1
|
Fenner JL, Newberry C, Todd C, Range RC. Anterior-Posterior Wnt Signaling Network Conservation between Indirect Developing Sea Urchin and Hemichordate Embryos. Integr Comp Biol 2024; 64:1214-1225. [PMID: 38769605 PMCID: PMC11579615 DOI: 10.1093/icb/icae047] [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: 02/28/2024] [Revised: 04/28/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024] Open
Abstract
How animal body plans evolved and diversified is a major question in evolutionary developmental biology. To address this question, it is important to characterize the exact molecular mechanisms that establish the major embryonic axes that give rise to the adult animal body plan. The anterior-posterior (AP) axis is the first axis to be established in most animal embryos, and in echinoderm sea urchin embryos its formation is governed by an integrated network of three different Wnt signaling pathways: Wnt/β-catenin, Wnt/JNK, and Wnt/PKC pathways. The extent to which this embryonic patterning mechanism is conserved among deuterostomes, or more broadly in metazoans, is an important open question whose answers could lead to a deeper appreciation of the evolution of the AP axis. Because Ambulacrarians (echinoderms and hemichordates) reside in a key phylogenetic position as the sister group to chordates, studies in these animals can help inform on how chordate body plans may have evolved. Here, we assayed the spatiotemporal gene expression of a subset of sea urchin AP Wnt patterning gene orthologs in the hemichordate, Schizocardium californicum. Our results show that positioning of the anterior neuroectoderm (ANE) to a territory around the anterior pole during early AP formation is spatially and temporally similar between indirect developing hemichordates and sea urchins. Furthermore, we show that the expression of wnt8 and frizzled5/8, two known drivers of ANE patterning in sea urchins, is similar in hemichordate embryos. Lastly, our results highlight divergence in embryonic expression of several early expressed Wnt genes (wnt1, wnt2, and wnt4). These results suggest that expression of the sea urchin AP Wnt signaling network is largely conserved in indirect developing hemichordates setting the foundation for future functional studies in S. californicum.
Collapse
Affiliation(s)
- Jennifer L Fenner
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Callum Newberry
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Callie Todd
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Ryan C Range
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
2
|
Nagy N, Kovacs T, Stavely R, Halasy V, Soos A, Szocs E, Hotta R, Graham H, Goldstein AM. Avian ceca are indispensable for hindgut enteric nervous system development. Development 2021; 148:dev199825. [PMID: 34792104 PMCID: PMC8645208 DOI: 10.1242/dev.199825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/22/2021] [Indexed: 11/20/2022]
Abstract
The enteric nervous system (ENS), which is derived from enteric neural crest cells (ENCCs), represents the neuronal innervation of the intestine. Compromised ENCC migration can lead to Hirschsprung disease, which is characterized by an aganglionic distal bowel. During the craniocaudal migration of ENCCs along the gut, we find that their proliferation is greatest as the ENCC wavefront passes through the ceca, a pair of pouches at the midgut-hindgut junction in avian intestine. Removal of the ceca leads to hindgut aganglionosis, suggesting that they are required for ENS development. Comparative transcriptome profiling of the cecal buds compared with the interceca region shows that the non-canonical Wnt signaling pathway is preferentially expressed within the ceca. Specifically, WNT11 is highly expressed, as confirmed by RNA in situ hybridization, leading us to hypothesize that cecal expression of WNT11 is important for ENCC colonization of the hindgut. Organ cultures using embryonic day 6 avian intestine show that WNT11 inhibits enteric neuronal differentiation. These results reveal an essential role for the ceca during hindgut ENS formation and highlight an important function for non-canonical Wnt signaling in regulating ENCC differentiation.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Tamas Kovacs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Rhian Stavely
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| | - Viktoria Halasy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Adam Soos
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Emoke Szocs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Ryo Hotta
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| | - Hannah Graham
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| |
Collapse
|
3
|
Frizzled 7 Activates β-Catenin-Dependent and β-Catenin-Independent Wnt Signalling Pathways During Developmental Morphogenesis: Implications for Therapeutic Targeting in Colorectal Cancer. Handb Exp Pharmacol 2021. [PMID: 34455486 DOI: 10.1007/164_2021_524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
Frizzled7 activates β-catenin-dependent and β-catenin-independent Wnt signalling pathways, is highly conserved through evolution from the ancient phylum hydra to man, plays essential roles in stem cells, tissue homeostasis and regeneration in the adult, and is upregulated in diverse cancers. Much of what is known about the core components of the Wnt signalling pathways was derived from studying the function of Frizzled7 orthologues in the development of lower organism. As we interrogate Frizzled7 signalling and function for therapeutic targeting in cancer, it is timely to revisit lower organisms to gain insight into the context dependent and dynamic nature of Wnt signalling for effective drug design.
Collapse
|
4
|
Alrefaei AF, Münsterberg AE, Wheeler GN. Expression analysis of chick Frizzled receptors during spinal cord development. Gene Expr Patterns 2021; 39:119167. [PMID: 33460819 DOI: 10.1016/j.gep.2021.119167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 11/29/2022]
Abstract
Frizzleds (Fzds) are transmembrane receptors that can transduce signals dependent upon binding of Wnts, a large family of secreted glycoproteins homologous to the Drosophila wingless gene. FZDs are critical for a wide variety of normal and pathological developmental processes. In the nervous system, Wnts and Frizzleds play an important role in anterior-posterior patterning, cell fate decisions, proliferation, and synaptogenesis. Here, we preformed a comprehensive expression profile of Wnt receptors (FZD) by using situ hybridization to identify FZDs that are expressed in dorsal-ventral regions of the neural tube development. Our data show specific expression for FZD1,2,3,7,9 and 10 in the chick developing spinal cord. This expression profile of cFZD receptors offers the basis for functional studies in the future to determine roles for the different FZD receptors and their interactions with Wnts during dorsal-ventral neural tube development in vivo. Furthermore, we also show that co-overexpression of Wnt1/3a by in vivo electroporation affects FZD7/10 expression in the neural tube. This illustrates an example of Wnts-FZDs interactions during spinal cord neurogenesis.
Collapse
Affiliation(s)
| | - Andrea E Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
5
|
Koni M, Pinnarò V, Brizzi MF. The Wnt Signalling Pathway: A Tailored Target in Cancer. Int J Mol Sci 2020; 21:E7697. [PMID: 33080952 PMCID: PMC7589708 DOI: 10.3390/ijms21207697] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the greatest public health challenges. According to the World Health Organization (WHO), 9.6 million cancer deaths have been reported in 2018. The most common cancers include lung, breast, colorectal, prostate, skin (non-melanoma) and stomach cancer. The unbalance of physiological signalling pathways due to the acquisition of mutations in tumour cells is considered the most common cancer driver. The Wingless-related integration site (Wnt)/β-catenin pathway is crucial for tissue development and homeostasis in all animal species and its dysregulation is one of the most relevant events linked to cancer development and dissemination. The canonical and the non-canonical Wnt/β-catenin pathways are known to control both physiological and pathological processes, including cancer. Herein, the impact of the Wnt/β-catenin cascade in driving cancers from different origin has been examined. Finally, based on the impact of Extracellular Vesicles (EVs) on tumour growth, invasion and chemoresistance, and their role as tumour diagnostic and prognostic tools, an overview of the current knowledge linking EVs to the Wnt/β-catenin pathway is also discussed.
Collapse
Affiliation(s)
| | | | - Maria Felice Brizzi
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy; (M.K.); (V.P.)
| |
Collapse
|
6
|
Kratzer MC, Becker SFS, Grund A, Merks A, Harnoš J, Bryja V, Giehl K, Kashef J, Borchers A. The Rho guanine nucleotide exchange factor Trio is required for neural crest cell migration and interacts with Dishevelled. Development 2020; 147:dev.186338. [PMID: 32366678 DOI: 10.1242/dev.186338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/08/2020] [Indexed: 01/31/2023]
Abstract
Directional migration during embryogenesis and tumor progression faces the challenge that numerous external signals need to converge to precisely control cell movement. The Rho guanine exchange factor (GEF) Trio is especially well suited to relay signals, as it features distinct catalytic domains to activate Rho GTPases. Here, we show that Trio is required for Xenopus cranial neural crest (NC) cell migration and cartilage formation. Trio cell-autonomously controls protrusion formation of NC cells and Trio morphant NC cells show a blebbing phenotype. Interestingly, the Trio GEF2 domain is sufficient to rescue protrusion formation and migration of Trio morphant NC cells. We show that this domain interacts with the DEP/C-terminus of Dishevelled (DVL). DVL - but not a deletion construct lacking the DEP domain - is able to rescue protrusion formation and migration of Trio morphant NC cells. This is likely mediated by activation of Rac1, as we find that DVL rescues Rac1 activity in Trio morphant embryos. Thus, our data provide evidence for a novel signaling pathway, whereby Trio controls protrusion formation of cranial NC cells by interacting with DVL to activate Rac1.
Collapse
Affiliation(s)
- Marie-Claire Kratzer
- Philipps-Universität Marburg, Faculty of Biology, Molecular Embryology, 35043 Marburg, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg, Germany
| | - Sarah F S Becker
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104/INSERM U1258, Université de Strasbourg, F-67400 Illkirch, CU Strasbourg, France
| | - Anita Grund
- Philipps-Universität Marburg, Faculty of Biology, Molecular Embryology, 35043 Marburg, Germany
| | - Anne Merks
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Jakub Harnoš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 62500, Czech Republic
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 62500, Czech Republic.,Department of Cytokinetics, Institute of Biophysics of the Academy of Sciences of the Czech Republic v.v.i., Brno 61265, Czech Republic
| | - Klaudia Giehl
- Signal Transduction of Cellular Motility, Internal Medicine V, Justus Liebig University Giessen, D-35392 Giessen, Germany
| | - Jubin Kashef
- Institute for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology (KIT), 76344 Eggenstein-Leopoldshafen, Germany
| | - Annette Borchers
- Philipps-Universität Marburg, Faculty of Biology, Molecular Embryology, 35043 Marburg, Germany .,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
7
|
Bertke MM, Dubiak KM, Cronin L, Zeng E, Huber PW. A deficiency in SUMOylation activity disrupts multiple pathways leading to neural tube and heart defects in Xenopus embryos. BMC Genomics 2019; 20:386. [PMID: 31101013 PMCID: PMC6525467 DOI: 10.1186/s12864-019-5773-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 05/03/2019] [Indexed: 02/08/2023] Open
Abstract
Background Adenovirus protein, Gam1, triggers the proteolytic destruction of the E1 SUMO-activating enzyme. Microinjection of an empirically determined amount of Gam1 mRNA into one-cell Xenopus embryos can reduce SUMOylation activity to undetectable, but nonlethal, levels, enabling an examination of the role of this post-translational modification during early vertebrate development. Results We find that SUMOylation-deficient embryos consistently exhibit defects in neural tube and heart development. We have measured differences in gene expression between control and embryos injected with Gam1 mRNA at three developmental stages: early gastrula (immediately following the initiation of zygotic transcription), late gastrula (completion of the formation of the three primary germ layers), and early neurula (appearance of the neural plate). Although changes in gene expression are widespread and can be linked to many biological processes, three pathways, non-canonical Wnt/PCP, snail/twist, and Ets-1, are especially sensitive to the loss of SUMOylation activity and can largely account for the predominant phenotypes of Gam1 embryos. SUMOylation appears to generate different pools of a given transcription factor having different specificities with this post-translational modification involved in the regulation of more complex, as opposed to housekeeping, processes. Conclusions We have identified changes in gene expression that underlie the neural tube and heart phenotypes resulting from depressed SUMOylation activity. Notably, these developmental defects correspond to the two most frequently occurring congenital birth defects in humans, strongly suggesting that perturbation of SUMOylation, either globally or of a specific protein, may frequently be the origin of these pathologies. Electronic supplementary material The online version of this article (10.1186/s12864-019-5773-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michelle M Bertke
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA.,Present Address: College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Kyle M Dubiak
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Laura Cronin
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Erliang Zeng
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana, 46556, USA.,Present Address: Division of Biostatistics and Computational Biology, Iowa Institute for Oral Health Research, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Preventive & Community Dentistry, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Biostatistics, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA
| | - Paul W Huber
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA. .,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana, USA. .,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, 46556, USA.
