1
|
Kang K, Shi Q, Wang X, Chen YG. Dishevelled phase separation promotes Wnt signalosome assembly and destruction complex disassembly. J Cell Biol 2022; 221:213667. [PMID: 36342472 PMCID: PMC9811998 DOI: 10.1083/jcb.202205069] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/21/2022] [Accepted: 09/19/2022] [Indexed: 11/09/2022] Open
Abstract
The amplitude of Wnt/β-catenin signaling is precisely controlled by the assembly of the cell surface-localized Wnt receptor signalosome and the cytosolic β-catenin destruction complex. How these two distinct complexes are coordinately controlled remains largely unknown. Here, we demonstrated that the signalosome scaffold protein Dishevelled 2 (Dvl2) undergoes liquid-liquid phase separation (LLPS). Dvl2 LLPS is mediated by an intrinsically disordered region and facilitated by components of the signalosome, such as the receptor Fzd5. Assembly of the signalosome is initiated by rapid recruitment of Dvl2 to the membrane, followed by slow and dynamic recruitment of Axin1. Axin LLPS mediates assembly of the β-catenin destruction complex, and Dvl2 attenuates LLPS of Axin. Compared with the destruction complex, Axin partitions into the signalosome at a lower concentration and exhibits a higher mobility. Together, our results revealed that Dvl2 LLPS is crucial for controlling the assembly of the Wnt receptor signalosome and disruption of the phase-separated β-catenin destruction complex.
Collapse
Affiliation(s)
- Kexin Kang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiaoni Shi
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xu Wang
- Guangzhou Laboratory, Guangzhou, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China,Guangzhou Laboratory, Guangzhou, China,School of Basic Medicine, Nanchang University, Nanchang, China,Correspondence to Ye-Guang Chen:
| |
Collapse
|
2
|
Nong J, Kang K, Shi Q, Zhu X, Tao Q, Chen YG. Phase separation of Axin organizes the β-catenin destruction complex. J Cell Biol 2021; 220:211840. [PMID: 33651074 PMCID: PMC7931644 DOI: 10.1083/jcb.202012112] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
In Wnt/β-catenin signaling, the β-catenin protein level is deliberately controlled by the assembly of the multiprotein β-catenin destruction complex composed of Axin, adenomatous polyposis coli (APC), glycogen synthase kinase 3β (GSK3β), casein kinase 1α (CK1α), and others. Here we provide compelling evidence that formation of the destruction complex is driven by protein liquid–liquid phase separation (LLPS) of Axin. An intrinsically disordered region in Axin plays an important role in driving its LLPS. Phase-separated Axin provides a scaffold for recruiting GSK3β, CK1α, and β-catenin. APC also undergoes LLPS in vitro and enhances the size and dynamics of Axin phase droplets. The LLPS-driven assembly of the destruction complex facilitates β-catenin phosphorylation by GSK3β and is critical for the regulation of β-catenin protein stability and thus Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Junxiu Nong
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Kexin Kang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiaoni Shi
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuechen Zhu
- The Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qinghua Tao
- The Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,The Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
3
|
Dionellis VS, Norkin M, Karamichali A, Rossetti GG, Huelsken J, Ordonez-Moran P, Halazonetis TD. Genomic Instability Profiles at the Single Cell Level in Mouse Colorectal Cancers of Defined Genotypes. Cancers (Basel) 2021; 13:cancers13061267. [PMID: 33809306 PMCID: PMC7999300 DOI: 10.3390/cancers13061267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022] Open
Abstract
The genomes of many human CRCs have been sequenced, revealing a large number of genetic alterations. However, the molecular mechanisms underlying the accumulation of these alterations are still being debated. In this study, we examined colorectal tumours that developed in mice with Apclox/lox, LSL-KrasG12D, and Tp53lox/lox targetable alleles. Organoids were derived from single cells and the spectrum of mutations was determined by exome sequencing. The number of single nucleotide substitutions (SNSs) correlated with the age of the tumour, but was unaffected by the number of targeted cancer-driver genes. Thus, tumours that expressed mutant Apc, Kras, and Tp53 alleles had as many SNSs as tumours that expressed only mutant Apc. In contrast, the presence of large-scale (>10 Mb) copy number alterations (CNAs) correlated strongly with Tp53 inactivation. Comparison of the SNSs and CNAs present in organoids derived from the same tumour revealed intratumoural heterogeneity consistent with genomic lesions accumulating at significantly higher rates in tumour cells compared to normal cells. The rate of acquisition of SNSs increased from the early stages of cancer development, whereas large-scale CNAs accumulated later, after Tp53 inactivation. Thus, a significant fraction of the genomic instability present in cancer cells cannot be explained by aging processes occurring in normal cells before oncogenic transformation.
