51
|
The Impact of Drosophila Awd/NME1/2 Levels on Notch and Wg Signaling Pathways. Int J Mol Sci 2020; 21:ijms21197257. [PMID: 33019537 PMCID: PMC7582475 DOI: 10.3390/ijms21197257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/18/2020] [Accepted: 09/29/2020] [Indexed: 01/09/2023] Open
Abstract
Awd, the Drosophila homologue of NME1/2 metastasis suppressors, plays key roles in many signaling pathways. Mosaic analysis of the null awdJ2A4 allele showed that loss of awd gene function blocks Notch signaling and the expression of its target genes including the Wingless (Wg/Wnt1) morphogen. We also showed that RNA interference (RNAi)-mediated awd silencing (awdi) in larval wing disc leads to chromosomal instability (CIN) and to Jun amino-terminal kinases (JNK)-mediated cell death. Here we show that this cell death is independent of p53 activity. Based on our previous finding showing that forced survival of awdi-CIN cells leads to aneuploidy without the hyperproliferative effect, we investigated the Wg expression in awdi wing disc cells. Interestingly, the Wg protein is expressed in its correct dorso-ventral domain but shows an altered cellular distribution which impairs its signaling. Further, we show that RNAi-mediated knock down of awd in wing discs does not affect Notch signaling. Thus, our analysis of the hypomorphic phenotype arising from awd downregulation uncovers a dose-dependent effect of Awd in Notch and Wg signaling.
Collapse
|
52
|
Li X, Ortiz MA, Kotula L. The physiological role of Wnt pathway in normal development and cancer. Exp Biol Med (Maywood) 2020; 245:411-426. [PMID: 31996036 PMCID: PMC7082880 DOI: 10.1177/1535370220901683] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the decades, many studies have illustrated the critical roles of Wnt signaling pathways in both developmental processes as well as tumorigenesis. Due to the complexity of Wnt signaling regulation, there are still questions to be addressed about ways cells are able to manipulate different types of Wnt pathways in order to fulfill the requirements for normal or cancer development. In this review, we will describe different types of Wnt signaling pathways and their roles in both normal developmental processes and their role in cancer development and progression. Additionally, we will briefly introduce new strategies currently in clinical trials targeting Wnt signaling pathway components for cancer therapy.
Collapse
Affiliation(s)
- Xiang Li
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Maria A Ortiz
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Leszek Kotula
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
53
|
Zhong Z, Virshup DM. Wnt Signaling and Drug Resistance in Cancer. Mol Pharmacol 2020; 97:72-89. [PMID: 31787618 DOI: 10.1124/mol.119.117978] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022] Open
Abstract
Wnts are secreted proteins that bind to cell surface receptors to activate downstream signaling cascades. Normal Wnt signaling plays key roles in embryonic development and adult tissue homeostasis. The secretion of Wnt ligands, the turnover of Wnt receptors, and the signaling transduction are tightly regulated and fine-tuned to keep the signaling output "just right." Hyperactivated Wnt signaling due to recurrent genetic alterations drives several human cancers. Elevated Wnt signaling also confers resistance to multiple conventional and targeted cancer therapies through diverse mechanisms including maintaining the cancer stem cell population, enhancing DNA damage repair, facilitating transcriptional plasticity, and promoting immune evasion. Different classes of Wnt signaling inhibitors targeting key nodes of the pathway have been developed and show efficacy in treating Wnt-driven cancers and subverting Wnt-mediated therapy resistance in preclinical studies. Several of these inhibitors have advanced to clinical trials, both singly and in combination with other existing US Food and Drug Administration-approved anti-cancer modalities. In the near future, pharmacological inhibition of Wnt signaling may be a real choice for patients with cancer. SIGNIFICANCE STATEMENT: The latest insights in Wnt signaling, ranging from basic biology to therapeutic implications in cancer, are reviewed. Recent studies extend understanding of this ancient signaling pathway and describe the development and improvement of anti-Wnt therapeutic modalities for cancer.
Collapse
Affiliation(s)
- Zheng Zhong
- Department of Physiology, National University of Singapore, Singapore, Singapore (Z.Z.); Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore (Z.Z., D.M.V.); and Department of Pediatrics, Duke University, Durham, North Carolina (D.M.V.)
| | - David M Virshup
- Department of Physiology, National University of Singapore, Singapore, Singapore (Z.Z.); Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore (Z.Z., D.M.V.); and Department of Pediatrics, Duke University, Durham, North Carolina (D.M.V.)
| |
Collapse
|
54
|
Dzobo K, Thomford NE, Senthebane DA. Targeting the Versatile Wnt/β-Catenin Pathway in Cancer Biology and Therapeutics: From Concept to Actionable Strategy. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:517-538. [PMID: 31613700 DOI: 10.1089/omi.2019.0147] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This expert review offers a critical synthesis of the latest insights and approaches at targeting the Wnt/β-catenin pathway in various cancers such as colorectal cancer, melanoma, leukemia, and breast and lung cancers. Notably, from organogenesis to cancer, the Wnt/β-catenin signaling displays varied and highly versatile biological functions in animals, with virtually all tissues requiring the Wnt/β-catenin signaling in one way or the other. Aberrant expression of the members of the Wnt/β-catenin has been implicated in many pathological conditions, particularly in human cancers. Mutations in the Wnt/β-catenin pathway genes have been noted in diverse cancers. Biochemical and genetic data support the idea that inhibition of Wnt/β-catenin signaling is beneficial in cancer therapeutics. The interaction of this important pathway with other signaling systems is also noteworthy, but remains as an area for further research and discovery. In addition, formation of different complexes by components of the Wnt/β-catenin pathway and the precise roles of these complexes in the cytoplasmic milieu are yet to be fully elucidated. This article highlights the latest medical technologies in imaging, single-cell omics, use of artificial intelligence (e.g., machine learning techniques), genome sequencing, quantum computing, molecular docking, and computational softwares in modeling interactions between molecules and predicting protein-protein and compound-protein interactions pertinent to the biology and therapeutic value of the Wnt/β-catenin signaling pathway. We discuss these emerging technologies in relationship to what is currently needed to move from concept to actionable strategies in translating the Wnt/β-catenin laboratory discoveries to Wnt-targeted cancer therapies and diagnostics in the clinic.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dimakatso A Senthebane
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
55
|
Exocyst-mediated apical Wg secretion activates signaling in the Drosophila wing epithelium. PLoS Genet 2019; 15:e1008351. [PMID: 31527874 PMCID: PMC6764796 DOI: 10.1371/journal.pgen.1008351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 09/27/2019] [Accepted: 08/07/2019] [Indexed: 01/19/2023] Open
Abstract
Wnt proteins are secreted signaling factors that regulate cell fate specification and patterning decisions throughout the animal kingdom. In the Drosophila wing epithelium, Wingless (Wg, the homolog of Wnt1) is secreted from a narrow strip of cells at the dorsal-ventral boundary. However, the route of Wg secretion in polarized epithelial cells remains poorly understood and key proteins involved in this process are still unknown. Here, we performed an in vivo RNAi screen and identified members of the exocyst complex to be required for apical but not basolateral Wg secretion. Specifically blocking the apical Wg secretion leads to reduced downstream signaling. Using an in vivo ‘temporal-rescue’ assay, our results further indicate that apically secreted Wg activates target genes that require high signaling activity. In conclusion, our results demonstrate that the exocyst is required for an apical route of Wg secretion from polarized wing epithelial cells. Regulation of Wnt signaling and the production of Wnt ligands is crucial for proper development and homeostasis, as dysregulation leads to developmental defects and diseases such as cancer. This study addresses the question of how functional Wnt ligands are secreted by epithelial cells. By using the polarized epithelium of the developing Drosophila wing as a model system to study Wnt/Wg secretion, the authors performed a large-scale RNAi screen and identified proteins of the exocyst complex to be required for Wnt signaling. The study shows that exocyst complex preferentially regulates apical secretion of Wg proteins. Taken together, this study identifies routes and regulators for secretion of signaling-active Wnt proteins from polarized epithelial cells.
Collapse
|
56
|
Petko J, Thileepan M, Sargen M, Canfield V, Levenson R. Alternative splicing of the Wnt trafficking protein, Wntless and its effects on protein-protein interactions. BMC Mol Cell Biol 2019; 20:22. [PMID: 31286866 PMCID: PMC6615345 DOI: 10.1186/s12860-019-0208-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022] Open
Abstract
Background Wntless (Wls) is a protein that regulates secretion of Wnt signaling molecules from Wnt-producing cells. Wnt signaling is known to be critical for several developmental and homeostatic processes. However, Wnt-independent functions of Wls are now being elucidated. Primates express an alternative splice variant of Wls (here termed WlsX). WlsX contains an alternatively spliced COOH-terminus, and does not appear to be able to sustain significant levels of WNT secretion because of its inability to undergo retrograde trafficking to the endoplasmic reticulum. The functional significance for this alternatively spliced form of Wls has not yet been elucidated. We previously identified a cohort of Wls interacting proteins using a combination of yeast 2-hybrid and candidate gene approaches. Results In the present study, we analyzed the interaction of WlsX with previously identified Wls interactors, and additionally screened for novel protein interactors of WlsX utilizing a membrane yeast two hybrid screen. Three novel Wls interactors, Glycoprotein M6A (GPM6A), Alkylglycerol Monooxygenase (AGMO), and ORAI1 were identified. Each of these novel WlsX interactors, as well as all other Wls interacting proteins identified previously, with the exception of the mu-opioid receptor, were found to interact with both Wls and WlsX splice forms. We show that WlsX can form homodimers, but that WlsX may not interact with Wls. Conclusions WlsX has the ability to form homodimers and to interact with most known Wls interacting proteins. Taken together, our results suggest that Wls and WlsX may have overlapping, but distinct functions, including sensitivity to opioid drugs. While studies have focused on the ability of Wls interacting proteins to affect Wnt secretion, future efforts will explore the reciprocal regulation of these proteins by Wls, possibly via Wnt-independent mechanisms.