| |
Collapse
|
8
|
Ye Z, Xu J, Feng X, Jia Y, Fu Z, Hong Y, Li H, Lu K, Lin J, Song M, Wang L, Yuan C. Spatiotemporal expression pattern of Sjfz7 and its expression comparison with other frizzled family genes in developmental stages of Schistosoma japonicum. Gene Expr Patterns 2019; 32:44-52. [PMID: 30851426 DOI: 10.1016/j.gep.2019.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/19/2019] [Accepted: 02/26/2019] [Indexed: 12/27/2022]
Abstract
Wnts are secreted signaling molecules that are implicated in a variety of growth-related processes. Frizzled proteins have been identified as receptors for Wnt ligands in vertebrates and invertebrates, but a functional role for dioecious flatworm Frizzleds has not been determined. To evaluate the endogenous role of Frizzled proteins during development, we have identified and characterized a Schistosoma japonicum frizzled gene (Sjfz7). We found that Sjfz7 encodes a 698 amino acid protein with typical characteristics of Frizzled proteins. The immunohistochemical localization pattern showed that Sjfz7 protein was extensively distributed in almost all tissues of S. japonicum, including subtegumental muscle cells, parenchymal cells, intestinal epithelial cells and male and female germ cells. This indicated that Sjfz7-mediated Wnt signaling might be associated with the development of musculature, intestinal tract and reproductive organs in schistosome. Comparing mRNA levels between frizzled family members showed that Sjfz7 mRNA was consistently higher in the developmental stages analyzed, suggesting that Sjfz7 may be responsible for more functional tasks than other frizzled family members. Comparing frizzled mRNA levels between not fully developed and normal worms suggested that Wnt signaling might be abnormal in not fully developed worms.
Collapse
Affiliation(s)
- Zhongxue Ye
- Northeast Agricultural University, College of Veterinary Medicine, Harbin, 150030, China
| | - Jingxiu Xu
- Northeast Agricultural University, College of Veterinary Medicine, Harbin, 150030, China
| | - Xingang Feng
- Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Yingying Jia
- Jilin Agricultural University, College of Animal Science and Technology, Changchun, 130118, China
| | - Zhiqiang Fu
- Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Yang Hong
- Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Hao Li
- Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Ke Lu
- Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Jiaojiao Lin
- Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Mingxin Song
- Northeast Agricultural University, College of Veterinary Medicine, Harbin, 150030, China
| | - Liqun Wang
- Northeast Agricultural University, College of Veterinary Medicine, Harbin, 150030, China
| | - Chunxiu Yuan
- Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China.
| |
Collapse
|
9
|
Abstract
Wnt signalling regulates cardiogenesis during specification of heart tissue and the morphogenetic movements necessary to form the linear heart. Wnt11-mediated non-canonical signalling promotes early cardiac development whilst Wnt11-R, which is expressed later, also signals through the non-canonical pathway to promote heart development. It is unclear which Frizzled proteins mediate these interactions. Frizzled-7 (fzd7) is expressed during gastrulation in the mesodermal cells fated to become heart, and then in the primary heart field. This expression is complementary to the expression of wnt11 and wnt11-R. We further show co-localisation of fzd7 with other early- and late-heart-specific markers using double in situ hybridisation. We have used loss of function analysis to determine the role of fzd7 during heart development. Morpholino antisense oligonucleotide-mediated knockdown of Fzd7 results in effects on heart development, similar to that caused by Wnt11 loss of function. Surprisingly, overexpression of dominant-negative Fzd7 cysteine rich domain (Fzd7 CRD) results in a cardia bifida phenotype, similar to the loss of wnt11-R phenotype. Overexpression of Fzd7 and activation of non-canonical wnt signalling can rescue the effect of Fzd7 CRD. We propose that Fzd7 has an important role during Xenopus heart development. Summary: Wnt signalling has been shown to be important in heart development. Here, we demonstrate that the wnt receptor fzd7 is required in mediating these Wnt signals.
Collapse
Affiliation(s)
- Muhammad Abu-Elmagd
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216 Jeddah 21589, Kingdom of Saudi Arabia.,School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Joanna Mulvaney
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
10
|
VanderVorst K, Hatakeyama J, Berg A, Lee H, Carraway KL. Cellular and molecular mechanisms underlying planar cell polarity pathway contributions to cancer malignancy. Semin Cell Dev Biol 2017; 81:78-87. [PMID: 29107170 DOI: 10.1016/j.semcdb.2017.09.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 07/20/2017] [Accepted: 09/06/2017] [Indexed: 12/18/2022]
Abstract
While the mutational activation of oncogenes drives tumor initiation and growth by promoting cellular transformation and proliferation, increasing evidence suggests that the subsequent re-engagement of largely dormant developmental pathways contributes to cellular phenotypes associated with the malignancy of solid tumors. Genetic studies from a variety of model organisms have defined many of the components that maintain epithelial planar cell polarity (PCP), or cellular polarity in the axis orthogonal to the apical-basal axis. These same components comprise an arm of non-canonical Wnt signaling that mediates cell motility events such as convergent extension movements essential to proper development. In this review, we summarize the increasing evidence that the Wnt/PCP signaling pathway plays active roles in promoting the proliferative and migratory properties of tumor cells, emphasizing the importance of subcellular localization of PCP components and protein-protein interactions in regulating cellullar properties associated with malignancy. Specifically, we discuss the increased expression of Wnt/PCP pathway components in cancer and the functional consequences of aberrant pathway activation, focusing on Wnt ligands, Frizzled (Fzd) receptors, the tetraspanin-like proteins Vangl1 and Vangl2, and the Prickle1 (Pk1) scaffold protein. In addition, we discuss negative regulation of the Wnt/PCP pathway, with particular emphasis on the Nrdp1 E3 ubiquitin ligase. We hypothesize that engagement of the Wnt/PCP pathway after tumor initiation drives malignancy by promoting cellular proliferation and invasiveness, and that the ability of Wnt/PCP signaling to supplant oncogene addiction may contribute to tumor resistance to oncogenic pathway-directed therapeutic agents.
Collapse
Affiliation(s)
- Kacey VanderVorst
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, United States
| | - Jason Hatakeyama
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, United States
| | - Anastasia Berg
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, United States
| | - Hyun Lee
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, United States
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, United States.
| |
Collapse
|
11
|
Xiao Q, Chen Z, Jin X, Mao R, Chen Z. The many postures of noncanonical Wnt signaling in development and diseases. Biomed Pharmacother 2017. [PMID: 28651237 DOI: 10.1016/j.biopha.2017.06.061] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Wnt signaling regulates many aspects of vertebrate development. Its dysregulation causes developmental defects and diseases including cancer. The signaling can be categorized in two pathways: canonical and noncanonical. Canonical pathway plays a key role in regulating proliferation and differentiation of cells whilst noncanonical Wnt signaling mainly controls cellular polarity and motility. During development, noncanonical Wnt signaling is required for tissue formation. Recent studies have shown that noncanonical Wnt signaling is involved in adult tissue development and cancer progression. In this review, we try to describe and discuss the mechanisms behind the biological effects of noncanonical Wnt signaling, diseases caused by its dysregulation, and implications in adult tissue development biology.
Collapse
Affiliation(s)
- Qian Xiao
- Senior Research Scientist, Department of Pharmacology, School of Medicine, Yale University, New Haven, USA
| | - Zhengxi Chen
- PhD, Department of Orthodontics, Ninth People's Hospital, School of Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaozhuang Jin
- PhD, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Runyi Mao
- MDS student, Department of Oral and Maxillofacial Surgery, Ninth People's Hospital, School of Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenqi Chen
- Professor, Department of Orthodontics, Ninth People's Hospital, School of Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
12
|
Ulmer B, Tingler M, Kurz S, Maerker M, Andre P, Mönch D, Campione M, Deißler K, Lewandoski M, Thumberger T, Schweickert A, Fainsod A, Steinbeißer H, Blum M. A novel role of the organizer gene Goosecoid as an inhibitor of Wnt/PCP-mediated convergent extension in Xenopus and mouse. Sci Rep 2017; 7:43010. [PMID: 28220837 PMCID: PMC5318956 DOI: 10.1038/srep43010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/18/2017] [Indexed: 12/12/2022] Open
Abstract
Goosecoid (Gsc) expression marks the primary embryonic organizer in vertebrates and beyond. While functions have been assigned during later embryogenesis, the role of Gsc in the organizer has remained enigmatic. Using conditional gain-of-function approaches in Xenopus and mouse to maintain Gsc expression in the organizer and along the axial midline, neural tube closure defects (NTDs) arose and dorsal extension was compromised. Both phenotypes represent convergent extension (CE) defects, arising from impaired Wnt/planar cell polarity (PCP) signaling. Dvl2 recruitment to the cell membrane was inhibited by Gsc in Xenopus animal cap assays and key Wnt/PCP factors (RhoA, Vangl2, Prickle, Wnt11) rescued Gsc-mediated NTDs. Re-evaluation of endogenous Gsc functions in MO-mediated gene knockdown frog and knockout mouse embryos unearthed PCP/CE-related phenotypes as well, including cartilage defects in Xenopus and misalignment of inner ear hair cells in mouse. Our results assign a novel function to Gsc as an inhibitor of Wnt/PCP-mediated CE. We propose that in the organizer Gsc represses CE as well: Gsc-expressing prechordal cells, which leave the organizer first, migrate and do not undergo CE like the Gsc-negative notochordal cells, which subsequently emerge from the organizer. In this model, Gsc provides a switch between cell migration and CE, i.e. cell intercalation.