Collapse
Affiliation(s)
- Vasilis S. Dionellis
- Department of Molecular Biology, University of Geneva, 1211 Geneva, Switzerland; (V.S.D.); (A.K.); (G.G.R.)
| | - Maxim Norkin
- Cancer Stem Cell Laboratory, Swiss Institute of Technology Lausanne (EPFL), ISREC, 1015 Lausanne, Switzerland;
| | - Angeliki Karamichali
- Department of Molecular Biology, University of Geneva, 1211 Geneva, Switzerland; (V.S.D.); (A.K.); (G.G.R.)
| | - Giacomo G. Rossetti
- Department of Molecular Biology, University of Geneva, 1211 Geneva, Switzerland; (V.S.D.); (A.K.); (G.G.R.)
| | - Joerg Huelsken
- Cancer Stem Cell Laboratory, Swiss Institute of Technology Lausanne (EPFL), ISREC, 1015 Lausanne, Switzerland;
- Correspondence: (J.H.); (P.O.-M.); (T.D.H.)
| | - Paloma Ordonez-Moran
- Division of Cancer & Stem Cells, School of Medicine, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- Correspondence: (J.H.); (P.O.-M.); (T.D.H.)
| | - Thanos D. Halazonetis
- Department of Molecular Biology, University of Geneva, 1211 Geneva, Switzerland; (V.S.D.); (A.K.); (G.G.R.)
- Correspondence: (J.H.); (P.O.-M.); (T.D.H.)
| |
Collapse
|
4
|
Ge T, Liu T, Guo L, Chen Z, Lou G. MicroRNA-302 represses epithelial-mesenchymal transition and cisplatin resistance by regulating ATAD2 in ovarian carcinoma. Exp Cell Res 2020; 396:112241. [PMID: 32835657 DOI: 10.1016/j.yexcr.2020.112241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 12/21/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is an important contributor to drug resistance in ovarian cancer. The aims of this study were to explore the potential role of the miR-302 cluster in modulating EMT and cisplatin resistance in ovarian cancer. We used qRT-PCR and western blotting to show that miR-302 expression was lower in chemoresistant than in chemosensitive cells, and miR-302 was upregulated in chemosensitive, but not chemoresistant ovarian cancer cells in response to cisplatin treatment. We identified ATAD2 as a target of miR-302 and showed that ectopic expression of miR-302 increased cisplatin sensitivity and inhibited EMT and the invasiveness of cisplatin-resistant cells in vitro by targeting ATAD2. Knockdown of ATAD2 restored cisplatin sensitivity and reversed EMT/metastasis in cisplatin-resistant cells, as shown by western blotting and invasion/migration assays. The effect of miR-302 overexpression on EMT and invasiveness was mediated by the modulation of β-catenin nuclear expression. Immunofluorescence analysis showed that ATAD2 overexpression reversed the miR-302-induced downregulation of nuclear β-catenin in cisplatin resistant cells. A xenograft tumor model was used to show that miR-302 increases the antitumor effect of cisplatin in vivo. Taken together, these results identify a potential regulatory axis involving miR-302 and ATAD2 with a role in chemoresistance, indicating that activation of miR-302 or inactivation of ATAD2 could serve as a novel approach to reverse cisplatin resistance in ovarian cancer.
Collapse
Affiliation(s)
- Tingting Ge
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianbo Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Liyuan Guo
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhuo Chen
- Department of Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ge Lou
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
5
|
Zhang Y, Li X, Zhang Y, Wang L, Xu J, Du J, Guan Y. Pegylated interferon-α inhibits the proliferation of hepatocellular carcinoma cells by downregulating miR-155. Ann Hepatol 2020; 18:494-500. [PMID: 31036495 DOI: 10.1016/j.aohep.2018.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/17/2018] [Accepted: 11/23/2018] [Indexed: 02/08/2023]
Abstract
INTRODUCTION AND AIMS Interferon-α (IFN) has shown potential benefits in patients with hepatocellular carcinoma (HCC), and these effects may be mediated by inhibiting cancer cell proliferation. However, the detailed mechanisms underlying the anti-proliferative effects of IFN remain obscure. In this study, we evaluate the role of the novel oncogenic microRNA (miRNA) miR-155 in the anti-proliferative effects of pegylated interferon-α (PEG-IFN) on HCC cells. METHODS The effects of PEG-IFN on HepG2 cell proliferation, migration and invasion were determined using the MTT assay, flow cytometry analysis and the Transwell assay, respectively. Reverse transcription quantitative polymerase chain reaction was used to analyze miR-155 expression. The levels of proteins involved in Wnt/β-catenin signal transduction were determined by western blot analysis and immunofluorescence staining. Mimics of miR-155 were transfected into HepG2 cells to assess the role of miR-155 in the PEG-IFN-induced anti-proliferative effect. RESULTS PEG-IFN significantly inhibited the proliferation, migration and invasion of HepG2 cells in a dose-dependent manner by inhibiting cell cycle progression. In parallel with reduced cell proliferation, migration and invasion, miR-155 was efficiently downregulated by PEG-IFN in a dose-dependent manner. Moreover, the transfection of miR-155 decreased the inhibitory effect of PEG-IFN on HepG2cell proliferation, migration and invasion, as well as the downregulation of proteins in the Wnt/β-catenin pathway. CONCLUSIONS The anti-proliferative effects of PEG-IFN on HCC are at least partially attributable to the downregulation of miR-155.