Collapse
Affiliation(s)
| | | | - Molly Sargen
- Biology Department, Penn State York, York, Pa, USA
| | - Victor Canfield
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Robert Levenson
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
57
|
Abstract
Wnt proteins are secreted glycoproteins that regulate multiple processes crucial to the development and tissue homeostasis of multicellular organisms, including tissue patterning, proliferation, cell fate specification, cell polarity and migration. To elicit these effects, Wnts act as autocrine as well as paracrine signalling molecules between Wnt-producing and Wnt-receiving cells. More than 40 years after the discovery of the Wg/Wnt pathway, it is still unclear how they are transported to fulfil their paracrine signalling functions. Several mechanisms have been proposed to mediate intercellular Wnt transport, including Wnt-binding proteins, lipoproteins, exosomes and cytonemes. In this Review, we describe the evidence for each proposed mechanism, and discuss how they may contribute to Wnt dispersal in tissue-specific and context-dependent manners, to regulate embryonic development precisely and maintain the internal steady state within a defined tissue.
Collapse
Affiliation(s)
- Daniel Routledge
- Living Systems Institute, Biosciences, College of Life and Environmental Science, University of Exeter, Exeter EX4 4QD, UK
| | - Steffen Scholpp
- Living Systems Institute, Biosciences, College of Life and Environmental Science, University of Exeter, Exeter EX4 4QD, UK
| |
Collapse
|
58
|
Jiang Y, Lin X, Kapoor A, He L, Wei F, Gu Y, Mei W, Zhao K, Yang H, Tang D. FAM84B promotes prostate tumorigenesis through a network alteration. Ther Adv Med Oncol 2019; 11:1758835919846372. [PMID: 31205500 PMCID: PMC6535720 DOI: 10.1177/1758835919846372] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/13/2019] [Indexed: 01/04/2023] Open
Abstract
Background: The aim of this study was to investigate the contributions of FAM84B in prostate tumorigenesis and progression. Methods: A FAM84B mutant with deletion of its HRASLS domain (ΔHRASLS) was constructed. DU145 prostate cancer (PC) cells stably expressing an empty vector (EV), FAM84B, or FAM84B (ΔHRASLS) were produced. These lines were examined for proliferation, invasion, and growth in soft agar in vitro. DU145 EV and FAM84B cells were investigated for tumor growth and lung metastasis in NOD/SCID mice. The transcriptome of DU145 EV xenografts (n = 2) and DU145 FAM84B tumors (n = 2) was determined using RNA sequencing, and analyzed for pathway alterations. The FAM84B-affected network was evaluated for an association with PC recurrence. Results: FAM84B but not FAM84B (ΔHRASLS) increased DU145 cell invasion and growth in soft agar. Co-immunoprecipitation and co-localization analyses revealed an interaction between FAM84B and FAM84B (ΔHRASLS), suggesting an intramolecular association among FAM84B molecules. FAM84B significantly enhanced DU145 cell-derived xenografts and lung metastasis. In comparison with DU145 EV cell-produced tumors, those generated by DU145 FAM84B cells showed a large number of differentially expressed genes (DEGs; n = 4976). A total of 51 pathways were enriched in these DEGs, which function in the Golgi-to-endoplasmic reticulum processes, cell cycle checkpoints, mitochondrial events, and protein translation. A novel 27-gene signature (SigFAM) was derived from these DEGs; SigFAM robustly stratifies PC recurrence in two large PC populations (n = 490, p = 0; n = 140, p = 4e−11), and remains an independent risk factor of PC recurrence after adjusting for age at diagnosis, Gleason scores, surgical margin, and tumor stages. Conclusions: FAM84B promotes prostate tumorigenesis through a complex network that predicts PC recurrence.
Collapse
Affiliation(s)
- Yanzhi Jiang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China Department of Medicine, McMaster University, Hamilton, ON, Canada Father Sean O'Sullivan Research Institute, St. Joseph's Hospital, Hamilton, ON. Canada Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, ON, Canada Hamilton Urologic Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON, Canada
| | - Xiaozeng Lin
- Department of Medicine, McMaster University, Hamilton, ON, Canada Father Sean O'Sullivan Research Institute, St. Joseph's Hospital/Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, ON, Canada Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON, Canada
| | - Anil Kapoor
- Father Sean O'Sullivan Research Institute, St. Joseph's Hospital, Hamilton, ON, Canada Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON, Canada Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Lizhi He
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Fengxiang Wei
- The Genetics Laboratory, Longgang District Maternity and Child Healthcare Hospital, Longgang District, Shenzhen, Guangdong, China
| | - Yan Gu
- Department of Medicine, McMaster University, Hamilton, ON, Canada Father Sean O'Sullivan Research Institute, St. Joseph's Hospital/Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, ON, Canada Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON, Canada
| | - Wenjuan Mei
- Department of Medicine, McMaster University, Hamilton, ON, Canada Father Sean O'Sullivan Research Institute, St. Joseph's Hospital Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, ON, Canada Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON, Canada Department of Nephrology, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Kuncheng Zhao
- Department of Medicine, McMaster University, Hamilton, ON, Canada Father Sean O'Sullivan Research Institute, St. Joseph's Hospital/Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, ON, Canada Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON, Canada
| | - Huixiang Yang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Damu Tang
- Department of Medicine, McMaster University, T3310, St. Joseph's Hospital, 50 Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada
| |
Collapse
|
59
|
Li H, Li Q, Ma Z, Zhou Z, Fan J, Jin Y, Wu Y, Cheng F, Liang P. AID modulates carcinogenesis network via DNA demethylation in bladder urothelial cell carcinoma. Cell Death Dis 2019; 10:251. [PMID: 30874539 PMCID: PMC6420503 DOI: 10.1038/s41419-019-1472-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/12/2018] [Accepted: 02/18/2019] [Indexed: 12/25/2022]
Abstract
Bladder cancer is one of the most common malignant diseases in the urinary system, with poor survival after metastasis. Activation-induced cytidine deaminase (AID), a versatile enzyme involved in antibody diversification, is an oncogenic gene that induces somatic hypermutation and class-switch recombination (CSR). However, the contribution of AID-mediated DNA demethylation to bladder urothelial cell carcinoma (BUCC) remains unclear. Herein, we evaluated the impact on BUCC caused by AID and explored the gene network downstream of AID by using a proteomic approach. Lentiviral vector containing AID-specific shRNA significantly reduced AID expression in T24 and 5637 cells. Silencing AID expression remarkably inhibited tumour malignancies, including cell proliferation, invasion and migration. We used Isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics analysis technology to study the underpinning mechanism in monoclonal T24 cells, with or without AID knockdown. Among the 6452 proteins identified, 99 and 142 proteins in shAICDA-T24 cells were significantly up- or downregulated, respectively (1.2-fold change) compared with the NC-T24 control. After a pipeline of bioinformatics analyses, we identified three tumour-associated factors, namely, matrix metallopeptidase 14 (MMP14), C–X–C motif chemokine ligand 12 and wntless Wnt ligand secretion mediator, which were further confirmed in human BUCC tissues. Nonetheless, only MMP14 was sensitive to the DNA demethylation molecule 5-aza-2’-deoxycytidine (5-azadC; 5 μM), which reversed the inhibition of carcinogenesis by AID silence in T24 and 5637 cells. Overall, AID is an oncogene that mediates tumourigenesis via DNA demethylation. Our findings provide novel insights into the clinical treatment for BUCC.
Collapse
Affiliation(s)
- Haoyong Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qi Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.,Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Zhe Ma
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Zhiyan Zhou
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Jinfeng Fan
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Yingxia Jin
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yaoxi Wu
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| | - Peiyu Liang
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China.
| |
Collapse
|
60
|
Moti N, Yu J, Boncompain G, Perez F, Virshup DM. Wnt traffic from endoplasmic reticulum to filopodia. PLoS One 2019; 14:e0212711. [PMID: 30794657 PMCID: PMC6386245 DOI: 10.1371/journal.pone.0212711] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/07/2019] [Indexed: 11/26/2022] Open
Abstract
Wnts are a family of secreted palmitoleated glycoproteins that play key roles in cell to cell communication during development and regulate stem cell compartments in adults. Wnt receptors, downstream signaling cascades and target pathways have been extensively studied while less is known about how Wnts are secreted and move from producing cells to receiving cells. We used the synchronization system called Retention Using Selective Hook (RUSH) to study Wnt trafficking from endoplasmic reticulum to Golgi and then to plasma membrane and filopodia in real time. Inhibition of porcupine (PORCN) or knockout of Wntless (WLS) blocked Wnt exit from the ER. Wnt-containing vesicles paused at sub-cortical regions of the plasma membrane before exiting the cell. Wnt-containing vesicles were associated with filopodia extending to adjacent cells. These data visualize and confirm the role of WLS and PORCN in ER exit of Wnts and support the role of filopodia in Wnt signaling.
Collapse
Affiliation(s)
- Naushad Moti
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Jia Yu
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Gaelle Boncompain
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR144 “Cell Biology and Cancer”, Paris, France
| | - Franck Perez
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR144 “Cell Biology and Cancer”, Paris, France
| | - David M. Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
- Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
61
|
Hosseini V, Dani C, Geranmayeh MH, Mohammadzadeh F, Nazari Soltan Ahmad S, Darabi M. Wnt lipidation: Roles in trafficking, modulation, and function. J Cell Physiol 2018; 234:8040-8054. [PMID: 30341908 DOI: 10.1002/jcp.27570] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/17/2018] [Indexed: 12/30/2022]
Abstract
The Wnt signaling pathway consists of various downstream target proteins that have substantial roles in mammalian cell proliferation, differentiation, and development. Its aberrant activity can lead to uncontrolled proliferation and tumorigenesis. The posttranslational connection of fatty acyl chains to Wnt proteins provides the unique capacity for regulation of Wnt activity. In spite of the past belief that Wnt molecules are subject to dual acylation, it has been shown that these proteins have only one acylation site and undergo monounsaturated fatty acylation. The Wnt monounsaturated fatty acyl chain is more than just a hydrophobic coating and appears to be critical for Wnt signaling, transport, and receptor activation. Here, we provide an overview of recent findings in Wnt monounsaturated fatty acylation and the mechanism by which this lipid moiety regulates Wnt activity from the site of production to its receptor interactions.