Collapse
Affiliation(s)
- Bärbel Ulmer
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Melanie Tingler
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Sabrina Kurz
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Markus Maerker
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Philipp Andre
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Dina Mönch
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Marina Campione
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Kirsten Deißler
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Mark Lewandoski
- Genetics of Vertebrate Development Section, Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | | | - Axel Schweickert
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University, Jerusalem 9112102, Israel
| | - Herbert Steinbeißer
- Institute of Human Genetics, University Hospital Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Martin Blum
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
13
|
Mazzotta S, Neves C, Bonner RJ, Bernardo AS, Docherty K, Hoppler S. Distinctive Roles of Canonical and Noncanonical Wnt Signaling in Human Embryonic Cardiomyocyte Development. Stem Cell Reports 2016; 7:764-776. [PMID: 27641648 PMCID: PMC5063467 DOI: 10.1016/j.stemcr.2016.08.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 08/11/2016] [Accepted: 08/11/2016] [Indexed: 11/27/2022] Open
Abstract
Wnt signaling is a key regulator of vertebrate heart development; however, specific roles for human cardiomyocyte development remain uncertain. Here we use human embryonic stem cells (hESCs) to analyze systematically in human cardiomyocyte development the expression of endogenous Wnt signaling components, monitor pathway activity, and dissect stage-specific requirements for canonical and noncanonical Wnt signaling mechanisms using small-molecule inhibitors. Our analysis suggests that WNT3 and WNT8A, via FZD7 and canonical signaling, regulate BRACHYURY expression and mesoderm induction; that WNT5A/5B, via ROR2 and noncanonical signaling, regulate MESP1 expression and cardiovascular development; and that later in development WNT2, WNT5A/5B, and WNT11, via FZD4 and FZD6, regulate functional cardiomyocyte differentiation via noncanonical Wnt signaling. Our findings confirm in human development previously proposed roles for canonical Wnt signaling in sequential stages of vertebrate cardiomyogenesis, and identify more precise roles for noncanonical signaling and for individual Wnt signal and Wnt receptor genes in human cardiomyocyte development. hESCs were used to study Wnt signaling during human cardiomyocyte development Previously proposed roles for canonical Wnt signaling were confirmed in human Specific roles for noncanonical Wnt signaling were identified in cardiomyogenesis Individual Wnt signal and receptor genes were identified in human cardiomyogenesis
Collapse
Affiliation(s)
- Silvia Mazzotta
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Carlos Neves
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Rory J Bonner
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Andreia S Bernardo
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK; Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, West Forvie Building, Robinson Way, Cambridge CB2 0SZ, UK
| | - Kevin Docherty
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Stefan Hoppler
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
14
|
Frizzled7: A Promising Achilles' Heel for Targeting the Wnt Receptor Complex to Treat Cancer. Cancers (Basel) 2016; 8:cancers8050050. [PMID: 27196929 PMCID: PMC4880867 DOI: 10.3390/cancers8050050] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
Frizzled7 is arguably the most studied member of the Frizzled family, which are the cognate Wnt receptors. Frizzled7 is highly conserved through evolution, from Hydra through to humans, and is expressed in diverse organisms, tissues and human disease contexts. Frizzled receptors can homo- or hetero-polymerise and associate with several co-receptors to transmit Wnt signalling. Notably, Frizzled7 can transmit signalling via multiple Wnt transduction pathways and bind to several different Wnt ligands, Frizzled receptors and co-receptors. These promiscuous binding and functional properties are thought to underlie the pivotal role Frizzled7 plays in embryonic developmental and stem cell function. Recent studies have identified that Frizzled7 is upregulated in diverse human cancers, and promotes proliferation, progression and invasion, and orchestrates cellular transitions that underscore cancer metastasis. Importantly, Frizzled7 is able to regulate Wnt signalling activity even in cancer cells which have mutations to down-stream signal transducers. In this review we discuss the various aspects of Frizzled7 signalling and function, and the implications these have for therapeutic targeting of Frizzled7 in cancer.
Collapse
|
15
|
Brinkmann EM, Mattes B, Kumar R, Hagemann AIH, Gradl D, Scholpp S, Steinbeisser H, Kaufmann LT, Özbek S. Secreted Frizzled-related Protein 2 (sFRP2) Redirects Non-canonical Wnt Signaling from Fz7 to Ror2 during Vertebrate Gastrulation. J Biol Chem 2016; 291:13730-42. [PMID: 27129770 DOI: 10.1074/jbc.m116.733766] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Indexed: 02/04/2023] Open
Abstract
Convergent extension movements during vertebrate gastrulation require a balanced activity of non-canonical Wnt signaling pathways, but the factors regulating this interplay on the molecular level are poorly characterized. Here we show that sFRP2, a member of the secreted frizzled-related protein (sFRP) family, is required for morphogenesis and papc expression during Xenopus gastrulation. We further provide evidence that sFRP2 redirects non-canonical Wnt signaling from Frizzled 7 (Fz7) to the receptor tyrosine kinase-like orphan receptor 2 (Ror2). During this process, sFRP2 promotes Ror2 signal transduction by stabilizing Wnt5a-Ror2 complexes at the membrane, whereas it inhibits Fz7 signaling, probably by blocking Fz7 receptor endocytosis. The cysteine-rich domain of sFRP2 is sufficient for Ror2 activation, and related sFRPs can substitute for this function. Notably, direct interaction of the two receptors via their cysteine-rich domains also promotes Ror2-mediated papc expression but inhibits Fz7 signaling. We propose that sFRPs can act as a molecular switch, channeling the signal input for different non-canonical Wnt pathways during vertebrate gastrulation.
Collapse
Affiliation(s)
- Eva-Maria Brinkmann
- From the Institute of Human Genetics, Department of Developmental Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Benjamin Mattes
- the Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, 76344 Karlsruhe, Germany
| | - Rahul Kumar
- From the Institute of Human Genetics, Department of Developmental Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Anja I H Hagemann
- the Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, 76344 Karlsruhe, Germany
| | - Dietmar Gradl
- the Zoological Institute, Department of Cell and Developmental Biology, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany, and
| | - Steffen Scholpp
- the Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, 76344 Karlsruhe, Germany
| | - Herbert Steinbeisser
- From the Institute of Human Genetics, Department of Developmental Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Lilian T Kaufmann
- From the Institute of Human Genetics, Department of Developmental Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany,
| | - Suat Özbek
- the Centre of Organismal Studies, Department of Molecular Evolution and Genomics, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Beaton H, Andrews D, Parsons M, Murphy M, Gaffney A, Kavanagh D, McKay GJ, Maxwell AP, Taylor CT, Cummins EP, Godson C, Higgins DF, Murphy P, Crean J. Wnt6 regulates epithelial cell differentiation and is dysregulated in renal fibrosis. Am J Physiol Renal Physiol 2016; 311:F35-45. [PMID: 27122540 DOI: 10.1152/ajprenal.00136.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/22/2016] [Indexed: 02/07/2023] Open
Abstract
Diabetic nephropathy is the most common microvascular complication of diabetes mellitus, manifesting as mesangial expansion, glomerular basement membrane thickening, glomerular sclerosis, and progressive tubulointerstitial fibrosis leading to end-stage renal disease. Here we describe the functional characterization of Wnt6, whose expression is progressively lost in diabetic nephropathy and animal models of acute tubular injury and renal fibrosis. We have shown prominent Wnt6 and frizzled 7 (FzD7) expression in the mesonephros of the developing mouse kidney, suggesting a role for Wnt6 in epithelialization. Importantly, TCF/Lef reporter activity is also prominent in the mesonephros. Analysis of Wnt family members in human renal biopsies identified differential expression of Wnt6, correlating with severity of the disease. In animal models of tubular injury and fibrosis, loss of Wnt6 was evident. Wnt6 signals through the canonical pathway in renal epithelial cells as evidenced by increased phosphorylation of GSK3β (Ser9), nuclear accumulation of β-catenin and increased TCF/Lef transcriptional activity. FzD7 was identified as a putative receptor of Wnt6. In vitro Wnt6 expression leads to de novo tubulogenesis in renal epithelial cells grown in three-dimensional culture. Importantly, Wnt6 rescued epithelial cell dedifferentiation in response to transforming growth factor-β (TGF-β); Wnt6 reversed TGF-β-mediated increases in vimentin and loss of epithelial phenotype. Wnt6 inhibited TGF-β-mediated p65-NF-κB nuclear translocation, highlighting cross talk between the two pathways. The critical role of NF-κB in the regulation of vimentin expression was confirmed in both p65(-/-) and IKKα/β(-/-) embryonic fibroblasts. We propose that Wnt6 is involved in epithelialization and loss of Wnt6 expression contributes to the pathogenesis of renal fibrosis.
Collapse
Affiliation(s)
- Hayley Beaton
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Darrell Andrews
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Martin Parsons
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Mary Murphy
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Andrew Gaffney
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - David Kavanagh
- Nephrology Research Group, Centre for Public Health, Queens University Belfast, Royal Victoria Hospital, Belfast, United Kingdom; and
| | - Gareth J McKay
- Nephrology Research Group, Centre for Public Health, Queens University Belfast, Royal Victoria Hospital, Belfast, United Kingdom; and
| | - Alexander P Maxwell
- Nephrology Research Group, Centre for Public Health, Queens University Belfast, Royal Victoria Hospital, Belfast, United Kingdom; and
| | - Cormac T Taylor
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Eoin P Cummins
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Catherine Godson
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Debra F Higgins
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Paula Murphy
- Zoology Department, School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | - John Crean
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland;
| |
Collapse
|
17
|
Podleschny M, Grund A, Berger H, Rollwitz E, Borchers A. A PTK7/Ror2 Co-Receptor Complex Affects Xenopus Neural Crest Migration. PLoS One 2015; 10:e0145169. [PMID: 26680417 PMCID: PMC4683079 DOI: 10.1371/journal.pone.0145169] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 11/30/2015] [Indexed: 12/15/2022] Open
Abstract
Neural crest cells are a highly migratory pluripotent cell population that generates a wide array of different cell types and failure in their migration can result in severe birth defects and malformation syndromes. Neural crest migration is controlled by various means including chemotaxis, repellent guidance cues and cell-cell interaction. Non-canonical Wnt PCP (planar cell polarity) signaling has previously been shown to control cell-contact mediated neural crest cell guidance. PTK7 (protein tyrosine kinase 7) is a transmembrane pseudokinase and a known regulator of Wnt/PCP signaling, which is expressed in Xenopus neural crest cells and required for their migration. PTK7 functions as a Wnt co-receptor; however, it remains unclear by which means PTK7 affects neural crest migration. Expressing fluorescently labeled proteins in Xenopus neural crest cells we find that PTK7 co-localizes with the Ror2 Wnt-receptor. Further, co-immunoprecipitation experiments demonstrate that PTK7 interacts with Ror2. The PTK7/Ror2 interaction is likely relevant for neural crest migration, because Ror2 expression can rescue the PTK7 loss of function migration defect. Live cell imaging of explanted neural crest cells shows that PTK7 loss of function affects the formation of cell protrusions as well as cell motility. Co-expression of Ror2 can rescue these defects. In vivo analysis demonstrates that a kinase dead Ror2 mutant cannot rescue PTK7 loss of function. Thus, our data suggest that Ror2 can substitute for PTK7 and that the signaling function of its kinase domain is required for this effect.