Collapse
Affiliation(s)
- Ying Zhang
- Sixth Department of Liver Diseases, Dalian Sixth People Hospital, Dalian Medical University, Dalian, Liaoning, PR China.
| | - Xuefeng Li
- Department of Infectious Diseases, The Second Hospital of Dalian Medical University, Dalian Medical University, Dalian, Liaoning, PR China
| | - Yong Zhang
- Sixth Department of Liver Diseases, Dalian Sixth People Hospital, Dalian Medical University, Dalian, Liaoning, PR China
| | - Lin Wang
- Sixth Department of Liver Diseases, Dalian Sixth People Hospital, Dalian Medical University, Dalian, Liaoning, PR China
| | - Jiao Xu
- Sixth Department of Liver Diseases, Dalian Sixth People Hospital, Dalian Medical University, Dalian, Liaoning, PR China
| | - Jinghua Du
- Sixth Department of Liver Diseases, Dalian Sixth People Hospital, Dalian Medical University, Dalian, Liaoning, PR China
| | - Yonghai Guan
- Department of Infectious Diseases, The Second Hospital of Dalian Medical University, Dalian Medical University, Dalian, Liaoning, PR China
| |
Collapse
|
6
|
LY75 Ablation Mediates Mesenchymal-Epithelial Transition (MET) in Epithelial Ovarian Cancer (EOC) Cells Associated with DNA Methylation Alterations and Suppression of the Wnt/β-Catenin Pathway. Int J Mol Sci 2020; 21:ijms21051848. [PMID: 32156068 PMCID: PMC7084525 DOI: 10.3390/ijms21051848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/26/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Growing evidence demonstrates that epithelial-mesenchymal transition (EMT) plays an important role in epithelial ovarian cancer (EOC) progression and spreading; however, its molecular mechanisms remain poorly defined. We have previously shown that the antigen receptor LY75 can modulate EOC cell phenotype and metastatic potential, as LY75 depletion directed mesenchymal-epithelial transition (MET) in EOC cell lines with mesenchymal phenotype. We used the LY75-mediated modulation of EMT as a model to investigate for DNA methylation changes during EMT in EOC cells, by applying the reduced representation bisulfite sequencing (RRBS) methodology. Numerous genes have displayed EMT-related DNA methylation patterns alterations in their promoter/exon regions. Ten selected genes, whose DNA methylation alterations were further confirmed by alternative methods, were further identified, some of which could represent new EOC biomarkers/therapeutic targets. Moreover, our methylation data were strongly indicative for the predominant implication of the Wnt/β-catenin pathway in the EMT-induced DNA methylation variations in EOC cells. Consecutive experiments, including alterations in the Wnt/β-catenin pathway activity in EOC cells with a specific inhibitor and the identification of LY75-interacting partners by a proteomic approach, were strongly indicative for the direct implication of the LY75 receptor in modulating the Wnt/β-catenin signaling in EOC cells.
Collapse
|
7
|
Schaefer KN, Pronobis MI, Williams CE, Zhang S, Bauer L, Goldfarb D, Yan F, Major MB, Peifer M. Wnt regulation: exploring Axin-Disheveled interactions and defining mechanisms by which the SCF E3 ubiquitin ligase is recruited to the destruction complex. Mol Biol Cell 2020; 31:992-1014. [PMID: 32129710 PMCID: PMC7346726 DOI: 10.1091/mbc.e19-11-0647] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Wnt signaling plays key roles in embryonic development and adult stem cell homeostasis and is altered in human cancer. Signaling is turned on and off by regulating stability of the effector β-catenin (β-cat). The multiprotein destruction complex binds and phosphorylates β-cat and transfers it to the SCF-TrCP E3-ubiquitin ligase for ubiquitination and destruction. Wnt signals act though Dishevelled to turn down the destruction complex, stabilizing β-cat. Recent work clarified underlying mechanisms, but important questions remain. We explore β-cat transfer from the destruction complex to the E3 ligase, and test models suggesting Dishevelled and APC2 compete for association with Axin. We find that Slimb/TrCP is a dynamic component of the destruction complex biomolecular condensate, while other E3 proteins are not. Recruitment requires Axin and not APC, and Axin’s RGS domain plays an important role. We find that elevating Dishevelled levels in Drosophila embryos has paradoxical effects, promoting the ability of limiting levels of Axin to turn off Wnt signaling. When we elevate Dishevelled levels, it forms its own cytoplasmic puncta, but these do not recruit Axin. Superresolution imaging in mammalian cells raises the possibility that this may result by promoting Dishevelled:Dishevelled interactions at the expense of Dishevelled: Axin interactions when Dishevelled levels are high.
Collapse
Affiliation(s)
- Kristina N Schaefer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Mira I Pronobis
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Clara E Williams
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Shiping Zhang
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Lauren Bauer
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Dennis Goldfarb
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110.,Institute for Informatics, Washington University School of Medicine, St. Louis, MO 63110
| | - Feng Yan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - M Ben Major
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110.,Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
8
|
Villegas SN, Ferres-Marco D, Domínguez M. Using Drosophila Models and Tools to Understand the Mechanisms of Novel Human Cancer Driver Gene Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1167:15-35. [PMID: 31520347 DOI: 10.1007/978-3-030-23629-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
The formation, overgrowth and metastasis of tumors comprise a complex series of cellular and molecular events resulting from the combined effects of a variety of aberrant signaling pathways, mutations, and epigenetic alterations. Modeling this complexity in vivo requires multiple genes to be manipulated simultaneously, which is technically challenging. Here, we analyze how Drosophila research can further contribute to identifying pathways and elucidating mechanisms underlying novel cancer driver (risk) genes associated with tumor growth and metastasis in humans.