Collapse
Affiliation(s)
- Vahid Hosseini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hossein Geranmayeh
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Mohammadzadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| |
Collapse
|
62
|
Seo J, Kee HJ, Choi HJ, Lee JE, Park SY, Lee SH, Jeong MH, Guk G, Lee S, Choi KC, Choi YY, Kim H, Noh SH, Yoon HG, Cheong JH. Inhibition of Wntless/GPR177 suppresses gastric tumorigenesis. BMB Rep 2018; 51:255-260. [PMID: 29555015 PMCID: PMC5988581 DOI: 10.5483/bmbrep.2018.51.5.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Indexed: 12/23/2022] Open
Abstract
Wntless/GPR177 functions as WNT ligand carrier protein and activator of WNT/β-catenin signaling, however, its molecular role in gastric cancer (GC) has remained elusive. We investigated the role of GPR177 in gastric tumorigenesis and provided the therapeutic potential of a clinical development of anti-GPR177 monoclonal antibodies. GPR177 mRNA expression was assessed in GC transcriptome data sets (GSE15459, n = 184; GSE66229, n = 300); protein expression was assessed in independent patient tumor tissues (Yonsei TMA, n = 909). GPR177 expression were associated with unfavorable prognosis [log-rank test, GSE15459 (P = 0.00736), GSE66229 (P = 0.0142), and Yonsei TMA (P = 0.0334)] and identified as an independent risk predictor of clinical outcomes: GSE15459 [hazard ratio (HR) 1.731 (95% confidence interval; CI; 1.103–2.715), P = 0.017], GSE66229 [HR 1.54 (95% CI, 1.10–2.151), P = 0.011], and Yonsei TMA [HR 1.254 (95% CI, 1.049–1.500), P = 0.013]. Either antibody treatment or GPR177 knockdown suppressed proliferation of GC cells and sensitized cells to apoptosis. And also inhibition of GPR177 suppresses in vitro and in vivo tumorogenesis in GC cells and inhibits WNT/β-catenin signaling. Finally, targeting and inhibition of GPR177 with antibody suppressed tumorigenesis in PDX model. Together, these results suggest GPR177 as a novel candidate for prognostic marker as well as a promising target for treatment of GC patients.
Collapse
Affiliation(s)
- Jaesung Seo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hyun Jung Kee
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hye Ji Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea; Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jae Eun Lee
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Soo-Yeon Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seung-Hyun Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Mi-Hyeon Jeong
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Garam Guk
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - SooYeon Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Kyung-Chul Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yoon Young Choi
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Sung Hoon Noh
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jae-Ho Cheong
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea; Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
63
|
McGough IJ, de Groot REA, Jellett AP, Betist MC, Varandas KC, Danson CM, Heesom KJ, Korswagen HC, Cullen PJ. SNX3-retromer requires an evolutionary conserved MON2:DOPEY2:ATP9A complex to mediate Wntless sorting and Wnt secretion. Nat Commun 2018; 9:3737. [PMID: 30213940 PMCID: PMC6137200 DOI: 10.1038/s41467-018-06114-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/13/2018] [Indexed: 12/28/2022] Open
Abstract
Wntless transports Wnt morphogens to the cell surface and is required for Wnt secretion and morphogenic gradients formation. Recycling of endocytosed Wntless requires the sorting nexin-3 (SNX3)-retromer-dependent endosome-to-Golgi transport pathway. Here we demonstrate the essential role of SNX3-retromer assembly for Wntless transport and report that SNX3 associates with an evolutionary conserved endosome-associated membrane re-modelling complex composed of MON2, DOPEY2 and the putative aminophospholipid translocase, ATP9A. In vivo suppression of Ce-mon-2, Ce-pad-1 or Ce-tat-5 (respective MON2, DOPEY2 and ATP9A orthologues) phenocopy a loss of SNX3-retromer function, leading to enhanced lysosomal degradation of Wntless and a Wnt phenotype. Perturbed Wnt signalling is also observed upon overexpression of an ATPase-inhibited TAT-5(E246Q) mutant, suggesting a role for phospholipid flippase activity during SNX3-retromer-mediated Wntless sorting. Together, these findings provide in vitro and in vivo mechanistic details to describe SNX3-retromer-mediated transport during Wnt secretion and the formation of Wnt-morphogenic gradients.
Collapse
Affiliation(s)
- Ian J McGough
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
| | - Reinoud E A de Groot
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands
| | - Adam P Jellett
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Marco C Betist
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands
| | - Katherine C Varandas
- Program in Cell Biology, University of California, San Francisco, 16th Street, San Francisco, CA, 94158, USA
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Chris M Danson
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Kate J Heesom
- Proteomics Facility, School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Hendrik C Korswagen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| | - Peter J Cullen
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
64
|
Grainger S, Willert K. Mechanisms of Wnt signaling and control. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10:e1422. [PMID: 29600540 PMCID: PMC6165711 DOI: 10.1002/wsbm.1422] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/14/2018] [Accepted: 02/22/2018] [Indexed: 01/17/2023]
Abstract
The Wnt signaling pathway is a highly conserved system that regulates complex biological processes across all metazoan species. At the cellular level, secreted Wnt proteins serve to break symmetry and provide cells with positional information that is critical to the patterning of the entire body plan. At the organismal level, Wnt signals are employed to orchestrate fundamental developmental processes, including the specification of the anterior-posterior body axis, induction of the primitive streak and ensuing gastrulation movements, and the generation of cell and tissue diversity. Wnt functions extend into adulthood where they regulate stem cell behavior, tissue homeostasis, and damage repair. Disruption of Wnt signaling activity during embryonic development or in adults results in a spectrum of abnormalities and diseases, including cancer. The molecular mechanisms that underlie the myriad of Wnt-regulated biological effects have been the subject of intense research for over three decades. This review is intended to summarize our current understanding of how Wnt signals are generated and interpreted. This article is categorized under: Biological Mechanisms > Cell Signaling Developmental Biology > Stem Cell Biology and Regeneration.
Collapse
Affiliation(s)
- Stephanie Grainger
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla California
| | - Karl Willert
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla California
| |
Collapse
|
65
|
Leibing T, Géraud C, Augustin I, Boutros M, Augustin HG, Okun JG, Langhans C, Zierow J, Wohlfeil SA, Olsavszky V, Schledzewski K, Goerdt S, Koch P. Angiocrine Wnt signaling controls liver growth and metabolic maturation in mice. Hepatology 2018; 68:707-722. [PMID: 29059455 PMCID: PMC6099291 DOI: 10.1002/hep.29613] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/08/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022]
Abstract
UNLABELLED Postnatal liver development is characterized by hepatocyte growth, proliferation, and functional maturation. Notably, canonical Wnt signaling in hepatocytes has been identified as an important regulator of final adult liver size and metabolic liver zonation. The cellular origin of Wnt ligands responsible for homeostatic liver/body weight ratio (LW/BW) remained unclear, which was also attributable to a lack of suitable endothelial Cre driver mice. To comprehensively analyze the effects of hepatic angiocrine Wnt signaling on liver development and metabolic functions, we used endothelial subtype-specific Stab2-Cre driver mice to delete Wls from hepatic endothelial cells (HECs). The resultant Stab2-Cretg/wt ;Wlsfl/fl (Wls-HECKO) mice were viable, but showed a significantly reduced LW/BW. Specifically, ablation of angiocrine Wnt signaling impaired metabolic zonation in the liver, as shown by loss of pericentral, β-catenin-dependent target genes such as glutamine synthase (Glul), RhBg, Axin2, and cytochrome P450 2E1, as well as by extended expression of periportal genes such as arginase 1. Furthermore, endothelial subtype-specific expression of a c-terminally YFP-tagged Wls fusion protein in Wls-HECKO mice (Stab2-Cretg/wt ;Wlsfl/fl ;Rosa26:Wls-YFPfl/wt [Wls-rescue]) restored metabolic liver zonation. Interestingly, lipid metabolism was altered in Wls-HECKO mice exhibiting significantly reduced plasma cholesterol levels, while maintaining normal plasma triglyceride and blood glucose concentrations. On the contrary, zonal expression of Endomucin, LYVE1, and other markers of HEC heterogeneity were not altered in Wls-HECKO livers. CONCLUSION Angiocrine Wnt signaling controls liver growth as well as development of metabolic liver zonation in mice, whereas intrahepatic HEC zonation is not affected. (Hepatology 2017).