Collapse
Affiliation(s)
- Martina Podleschny
- Faculty of Biology, Molecular Embryology, Philipps-Universität Marburg, 35043 Marburg, Germany
| | - Anita Grund
- Faculty of Biology, Molecular Embryology, Philipps-Universität Marburg, 35043 Marburg, Germany
| | - Hanna Berger
- Faculty of Biology, Molecular Embryology, Philipps-Universität Marburg, 35043 Marburg, Germany
| | - Erik Rollwitz
- Faculty of Biology, Molecular Embryology, Philipps-Universität Marburg, 35043 Marburg, Germany
| | - Annette Borchers
- Faculty of Biology, Molecular Embryology, Philipps-Universität Marburg, 35043 Marburg, Germany
- * E-mail:
| |
Collapse
|
18
|
Vitorino M, Silva AC, Inácio JM, Ramalho JS, Gur M, Fainsod A, Steinbeisser H, Belo JA. Xenopus Pkdcc1 and Pkdcc2 Are Two New Tyrosine Kinases Involved in the Regulation of JNK Dependent Wnt/PCP Signaling Pathway. PLoS One 2015; 10:e0135504. [PMID: 26270962 PMCID: PMC4536202 DOI: 10.1371/journal.pone.0135504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 07/22/2015] [Indexed: 12/28/2022] Open
Abstract
Protein Kinase Domain Containing, Cytoplasmic (PKDCC) is a protein kinase which has been implicated in longitudinal bone growth through regulation of chondrocytes formation. Nevertheless, the mechanism by which this occurs remains unknown. Here, we identified two new members of the PKDCC family, Pkdcc1 and Pkdcc2 from Xenopus laevis. Interestingly, our knockdown experiments revealed that these two proteins are both involved on blastopore and neural tube closure during gastrula and neurula stages, respectively. In vertebrates, tissue polarity and cell movement observed during gastrulation and neural tube closure are controlled by Wnt/Planar Cell Polarity (PCP) molecular pathway. Our results showed that Pkdcc1 and Pkdcc2 promote the recruitment of Dvl to the plasma membrane. But surprisingly, they revealed different roles in the induction of a luciferase reporter under the control of Atf2 promoter. While Pkdcc1 induces Atf2 expression, Pkdcc2 does not, and furthermore inhibits its normal induction by Wnt11 and Wnt5a. Altogether our data show, for the first time, that members of the PKDCC family are involved in the regulation of JNK dependent Wnt/PCP signaling pathway.
Collapse
Affiliation(s)
- Marta Vitorino
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- Center for Biomedical Research (CBMR), Universidade do Algarve, Campus de Gambelas, Faro, Portugal
| | - Ana Cristina Silva
- Center for Biomedical Research (CBMR), Universidade do Algarve, Campus de Gambelas, Faro, Portugal
| | - José Manuel Inácio
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- Center for Biomedical Research (CBMR), Universidade do Algarve, Campus de Gambelas, Faro, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - José Silva Ramalho
- CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Michal Gur
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, P.O. Box 12272, Jerusalem, 91120, Israel
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, P.O. Box 12272, Jerusalem, 91120, Israel
| | | | - José António Belo
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- Center for Biomedical Research (CBMR), Universidade do Algarve, Campus de Gambelas, Faro, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
- * E-mail:
| |
Collapse
|
19
|
Song JL, Nigam P, Tektas SS, Selva E. microRNA regulation of Wnt signaling pathways in development and disease. Cell Signal 2015; 27:1380-91. [PMID: 25843779 PMCID: PMC4437805 DOI: 10.1016/j.cellsig.2015.03.018] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 12/19/2022]
Abstract
Wnt signaling pathways and microRNAs (miRNAs) are critical regulators of development. Aberrant Wnt signaling pathways and miRNA levels lead to developmental defects and diverse human pathologies including but not limited to cancer. Wnt signaling pathways regulate a plethora of cellular processes during embryonic development and maintain homeostasis of adult tissues. A majority of Wnt signaling components are regulated by miRNAs which are small noncoding RNAs that are expressed in both animals and plants. In animal cells, miRNAs fine tune gene expression by pairing primarily to the 3'untranslated region of protein coding mRNAs to repress target mRNA translation and/or induce target degradation. miRNA-mediated regulation of signaling transduction pathways is important in modulating dose-sensitive response of cells to signaling molecules. This review discusses components of the Wnt signaling pathways that are regulated by miRNAs in the context of development and diseases. A fundamental understanding of miRNA functions in Wnt signaling transduction pathways may yield new insight into crosstalks of regulatory mechanisms essential for development and disease pathophysiology leading to novel therapeutics.
Collapse
Affiliation(s)
- Jia L Song
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| | - Priya Nigam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Senel S Tektas
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Erica Selva
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
20
|
Luu O, Damm EW, Parent SE, Barua D, Smith THL, Wen JWH, Lepage SE, Nagel M, Ibrahim-Gawel H, Huang Y, Bruce AEE, Winklbauer R. PAPC mediates self/non-self-distinction during Snail1-dependent tissue separation. ACTA ACUST UNITED AC 2015; 208:839-56. [PMID: 25778923 PMCID: PMC4362454 DOI: 10.1083/jcb.201409026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In Xenopus and zebrafish gastrulae, PAPC attenuates planar cell polarity signaling and controls formation of an adhesive, yet flexible, contact at the ectoderm–mesoderm boundary. Cleft-like boundaries represent a type of cell sorting boundary characterized by the presence of a physical gap between tissues. We studied the cleft-like ectoderm–mesoderm boundary in Xenopus laevis and zebrafish gastrulae. We identified the transcription factor Snail1 as being essential for tissue separation, showed that its expression in the mesoderm depends on noncanonical Wnt signaling, and demonstrated that it enables paraxial protocadherin (PAPC) to promote tissue separation through two novel functions. First, PAPC attenuates planar cell polarity signaling at the ectoderm–mesoderm boundary to lower cell adhesion and facilitate cleft formation. Second, PAPC controls formation of a distinct type of adhesive contact between mesoderm and ectoderm cells that shows properties of a cleft-like boundary at the single-cell level. It consists of short stretches of adherens junction–like contacts inserted between intermediate-sized contacts and large intercellular gaps. These roles of PAPC constitute a self/non–self-recognition mechanism that determines the site of boundary formation at the interface between PAPC-expressing and -nonexpressing cells.
Collapse
Affiliation(s)
- Olivia Luu
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Erich W Damm
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Serge E Parent
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Debanjan Barua
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Tamara H L Smith
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Jason W H Wen
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Stephanie E Lepage
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Martina Nagel
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | | | - Yunyun Huang
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Ashley E E Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| | - Rudolf Winklbauer
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
| |
Collapse
|
21
|
MacMillan CD, Leong HS, Dales DW, Robertson AE, Lewis JD, Chambers AF, Tuck AB. Stage of breast cancer progression influences cellular response to activation of the WNT/planar cell polarity pathway. Sci Rep 2014; 4:6315. [PMID: 25204426 PMCID: PMC4159636 DOI: 10.1038/srep06315] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/11/2014] [Indexed: 12/20/2022] Open
Abstract
Planar cell polarity (PCP) signaling has been shown in different studies to either promote or inhibit the malignancy of breast cancer. Using the 21T cell lines, which were derived from an individual patient and represent distinct stages of progression, we show that the prototypical PCP ligand, WNT5A, is expressed highest in 21MT-1 cells (invasive mammary carcinoma) and lowest in 21PT (atypical ductal hyperplasia) and 21NT (ductal carcinoma in situ) cells. Overexpression of WNT5A decreased spherical colony formation and increased invasion and in vivo extravasation only in 21NT cells; whereas overexpression increased migration of both 21PT and 21NT cells. WNT5A overexpression also increased RHOA expression of both cell lines and subsequent RHOA knockdown blocked WNT5A-induced migration, but only partially blocked WNT5A-induced invasion of 21NT cells. PCP can signal through VANGL1 to modulate AP-1 target genes (e.g. MMP3) and induce invasion. VANGL1 knockdown inhibited WNT5A-induced invasion of 21NT cells, but had no effect on WNT5A-induced migration of either 21PT or 21NT cells. WNT5A-induced MMP3 expression was seen only in 21NT cells, an effect that was VANGL1 dependent, but independent of AP-1. We thus provide evidence that PCP signaling can act in a context dependent manner to promote breast cancer progression.
Collapse
Affiliation(s)
- Connor D MacMillan
- 1] Department of Pathology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [2] London Regional Cancer Program, London Health Sciences Centre, London, ON [3] Department of Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - Hon S Leong
- 1] London Regional Cancer Program, London Health Sciences Centre, London, ON [2] Department of Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - David W Dales
- London Regional Cancer Program, London Health Sciences Centre, London, ON
| | - Amy E Robertson
- 1] London Regional Cancer Program, London Health Sciences Centre, London, ON [2] Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - John D Lewis
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB
| | - Ann F Chambers
- 1] Department of Pathology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [2] London Regional Cancer Program, London Health Sciences Centre, London, ON [3] Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [4] Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - Alan B Tuck
- 1] Department of Pathology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [2] London Regional Cancer Program, London Health Sciences Centre, London, ON [3] Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| |
Collapse
|
22
|
Tuttle AM, Hoffman TL, Schilling TF. Rabconnectin-3a regulates vesicle endocytosis and canonical Wnt signaling in zebrafish neural crest migration. PLoS Biol 2014; 12:e1001852. [PMID: 24802872 PMCID: PMC4011682 DOI: 10.1371/journal.pbio.1001852] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 03/28/2014] [Indexed: 12/15/2022] Open
Abstract
Cell migration requires dynamic regulation of cell-cell signaling and cell adhesion. Both of these processes involve endocytosis, lysosomal degradation, and recycling of ligand-receptor complexes and cell adhesion molecules from the plasma membrane. Neural crest (NC) cells in vertebrates are highly migratory cells, which undergo an epithelial-mesenchymal transition (EMT) to leave the neural epithelium and migrate throughout the body to give rise to many different derivatives. Here we show that the v-ATPase interacting protein, Rabconnectin-3a (Rbc3a), controls intracellular trafficking events and Wnt signaling during NC migration. In zebrafish embryos deficient in Rbc3a, or its associated v-ATPase subunit Atp6v0a1, many NC cells fail to migrate and misregulate expression of cadherins. Surprisingly, endosomes in Rbc3a- and Atp6v0a1-deficient NC cells remain immature but still acidify. Rbc3a loss-of-function initially downregulates several canonical Wnt targets involved in EMT, but later Frizzled-7 accumulates at NC cell membranes, and nuclear B-catenin levels increase. Presumably due to this later Wnt signaling increase, Rbc3a-deficient NC cells that fail to migrate become pigment progenitors. We propose that Rbc3a and Atp6v0a1 promote endosomal maturation to coordinate Wnt signaling and intracellular trafficking of Wnt receptors and cadherins required for NC migration and cell fate determination. Our results suggest that different v-ATPases and associated proteins may play cell-type-specific functions in intracellular trafficking in many contexts.