Collapse
Affiliation(s)
- Santiago Nahuel Villegas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC) and Universidad Miguel Hernández (UMH), Alicante, Spain.
| | - Dolors Ferres-Marco
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC) and Universidad Miguel Hernández (UMH), Alicante, Spain.
| | - María Domínguez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC) and Universidad Miguel Hernández (UMH), Alicante, Spain
| |
Collapse
|
9
|
Mohamed NE, Hay T, Reed KR, Smalley MJ, Clarke AR. APC2 is critical for ovarian WNT signalling control, fertility and tumour suppression. BMC Cancer 2019; 19:677. [PMID: 31291912 PMCID: PMC6617595 DOI: 10.1186/s12885-019-5867-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/24/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Canonical WNT signalling plays a critical role in the regulation of ovarian development; mis-regulation of this key pathway in the adult ovary is associated with subfertility and tumourigenesis. The roles of Adenomatous polyposis coli 2 (APC2), a little-studied WNT signalling pathway regulator, in ovarian homeostasis, fertility and tumourigenesis have not previously been explored. Here, we demonstrate essential roles of APC2 in regulating ovarian WNT signalling and ovarian homeostasis. METHODS A detailed analysis of ovarian histology, gene expression, ovulation and hormone levels was carried out in 10 week old and in aged constitutive APC2-knockout (Apc2-/-) mice (mixed background). Statistical significance for qRT-PCR data was determined from 95% confidence intervals. Significance testing was performed using 2-tailed Student's t-test, when 2 experimental cohorts were compared. When more were compared, ANOVA test was used, followed by a post-hoc test (LSD or Games-Howell). P-values of < 0.05 were considered statistically significant. RESULTS APC2-deficiency resulted in activation of ovarian WNT signalling and sub-fertility driven by intra-ovarian defects. Follicular growth was perturbed, resulting in a reduced rate of ovulation and corpora lutea formation, which could not be rescued by administration of gonadotrophins. Defects in steroidogenesis and follicular vascularity contributed to the subfertility phenotype. Tumour incidence was assessed in aged APC2-deficient mice, which also carried a hypomorphic Apc allele. APC2-deficiency in these mice resulted in predisposition to granulosa cell tumour (GCT) formation, accompanied by acute tumour-associated WNT-signalling activation and a histologic pattern and molecular signature seen in human adult GCTs. CONCLUSIONS Our work adds APC2 to the growing list of WNT-signalling members that regulate ovarian homeostasis, fertility and suppress GCT formation. Importantly, given that the APC2-deficient mouse develops tumours that recapitulate the molecular signature and histological features of human adult GCTs, this mouse has excellent potential as a pre-clinical model to study ovarian subfertility and transitioning to GCT, tumour biology and for therapeutic testing.
Collapse
Affiliation(s)
- Noha-Ehssan Mohamed
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
- Hormones Evaluation Department, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
- Present address: CRUK Beatson Institute, Switchback road, Bearsden, Glasgow, G61 1BD UK
| | - Trevor Hay
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| | - Karen R. Reed
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| | - Matthew J. Smalley
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| | - Alan R. Clarke
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| |
Collapse
|
10
|
Schaefer KN, Peifer M. Wnt/Beta-Catenin Signaling Regulation and a Role for Biomolecular Condensates. Dev Cell 2019; 48:429-444. [PMID: 30782412 PMCID: PMC6386181 DOI: 10.1016/j.devcel.2019.01.025] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/06/2018] [Accepted: 01/29/2019] [Indexed: 12/19/2022]
Abstract
Wnt/β-Catenin signaling plays key roles in tissue homeostasis and cell fate decisions in embryonic and post-embryonic development across the animal kingdom. As a result, pathway mutations are associated with developmental disorders and many human cancers. The multiprotein destruction complex keeps signaling off in the absence of Wnt ligands and needs to be downregulated for pathway activation. We discuss new insights into destruction complex activity and regulation, highlighting parallels to the control of other cell biological processes by biomolecular condensates that form by phase separation to suggest that the destruction complex acts as a biomolecular condensate in Wnt pathway regulation.
Collapse
Affiliation(s)
- Kristina N Schaefer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
11
|
Molinar-Inglis O, Oliver SL, Rudich P, Kunttas E, McCartney BM. APC2 associates with the actin cortex through a multipart mechanism to regulate cortical actin organization and dynamics in the Drosophila ovary. Cytoskeleton (Hoboken) 2018; 75:323-335. [PMID: 30019417 DOI: 10.1002/cm.21471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 05/24/2018] [Accepted: 06/19/2018] [Indexed: 01/20/2023]
Abstract
The actin cortex that lines the plasma membrane of most eukaryotic cells resists external mechanical forces and plays critical roles in a variety of cellular processes including morphogenesis, cytokinesis, and cell migration. Despite its ubiquity and significance, we understand relatively little about the composition, dynamics, and structure of the actin cortex. Adenomatous polyposis coli (APC) proteins regulate the actin and microtubule cytoskeletons through a variety of mechanisms, and in some contexts, APC proteins are cortically enriched. Here we show that APC2 regulates cortical actin dynamics in the follicular epithelium and the nurse cells of the Drosophila ovary and in addition affects the distribution of cortical actin at the apical side of the follicular epithelium. To understand how APC2 influences these properties of the actin cortex, we investigated the mechanisms controlling the cortical localization of APC2 in S2 cultured cells. We previously showed that the N-terminal half of APC2 containing the Armadillo repeats and the C-terminal 30 amino acids (C30) are together necessary and sufficient for APC2's cortical localization. Our work presented here supports a model that cortical localization of APC2 is governed in part by self-association through the N-terminal APC Self-Association Domain (ASAD) and a highly conserved coiled-coil within the C30 domain.