Collapse
Affiliation(s)
- Thomas Leibing
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Cyrill Géraud
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Iris Augustin
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics and Heidelberg University, Faculty of Medicine Mannheim, Department of Cell and Molecular BiologyHeidelbergGermany,Molecular Cell Biology and Plant Cell TechnologyUniversity of Applied Sciences Weihenstephan‐TriesdorfFreisingGermany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics and Heidelberg University, Faculty of Medicine Mannheim, Department of Cell and Molecular BiologyHeidelbergGermany
| | - Hellmut G. Augustin
- Division of Vascular Oncology and Metastasis (DKFZ‐ZMBH Alliance)DKFZHeidelbergGermany,Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Jürgen G. Okun
- Department of General Pediatrics, Division of Inherited Metabolic DiseasesUniversity Children's HospitalHeidelbergGermany
| | - Claus‐Dieter Langhans
- Department of General Pediatrics, Division of Inherited Metabolic DiseasesUniversity Children's HospitalHeidelbergGermany
| | - Johanna Zierow
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Sebastian A. Wohlfeil
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Victor Olsavszky
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Kai Schledzewski
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Sergij Goerdt
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany,European Center for AngioscienceMedical Faculty Mannheim, University of HeidelbergMannheimGermany
| | - Philipp‐Sebastian Koch
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| |
Collapse
|
66
|
Mattes B, Dang Y, Greicius G, Kaufmann LT, Prunsche B, Rosenbauer J, Stegmaier J, Mikut R, Özbek S, Nienhaus GU, Schug A, Virshup DM, Scholpp S. Wnt/PCP controls spreading of Wnt/β-catenin signals by cytonemes in vertebrates. eLife 2018; 7:36953. [PMID: 30060804 PMCID: PMC6086664 DOI: 10.7554/elife.36953] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022] Open
Abstract
Signaling filopodia, termed cytonemes, are dynamic actin-based membrane structures that regulate the exchange of signaling molecules and their receptors within tissues. However, how cytoneme formation is regulated remains unclear. Here, we show that Wnt/planar cell polarity (PCP) autocrine signaling controls the emergence of cytonemes, and that cytonemes subsequently control paracrine Wnt/β-catenin signal activation. Upon binding of the Wnt family member Wnt8a, the receptor tyrosine kinase Ror2 becomes activated. Ror2/PCP signaling leads to the induction of cytonemes, which mediate the transport of Wnt8a to neighboring cells. In the Wnt-receiving cells, Wnt8a on cytonemes triggers Wnt/β-catenin-dependent gene transcription and proliferation. We show that cytoneme-based Wnt transport operates in diverse processes, including zebrafish development, murine intestinal crypt and human cancer organoids, demonstrating that Wnt transport by cytonemes and its control via the Ror2 pathway is highly conserved in vertebrates. Communication helps the cells that make up tissues and organs to work together as a team. One way that cells share information with each other as tissues grow and develop is by exchanging signaling proteins. These interact with receptors on the surface of other cells; this causes the cell to change how it behaves. The Wnt family of signaling proteins orchestrate organ development. Wnt proteins influence which types of cells develop, how fast they divide, and how and when they move. Relatively few cells, or small groups of cells, in developing tissues produce Wnt proteins, while larger groups nearby respond to the signals. We do not fully understand how Wnt proteins travel between cells, but recent work revealed an unexpected mechanism – cells seem to hand-deliver their messages. Finger-like structures called cytonemes grow out of the cell membrane and carry Wnt proteins to their destination. If the cytonemes do not form properly the target cells do not behave correctly, which can lead to severe tissue malformation. Mattes et al. have now investigated how cytonemes form using a combination of state-of-the-art genetic and high-resolution imaging techniques. In initial experiments involving zebrafish cells that were grown in the laboratory, Mattes et al. found that the Wnt proteins kick start their own transport; before they travel to their destination, they act on the cells that made them. A Wnt protein called Wnt8a activates the receptor Ror2 on the surface of the signal-producing cell. Ror2 then triggers signals inside the cell that begin the assembly of the cytonemes. The more Ror2 is activated, the more cytonemes the cell makes, and the more Wnt signals it can send out. This mechanism operates in various tissues: Ror2 also controls the cytoneme transport process in living zebrafish embryos, the mouse intestine and human stomach tumors. This knowledge will help researchers to develop new ways to control Wnt signaling, which could help to produce new treatments for diseases ranging from cancers (for example in the stomach and bowel) to degenerative diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Benjamin Mattes
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Yonglong Dang
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gediminas Greicius
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Benedikt Prunsche
- Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Jakob Rosenbauer
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany
| | - Johannes Stegmaier
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Suat Özbek
- Centre of Organismal Studies, University of Heidelberg, Karlsruhe, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Department of Physics, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Alexander Schug
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany.,Steinbuch Centre for Computing, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - David M Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
67
|
A versatile nanobody-based toolkit to analyze retrograde transport from the cell surface. Proc Natl Acad Sci U S A 2018; 115:E6227-E6236. [PMID: 29915061 DOI: 10.1073/pnas.1801865115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Retrograde transport of membranes and proteins from the cell surface to the Golgi and beyond is essential to maintain homeostasis, compartment identity, and physiological functions. To study retrograde traffic biochemically, by live-cell imaging or by electron microscopy, we engineered functionalized anti-GFP nanobodies (camelid VHH antibody domains) to be bacterially expressed and purified. Tyrosine sulfation consensus sequences were fused to the nanobody for biochemical detection of trans-Golgi arrival, fluorophores for fluorescence microscopy and live imaging, and APEX2 (ascorbate peroxidase 2) for electron microscopy and compartment ablation. These functionalized nanobodies are specifically captured by GFP-modified reporter proteins at the cell surface and transported piggyback to the reporters' homing compartments. As an application of this tool, we have used it to determine the contribution of adaptor protein-1/clathrin in retrograde transport kinetics of the mannose-6-phosphate receptors from endosomes back to the trans-Golgi network. Our experiments establish functionalized nanobodies as a powerful tool to demonstrate and quantify retrograde transport pathways.
Collapse
|
68
|
Azevedo C, Macedo MH, Sarmento B. Strategies for the enhanced intracellular delivery of nanomaterials. Drug Discov Today 2018; 23:944-959. [PMID: 28919437 PMCID: PMC7108348 DOI: 10.1016/j.drudis.2017.08.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/13/2017] [Accepted: 08/23/2017] [Indexed: 11/25/2022]
Abstract
The intracellular delivery of nanomaterials and drugs has been attracting increasing research interest, mainly because of their important effects and functions in several organelles. Targeting specific organelles can help treat or decrease the symptoms of diabetes, cancer, infectious, and autoimmune diseases. Tuning biological and chemical properties enables the creation of functionalized nanomaterials with enhanced intracellular uptake, ability to escape premature lysosome degradation, and to reach a specific target. Here, we provide an update of recent advances in the intracellular delivery mechanisms that could help drugs reach their target more efficiently.
Collapse
Affiliation(s)
- Cláudia Azevedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Maria Helena Macedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde, Gandra, Portugal.
| |
Collapse
|
69
|
Abstract
Spatial organization of membrane domains within cells and cells within tissues is key to the development of organisms and the maintenance of adult tissue. Cell polarization is crucial for correct cell-cell signalling, which, in turn, promotes cell differentiation and tissue patterning. However, the mechanisms linking internal cell polarity to intercellular signalling are just beginning to be unravelled. The Hedgehog (Hh) and Wnt pathways are major directors of development and their malfunction can cause severe disorders like cancer. Here we discuss parallel advances into understanding the mechanism of Hedgehog and Wnt signal dissemination and reception. We hypothesize that cell polarization of the signal-sending and signal-receiving cells is crucial for proper signal spreading and activation of the pathway and, thus, fundamental for development of multicellular organisms.
Collapse
|
70
|
Liao CP, Li H, Lee HH, Chien CT, Pan CL. Cell-Autonomous Regulation of Dendrite Self-Avoidance by the Wnt Secretory Factor MIG-14/Wntless. Neuron 2018; 98:320-334.e6. [DOI: 10.1016/j.neuron.2018.03.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/06/2018] [Accepted: 03/16/2018] [Indexed: 11/26/2022]
|
71
|
Leucht P, Lee S, Yim N. Wnt signaling and bone regeneration: Can't have one without the other. Biomaterials 2018; 196:46-50. [PMID: 29573821 DOI: 10.1016/j.biomaterials.2018.03.029] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 12/25/2022]
Abstract
Advances in the understanding of the complexities of the Wnt signaling pathway during development and tissue homeostasis have made the Wnt pathway one of the prime candidates for translational applications during tissue regeneration. Wnts are key components of the stem cell niche and are short range signaling molecules responsible for cellular decisions such as proliferation and differentiation. Systemic treatment using biologics targeting the Wnt signaling pathway have shown promising early results and will likely enter the clinical arena in the near future. This comprehensive review summarizes the intricacies how Wnts function in the context of the bone regeneration.
Collapse
Affiliation(s)
- Philipp Leucht
- NYU Langone Health, Departments of Orthopaedic Surgery and Cell Biology, New York, NY, USA.
| | - Sooyeon Lee
- NYU Langone Health, Departments of Orthopaedic Surgery and Cell Biology, New York, NY, USA
| | - Nury Yim
- NYU Langone Health, Departments of Orthopaedic Surgery and Cell Biology, New York, NY, USA
| |
Collapse
|
72
|
Brunt L, Scholpp S. The function of endocytosis in Wnt signaling. Cell Mol Life Sci 2018; 75:785-795. [PMID: 28913633 PMCID: PMC5809524 DOI: 10.1007/s00018-017-2654-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/17/2017] [Accepted: 09/06/2017] [Indexed: 10/31/2022]
Abstract
Wnt growth factors regulate one of the most important signaling networks during development, tissue homeostasis and disease. Despite the biological importance of Wnt signaling, the mechanism of endocytosis during this process is ill described. Wnt molecules can act as paracrine signals, which are secreted from the producing cells and transported through neighboring tissue to activate signaling in target cells. Endocytosis of the ligand is important at several stages of action: One central function of endocytic trafficking in the Wnt pathway occurs in the source cell. Furthermore, the β-catenin-dependent Wnt ligands require endocytosis for signal activation and to regulate gene transcription in the responding cells. Alternatively, Wnt/β-catenin-independent signaling regulates endocytosis of cell adherence plaques to control cell migration. In this comparative review, we elucidate these three fundamental interconnected functions, which together regulate cellular fate and cellular behavior. Based on established hypotheses and recent findings, we develop a revised picture for the complex function of endocytosis in the Wnt signaling network.
Collapse
Affiliation(s)
- Lucy Brunt
- Living Systems Institute, School of Biosciences, College of Life and Environmental Science, University of Exeter, Exeter, EX4 4QD, UK
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Science, University of Exeter, Exeter, EX4 4QD, UK.
- Institute of Toxicology and Genetics (ITG), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
73
|
Glaeser K, Urban M, Fenech E, Voloshanenko O, Kranz D, Lari F, Christianson JC, Boutros M. ERAD-dependent control of the Wnt secretory factor Evi. EMBO J 2018; 37:embj.201797311. [PMID: 29378775 PMCID: PMC5813261 DOI: 10.15252/embj.201797311] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/01/2017] [Accepted: 01/02/2018] [Indexed: 12/21/2022] Open
Abstract
Active regulation of protein abundance is an essential strategy to modulate cellular signaling pathways. Within the Wnt signaling cascade, regulated degradation of β-catenin by the ubiquitin-proteasome system (UPS) affects the outcome of canonical Wnt signaling. Here, we found that abundance of the Wnt cargo receptor Evi (Wls/GPR177), which is required for Wnt protein secretion, is also regulated by the UPS through endoplasmic reticulum (ER)-associated degradation (ERAD). In the absence of Wnt ligands, Evi is ubiquitinated and targeted for ERAD in a VCP-dependent manner. Ubiquitination of Evi involves the E2-conjugating enzyme UBE2J2 and the E3-ligase CGRRF1. Furthermore, we show that a triaging complex of Porcn and VCP determines whether Evi enters the secretory or the ERAD pathway. In this way, ERAD-dependent control of Evi availability impacts the scale of Wnt protein secretion by adjusting the amount of Evi to meet the requirement of Wnt protein export. As Wnt and Evi protein levels are often dysregulated in cancer, targeting regulatory ERAD components might be a useful approach for therapeutic interventions.