Collapse
Affiliation(s)
- Adam M. Tuttle
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
| | - Trevor L. Hoffman
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Gene-expression analysis of hair cell regeneration in the zebrafish lateral line. Proc Natl Acad Sci U S A 2014; 111:E1383-92. [PMID: 24706903 DOI: 10.1073/pnas.1402898111] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Deafness caused by the terminal loss of inner ear hair cells is one of the most common sensory diseases. However, nonmammalian animals (e.g., birds, amphibians, and fish) regenerate damaged hair cells. To understand better the reasons underpinning such disparities in regeneration among vertebrates, we set out to define at high resolution the changes in gene expression associated with the regeneration of hair cells in the zebrafish lateral line. We performed RNA-Seq analyses on regenerating support cells purified by FACS. The resulting expression data were subjected to pathway enrichment analyses, and the differentially expressed genes were validated in vivo via whole-mount in situ hybridizations. We discovered that cell cycle regulators are expressed hours before the activation of Wnt/β-catenin signaling following hair cell death. We propose that Wnt/β-catenin signaling is not involved in regulating the onset of proliferation but governs proliferation at later stages of regeneration. In addition, and in marked contrast to mammals, our data clearly indicate that the Notch pathway is significantly down-regulated shortly after injury, thus uncovering a key difference between the zebrafish and mammalian responses to hair cell injury. Taken together, our findings lay the foundation for identifying differences in signaling pathway regulation that could be exploited as potential therapeutic targets to promote either sensory epithelium or hair cell regeneration in mammals.
Collapse
|
24
|
Zhang Z, Rankin SA, Zorn AM. Different thresholds of Wnt-Frizzled 7 signaling coordinate proliferation, morphogenesis and fate of endoderm progenitor cells. Dev Biol 2013; 378:1-12. [PMID: 23562607 DOI: 10.1016/j.ydbio.2013.02.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/06/2013] [Accepted: 02/22/2013] [Indexed: 11/27/2022]
Abstract
Wnt signaling has multiple dynamic roles during development of the gastrointestinal and respiratory systems. Differential Wnt signaling is thought to be a critical step in Xenopus endoderm patterning such that during late gastrula and early somite stages of embryogenesis, Wnt activity must be suppressed in the anterior to allow the specification of foregut progenitors. However, the foregut endoderm also expresses the Wnt-receptor Frizzled 7 (Fzd7) as well as several Wnt ligands suggesting that the current model may be too simple. In this study, we show that Fzd7 is required to transduce a low level of Wnt signaling that is essential to maintain foregut progenitors. Foregut-specific Fzd7-depletion from the Xenopus foregut resulted in liver and pancreas agenesis. Fzd7-depleted embryos failed to maintain the foregut progenitor marker hhex and exhibited decreased proliferation; in addition the foregut cells were enlarged with a randomized orientation. We show that in the foregut Fzd7 signals via both the Wnt/β-catenin and Wnt/JNK pathways and that different thresholds of Wnt-Fzd7 activity coordinate progenitor cell fate, proliferation and morphogenesis.
Collapse
Affiliation(s)
- Zheng Zhang
- Perinatal Institute, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center and the College of Medicine, University of Cincinnati, Cincinnati OH 45229, USA
| | | | | |
Collapse
|
25
|
Range RC, Angerer RC, Angerer LM. Integration of canonical and noncanonical Wnt signaling pathways patterns the neuroectoderm along the anterior-posterior axis of sea urchin embryos. PLoS Biol 2013; 11:e1001467. [PMID: 23335859 PMCID: PMC3545869 DOI: 10.1371/journal.pbio.1001467] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/29/2012] [Indexed: 11/29/2022] Open
Abstract
Three different Wnt signaling pathways function to restrict the anterior neuroectoderm state to the anterior end of the sea urchin embryo, a mechanism of anterior fate restriction that could be conserved among deuterostomes. Patterning the neuroectoderm along the anterior–posterior (AP) axis is a critical event in the early development of deuterostome embryos. However, the mechanisms that regulate the specification and patterning of the neuroectoderm are incompletely understood. Remarkably, the anterior neuroectoderm (ANE) of the deuterostome sea urchin embryo expresses many of the same transcription factors and secreted modulators of Wnt signaling, as does the early vertebrate ANE (forebrain/eye field). Moreover, as is the case in vertebrate embryos, confining the ANE to the anterior end of the embryo requires a Wnt/β-catenin-dependent signaling mechanism. Here we use morpholino- or dominant negative-mediated interference to demonstrate that the early sea urchin embryo integrates information not only from Wnt/β-catenin but also from Wnt/Fzl5/8-JNK and Fzl1/2/7-PKC pathways to provide precise spatiotemporal control of neuroectoderm patterning along its AP axis. Together, through the Wnt1 and Wnt8 ligands, they orchestrate a progressive posterior-to-anterior wave of re-specification that restricts the initial, ubiquitous, maternally specified, ANE regulatory state to the most anterior blastomeres. There, the Wnt receptor antagonist, Dkk1, protects this state through a negative feedback mechanism. Because these different Wnt pathways converge on the same cell fate specification process, our data suggest they may function as integrated components of an interactive Wnt signaling network. Our findings provide strong support for the idea that the sea urchin ANE regulatory state and the mechanisms that position and define its borders represent an ancient regulatory patterning system that was present in the common echinoderm/vertebrate ancestor. The initial regulatory state of most cells in many deuterostome embryos, including those of vertebrates and sea urchins, supports anterior neural fate specification. It is important to restrict this neurogenic potential to the anterior end of the embryo during early embryogenesis, but the molecular mechanisms by which this re-specification of posterior fate occurs are incompletely understood in any embryo. The sea urchin embryo is ideally suited to study this process because, in contrast to vertebrates, anterior–posterior neuroectoderm patterning occurs independently of dorsal-ventral axis patterning and takes place before the complex cell movements of gastrulation. In this study, we show that a linked, three-step process involving at least three different Wnt signaling pathways provides precise spatiotemporal restriction of the anterior neuroectoderm regulatory state to the anterior end of the sea urchin embryo. Because these three pathways impinge on the same developmental process, they could be functioning as an integrated Wnt signaling network. Moreover, striking parallels among gene expression patterns and functional studies suggest that this mechanism of anterior fate restriction could be highly conserved among deuterostomes.
Collapse
Affiliation(s)
- Ryan C. Range
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert C. Angerer
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lynne M. Angerer
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
26
|
The WNT signaling pathway from ligand secretion to gene transcription: molecular mechanisms and pharmacological targets. Pharmacol Ther 2013; 138:66-83. [PMID: 23328704 DOI: 10.1016/j.pharmthera.2013.01.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 12/20/2012] [Indexed: 12/22/2022]
Abstract
Wingless/integrase-1 (WNT) signaling is a key pathway regulating various aspects of embryonic development; however it also underlies several pathological conditions in man, including various cancers and fibroproliferative diseases in several organs. Investigating the molecular processes involved in (canonical) WNT signaling will open new avenues for generating new therapeutics to specifically target diseases in which WNT signaling is aberrantly regulated. Here we describe the complexity of WNT signal transduction starting from the processes involved in WNT ligand biogenesis and secretion by WNT producing cells followed by a comprehensive overview of the molecular signaling events ultimately resulting in enhanced transcription of specific genes in WNT receiving cells. Finally, the possible targets for therapeutic intervention and the available pharmacological inhibitors for this complex signaling pathway are discussed.
Collapse
|
27
|
Dawson K, Aflaki M, Nattel S. Role of the Wnt-Frizzled system in cardiac pathophysiology: a rapidly developing, poorly understood area with enormous potential. J Physiol 2012. [PMID: 23207593 DOI: 10.1113/jphysiol.2012.235382] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract The Wnt-Frizzled (Fzd) G-protein-coupled receptor system, involving 19 distinct Wnt ligands and 10 Fzd receptors, plays key roles in the development and functioning of many organ systems. There is increasing evidence that Wnt-Fzd signalling is important in regulating cardiac function. Wnt-Fzd signalling primarily involves a canonical pathway, with dishevelled-1-dependent nuclear translocation of β-catenin that derepresses Wnt-sensitive gene transcription, but can also include non-canonical pathways via phospholipase-C/Ca(2+) mobilization and dishevelled-protein activation of small GTPases. Wnt-Fzd effects vary with specific ligand/receptor interactions and associated downstream pathways. This paper reviews the biochemistry and physiology of the Wnt-Fzd complex, and presents current knowledge of Wnt signalling in cardiac remodelling processes such as hypertrophy and fibrosis, as well as disease states such as myocardial infarction (MI), heart failure and arrhythmias. Wnt signalling is activated during hypertrophy; inhibiting Wnt signalling by activating glycogen synthase kinase attenuates the hypertrophic response. Wnt signalling has complex and time-dependent actions post-MI, so that either beneficial or harmful effects might result from Wnt-directed interventions. Stem cell biology, a promising area for therapeutic intervention, is highly regulated by Wnt signalling. The Wnt system regulates fibroblast function, and is prominently altered in arrhythmogenic ventricular cardiomyopathy, a familial disease involving excess deposition of fibroadipose tissue. Wnt signalling controls connexin43 expression, thereby contributing to the regulation of cardiac electrical stability and arrhythmia generation. Although much has been learned about Wnt-Fzd signalling in hypertrophy and infarction, its role is poorly understood for a broad range of other heart disorders. Much more needs to be learned for its contributions to be fully appreciated, and to permit more effective exploitation of its enormous potential in therapeutic development.