Collapse
Affiliation(s)
- Olivia Molinar-Inglis
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Stacie L Oliver
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Paige Rudich
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Ezgi Kunttas
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Brooke M McCartney
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Daly CS, Shaw P, Ordonez LD, Williams GT, Quist J, Grigoriadis A, Van Es JH, Clevers H, Clarke AR, Reed KR. Functional redundancy between Apc and Apc2 regulates tissue homeostasis and prevents tumorigenesis in murine mammary epithelium. Oncogene 2017; 36:1793-1803. [PMID: 27694902 PMCID: PMC5219933 DOI: 10.1038/onc.2016.342] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 08/02/2016] [Accepted: 08/05/2016] [Indexed: 02/08/2023]
Abstract
Aberrant Wnt signaling within breast cancer is associated with poor prognosis, but regulation of this pathway in breast tissue remains poorly understood and the consequences of immediate or long-term dysregulation remain elusive. The exact contribution of the Wnt-regulating proteins adenomatous polyposis coli (APC) and APC2 in the pathogenesis of human breast cancer are ill-defined, but our analysis of publically available array data sets indicates that tumors with concomitant low expression of both proteins occurs more frequently in the 'triple negative' phenotype, which is a subtype of breast cancer with particularly poor prognosis. We have used mouse transgenics to delete Apc and/or Apc2 from mouse mammary epithelium to elucidate the significance of these proteins in mammary homeostasis and delineate their influences on Wnt signaling and tumorigenesis. Loss of either protein alone failed to affect Wnt signaling levels or tissue homeostasis. Strikingly, concomitant loss led to local disruption of β-catenin status, disruption in epithelial integrity, cohesion and polarity, increased cell division and a distinctive form of ductal hyperplasia with 'squamoid' ghost cell nodules in young animals. Upon aging, the development of Wnt activated mammary carcinomas with squamous differentiation was accompanied by a significantly reduced survival. This novel Wnt-driven mammary tumor model highlights the importance of functional redundancies existing between the Apc proteins both in normal homeostasis and in tumorigenesis.
Collapse
Affiliation(s)
- C S Daly
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, Wales, UK
| | - P Shaw
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, Wales, UK
| | - L D Ordonez
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, Wales, UK
| | - G T Williams
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - J Quist
- Breast Cancer Now Unit, King's College London, Guy's Hospital London, London, UK
- Cancer Bioinformatics, King's College London, Guy's Hospital London, London, UK
| | - A Grigoriadis
- Breast Cancer Now Unit, King's College London, Guy's Hospital London, London, UK
- Cancer Bioinformatics, King's College London, Guy's Hospital London, London, UK
| | - J H Van Es
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | - H Clevers
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | - A R Clarke
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, Wales, UK
| | - K R Reed
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, Wales, UK
| |
Collapse
|
13
|
Armc5 deletion causes developmental defects and compromises T-cell immune responses. Nat Commun 2017; 8:13834. [PMID: 28169274 PMCID: PMC5309699 DOI: 10.1038/ncomms13834] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 11/04/2016] [Indexed: 01/10/2023] Open
Abstract
Armadillo repeat containing 5 (ARMC5) is a cytosolic protein with no enzymatic activities. Little is known about its function and mechanisms of action, except that gene mutations are associated with risks of primary macronodular adrenal gland hyperplasia. Here we map Armc5 expression by in situ hybridization, and generate Armc5 knockout mice, which are small in body size. Armc5 knockout mice have compromised T-cell proliferation and differentiation into Th1 and Th17 cells, increased T-cell apoptosis, reduced severity of experimental autoimmune encephalitis, and defective immune responses to lymphocytic choriomeningitis virus infection. These mice also develop adrenal gland hyperplasia in old age. Yeast 2-hybrid assays identify 16 ARMC5-binding partners. Together these data indicate that ARMC5 is crucial in fetal development, T-cell function and adrenal gland growth homeostasis, and that the functions of ARMC5 probably depend on interaction with multiple signalling pathways. Mutations in ARMC5 are associated with risk of primary macronodular adrenal gland hyperplasia. Here the authors show that mice lacking Armc5 have adrenal gland hyperplasia and defective T-cell proliferation, differentiation, survival and in vivo T-cell-mediated immune responses.