Collapse
Affiliation(s)
- Kathrin Glaeser
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Manuela Urban
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Emma Fenech
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Oksana Voloshanenko
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Dominique Kranz
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Federica Lari
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | | | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
74
|
Billmann M, Chaudhary V, ElMaghraby MF, Fischer B, Boutros M. Widespread Rewiring of Genetic Networks upon Cancer Signaling Pathway Activation. Cell Syst 2017; 6:52-64.e4. [PMID: 29199019 PMCID: PMC5791663 DOI: 10.1016/j.cels.2017.10.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/30/2017] [Accepted: 10/25/2017] [Indexed: 12/14/2022]
Abstract
Cellular signaling networks coordinate physiological processes in all multicellular organisms. Within networks, modules switch their function to control signaling activity in response to the cellular context. However, systematic approaches to map the interplay of such modules have been lacking. Here, we generated a context-dependent genetic interaction network of a metazoan's signaling pathway. Using Wnt signaling in Drosophila as a model, we measured >290,000 double perturbations of the pathway in a baseline state, after activation by Wnt ligand or after loss of the tumor suppressor APC. We found that genetic interactions within the Wnt network globally rewired after pathway activation. We derived between-state networks that showed how genes changed their function between state-specific networks. This related pathway inhibitors across states and identified genes required for pathway activation. For instance, we predicted and confirmed the ER-resident protein Catsup to be required for ligand-mediated Wnt signaling activation. Together, state-dependent and between-state genetic interaction networks identify responsive functional modules that control cellular pathways. Genetic interaction networks of Wnt signaling in three cellular states Networks rewire upon activation of Wnt pathway by ligand or by loss of APC Interaction profiles identify known and novel state-dependent pathway modules State-specific and between-state profile similarity identify signaling regulators
Collapse
Affiliation(s)
- Maximilian Billmann
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department of Cell and Molecular Biology, Faculty of Medicine Mannheim, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Varun Chaudhary
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department of Cell and Molecular Biology, Faculty of Medicine Mannheim, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Mostafa F ElMaghraby
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department of Cell and Molecular Biology, Faculty of Medicine Mannheim, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Bernd Fischer
- German Cancer Research Center (DKFZ), Computational Genome Biology Group, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department of Cell and Molecular Biology, Faculty of Medicine Mannheim, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| |
Collapse
|
75
|
Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017; 169:985-999. [PMID: 28575679 DOI: 10.1016/j.cell.2017.05.016] [Citation(s) in RCA: 3032] [Impact Index Per Article: 379.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/07/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022]
Abstract
The WNT signal transduction cascade is a main regulator of development throughout the animal kingdom. Wnts are also key drivers of most types of tissue stem cells in adult mammals. Unsurprisingly, mutated Wnt pathway components are causative to multiple growth-related pathologies and to cancer. Here, we describe the core Wnt/β-catenin signaling pathway, how it controls stem cells, and contributes to disease. Finally, we discuss strategies for Wnt-based therapies.
Collapse
|
76
|
Sun J, Yu S, Zhang X, Capac C, Aligbe O, Daudelin T, Bonder EM, Gao N. A Wntless-SEC12 complex on the ER membrane regulates early Wnt secretory vesicle assembly and mature ligand export. J Cell Sci 2017; 130:2159-2171. [PMID: 28515233 DOI: 10.1242/jcs.200634] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/11/2017] [Indexed: 01/02/2023] Open
Abstract
Wntless (Wls) transports Wnt molecules for secretion; however, the cellular mechanism underlying the initial assembly of Wnt secretory vesicles is still not fully defined. Here, we performed proteomic and mutagenic analyses of mammalian Wls, and report a mechanism for formation of early Wnt secretory vesicles on ER membrane. Wls forms a complex with SEC12 (also known as PREB), an ER membrane-localized guanine nucleotide-exchange factor (GEF) activator of the SAR1 (the SAR1A isoform) small GTPase. Compared to palmitoylation-deficient Wnt molecules, binding of mature Wnt to Wls increases Wls-SEC12 interaction and promotes association of Wls with SAR1, the key activator of the COPII machinery. Incorporation of Wls into this exporting ER compartment is affected by Wnt ligand binding and SEC12 binding to Wls, as well as the structural integrity and, potentially, the folding of the cytosolic tail of Wls. In contrast, Wls-SEC12 binding is stable, with the interacting interface biochemically mapped to cytosolic segments of individual proteins. Mutant Wls that fails to communicate with the COPII machinery cannot effectively support Wnt secretion. These data suggest that formation of early Wnt secretory vesicles is carefully regulated to ensure proper export of functional ligands.
Collapse
Affiliation(s)
- Jiaxin Sun
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Xiao Zhang
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Catherine Capac
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | | | - Timothy Daudelin
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA .,Rutgers Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
77
|
Schmid SC, Sathe A, Guerth F, Seitz AK, Heck MM, Maurer T, Schwarzenböck SM, Krause BJ, Schulz WA, Stoehr R, Gschwend JE, Retz M, Nawroth R. Wntless promotes bladder cancer growth and acts synergistically as a molecular target in combination with cisplatin. Urol Oncol 2017; 35:544.e1-544.e10. [PMID: 28501564 DOI: 10.1016/j.urolonc.2017.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 04/04/2017] [Accepted: 04/15/2017] [Indexed: 11/29/2022]
Abstract
PURPOSE To analyze the contribution of Wnt signaling pathway to bladder cancer growth in order to identify suitable target molecules for therapy. MATERIAL AND METHODS Expression of Wnt 2/4/7, LRP5/6, TCF1/2/4, LEF-1, and β-actin was detected by reverse transcription polymerase chain reaction in a panel of 9 and for Wntless (WLS) in 17 bladder cancer cell lines. Protein expression of WLS was detected in 6 cell lines. Wnt/β-catenin activity was analyzed using the TOPflash/FOPflash luciferase reporter assay. Expression level of β-catenin, WIF1, Dickkopf proteins (DKK), HSulf-2, sFRP4, and WLS was modulated by transfecting or infecting cells transiently or stably with respective shRNAs, siRNAs, or cDNAs. For protein detection, whole cell lysates were applied to sodium dodecyl sulfate polyacrylamide gel electrophoresis followed by immunoblots. Effects on cell growth were determined by cell viability assays and BrdU/APC incorporation/staining. For 3-dimensional tumor growth, the chicken chorioallantoic membrane model was used. Tumor growth was characterized by weight. RESULTS Expression of molecular components and activation of the Wnt signaling pathway could be detected in all cell lines. Expression level of β-catenin, WIF1, DKK, WLS, and HSulf-2 influenced Wnt activity. Expression of WLS was confirmed in 17 cell lines by reverse transcription polymerase chain reaction and in 6 cell lines by immunoblotting. WLS positively regulates Wnt signaling, cell proliferation, and tumor growth in vitro and in vivo. These effects could be reversed by the expression of the Wnt antagonist WIF1 and DKK. Synergistic activity of cisplatin and WLS inactivation by genetic silencing could be observed on cell viability. CONCLUSION The Wnt signaling pathway is ubiquitously activated in bladder cancer and regulates tumor growth. WLS might be a target protein for novel therapies in combination with established chemotherapy regimens.
Collapse
Affiliation(s)
- Sebastian C Schmid
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Anuja Sathe
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ferdinand Guerth
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Anna-Katharina Seitz
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Matthias M Heck
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Maurer
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Bernd J Krause
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Wolfgang A Schulz
- Department of Urology, Heinrich-Heine-University, Du¨sseldorf, Germany
| | - Robert Stoehr
- Department of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Jürgen E Gschwend
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Margitta Retz
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Roman Nawroth
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.
| |
Collapse
|
78
|
Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene 2017; 36:1461-1473. [PMID: 27617575 PMCID: PMC5357762 DOI: 10.1038/onc.2016.304] [Citation(s) in RCA: 1856] [Impact Index Per Article: 232.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/07/2016] [Accepted: 07/17/2016] [Indexed: 12/14/2022]
Abstract
Wnt signaling is one of the key cascades regulating development and stemness, and has also been tightly associated with cancer. The role of Wnt signaling in carcinogenesis has most prominently been described for colorectal cancer, but aberrant Wnt signaling is observed in many more cancer entities. Here, we review current insights into novel components of Wnt pathways and describe their impact on cancer development. Furthermore, we highlight expanding functions of Wnt signaling for both solid and liquid tumors. We also describe current findings how Wnt signaling affects maintenance of cancer stem cells, metastasis and immune control. Finally, we provide an overview of current strategies to antagonize Wnt signaling in cancer and challenges that are associated with such approaches.
Collapse
Affiliation(s)
- T Zhan
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg, Germany
- Heidelberg University, Department of Internal Medicine II, Medical Faculty Mannheim, Mannheim, Germany
| | - N Rindtorff
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg, Germany
| | - M Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
79
|
Varandas KC, Irannejad R, von Zastrow M. Retromer Endosome Exit Domains Serve Multiple Trafficking Destinations and Regulate Local G Protein Activation by GPCRs. Curr Biol 2016; 26:3129-3142. [PMID: 27839977 DOI: 10.1016/j.cub.2016.09.052] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 09/08/2016] [Accepted: 09/26/2016] [Indexed: 02/01/2023]
Abstract
Retromer mediates sequence-directed cargo exit from endosomes to support both endosome-to-Golgi (retrograde transport) and endosome-to-plasma membrane (recycling) itineraries. It is not known whether these trafficking functions require cargos to exit endosomes separately via distinct transport intermediates or whether the same retromer-coated carriers can support both itineraries. We addressed this question by comparing human Wntless (Wls) and β2 adrenergic receptor (β2AR), which require retromer physiologically for retrograde transport and recycling, respectively. We show here by direct visualization in living cells that both cargos transit primarily the same endosomes and exit via shared transport vesicles generated from a retromer-coated endosome domain. While both Wls and β2AR clearly localize to the same retromer-coated endosome domains, Wls is consistently enriched more strongly. This enrichment difference is determined by distinct motifs present in the cytoplasmic tail of each cargo, with Wls using tandem Φ-X-[L/M] motifs and β2AR using a PDZ motif. Exchanging these determinants reverses the enrichment phenotype of each cargo but does not change cargo itinerary, verifying the multifunctional nature of retromer and implying that additional sorting must occur downstream. Quantitative differences in the degree of cargo enrichment instead underlie a form of kinetic sorting that impacts the rate of cargo delivery via both itineraries and determines the ability of β2AR to activate its cognate G protein transducer locally from endosomes. We propose that mammalian retromer forms a multifunctional membrane coat that supports shared cargo exit for divergent trafficking itineraries and regulates signaling from endosomes.