Collapse
Affiliation(s)
- Kristin Dawson
- S. Nattel: 5000 Belanger St. E, Montreal, Quebec, Canada H1T 1C8.
| | | | | |
Collapse
|
28
|
Kraft B, Berger CD, Wallkamm V, Steinbeisser H, Wedlich D. Wnt-11 and Fz7 reduce cell adhesion in convergent extension by sequestration of PAPC and C-cadherin. ACTA ACUST UNITED AC 2012; 198:695-709. [PMID: 22908314 PMCID: PMC3514027 DOI: 10.1083/jcb.201110076] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Wnt-11/frizzled-7 reduces the lateral clustering of C-cadherin by capturing the
protocadherin PAPC and C-cadherin into distinct adhesion-modulating
complexes. Wnt-11/planar cell polarity signaling polarizes mesodermal cells undergoing
convergent extension during Xenopus laevis gastrulation. These
shape changes associated with lateral intercalation behavior require a dynamic
modulation of cell adhesion. In this paper, we report that Wnt-11/frizzled-7
(Fz7) controls cell adhesion by forming separate adhesion-modulating complexes
(AMCs) with the paraxial protocadherin (PAPC; denoted as AMCP) and C-cadherin
(denoted as AMCC) via distinct Fz7 interaction domains. When PAPC was part of a
Wnt-11–Fz7 complex, its Dynamin1- and clathrin-dependent internalization
was blocked. This membrane stabilization of AMCP (Fz7/PAPC) by Wnt-11 prevented
C-cadherin clustering, resulting in reduced cell adhesion and modified cell
sorting activity. Importantly, Wnt-11 did not influence C-cadherin
internalization; instead, it promoted the formation of AMCC (Fz7/Cadherin),
which competed with cis-dimerization of C-cadherin. Because PAPC and C-cadherin
did not directly interact and did not form a joint complex with Fz7, we suggest
that Wnt-11 triggers the formation of two distinct complexes, AMCC and AMCP,
that act in parallel to reduce cell adhesion by hampering lateral clustering of
C-cadherin.
Collapse
Affiliation(s)
- Bianca Kraft
- Cell and Developmental Biology, Zoological Institute, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | | | | | | | | |
Collapse
|
29
|
Abstract
In addition to activating β-catenin/TCF transcriptional complexes, Wnt proteins can elicit a variety of other responses. These are often lumped together under the denominator "alternative" or "non-canonical" Wnt signaling, but they likely comprise distinct signaling events. In this article, I discuss how the use of different ligand and receptor combinations is thought to give rise to these alternative Wnt-signaling responses. Although many of the biochemical details remain to be resolved, it is evident that alternative Wnt signaling plays important roles in regulating tissue morphogenesis during embryonic development.
Collapse
Affiliation(s)
- Renée van Amerongen
- Department of Developmental Biology, Stanford University, Stanford, California 94305, USA.
| |
Collapse
|
30
|
Dellinger TH, Planutis K, Tewari KS, Holcombe RF. Role of canonical Wnt signaling in endometrial carcinogenesis. Expert Rev Anticancer Ther 2012; 12:51-62. [PMID: 22149432 DOI: 10.1586/era.11.194] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
While the role of Wnt signaling is well established in colorectal carcinogenesis, its function in gynecologic cancers has not been elucidated. Here, we describe the current state of knowledge of canonical Wnt signaling in endometrial cancer (EC), and its implications for future therapeutic targets. Deregulation of the Wnt/β-catenin signaling pathway in EC occurs by inactivating β-catenin mutations in approximately 10-45% of ECs, and via downregulation of Wnt antagonists by epigenetic silencing. The Wnt pathway is intimately involved with estrogen and progesterone, and emerging data implicate it in other important signaling pathways, such as mTOR and Hedgehog. While no therapeutic agents targeting the Wnt signaling pathway are currently in clinical trials, the preclinical data presented suggest a role for Wnt signaling in uterine carcinogenesis, with further research warranted to elucidate the mechanism of action and to proceed towards targeted cancer drug development.
Collapse
Affiliation(s)
- Thanh H Dellinger
- Divison of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of California, Irvine, Medical Center, 101 The City Drive, Building 56, Room 260, Orange, CA 92868, USA.
| | | | | | | |
Collapse
|
31
|
Nishioka M, Ueno K, Hazama S, Okada T, Sakai K, Suehiro Y, Okayama N, Hirata H, Oka M, Imai K, Dahiya R, Hinoda Y. Possible involvement of Wnt11 in colorectal cancer progression. Mol Carcinog 2011; 52:207-17. [PMID: 22161723 DOI: 10.1002/mc.21845] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 10/06/2011] [Accepted: 10/27/2011] [Indexed: 01/09/2023]
Abstract
Our previous report revealed that the expression of Frizzled-7 (FZD7) in colorectal cancer (CRC) and its possible role in CRC progression. In this study we measured the expression levels of candidate FZD7 ligands, Wnt3 and Wnt11 in colon cancer cell lines (n = 7) and primary CRC tissues (n = 133) by quantitative RT-PCR. We also examined the functional effects of Wnt11 with the use of Wnt11 transfectants of colon cancer HCT-116 cells. Wnt11 transfectants showed the increased proliferation and migration/invasion activities compared to mock cells. Western blot analysis of transfectants revealed that phosphorylation of JNK and c-jun was increased after Wnt11 transfection. Wnt11 mRNA expression was significantly higher in the stage I, II, III, or IV tumor tissues than in non-tumor tissues (overall P < 0.003), while there was no significant difference in Wnt3 mRNA expression between tumor and non-tumor tissues. In addition, Wnt11 mRNA expression was significantly higher in patients with recurrence or death after surgery than in those with no recurrence (disease free) after surgery (P = 0.018). We also compared the expression levels of Wnt11 mRNA with those of FZD7 mRNA in the same CRC samples. Wnt11 mRNA expression was significantly higher in patients with higher FZD7 mRNA levels than in those with lower FZD7 mRNA levels (P = 0.0005). The expression levels of Wnt11 mRNA were correlated with those of FZD7 mRNA (P < 0.0001). These data suggest that Wnt11 may play an important role in CRC progression.
Collapse
Affiliation(s)
- Mitsuaki Nishioka
- Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang B, Tran U, Wessely O. Expression of Wnt signaling components during Xenopus pronephros development. PLoS One 2011; 6:e26533. [PMID: 22028899 PMCID: PMC3197532 DOI: 10.1371/journal.pone.0026533] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 09/28/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The formation of the vertebrate kidney is tightly regulated and relies on multiple evolutionarily conserved inductive events. These are present in the complex metanephric kidney of higher vertebrates, but also in the more primitive pronephric kidney functional in the larval stages of amphibians and fish. Wnts have long been viewed as central in this process. Canonical β-Catenin-dependent Wnt signaling establishes kidney progenitors and non-canonical β-Catenin-independent Wnt signaling participate in the morphogenetic processes that form the highly sophisticated nephron structure. While some individual Wnt signaling components have been studied extensively in the kidney, the overall pathway has not yet been analyzed in depth. METHODOLOGY/PRINCIPAL FINDINGS Here we report a detailed expression analysis of all Wnt ligands, receptors and several downstream Wnt effectors during pronephros development in Xenopus laevis using in situ hybridization. Out of 19 Wnt ligands, only three, Wnt4, Wnt9a and Wnt11, are specifically expressed in the pronephros. Others such as Wnt8a are present, but in a broader domain comprising adjacent tissues in addition to the kidney. The same paradigm is observed for the Wnt receptors and its downstream signaling components. Fzd1, Fzd4, Fzd6, Fzd7, Fzd8 as well as Celsr1 and Prickle1 show distinct expression domains in the pronephric kidney, whereas the non-traditional Wnt receptors, Ror2 and Ryk, as well as the majority of the effector molecules are rather ubiquitous. In addition to this spatial regulation, the timing of expression is also tightly regulated. In particular, non-canonical Wnt signaling seems to be restricted to later stages of pronephros development. CONCLUSION/SIGNIFICANCE Together these data suggest a complex cross talk between canonical and non-canonical Wnt signaling is required to establish a functional pronephric kidney.
Collapse
Affiliation(s)
- Bo Zhang
- Lerner Research Institute/Cleveland Clinic, Department of Cell Biology, Cleveland, Ohio, United States of America
- Louisiana State University (LSU) Health Sciences Center, Department of Cell Biology & Anatomy, New Orleans, Louisiana, United States of America
| | - Uyen Tran
- Lerner Research Institute/Cleveland Clinic, Department of Cell Biology, Cleveland, Ohio, United States of America
| | - Oliver Wessely
- Lerner Research Institute/Cleveland Clinic, Department of Cell Biology, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
33
|
Luna-Ulloa LB, Hernández-Maqueda JG, Santoyo-Ramos P, Castañeda-Patlán MC, Robles-Flores M. Protein kinase C ζ is a positive modulator of canonical Wnt signaling pathway in tumoral colon cell lines. Carcinogenesis 2011; 32:1615-24. [PMID: 21859831 DOI: 10.1093/carcin/bgr190] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The colonic epithelium is a continuously renewing tissue with a dynamic turnover of cells. Wnt pathway is a key regulator of its homeostasis and is altered in a large proportion of colon cancers. Protein kinase C (PKC) family of serine/threonine kinases are also involved in colon tumor formation and progression; however, the molecular role played by them in the Wnt pathway, is poorly understood. Reciprocal coimmunoprecipitation and immunofluorescence studies revealed that PKCζ interacts with β-catenin mainly in tumoral colon cells, which overexpressed this PKC isoform. The pharmacological inhibition, the small interference RNA-mediated knockdown of PKCζ or the expression of a dominant-negative form of it in tumoral SW480 cells, blocked in a dose-dependent manner the constitutive transcriptional activity mediated by β-catenin, the cell proliferation and the expression of the Wnt target gene c-myc. Remarkably, the PKCζ stably depleted cells exhibited diminished tumorigenic activity in grafted mice. We show that PKCζ functions in a mechanism that does not involve β-catenin degradation since the effects produced by PKCζ inhibition were also obtained in the presence of proteosome inhibitor and in cells expressing a β-catenin degradation-resistant mutant. It was found that PKCζ activity regulates the nuclear localization of β-catenin since PKCζ inhibition induces a rapid export of β-catenin from the nucleus to the cytoplasm in a Leptomycin B sensitive manner. Taken together, our results indicate that the atypical PKCζ plays an important role in the positive regulation of canonical Wnt pathway.
Collapse
Affiliation(s)
- Luis Bernardo Luna-Ulloa
- Department of Biochemistry, Faculty of Medicine, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico, 04510, Mexico
| | | | | | | | | |
Collapse
|
34
|
Abstract
In all multicellular organisms, epithelial cells are not only polarized along the apical-basal axis, but also within the epithelial plane, giving cells a sense of direction. Planar cell polarity (PCP) signaling regulates establishment of polarity within the plane of an epithelium. The outcomes of PCP signaling are diverse and include the determination of cell fates, the generation of asymmetric but highly aligned structures, such as the stereocilia in the human inner ear or the hairs on a fly wing, or the directional migration of cells during convergence and extension during vertebrate gastrulation. In humans, aberrant PCP signaling can result in severe developmental defects, such as open neural tubes (spina bifida), and can cause cystic kidneys. In this review, we discuss the basic mechanism and more recent findings of PCP signaling focusing on Drosophila melanogaster, the model organism in which most key PCP components were initially identified.