Collapse
|
14
|
Tang Q, Zhao H, Yang B, Li L, Shi Q, Jiang C, Liu H. WIF-1 gene inhibition and Wnt signal transduction pathway activation in NSCLC tumorigenesis. Oncol Lett 2017; 13:1183-1188. [PMID: 28454231 PMCID: PMC5403432 DOI: 10.3892/ol.2017.5566] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 11/01/2016] [Indexed: 01/24/2023] Open
Abstract
The aim of the present study is to explore the differential expression of key molecules associated with Wnt signaling in both clinical non-small cell lung cancer (NSCLC) tissue and adjacent normal lung tissue, and to discuss the tumorigenic role of the activation of Wnt signaling pathways in NSCLC. A total of 52 NSCLC patients were employed in the present study. Lung cancer tissue samples and paracarcinoma tissue samples were obtained from these patients, who had undergone surgical resection of their primary cancer. The cases were diagnosed by hematoxylin and eosin staining. Using reverse transcription-quantitative polymerase chain reaction and immunohistochemical straining, the messenger RNA (mRNA) and protein expression levels of Wnt inhibitory factor-1 (WIF-1) and important molecules associated with Wnt signaling pathways were detected. Compared with normal tissues, a marked decreased in the mRNA and protein expression levels of WIF-1, and an increase in β-catenin and cyclin D1 expression, were observed in tumor tissues. This suggests that the activation of the Wnt/β-catenin signaling pathway may be closely associated with lymph nodal metastasis and lower pathological classification. However, no obvious difference could be observed in adenomatous polyposis coli (APC) expression levels between lung cancer tissues and adjacent tissues to the carcinoma. The activation of the Wnt/β-catenin signaling pathway in NSCLC could be initiated by WIF-1 gene inhibition without APC expression changes, and this may be different to the mechanism in other tumors.
Collapse
Affiliation(s)
- Qiong Tang
- Department of Respiratory Medicine, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Hui Zhao
- Department of Respiratory Medicine, Tianjin Union Medical Center, Tianjin 300121, P.R. China.,Department of Thoracic Surgery, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Bingjun Yang
- Department of Thoracic Surgery, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Li Li
- Department of Thoracic Surgery, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Qiulan Shi
- School of Foreign Languages, North China University of Science and Technology, Tangshan, Hebei 063001, P.R. China
| | - Chunyang Jiang
- Department of Thoracic Surgery, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Huibin Liu
- Office of Clinical Drug Trial Institution, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
15
|
Pronobis MI, Deuitch N, Posham V, Mimori-Kiyosue Y, Peifer M. Reconstituting regulation of the canonical Wnt pathway by engineering a minimal β-catenin destruction machine. Mol Biol Cell 2016; 28:41-53. [PMID: 27852897 PMCID: PMC5221518 DOI: 10.1091/mbc.e16-07-0557] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/01/2016] [Accepted: 11/07/2016] [Indexed: 02/05/2023] Open
Abstract
APC and Axin are key negative regulators of Wnt signaling in development and oncogenesis. They form a multiprotein complex targeting the key Wnt effector β-catenin for destruction. Essential components of APC and Axin required for their cooperative function are identified, and the data are used to design a minimal β-catenin–destruction machine. Negatively regulating key signaling pathways is critical to development and altered in cancer. Wnt signaling is kept off by the destruction complex, which is assembled around the tumor suppressors APC and Axin and targets β-catenin for destruction. Axin and APC are large proteins with many domains and motifs that bind other partners. We hypothesized that if we identified the essential regions required for APC:Axin cooperative function and used these data to design a minimal β-catenin-destruction machine, we would gain new insights into the core mechanisms of destruction complex function. We identified five key domains/motifs in APC or Axin that are essential for their function in reconstituting Wnt regulation. Strikingly, however, certain APC and Axin mutants that are nonfunctional on their own can complement one another in reducing β-catenin, revealing that the APC:Axin complex is a highly robust machine. We used these insights to design a minimal β-catenin-destruction machine, revealing that a minimized chimeric protein covalently linking the five essential regions of APC and Axin reconstitutes destruction complex internal structure, size, and dynamics, restoring efficient β-catenin destruction in colorectal tumor cells. On the basis of our data, we propose a new model of the mechanistic function of the destruction complex as an integrated machine.
Collapse
Affiliation(s)
- Mira I Pronobis
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Natalie Deuitch
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Vinya Posham
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yuko Mimori-Kiyosue
- Cellular Dynamics Analysis Unit, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 .,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
16
|
Kunttas-Tatli E, Von Kleeck RA, Greaves BD, Vinson D, Roberts DM, McCartney BM. The two SAMP repeats and their phosphorylation state in Drosophila Adenomatous polyposis coli-2 play mechanistically distinct roles in negatively regulating Wnt signaling. Mol Biol Cell 2015; 26:4503-18. [PMID: 26446838 PMCID: PMC4666143 DOI: 10.1091/mbc.e15-07-0515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/28/2015] [Indexed: 11/30/2022] Open
Abstract
The colon cancer tumor suppressor Adenomatous polyposis coli (APC) negatively regulates Wnt signaling destruction complex by binding to β-catenin and facilitating its phosphorylation and degradation. The two SAMP repeats and their phosphorylation state in Drosophila APC2 play distinct roles in negatively regulating Wnt signaling. The tumor suppressor Adenomatous polyposis coli (APC) plays a key role in regulating the canonical Wnt signaling pathway as an essential component of the β-catenin destruction complex. C-terminal truncations of APC are strongly implicated in both sporadic and familial forms of colorectal cancer. However, many questions remain as to how these mutations interfere with APC’s tumor suppressor activity. One set of motifs frequently lost in these cancer-associated truncations is the SAMP repeats that mediate interactions between APC and Axin. APC proteins in both vertebrates and Drosophila contain multiple SAMP repeats that lack high sequence conservation outside of the Axin-binding motif. In this study, we tested the functional redundancy between different SAMPs and how these domains are regulated, using Drosophila APC2 and its two SAMP repeats as our model. Consistent with sequence conservation–based predictions, we show that SAMP2 has stronger binding activity to Axin in vitro, but SAMP1 also plays an essential role in the Wnt destruction complex in vivo. In addition, we demonstrate that the phosphorylation of SAMP repeats is a potential mechanism to regulate their activity. Overall our findings support a model in which each SAMP repeat plays a mechanistically distinct role but they cooperate for maximal destruction complex function.
