, Diabetes & Metabolic Diseases Research Center, School of Medicine, State University of New York/Stony Brook, Stony Brook, New York 11794-8661.
| |
Collapse
|
46
|
Fischer T, Guimera J, Wurst W, Prakash N. Distinct but redundant expression of the Frizzled Wnt receptor genes at signaling centers of the developing mouse brain. Neuroscience 2007; 147:693-711. [PMID: 17582687 DOI: 10.1016/j.neuroscience.2007.04.060] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 04/05/2007] [Accepted: 04/05/2007] [Indexed: 01/07/2023]
Abstract
The establishment of the regional subdivisions of the vertebrate CNS is accomplished through the activity of different neuroepithelial organizing centers. The wingless/int (Wnt) family of secreted glycoproteins, among other factors, plays a crucial role in signaling from these centers. Wnt1 secreted from the boundary between the mid- and hindbrain, for instance, controls the development of this brain region and of associated neuronal populations. Different Wnts secreted from the caudomedial pallium, the cortical hem, pattern the adjacent hippocampal field. The first step in Wnt signal transduction is binding of the Wnt ligand to its receptors, the seven-pass transmembrane Frizzled proteins. Inactivation of different Frizzled genes in mice have revealed an extensive functional redundancy between these receptors. In order to discriminate between a possible participation of different Frizzled receptors in the transduction of Wnt signals at the mid-/hindbrain boundary and the cortical hem, we have performed a detailed expression study of the 10 known murine Frizzled genes at crucial stages of mouse embryonic development. Our analysis reveals a highly dynamic yet distinct expression pattern of individual Frizzled genes in the anterior neural tube of the developing mouse embryo. The overlapping spatio-temporal expression of at least two and up to six Frizzled genes in any region of the developing mouse brain, however, also suggests a vast functional redundancy of the murine Frizzled receptors. This redundancy has to be taken into consideration for future analyses of Frizzled receptor function at these signaling centers in the mouse.
Collapse
Affiliation(s)
- T Fischer
- GSF-National Research Center for Environment and Health, Technical University Munich, Institute of Developmental Genetics, Munich, Neuherberg, Germany
| | | | | | | |
Collapse
|
47
|
Merkel CE, Karner CM, Carroll TJ. Molecular regulation of kidney development: is the answer blowing in the Wnt? Pediatr Nephrol 2007; 22:1825-38. [PMID: 17554566 PMCID: PMC6949197 DOI: 10.1007/s00467-007-0504-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Revised: 04/10/2007] [Accepted: 04/11/2007] [Indexed: 01/25/2023]
Abstract
Development of the metanephric kidney is a complicated process regulated by reciprocal signals from the ureteric bud and the metanephric mesenchyme that regulate tubule formation and epithelial branching morphogenesis. Over the past several years, several studies have suggested that Wnt signaling is involved in multiple aspects of normal kidney development as well as injury response and cancer progression. We will review these data here.