Collapse
Affiliation(s)
- Saw Myat Thanda W Maung
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY, USA
| | | |
Collapse
|
35
|
Yeh JR, Zhang X, Nagano MC. Wnt5a is a cell-extrinsic factor that supports self-renewal of mouse spermatogonial stem cells. J Cell Sci 2011; 124:2357-66. [PMID: 21693582 DOI: 10.1242/jcs.080903] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The maintenance of spermatogonial stem cells (SSCs) provides the foundation for life-long spermatogenesis. Although glial-cell-line-derived neurotrophic factor and fibroblast growth factor 2 are crucial for self-renewal of SSCs, recent studies have suggested that other growth factors have important roles in controlling SSC fate. Because β-catenin-dependent Wnt signaling promotes self-renewal of various stem cell types, we hypothesized that this pathway contributes to SSC maintenance. Using transgenic reporter mice for β-catenin-dependent signaling, we found that this signaling was not active in SSCs in vitro and in most spermatogonia in vivo. Nonetheless, a pan-Wnt antagonist significantly reduced SSC activity in vitro, suggesting that some Wnt molecules exist in our serum-free culture system and contribute to SSC maintenance. Here, we report that Wnt5a promotes SSC activity. We found that Wnt5a-expressing fibroblasts supported SSC activity better than those not expressing Wnt5a in culture, and that recombinant Wnt5a stimulated SSC maintenance. Furthermore, Wnt5a promoted SSC survival in the absence of feeder cells, and this effect was abolished by inhibiting the Jun N-terminal kinase cascade. In addition, Wnt5a blocked β-catenin-dependent signaling. We detected the expression of Wnt5a and potential Wnt5a receptors in Sertoli cells and stem/progenitor spermatogonia, respectively. These results indicate that Wnt5a is a cell-extrinsic factor that supports SSC self-renewal through β-catenin-independent mechanisms.
Collapse
Affiliation(s)
- Jonathan R Yeh
- Department of Obstetrics and Gynecology and Division of Experimental Medicine, McGill University, Montreal, QC H3A 1A1, Canada
| | | | | |
Collapse
|
36
|
Jung B, Köhler A, Schambony A, Wedlich D. PAPC and the Wnt5a/Ror2 pathway control the invagination of the otic placode in Xenopus. BMC DEVELOPMENTAL BIOLOGY 2011; 11:36. [PMID: 21663658 PMCID: PMC3127988 DOI: 10.1186/1471-213x-11-36] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 06/10/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND Paraxial protocadherin (PAPC) plays a crucial role in morphogenetic movements during gastrulation and somitogenesis in mouse, zebrafish and Xenopus. PAPC influences cell-cell adhesion mediated by C-Cadherin. A putative direct adhesion activity of PAPC is discussed. PAPC also promotes cell elongation, tissue separation and coordinates cell mass movements. In these processes the signaling function of PAPC in activating RhoA/JNK and supporting Wnt-11/PCP by binding to frizzled 7 (fz7) is important. RESULTS Here we demonstrate by loss of function experiments in Xenopus embryos that PAPC regulates another type of morphogenetic movement, the invagination of the ear placode. Knockdown of PAPC by antisense morpholinos results in deformation of the otic vesicle without altering otocyst marker expression. Depletion of PAPC could be rescued by full-length PAPC, constitutive active RhoA and by the closely related PCNS but not by classical cadherins. Also the cytoplasmic deletion mutant M-PAPC, which influences cell adhesion, does not rescue the PAPC knockdown. Interestingly, depletion of Wnt5a or Ror2 which are also expressed in the otocyst phenocopies the PAPC morphant phenotype. CONCLUSIONS PAPC signaling via RhoA and Wnt5a/Ror2 activity are required to keep cells aligned in apical-basal orientation during invagination of the ear placode. Since neither the cytoplasmic deletion mutant M-PAPC nor a classical cadherin is able to rescue loss of PAPC we suggest that the signaling function of the protocadherin rather than its role as modulator of cell-cell adhesion is required during invagination of the ear placode.
Collapse
Affiliation(s)
- Barbara Jung
- Karlsruhe Institute of Technology, Department of Cell and Developmental Biology, Karlsruhe, Germany
| | | | | | | |
Collapse
|
37
|
Ohkawara B, Niehrs C. An ATF2-based luciferase reporter to monitor non-canonical Wnt signaling in Xenopus embryos. Dev Dyn 2011; 240:188-94. [PMID: 21128306 DOI: 10.1002/dvdy.22500] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Non-canonical/planar cell polarity (PCP) Wnt signaling plays important roles in embryonic development and tissue homeostasis, and is implicated in human disease. Monitoring Wnt/PCP signaling relies mostly on semi-quantitative bioassays or biochemical analysis. Here we describe a luciferase reporter assay based on an ATF2 response element, which faithfully monitors non-canonical Wnt signaling in Xenopus embryos. The assay is simple, quantitative, and robust. It can be used to detect non-canonical Wnt signaling changes following gain and loss of function of pathway components, including Wnt, Frizzled, Ror2, Disheveled, Rac1, MKK7, and JNK. Wnt/PCP signaling has recently been implicated in left-right asymmetry and our reporter assay suggests that in gastrula embryos there is a right-ward bias in Wnt/PCP signaling. We also mapped Wnt/PCP signaling in the early Xenopus embryo and find that it peaks in the dorso-vegetal region, paralleling Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Bisei Ohkawara
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | |
Collapse
|
38
|
The involvement of Eph–Ephrin signaling in tissue separation and convergence during Xenopus gastrulation movements. Dev Biol 2011; 350:441-50. [DOI: 10.1016/j.ydbio.2010.12.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 12/03/2010] [Accepted: 12/03/2010] [Indexed: 11/21/2022]
|
39
|
Kiryukhin DO, Shustikova LA, Kopantseva EE, Luchinskaya NN, Belyavskii AV. Involvement of the guanine nucleotide exchange factor xLARG in the epiboly of Xenopus laevis embryo animal pole cells. Biophysics (Nagoya-shi) 2010. [DOI: 10.1134/s0006350910060060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
40
|
Mueller RL, Huang C, Ho RK. Spatio-temporal regulation of Wnt and retinoic acid signaling by tbx16/spadetail during zebrafish mesoderm differentiation. BMC Genomics 2010; 11:492. [PMID: 20828405 PMCID: PMC2996988 DOI: 10.1186/1471-2164-11-492] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 09/09/2010] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND A complex network of signaling pathways and transcription factors regulates vertebrate mesoderm development. Zebrafish mutants provide a powerful tool for examining the roles of individual genes in such a network. spadetail (spt) is a mutant with a lesion in tbx16, a T-box transcription factor involved in mesoderm development; the mutant phenotype includes disrupted primitive red blood cell formation as well as disrupted somitogenesis. Despite much recent progress, the downstream targets of tbx16 remain incompletely understood. The current study was carried out to test whether any of the five major signaling pathways are regulated by tbx16 during two specific stages of mesoderm development: primitive red blood cell formation in the intermediate mesoderm and somite formation in the tail paraxial mesoderm. This test was performed using Gene Set Enrichment Analysis, which identifies coordinated changes in expression among a priori sets of genes associated with biological features or processes. RESULTS Our Gene Set Enrichment Analysis results identify Wnt and retinoic acid signaling as likely downstream targets of tbx16 in the developing zebrafish intermediate mesoderm, the site of primitive red blood cell formation. In addition, such results identify retinoic acid signaling as a downstream target of tbx16 in the developing zebrafish posterior somites. Finally, using candidate gene identification and in situ hybridization, we provide expression domain information for 25 additional genes downstream of tbx16 that are outside of both pathways; 23 were previously unknown downstream targets of tbx16, and seven had previously uncharacterized expression in zebrafish. CONCLUSIONS Our results suggest that (1) tbx16 regulates Wnt signaling in the developing zebrafish intermediate mesoderm, the site of primitive red blood cell formation, and (2) tbx16 regulates retinoic acid signaling at two distinct embryonic locations and developmental stages, which may imply ongoing spatio-temporal regulation throughout mesoderm development.
Collapse
Affiliation(s)
- Rachel Lockridge Mueller
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago IL 60637, USA
- Department of Biology, Colorado State University, Fort Collins CO 80523, USA
| | - Cheng Huang
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago IL 60637, USA
| | - Robert K Ho
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago IL 60637, USA
| |
Collapse
|
41
|
Camilli TC, Weeraratna AT. Striking the target in Wnt-y conditions: intervening in Wnt signaling during cancer progression. Biochem Pharmacol 2010; 80:702-11. [PMID: 20211149 PMCID: PMC2897923 DOI: 10.1016/j.bcp.2010.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 02/25/2010] [Accepted: 03/01/2010] [Indexed: 12/30/2022]
Abstract
Wnt signaling can be divided into three pathways, namely the canonical Wnt/beta-catenin pathway, and the non-canonical (or heretical) Wnt/Ca(2+) and planar cell polarity (PCP) pathways. Although the canonical Wnt/beta-catenin pathway is the best described in cancer, increasing data points to the importance of the heretical Wnt pathways in several aspects of tumor progression. The recent advances in understanding the players and mechanisms by which these Wnt pathways contribute to cancer progression have led to the identification of numerous molecules that are already, or could be considered, targets for cancer therapy.
Collapse
Affiliation(s)
- Tura C. Camilli
- Laboratory of Immunology and Research Resources Branch, National Institute on, Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ashani T. Weeraratna
- Laboratory of Immunology and Research Resources Branch, National Institute on, Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
42
|
Ror2/Frizzled complex mediates Wnt5a-induced AP-1 activation by regulating Dishevelled polymerization. Mol Cell Biol 2010; 30:3610-9. [PMID: 20457807 DOI: 10.1128/mcb.00177-10] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The receptor tyrosine kinase Ror2 acts as a receptor or coreceptor for Wnt5a to mediate Wnt5a-induced activation of the Wnt/JNK pathway and inhibition of the beta-catenin-dependent canonical Wnt pathway. However, little is known about how Ror2 cooperates with another receptor component(s) to mediate Wnt5a signaling. We show here that Ror2 regulates Wnt5a-induced polymerization of Dishevelled (Dvl) and that this Ror2-mediated regulation of Dvl is independent of the cytoplasmic region of Ror2. Ror2 can associate with Frizzled7 (Fz7) via its extracellular cysteine-rich domain to form a receptor complex that is required for the regulation of Dvl and activation of the AP-1 promoter after Wnt5a stimulation. Suppressed expression of Fz7 indeed results in the inhibition of Wnt5a-induced polymerization of Dvl and AP-1 activation. Interestingly, both the DIX and the DEP domains of Dvl are indispensable for Dvl polymerization and subsequent AP-1 activation after Wnt5a stimulation. We further show that polymerized Dvl is colocalized with Rac1 and that suppressed expression of Rac1 inhibits Wnt5a-induced AP-1 activation. Collectively, our results indicate that Ror2/Fz receptor complex plays an important role in the Wnt5a/Rac1/AP-1 pathway by regulating the polymerization of Dvl.