Collapse
Affiliation(s)
- Katherine C Varandas
- Program in Cell Biology, University of California, San Francisco, 16(th) Street, San Francisco, CA 94158, USA
| | - Roshanak Irannejad
- Department of Psychiatry, UCSF School of Medicine, 16(th) Street, San Francisco, CA 94158, USA
| | - Mark von Zastrow
- Program in Cell Biology, University of California, San Francisco, 16(th) Street, San Francisco, CA 94158, USA; Department of Psychiatry, UCSF School of Medicine, 16(th) Street, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 16(th) Street, San Francisco, CA 94158, USA.
| |
Collapse
|
80
|
Adipose- and muscle-derived Wnts trigger pancreatic β-cell adaptation to systemic insulin resistance. Sci Rep 2016; 6:31553. [PMID: 27527335 PMCID: PMC4985739 DOI: 10.1038/srep31553] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023] Open
Abstract
Wnt signaling molecules are associated with obesity, hyperlipidemia, and type 2 diabetes (T2D). Here, we show that two Wnt proteins, WNT3a and WNT4, are specifically secreted by skeletal muscle and adipose tissue during the development of insulin resistance and play an important role in cross-talk between insulin-resistant tissues and pancreatic beta cells. The activation of Frizzled receptor and Wnt signaling in pancreatic islets via circulating WNT3a in blood resulted in higher insulin secretion and an increase in beta cell proliferation, thus leading to islet adaptation in a pre-diabetic state. Interestingly, in fully developed T2D, the expression profiles of Wnt3a and Wnt4 in adipose tissue and muscle cells and blood plasma levels of these proteins were opposite to the pre-diabetic state, thus favoring the downregulation of Wnt signaling in beta cells and resulting in dysfunctional pancreatic islets. These results demonstrate that alterations in the secretion profile of a canonical Wnt activator (WNT3a) and inhibitor (WNT4) from insulin-resistant tissues during the development of T2D are responsible for triggering progression from a pre-diabetic to a diabetic state. We also show here that WNT3a and WNT4 are potent myokines, and their expression and secretion are regulated in response to nutritional and metabolic changes.
Collapse
|
81
|
Galli LM, Zebarjadi N, Li L, Lingappa VR, Burrus LW. Divergent effects of Porcupine and Wntless on WNT1 trafficking, secretion, and signaling. Exp Cell Res 2016; 347:171-183. [PMID: 27492485 DOI: 10.1016/j.yexcr.2016.07.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 07/12/2016] [Accepted: 07/31/2016] [Indexed: 12/30/2022]
Abstract
Loss-of-function studies have identified Porcupine (PORCN) and Wntless (WLS) as essential mediators of Wnt secretion and signaling. Whereas PORCN is thought to palmitoylate Wnt proteins, WLS is believed to transport palmitoylated Wnt proteins to the cell surface. However, little is known about how these two proteins cooperate to regulate Wnt palmitoylation, trafficking, secretion, and signaling. We first investigated possible interactions between PORCN, WLS, and WNT1, by carrying out co-immunoprecipitation studies. These studies demonstrate the existence of a complex containing PORCN and WLS. They further show that PORCN and WLS compete for binding to WNT1. Then, we used gain-of-function studies to investigate the cooperation between PORCN and WLS as well as possible biochemical interactions between PORCN, WLS, and WNT1. Consistent with the proposed roles for PORCN and WLS, we show that overexpression of PORCN promotes palmitoylation of WNT1 while overexpression of WLS does not. Overexpression of PORCN enhances the ability of WLS to promote WNT1 trafficking to the cell surface as well as secretion, but decreases the ability of WLS to activate WNT1 signaling in target cell. These observations suggest that the levels of WNT1 on the cell surface and in the media are not the sole determinants of the activation of Wnt signaling in target cells.
Collapse
Affiliation(s)
- Lisa M Galli
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Navid Zebarjadi
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Lydia Li
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | | | - Laura W Burrus
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA.
| |
Collapse
|
82
|
Robinson DG, Neuhaus JM. Receptor-mediated sorting of soluble vacuolar proteins: myths, facts, and a new model. JOURNAL OF EXPERIMENTAL BOTANY 2016; 67:4435-49. [PMID: 27262127 DOI: 10.1093/jxb/erw222] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
To prevent their being released to the cell exterior, acid hydrolases are recognized by receptors at some point in the secretory pathway and diverted towards the lytic compartment of the cell (lysosome or vacuole). In animal cells, the receptor is called the mannosyl 6-phosphate receptor (MPR) and it binds hydrolase ligands in the trans-Golgi network (TGN). These ligands are then sequestered into clathrin-coated vesicles (CCVs) because of motifs in the cytosolic tail of the MPR which interact first with monomeric adaptors (Golgi-localized, Gamma-ear-containing, ARF-binding proteins, GGAs) and then with tetrameric (adaptin) adaptor complexes. The CCVs then fuse with an early endosome, whose more acidic lumen causes the ligands to dissociate. The MPRs are then recycled back to the TGN via retromer-coated carriers. Plants have vacuolar sorting receptors (VSRs) which were originally identified in CCVs isolated from pea (Pisum sativum L.) cotyledons. It was therefore assumed that VSRs would have an analogous function in plants to MPRs in animals. Although this dogma has enjoyed wide support over the last 20 years there are many inconsistencies. Recently, results have been published which are quite contrary to it. It now emerges that VSRs and their ligands can interact very early in the secretory pathway, and dissociate in the TGN, which, in contrast to its mammalian counterpart, has a pH of 5.5. Multivesicular endosomes in plants lack proton pump complexes and consequently have an almost neutral internal pH, which discounts them as organelles of pH-dependent receptor-ligand dissociation. These data force a critical re-evaluation of the role of CCVs at the TGN, especially considering that vacuolar cargo ligands have never been identified in them. We propose that one population of TGN-derived CCVs participate in retrograde transport of VSRs from the TGN. We also present a new model to explain how secretory and vacuolar cargo proteins are effectively separated after entering the late Golgi/TGN compartments.
Collapse
Affiliation(s)
- David G Robinson
- Centre for Organismal Studies (COS), University of Heidelberg, Germany
| | - Jean-Marc Neuhaus
- Institute of Biology, Laboratory of Cell and Molecular Biology, University of Neuchatel, Switzerland
| |
Collapse
|
83
|
Langton PF, Kakugawa S, Vincent JP. Making, Exporting, and Modulating Wnts. Trends Cell Biol 2016; 26:756-765. [PMID: 27325141 DOI: 10.1016/j.tcb.2016.05.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/15/2016] [Accepted: 05/20/2016] [Indexed: 10/21/2022]
Abstract
Wnt proteins activate a conserved signalling pathway that controls development and tissue homeostasis in all metazoans. The intensity of Wnt signalling must be tightly controlled to avoid diseases caused by excess or ectopic signalling. Over the years, many proteins dedicated to Wnt function have been identified, including Porcupine, which appends a palmitoleate moiety that is essential for signalling activity. This lipid inevitably affects subcellular trafficking and solubility, as well as providing a target for post-translational modulation. We review here the life history of Wnts, starting with progression through the secretory pathway, continuing with release and spread in the extracellular space, and finishing with the various proteins that dampen or inactivate Wnts in the extracellular space.
Collapse
Affiliation(s)
- Paul F Langton
- The Henry Wellcome Integrated Signalling Laboratories, School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Satoshi Kakugawa
- Hakuhodo Medical Inc., 6-1-20 Akasaka Minato-ku, Tokyo 107-0052, Japan
| | - Jean-Paul Vincent
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK.
| |
Collapse
|
84
|
Scholz B, Korn C, Wojtarowicz J, Mogler C, Augustin I, Boutros M, Niehrs C, Augustin HG. Endothelial RSPO3 Controls Vascular Stability and Pruning through Non-canonical WNT/Ca(2+)/NFAT Signaling. Dev Cell 2016; 36:79-93. [PMID: 26766444 DOI: 10.1016/j.devcel.2015.12.015] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 11/16/2015] [Accepted: 12/11/2015] [Indexed: 12/15/2022]
Abstract
The WNT signaling enhancer R-spondin3 (RSPO3) is prominently expressed in the vasculature. Correspondingly, embryonic lethality of Rspo3-deficient mice is caused by vessel remodeling defects. Yet the mechanisms underlying vascular RSPO3 function remain elusive. Inducible endothelial Rspo3 deletion (Rspo3-iECKO) resulted in perturbed developmental and tumor vascular remodeling. Endothelial cell apoptosis and vascular pruning led to reduced microvessel density in Rspo3-iECKO mice. Rspo3-iECKO mice strikingly phenocopied the non-canonical WNT signaling-induced vascular defects of mice deleted for the WNT secretion factor Evi/Wls. An endothelial screen for RSPO3 and EVI/WLS co-regulated genes identified Rnf213, Usp18, and Trim30α. RNF213 targets filamin A and NFAT1 for proteasomal degradation attenuating non-canonical WNT/Ca(2+) signaling. Likewise, USP18 and TRIM5α inhibited NFAT1 activation. Consequently, NFAT protein levels were decreased in endothelial cells of Rspo3-iECKO mice and pharmacological NFAT inhibition phenocopied Rspo3-iECKO mice. The data identify endothelial RSPO3-driven non-canonical WNT/Ca(2+)/NFAT signaling as a critical maintenance pathway of the remodeling vasculature.