Collapse
Affiliation(s)
- Ezgi Kunttas-Tatli
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Ryan A Von Kleeck
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - Bradford D Greaves
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - David Vinson
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - David M Roberts
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - Brooke M McCartney
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
17
|
Pronobis MI, Rusan NM, Peifer M. A novel GSK3-regulated APC:Axin interaction regulates Wnt signaling by driving a catalytic cycle of efficient βcatenin destruction. eLife 2015; 4:e08022. [PMID: 26393419 PMCID: PMC4568445 DOI: 10.7554/elife.08022] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/20/2015] [Indexed: 01/11/2023] Open
Abstract
APC, a key negative regulator of Wnt signaling in development and oncogenesis, acts in the destruction complex with the scaffold Axin and the kinases GSK3 and CK1 to target βcatenin for destruction. Despite 20 years of research, APC's mechanistic function remains mysterious. We used FRAP, super-resolution microscopy, functional tests in mammalian cells and flies, and other approaches to define APC's mechanistic role in the active destruction complex when Wnt signaling is off. Our data suggest APC plays two roles: (1) APC promotes efficient Axin multimerization through one known and one novel APC:Axin interaction site, and (2) GSK3 acts through APC motifs R2 and B to regulate APC:Axin interactions, promoting high-throughput of βcatenin to destruction. We propose a new dynamic model of how the destruction complex regulates Wnt signaling and how this goes wrong in cancer, providing insights into how this multiprotein signaling complex is assembled and functions via multivalent interactions. DOI:http://dx.doi.org/10.7554/eLife.08022.001 An embryo starts off as a small ball of stem cells, each of which has the potential to become any type of cell in the body. Adult organs and tissues also contain small numbers of stem cells that can replace old or damaged cells. In both of these processes, stem cells need to ‘decide’ when they should start to change into a more specialized cell type, and which cell fate to choose (e.g., liver cell vs kidney cell). A signaling pathway involving Wnt proteins helps to direct many of these decisions. But if the ‘Wnt signaling pathway’ becomes activated at the wrong time, it can lead to cancer. For example, the first step in development of colon cancer is the inappropriate activation of Wnt signaling, and is most often caused by mutations in the gene that encodes a protein called APC. The APC protein is a tumor suppressor and normally inhibits Wnt signaling. However, even after over 20 years of effort, it remains largely mysterious how APC does this. APC is known to work with another protein called Axin as part of a large protein machine. This protein complex performs one of the first steps in a process that ultimately marks a key component of the Wnt signaling pathway for destruction. Pronobis et al. have now used a range of techniques to define APC's role in this so-called ‘destruction complex’. This analysis revealed the internal structure of a complex made from APC and Axin, and showed that cable- and sheet-like assemblies of Axin were intertwined with APC cables. Further experiments then revealed how APC and Axin proteins are added into or leave these complexes, and showed that this is critical for this protein machine to work. Pronobis et al.'s data also suggest that APC plays two roles, which make the destruction complex more efficient. Firstly, it can interact with Axin via two separate interaction sites that help to assemble the destruction complex. Secondly, specific features in APC allow it to interact with a third protein (called GSK3), which can then regulate how APC interacts with Axin. One of the next challenges will be to uncover how APC helps to transfer the components of Wnt signaling to the next step of their destruction, and to clear up the role played by GSK3. DOI:http://dx.doi.org/10.7554/eLife.08022.002
Collapse
Affiliation(s)
- Mira I Pronobis
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Nasser M Rusan
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, Bethesda, United States
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
18
|
Beaven R, Dzhindzhev NS, Qu Y, Hahn I, Dajas-Bailador F, Ohkura H, Prokop A. Drosophila CLIP-190 and mammalian CLIP-170 display reduced microtubule plus end association in the nervous system. Mol Biol Cell 2015; 26:1491-508. [PMID: 25694447 PMCID: PMC4395129 DOI: 10.1091/mbc.e14-06-1083] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 01/05/2015] [Accepted: 02/10/2015] [Indexed: 11/11/2022] Open
Abstract
Axons act like cables, electrically wiring the nervous system. Polar bundles of microtubules (MTs) form their backbones and drive their growth. Plus end-tracking proteins (+TIPs) regulate MT growth dynamics and directionality at their plus ends. However, current knowledge about +TIP functions, mostly derived from work in vitro and in nonneuronal cells, may not necessarily apply to the very different context of axonal MTs. For example, the CLIP family of +TIPs are known MT polymerization promoters in nonneuronal cells. However, we show here that neither Drosophila CLIP-190 nor mammalian CLIP-170 is a prominent MT plus end tracker in neurons, which we propose is due to low plus end affinity of the CAP-Gly domain-containing N-terminus and intramolecular inhibition through the C-terminus. Instead, both CLIP-190 and CLIP-170 form F-actin-dependent patches in growth cones, mediated by binding of the coiled-coil domain to myosin-VI. Because our loss-of-function analyses in vivo and in culture failed to reveal axonal roles for CLIP-190, even in double-mutant combinations with four other +TIPs, we propose that CLIP-190 and -170 are not essential axon extension regulators. Our findings demonstrate that +TIP functions known from nonneuronal cells do not necessarily apply to the regulation of the very distinct MT networks in axons.