Collapse
|
43
|
A flanking gene problem leads to the discovery of a Gprc5b splice variant predominantly expressed in C57Bl/6J mouse brain and in maturing neurons. PLoS One 2010; 5:e10351. [PMID: 20436672 PMCID: PMC2859937 DOI: 10.1371/journal.pone.0010351] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 03/22/2010] [Indexed: 01/19/2023] Open
Abstract
Background Gprc5b, a retinoic acid-inducible orphan G protein–coupled receptor (GPCR), is a member of the group C metabotropic glutamate receptor family proteins possibly involved in non-canonical Wnt signaling. Many GPCR transcripts are alternatively spliced, which diversifies this class of proteins in their cell- and tissue-specific signaling, regulatory and/or pharmacological properties. We previously generated p97FE65 isoform-specific knockout mice that showed learning/memory deficits. In this study, we further characterized the 97FE65 null mice using cDNA microarray and RT-PCR analyses. Methodology/Principal Findings We discovered a novel brain-specific C-terminal splice variant of Gprc5b, Gprc5b_v2, which was differentially expressed in p97FE65 wild type and null mouse brains. The null mice were generated in 129/Sv ES cells, and backcrossed to C57Bl/6J for ten generations. We found that expression of Gprc5b_v2 mRNA in the brains of p97FE65 null mice was dramatically down-regulated (more than 20 fold) compared to their wild type littermates. However, expression profiles of Gprc5b variants and SNP analysis surrounding the FE65 locus suggest that the down-regulation is unlikely due to the altered FE65 function, but rather is caused by gene retention from the 129/Sv ES cells. Consistently, in contrast to ubiquitously expressed Gprc5b_v1, Gprc5b_v2 was predominantly expressed in the brain tissues of C57Bl/6J mice. The alternative splicing of the 3′ terminal exon also altered the protein coding sequences, giving rise to the characteristic C-termini. Levels of Gprc5b_v2 mRNA were increased during neuronal maturation, paralleling the expression of synaptic proteins. Overexpression of both Gprc5b variants stimulated neurite-like outgrowth in a neuroblastoma cell line. Conclusions/Significance Our results suggest that Gprc5b-v2 may play a role during brain maturation and in matured brain, possibly through the regulation of neuronal morphology and protein-protein interaction. This study also highlights the fact that unexpected gene retention following repeated backcrosses can lead to important biological consequences.
Collapse
|
44
|
xGit2 and xRhoGAP 11A regulate convergent extension and tissue separation in Xenopus gastrulation. Dev Biol 2010; 344:26-35. [PMID: 20380829 DOI: 10.1016/j.ydbio.2010.03.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 03/01/2010] [Accepted: 03/31/2010] [Indexed: 11/22/2022]
Abstract
In a microarray-based screen for genes that are involved in tissue separation downstream of Paraxial Protocadherin (PAPC) and Frizzled-7 (Fz7)-mediated signaling we identified xGit2 and xRhoGAP 11A, two GTPase-activating proteins (GAP) for small GTPases. xGit2 and xRhoGAP 11A are expressed in the dorsal ectoderm, and their transcription is downregulated in the involuting dorsal mesoderm by PAPC and Fz7. Overexpression of xGit2 and xRhoGAP 11A inhibits Rho activity and impairs convergent extension movements as well as tissue separation behaviour. We propose that Rho activity in the involuting mesoderm is enhanced through inhibition of xGit2 and xRhoGAP 11A transcription by PAPC and Fz7. By this mechanism xRhoGAP 11A and xGit2 are restricted to the dorsal ectoderm, while Rho signaling is inhibited.
Collapse
|
45
|
van Eekelen M, Runtuwene V, Overvoorde J, den Hertog J. RPTPα and PTPε signaling via Fyn/Yes and RhoA is essential for zebrafish convergence and extension cell movements during gastrulation. Dev Biol 2010; 340:626-39. [DOI: 10.1016/j.ydbio.2010.02.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 02/15/2010] [Accepted: 02/16/2010] [Indexed: 10/19/2022]
|
46
|
van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development 2009; 136:3205-14. [PMID: 19736321 DOI: 10.1242/dev.033910] [Citation(s) in RCA: 914] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Wnt signaling is crucial for embryonic development in all animal species studied to date. The interaction between Wnt proteins and cell surface receptors can result in a variety of intracellular responses. A key remaining question is how these specific responses take shape in the context of a complex, multicellular organism. Recent studies suggest that we have to revise some of our most basic ideas about Wnt signal transduction. Rather than thinking about Wnt signaling in terms of distinct, linear, cellular signaling pathways, we propose a novel view that considers the integration of multiple, often simultaneous, inputs at the level of both Wnt-receptor binding and the downstream, intracellular response.
Collapse
Affiliation(s)
- Renée van Amerongen
- Department of Developmental Biology and Howard Hughes Medical Institute, Beckman Center, 279 Campus Drive, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
47
|
Freese JL, Pino D, Pleasure SJ. Wnt signaling in development and disease. Neurobiol Dis 2009; 38:148-53. [PMID: 19765659 DOI: 10.1016/j.nbd.2009.09.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/01/2009] [Accepted: 09/10/2009] [Indexed: 12/26/2022] Open
Abstract
The Wnt signaling pathway is one of the central morphogenic signaling pathways regulating early vertebrate development. In recent years, it has become clear that the Wnt pathway also regulates many aspects of nervous system development from the patterning stage through the regulation of neural plasticity. In this review, we first present an overview of the components of the Wnt signaling pathway and then go on to discuss the literature describing the multitude of roles of Wnts in nervous system. In the latter portion of the review, we turn to the ways that defects in Wnt signaling lead to neurologic disease.
Collapse
Affiliation(s)
- Jennifer L Freese
- Department of Neurology, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
48
|
Abstract
The evolutionarily conserved and developmentally important Wnt signaling pathway has traditionally been regarded as a critical player in tumorigenesis through the canonical Wnt/beta-catenin cascade. Nevertheless, accumulating evidence based on recent research has revealed the previously unacknowledged role of noncanonical Wnt/planar cell polarity (PCP) signaling in cancer progression, invasion and metastasis, and angiogenesis. This review describes the PCP signaling pathway and its ever-expanding components and modulators, highlights the most recent studies that provide insight into the link between PCP signaling and cancer, and, finally, proposes a model by which PCP signaling may promote cancer development. This review underscores the emerging theme that deregulated PCP signaling contributes to tumorigenesis, providing new potential targets for cancer therapy.
Collapse
Affiliation(s)
- Yingqun Wang
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
49
|
Abstract
It is well documented that mutations in the MYOCILIN gene may lead to juvenile- and adult-onset primary open-angle glaucoma. However, the functions of wild-type myocilin are still not well understood. To study the functions of human myocilin and its two proteolytic fragments, these proteins were expressed in HEK293 cells. Conditioned medium from myocilin-expressing cells, as well as purified myocilin, induced the formation of stress fibers in primary cultures of human trabecular meshwork or NIH 3T3 cells. Stress fiber-inducing activity of myocilin was blocked by antibodies against myocilin, as well as secreted inhibitors of Wnt signaling, secreted Frizzled-related protein 1 (sFRP1) or sFRP3, and beta-catenin small interfering RNA. Interaction of myocilin with sFRP1, sFRP3, and several Frizzled receptors was confirmed by immunoprecipitation experiments and by binding of myocilin to the surface of cells expressing cysteine-rich domains of different Frizzled and sFRPs. Treatment of NIH 3T3 cells with myocilin and its fragments induced intracellular redistribution of beta-catenin and its accumulation on the cellular membrane but did not induce nuclear accumulation of beta-catenin. Overexpression of myocilin in the eye angle tissues of transgenic mice stimulated accumulation of beta-catenin in these tissues. Myocilin and Wnt proteins may perform redundant functions in the mammalian eye, since myocilin modulates Wnt signaling by interacting with components of this signaling pathway.
Collapse
|
50
|
Louie SH, Yang XY, Conrad WH, Muster J, Angers S, Moon RT, Cheyette BNR. Modulation of the beta-catenin signaling pathway by the dishevelled-associated protein Hipk1. PLoS One 2009; 4:e4310. [PMID: 19183803 PMCID: PMC2629544 DOI: 10.1371/journal.pone.0004310] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 12/09/2008] [Indexed: 12/31/2022] Open
Abstract
Background Wnts are evolutionarily conserved ligands that signal through β-catenin-dependent and β-catenin–independent pathways to regulate cell fate, proliferation, polarity, and movements during vertebrate development. Dishevelled (Dsh/Dvl) is a multi-domain scaffold protein required for virtually all known Wnt signaling activities, raising interest in the identification and functions of Dsh-associated proteins. Methodology We conducted a yeast-2-hybrid screen using an N-terminal fragment of Dsh, resulting in isolation of the Xenopus laevis ortholog of Hipk1. Interaction between the Dsh and Hipk1 proteins was confirmed by co-immunoprecipitation assays and mass spectrometry, and further experiments suggest that Hipk1 also complexes with the transcription factor Tcf3. Supporting a nuclear function during X. laevis development, Myc-tagged Hipk1 localizes primarily to the nucleus in animal cap explants, and the endogenous transcript is strongly expressed during gastrula and neurula stages. Experimental manipulations of Hipk1 levels indicate that Hipk1 can repress Wnt/β-catenin target gene activation, as demonstrated by β-catenin reporter assays in human embryonic kidney cells and by indicators of dorsal specification in X. laevis embryos at the late blastula stage. In addition, a subset of Wnt-responsive genes subsequently requires Hipk1 for activation in the involuting mesoderm during gastrulation. Moreover, either over-expression or knock-down of Hipk1 leads to perturbed convergent extension cell movements involved in both gastrulation and neural tube closure. Conclusions These results suggest that Hipk1 contributes in a complex fashion to Dsh-dependent signaling activities during early vertebrate development. This includes regulating the transcription of Wnt/β-catenin target genes in the nucleus, possibly in both repressive and activating ways under changing developmental contexts. This regulation is required to modulate gene expression and cell movements that are essential for gastrulation.
Collapse
Affiliation(s)
- Sarah H. Louie
- Howard Hughes Medical Institute, Department of Pharmacology, and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Xiao Yong Yang
- Department of Psychiatry, and Graduate Program in Developmental Biology, Program in Biological Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - William H. Conrad
- Howard Hughes Medical Institute, Department of Pharmacology, and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jeanot Muster
- Howard Hughes Medical Institute, Department of Pharmacology, and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephane Angers
- Howard Hughes Medical Institute, Department of Pharmacology, and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Randall T. Moon
- Howard Hughes Medical Institute, Department of Pharmacology, and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Benjamin N. R. Cheyette
- Department of Psychiatry, and Graduate Program in Developmental Biology, Program in Biological Sciences, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|