Collapse
Affiliation(s)
- Beate Scholz
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Claudia Korn
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jessica Wojtarowicz
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Carolin Mogler
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Institute of Pathology, Heidelberg University, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Iris Augustin
- Division of Signaling and Functional Genomics, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Cell and Molecular Biology (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Cell and Molecular Biology (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; DNA Demethylation, DNA Repair and Reprogramming, Institute of Molecular Biology, Ackermannweg 4, 55128 Mainz, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; German Cancer Consortium, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
85
|
Xu H, Jiang W, Zhu F, Zhu C, Wei J, Wang J. Expression of Wntless in colorectal carcinomas is associated with invasion, metastasis, and poor survival. APMIS 2016; 124:522-8. [PMID: 27102079 DOI: 10.1111/apm.12534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 02/17/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Hanfeng Xu
- Department of Oncology; The Second Affiliated Hospital of Southeast University; Nanjing China
| | - Wen Jiang
- Department of Oncology; The Second Affiliated Hospital of Southeast University; Nanjing China
| | - Fang Zhu
- Department of Oncology; The Second Affiliated Hospital of Southeast University; Nanjing China
| | - Chuandong Zhu
- Department of Oncology; The Second Affiliated Hospital of Southeast University; Nanjing China
| | - Juan Wei
- Department of Oncology; The Second Affiliated Hospital of Southeast University; Nanjing China
| | - Jiandong Wang
- Department of Pathology; Jinling Hospital; Nanjing China
| |
Collapse
|
86
|
Tang BL. Are Wnts Retrogradely Transported to the ER? J Cell Physiol 2016; 231:2315-6. [DOI: 10.1002/jcp.25360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University Health System; NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| |
Collapse
|
87
|
Yamazaki Y, Palmer L, Alexandre C, Kakugawa S, Beckett K, Gaugue I, Palmer RH, Vincent JP. Godzilla-dependent transcytosis promotes Wingless signalling in Drosophila wing imaginal discs. Nat Cell Biol 2016; 18:451-7. [PMID: 26974662 PMCID: PMC4817240 DOI: 10.1038/ncb3325] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/05/2016] [Indexed: 12/18/2022]
Abstract
The apical and basolateral membranes of epithelia are insulated from each other, preventing the transfer of extracellular proteins from one side to the other. Thus, a signalling protein produced apically is not expected to reach basolateral receptors. Evidence suggests that Wingless, the main Drosophila Wnt, is secreted apically in the embryonic epidermis. However, in the wing imaginal disc epithelium, Wingless is mostly seen on the basolateral membrane where it spreads from secreting to receiving cells. Here we examine the apico-basal movement of Wingless in Wingless-producing cells of wing imaginal discs. We find that it is presented first on the apical surface before making its way to the basolateral surface, where it is released and allowed to interact with signalling receptors. We show that Wingless transcytosis involves dynamin-dependent endocytosis from the apical surface. Subsequent trafficking from early apical endosomes to the basolateral surface requires Godzilla, a member of the RNF family of membrane-anchored E3 ubiquitin ligases. Without such transport, Wingless signalling is strongly reduced in this tissue.
Collapse
Affiliation(s)
- Yasuo Yamazaki
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 9A, 40539 Gothenburg, Sweden
| | - Lucy Palmer
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Cyrille Alexandre
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Satoshi Kakugawa
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Karen Beckett
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Isabelle Gaugue
- Polarity, Division and Morphogenesis Team, Institut Curie, CNRS UMR 3215, INSERM U934, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 9A, 40539 Gothenburg, Sweden
| | - Jean-Paul Vincent
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| |
Collapse
|
88
|
Dysfunction of Wntless triggers the retrograde Golgi-to-ER transport of Wingless and induces ER stress. Sci Rep 2016; 6:19418. [PMID: 26887613 PMCID: PMC4757895 DOI: 10.1038/srep19418] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/14/2015] [Indexed: 01/02/2023] Open
Abstract
Secreted Wnts play diverse roles in a non-cell-autonomous fashion. However, the cell-autonomous effect of unsecreted Wnts remains unknown. Endoplasmic reticulum (ER) stress is observed in specialized secretory cells and participates in pathophysiological processes. The correlation between Wnt secretion and ER stress remains poorly understood. Here, we demonstrated that Drosophila miR-307a initiates ER stress specifically in wingless (wg)-expressing cells through targeting wntless (wls/evi). This phenotype could be mimicked by retromer loss-of-function or porcupine (porc) depletion, and rescued by wg knockdown, arguing that unsecreted Wg triggers ER stress. Consistently, we found that disrupting the secretion of human Wnt5a also induced ER stress in mammalian cells. Furthermore, we showed that a C-terminal KKVY-motif of Wg is required for its retrograde Golgi-to-ER transport, thus inducing ER stress. Next, we investigated if COPI, the regulator of retrograde transport, is responsible for unsecreted Wg to induce ER stress. To our surprise, we found that COPI acts as a novel regulator of Wg secretion. Taken together, this study reveals a previously unknown Golgi-to-ER retrograde route of Wg, and elucidates a correlation between Wnt secretion and ER stress during development.
Collapse
|
89
|
Yang K, Wang X, Zhang H, Wang Z, Nan G, Li Y, Zhang F, Mohammed MK, Haydon RC, Luu HH, Bi Y, He TC. The evolving roles of canonical WNT signaling in stem cells and tumorigenesis: implications in targeted cancer therapies. J Transl Med 2016; 96:116-36. [PMID: 26618721 PMCID: PMC4731283 DOI: 10.1038/labinvest.2015.144] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/06/2015] [Indexed: 02/07/2023] Open
Abstract
The canonical WNT/β-catenin signaling pathway governs a myriad of biological processes underlying the development and maintenance of adult tissue homeostasis, including regulation of stem cell self-renewal, cell proliferation, differentiation, and apoptosis. WNTs are secreted lipid-modified glycoproteins that act as short-range ligands to activate receptor-mediated signaling pathways. The hallmark of the canonical pathway is the activation of β-catenin-mediated transcriptional activity. Canonical WNTs control the β-catenin dynamics as the cytoplasmic level of β-catenin is tightly regulated via phosphorylation by the 'destruction complex', consisting of glycogen synthase kinase 3β (GSK3β), casein kinase 1α (CK1α), the scaffold protein AXIN, and the tumor suppressor adenomatous polyposis coli (APC). Aberrant regulation of this signaling cascade is associated with varieties of human diseases, especially cancers. Over the past decade, significant progress has been made in understanding the mechanisms of canonical WNT signaling. In this review, we focus on the current understanding of WNT signaling at the extracellular, cytoplasmic membrane, and intracellular/nuclear levels, including the emerging knowledge of cross-talk with other pathways. Recent progresses in developing novel WNT pathway-targeted therapies will also be reviewed. Thus, this review is intended to serve as a refresher of the current understanding about the physiologic and pathogenic roles of WNT/β-catenin signaling pathway, and to outline potential therapeutic opportunities by targeting the canonical WNT pathway.
Collapse
Affiliation(s)
- Ke Yang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xin Wang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Department of Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Zhang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China
| | - Zhongliang Wang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guoxin Nan
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yasha Li
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Fugui Zhang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China
| | - Maryam K. Mohammed
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Corresponding authors T.-C. He, MD, PhD, Molecular Oncology Laboratory, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL 60637, USA, Tel. (773) 702-7169; Fax (773) 834-4598, , Yang Bi, MD, PhD, Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University, Chongqing 400046, China, Tel. 011-86-23-63633113; Fax: 011-86-236362690,
| | - Tong-Chuan He
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China, Corresponding authors T.-C. He, MD, PhD, Molecular Oncology Laboratory, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL 60637, USA, Tel. (773) 702-7169; Fax (773) 834-4598, , Yang Bi, MD, PhD, Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University, Chongqing 400046, China, Tel. 011-86-23-63633113; Fax: 011-86-236362690,
| |
Collapse
|
90
|
Nile AH, Hannoush RN. Fatty acylation of Wnt proteins. Nat Chem Biol 2016; 12:60-9. [DOI: 10.1038/nchembio.2005] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/09/2015] [Indexed: 02/04/2023]
|
91
|
Wnt/β-catenin signaling plays an ever-expanding role in stem cell self-renewal, tumorigenesis and cancer chemoresistance. Genes Dis 2016; 3:11-40. [PMID: 27077077 PMCID: PMC4827448 DOI: 10.1016/j.gendis.2015.12.004] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Wnt signaling transduces evolutionarily conserved pathways which play important roles in initiating and regulating a diverse range of cellular activities, including cell proliferation, calcium homeostasis, and cell polarity. The role of Wnt signaling in controlling cell proliferation and stem cell self-renewal is primarily carried out through the canonical pathway, which is the best-characterized the multiple Wnt signaling branches. The past 10 years has seen a rapid expansion in our understanding of the complexity of this pathway, as many new components of Wnt signaling have been identified and linked to signaling regulation, stem cell functions, and adult tissue homeostasis. Additionally, a substantial body of evidence links Wnt signaling to tumorigenesis of cancer types and implicates it in the development of cancer drug resistance. Thus, a better understanding of the mechanisms by which dysregulation of Wnt signaling precedes the development and progression of human cancer may hasten the development of pathway inhibitors to augment current therapy. This review summarizes and synthesizes our current knowledge of the canonical Wnt pathway in development and disease. We begin with an overview of the components of the canonical Wnt signaling pathway and delve into the role this pathway has been shown to play in stemness, tumorigenesis, and cancer drug resistance. Ultimately, we hope to present an organized collection of evidence implicating Wnt signaling in tumorigenesis and chemoresistance to facilitate the pursuit of Wnt pathway modulators that may improve outcomes of cancers in which Wnt signaling contributes to aggressive disease and/or treatment resistance.
Collapse
|
92
|
Deshpande M, Rodal AA. The Crossroads of Synaptic Growth Signaling, Membrane Traffic and Neurological Disease: Insights from Drosophila. Traffic 2015; 17:87-101. [PMID: 26538429 DOI: 10.1111/tra.12345] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/29/2015] [Accepted: 10/29/2015] [Indexed: 12/13/2022]
Abstract
Neurons require target-derived autocrine and paracrine growth factors to maintain proper identity, innervation, homeostasis and survival. Neuronal growth factor signaling is highly dependent on membrane traffic, both for the packaging and release of the growth factors themselves, and for regulation of intracellular signaling by their transmembrane receptors. Here, we review recent findings from the Drosophila larval neuromuscular junction (NMJ) that illustrate how specific steps of intracellular traffic and inter-organelle interactions impinge on signaling, particularly in the bone morphogenic protein, Wingless and c-Jun-activated kinase pathways, regulating elaboration and stability of NMJ arbors, construction of synapses and synaptic transmission and homeostasis. These membrane trafficking and signaling pathways have been implicated in human motor neuron diseases including amyotrophic lateral sclerosis and hereditary spastic paraplegia, highlighting their importance for neuronal health and survival.
Collapse
Affiliation(s)
| | - Avital A Rodal
- Department of Biology, Brandeis University, Waltham, MA, USA
| |
Collapse
|
93
|
Kuan YS, Roberson S, Akitake CM, Fortuno L, Gamse J, Moens C, Halpern ME. Distinct requirements for Wntless in habenular development. Dev Biol 2015; 406:117-128. [PMID: 26116173 PMCID: PMC4639407 DOI: 10.1016/j.ydbio.2015.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 01/24/2023]
Abstract
Secreted Wnt proteins play pivotal roles in development, including regulation of cell proliferation, differentiation, progenitor maintenance and tissue patterning. The transmembrane protein Wntless (Wls) is necessary for secretion of most Wnts and essential for effective Wnt signaling. During a mutagenesis screen to identify genes important for development of the habenular nuclei in the dorsal forebrain, we isolated a mutation in the sole wls gene of zebrafish and confirmed its identity with a second, independent allele. Early embryonic development appears normal in homozygous wls mutants, but they later lack the ventral habenular nuclei, form smaller dorsal habenulae and otic vesicles, have truncated jaw and fin cartilages and lack swim bladders. Activation of a reporter for β-catenin-dependent transcription is decreased in wls mutants, indicative of impaired signaling by the canonical Wnt pathway, and expression of Wnt-responsive genes is reduced in the dorsal diencephalon. Wnt signaling was previously implicated in patterning of the zebrafish brain and in the generation of left-right (L-R) differences between the bilaterally paired dorsal habenular nuclei. Outside of the epithalamic region, development of the brain is largely normal in wls mutants and, despite their reduced size, the dorsal habenulae retain L-R asymmetry. We find that homozygous wls mutants show a reduction in two cell populations that contribute to the presumptive dorsal habenulae. The results support distinct temporal requirements for Wls in habenular development and reveal a new role for Wnt signaling in the regulation of dorsal habenular progenitors.