Collapse
Affiliation(s)
- Calli E. Merkel
- Departments of Internal Medicine (Nephrology) and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8856 USA
| | - Courtney M. Karner
- Departments of Internal Medicine (Nephrology) and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8856 USA
| | - Thomas J. Carroll
- Departments of Internal Medicine (Nephrology) and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8856 USA
| |
Collapse
|
48
|
Smallwood PM, Williams J, Xu Q, Leahy DJ, Nathans J. Mutational analysis of Norrin-Frizzled4 recognition. J Biol Chem 2006; 282:4057-68. [PMID: 17158104 DOI: 10.1074/jbc.m609618200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Norrin and Frizzled4 (Fz4) function as a ligand-receptor pair to control vascular development in the retina and inner ear. In mice and humans, mutations in either of the corresponding genes lead to defects in vascular development. The present work is aimed at defining the sequence determinants of binding specificity between Norrin and the Fz4 amino-terminal ligand-binding domain (the "cysteine-rich domain" (CRD)). The principal conclusions are as follows: 1) Norrin binds to the Fz4 CRD and does not detectably bind to the 14 other mammalian Frizzled and secreted Frizzled-related protein CRDs; 2) Norrin and Xenopus Wnt8 recognize largely overlapping regions of the Fz4 CRD; 3) surface determinants on the Fz4 and Fz8 CRDs that allow Norrin to distinguish between these two CRDs reside within several small regions on one face of the CRD; 4) Norrin function depends critically on three pairs of cysteines that form the highly conserved trio of disulfide bonds shared among all cystine knot proteins, but the remaining two putative disulfide bonds are less important; 5) Norrin-CRD binding depends on a largely contiguous group of amino acids in the extended beta-sheet domain of Norrin that are predicted to face away from the interface between the two monomers in the Norrin homodimer; 6) Norrin-CRD binding is strongly modulated by interactions involving charged amino acid side chains; and 7) Norrin-CRD binding is enhanced approximately 10-fold by the addition of heparin. These observations are discussed in the context of Frizzled signaling and the structure and function of other cystine knot proteins.
Collapse
Affiliation(s)
- Philip M Smallwood
- Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
The fruitfly, Drosophila melanogaster, has been of central importance in analysing the mechanics of cellular processes. Classic forward genetic screens in the fly have identified many of the genes that define critical cell signaling pathways, for example. Our understanding of the Wnt pathway, in particular, has benefited from the many advantages that the fly offers as a model system. Here, I review the history of these discoveries and highlight the utility of the fly in dissecting the molecular workings of Wnt signal transduction.
Collapse
Affiliation(s)
- A Bejsovec
- Department of Biology, Duke University, Durham, NC 27708-1000, USA.
| |
Collapse
|
50
|
Hou R, Liu L, Anees S, Hiroyasu S, Sibinga NES. The Fat1 cadherin integrates vascular smooth muscle cell growth and migration signals. ACTA ACUST UNITED AC 2006; 173:417-29. [PMID: 16682528 PMCID: PMC2063842 DOI: 10.1083/jcb.200508121] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The significance of cadherin superfamily proteins in vascular smooth muscle cell (VSMC) biology is undefined. Here we describe recent studies of the Fat1 protocadherin. Fat1 expression in VSMCs increases significantly after arterial injury or growth factor stimulation. Fat1 knockdown decreases VSMC migration in vitro, but surprisingly, enhances cyclin D1 expression and proliferation. Despite limited similarity to classical cadherins, the Fat1 intracellular domain (Fat1(IC)) interacts with beta-catenin, inhibiting both its nuclear localization and transcriptional activity. Fat1 undergoes cleavage and Fat1(IC) species localize to the nucleus; however, inhibition of the cyclin D1 promoter by truncated Fat1(IC) proteins corresponds to their presence outside the nucleus, which argues against repression of beta-catenin-dependent transcription by nuclear Fat1(IC). These findings extend recent observations about Fat1 and migration in other cell types, and demonstrate for the first time its anti-proliferative activity and interaction with beta-catenin. Because it is induced after arterial injury, Fat1 may control VSMC functions central to vascular remodeling by facilitating migration and limiting proliferation.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Binding Sites/genetics
- Cadherins/genetics
- Cadherins/metabolism
- Cadherins/physiology
- Carotid Arteries/metabolism
- Carotid Artery Injuries/physiopathology
- Cell Line
- Cell Movement/drug effects
- Cell Movement/genetics
- Cell Movement/physiology
- Cell Nucleus/metabolism
- Cell Proliferation
- Cells, Cultured
- Gene Expression/drug effects
- Growth Substances/pharmacology
- Male
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Protein Binding
- RNA, Small Interfering/genetics
- Rats
- Rats, Sprague-Dawley
- Transcription, Genetic/genetics
- Transfection
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Rong Hou
- Department of Medicine (Cardiovascular Division), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|