Collapse
Affiliation(s)
- Yung-Shu Kuan
- Department of Embryology, Carnegie Institution for Science, USA
| | - Sara Roberson
- Department of Embryology, Carnegie Institution for Science, USA
- Department of Biology, Johns Hopkins University, USA
| | - Courtney M. Akitake
- Department of Embryology, Carnegie Institution for Science, USA
- Department of Biology, Johns Hopkins University, USA
| | - Lea Fortuno
- Department of Embryology, Carnegie Institution for Science, USA
| | - Joshua Gamse
- Department of Biological Sciences, Vanderbilt University, USA
| | - Cecilia Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, USA
| | - Marnie E. Halpern
- Department of Embryology, Carnegie Institution for Science, USA
- Department of Biology, Johns Hopkins University, USA
| |
Collapse
|
94
|
Klinger SC, Siupka P, Nielsen MS. Retromer-Mediated Trafficking of Transmembrane Receptors and Transporters. MEMBRANES 2015; 5:288-306. [PMID: 26154780 PMCID: PMC4584283 DOI: 10.3390/membranes5030288] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/29/2015] [Indexed: 12/21/2022]
Abstract
Transport between the endoplasmatic reticulum, the Golgi-network, the endo-lysosomal system and the cell surface can be categorized as anterograde or retrograde, describing traffic that goes forward or backward, respectively. Traffic going from the plasma membrane to endosomes and lysosomes or the trans-Golgi network (TGN) constitutes the major retrograde transport routes. Several transmembrane proteins undergo retrograde transport as part of a recycling mechanism that contributes to reutilization and maintenance of a steady-state protein localization. In addition, some receptors are hijacked by exotoxins and used for entry and intracellular transport. The physiological relevance of retrograde transport cannot be overstated. Retrograde trafficking of the amyloid precursor protein determines the distribution between organelles, and hence the possibility of cleavage by γ-secretase. Right balancing of the pathways is critical for protection against Alzheimer’s disease. During embryonic development, retrograde transport of Wntless to the TGN is essential for the following release of Wnt from the plasma membrane. Furthermore, overexpression of Wntless has been linked to oncogenesis. Here, we review relevant aspects of the retrograde trafficking of mammalian transmembrane receptors and transporters, with focus on the retromer-mediated transport between endosomes and the TGN.
Collapse
Affiliation(s)
- Stine C Klinger
- The Lundbeck Foundation Initiative on Brain Barriers and Drug Delivery, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| | - Piotr Siupka
- The Lundbeck Foundation Initiative on Brain Barriers and Drug Delivery, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| | - Morten S Nielsen
- The Lundbeck Foundation Initiative on Brain Barriers and Drug Delivery, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
95
|
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
|
96
|
Wnts are dispensable for differentiation and self-renewal of adult murine hematopoietic stem cells. Blood 2015; 126:1086-94. [PMID: 26089398 DOI: 10.1182/blood-2014-09-598540] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 06/16/2015] [Indexed: 01/09/2023] Open
Abstract
Wnt signaling controls early embryonic hematopoiesis and dysregulated β-catenin is implicated in leukemia. However, the role of Wnts and their source in adult hematopoiesis is still unclear, and is clinically important as upstream Wnt inhibitors enter clinical trials. We blocked Wnt secretion in hematopoietic lineages by targeting Porcn, a membrane-bound O-acyltransferase that is indispensable for the activity and secretion of all vertebrate Wnts. Surprisingly, deletion of Porcn in Rosa-CreER(T2)/Porcn(Del), MX1-Cre/Porcn(Del), and Vav-Cre/Porcn(Del) mice had no effects on proliferation, differentiation, or self-renewal of adult hematopoietic stem cells. Targeting Wnt secretion in the bone marrow niche by treatment with a PORCN inhibitor, C59, similarly had no effect on hematopoiesis. These results exclude a role for hematopoietic PORCN-dependent Wnts in adult hematopoiesis. Clinical use of upstream Wnt inhibitors is not likely to be limited by effects on hematopoiesis.
Collapse
|
97
|
Das S, Yu S, Sakamori R, Vedula P, Feng Q, Flores J, Hoffman A, Fu J, Stypulkowski E, Rodriguez A, Dobrowolski R, Harada A, Hsu W, Bonder EM, Verzi MP, Gao N. Rab8a vesicles regulate Wnt ligand delivery and Paneth cell maturation at the intestinal stem cell niche. Development 2015; 142:2147-62. [PMID: 26015543 PMCID: PMC4483769 DOI: 10.1242/dev.121046] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/16/2015] [Indexed: 12/11/2022]
Abstract
Communication between stem and niche supporting cells maintains the homeostasis of adult tissues. Wnt signaling is a crucial regulator of the stem cell niche, but the mechanism that governs Wnt ligand delivery in this compartment has not been fully investigated. We identified that Wnt secretion is partly dependent on Rab8a-mediated anterograde transport of Gpr177 (wntless), a Wnt-specific transmembrane transporter. Gpr177 binds to Rab8a, depletion of which compromises Gpr177 traffic, thereby weakening the secretion of multiple Wnts. Analyses of generic Wnt/β-catenin targets in Rab8a knockout mouse intestinal crypts indicate reduced signaling activities; maturation of Paneth cells – a Wnt-dependent cell type – is severely affected. Rab8a knockout crypts show an expansion of Lgr5+ and Hopx+ cells in vivo. However, in vitro, the knockout enteroids exhibit significantly weakened growth that can be partly restored by exogenous Wnts or Gsk3β inhibitors. Immunogold labeling and surface protein isolation identified decreased plasma membrane localization of Gpr177 in Rab8a knockout Paneth cells and fibroblasts. Upon stimulation by exogenous Wnts, Rab8a-deficient cells show ligand-induced Lrp6 phosphorylation and transcriptional reporter activation. Rab8a thus controls Wnt delivery in producing cells and is crucial for Paneth cell maturation. Our data highlight the profound tissue plasticity that occurs in response to stress induced by depletion of a stem cell niche signal. Summary: In maturing mouse Paneth cells, Wnt secretion is partly dependent on a Rab8a-mediated anterograde transport of Gpr177. Rab8a is required for Paneth cell maturation.
Collapse
Affiliation(s)
- Soumyashree Das
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Ryotaro Sakamori
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Pavan Vedula
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Qiang Feng
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Juan Flores
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Andrew Hoffman
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | - Jiang Fu
- Department of Biomedical Genetics, Center for Oral Biology, James P. Wilmot Cancer Center, Stem Cell and Regenerative Medicine Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ewa Stypulkowski
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Alexis Rodriguez
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Wei Hsu
- Department of Biomedical Genetics, Center for Oral Biology, James P. Wilmot Cancer Center, Stem Cell and Regenerative Medicine Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Michael P Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
98
|
Lu D, Li Y, Liu QR, Wu Q, Zhang H, Xie P, Wang Q. Wls promotes the proliferation of breast cancer cells via Wnt signaling. Med Oncol 2015; 32:140. [DOI: 10.1007/s12032-015-0585-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/14/2015] [Indexed: 02/06/2023]
|
99
|
Abstract
Cholangiocarcinoma is a relatively rare cancer of the biliary ducts that is highly refractory to treatment. The factors that drive cholangiocarcinoma are poorly understood, though chronic liver fluke infection is a risk factor for disease. In this issue of the JCI, Boulter and colleagues demonstrate that the WNT/β-catenin signaling pathway is upregulated in patients with sporadic cholangiocarcinoma. The authors determined that macrophages generate WNT ligands in cholangiocarcinomas and depletion or inhibition of this cell population in animal models of cholangiocarcinoma reduced tumor burden and proliferation. Moreover, pharmacological inhibition of WNT secretion or β-catenin activity was efficacious in animal models. Together the results of this study suggest that targeting WNT has potential as a therapeutic strategy for cholangiocarcinoma.
Collapse
|
100
|
Yamamoto H, Awada C, Matsumoto S, Kaneiwa T, Sugimoto T, Takao T, Kikuchi A. Basolateral secretion of Wnt5a in polarized epithelial cells is required for apical lumen formation. J Cell Sci 2015; 128:1051-63. [PMID: 25593127 DOI: 10.1242/jcs.163683] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Wnt5a regulates planar cell polarity in epithelial cells, but it remains to be determined whether Wnt5a and its receptors are sorted apically or basolaterally, and how Wnt5a signaling is involved in apical and basolateral polarization. We found that Wnt5a was secreted basolaterally in polarized kidney epithelial cells. The basolateral secretion of Wnt5a required Wntless (Wls), clathrin and adaptor protein 1 (AP-1). Wnt5a receptors were also localized to the basolateral membranes, but their sorting did not require Wls. Wnt5a-induced signaling was stimulated more efficiently at the basolateral side than the apical side of epithelial cells. Knockdown of Wnt5a delayed apical lumen formation of the epithelial cyst, and these phenotypes were rescued by wild-type Wnt5a, but not by a Wnt5a mutant that is secreted apically. Although apoptosis was not required for apical lumen formation in a wild-type cyst, apoptosis was necessary for eliminating luminal cells in a Wnt5a-depleted cyst. These results suggest that Wnt5a and its receptors are sorted to their correct destination by different mechanisms and that the basolateral secretion of Wnt5a is necessary for apical lumen formation in the epithelial cyst.
Collapse
Affiliation(s)
- Hideki Yamamoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chihiro Awada
- Laboratory of Protein Profiling and Functional Proteomics, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tomoyuki Kaneiwa
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Sugimoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshifumi Takao
- Laboratory of Protein Profiling and Functional Proteomics, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|