Collapse
Affiliation(s)
- Robin Beaven
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Nikola S Dzhindzhev
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Yue Qu
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Ines Hahn
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| | | | - Hiroyuki Ohkura
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Andreas Prokop
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
19
|
Mannava AG, Tolwinski NS. Membrane bound GSK-3 activates Wnt signaling through disheveled and arrow. PLoS One 2015; 10:e0121879. [PMID: 25848770 PMCID: PMC4388798 DOI: 10.1371/journal.pone.0121879] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 02/17/2015] [Indexed: 01/22/2023] Open
Abstract
Wnt ligands and their downstream pathway components coordinate many developmental and cellular processes. In adults, they regulate tissue homeostasis through regulation of stem cells. Mechanistically, signal transduction through this pathway is complicated by pathway components having both positive and negative roles in signal propagation. Here we examine the positive role of GSK-3/Zw3 in promoting signal transduction at the plasma membrane. We find that targeting GSK-3 to the plasma membrane activates signaling in Drosophila embryos. This activation requires the presence of the co-receptor Arrow-LRP5/6 and the pathway activating protein Disheveled. Our results provide genetic evidence for evolutionarily conserved, separable roles for GSK-3 at the membrane and in the cytosol, and are consistent with a model where the complex cycles from cytosol to membrane in order to promote signaling at the membrane and to prevent it in the cytosol.
Collapse
Affiliation(s)
- Anirudh G. Mannava
- Yale-NUS College and Department of Biological Sciences, National University of Singapore, Block MD6, Centre for Translational Medicine, Yong Loo Lin School of Medicine, 14 Medical Drive, Level 10 South, 10-02M, Singapore 117599, Singapore
| | - Nicholas S. Tolwinski
- Yale-NUS College and Department of Biological Sciences, National University of Singapore, Block MD6, Centre for Translational Medicine, Yong Loo Lin School of Medicine, 14 Medical Drive, Level 10 South, 10-02M, Singapore 117599, Singapore
- * E-mail:
| |
Collapse
|
20
|
Kunttas-Tatli E, Roberts DM, McCartney BM. Self-association of the APC tumor suppressor is required for the assembly, stability, and activity of the Wnt signaling destruction complex. Mol Biol Cell 2014; 25:3424-36. [PMID: 25208568 PMCID: PMC4214788 DOI: 10.1091/mbc.e14-04-0885] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The tumor suppressor adenomatous polyposis coli (APC) is an essential negative regulator of Wnt signaling through its activity in the destruction complex with Axin, GSK3β, and CK1 that targets β-catenin/Armadillo (β-cat/Arm) for proteosomal degradation. The destruction complex forms macromolecular particles we termed the destructosome. Whereas APC functions in the complex through its ability to bind both β-cat and Axin, we hypothesize that APC proteins play an additional role in destructosome assembly through self-association. Here we show that a novel N-terminal coil, the APC self-association domain (ASAD), found in vertebrate and invertebrate APCs, directly mediates self-association of Drosophila APC2 and plays an essential role in the assembly and stability of the destructosome that regulates β-cat degradation in Drosophila and human cells. Consistent with this, removal of the ASAD from the Drosophila embryo results in β-cat/Arm accumulation and aberrant Wnt pathway activation. These results suggest that APC proteins are required not only for the activity of the destructosome, but also for the assembly and stability of this macromolecular machine.
Collapse
Affiliation(s)
- Ezgi Kunttas-Tatli
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - David M Roberts
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - Brooke M McCartney
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
21
|
Testing models of the APC tumor suppressor/β-catenin interaction reshapes our view of the destruction complex in Wnt signaling. Genetics 2014; 197:1285-302. [PMID: 24931405 DOI: 10.1534/genetics.114.166496] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Wnt pathway is a conserved signal transduction pathway that contributes to normal development and adult homeostasis, but is also misregulated in human diseases such as cancer. The tumor suppressor adenomatous polyposis coli (APC) is an essential negative regulator of Wnt signaling inactivated in >80% of colorectal cancers. APC participates in a multiprotein "destruction complex" that targets the proto-oncogene β-catenin for ubiquitin-mediated proteolysis; however, the mechanistic role of APC in the destruction complex remains unknown. Several models of APC function have recently been proposed, many of which have emphasized the importance of phosphorylation of high-affinity β-catenin-binding sites [20-amino-acid repeats (20Rs)] on APC. Here we test these models by generating a Drosophila APC2 mutant lacking all β-catenin-binding 20Rs and performing functional studies in human colon cancer cell lines and Drosophila embryos. Our results are inconsistent with current models, as we find that β-catenin binding to the 20Rs of APC is not required for destruction complex activity. In addition, we generate an APC2 mutant lacking all β-catenin-binding sites (including the 15Rs) and find that a direct β-catenin/APC interaction is also not essential for β-catenin destruction, although it increases destruction complex efficiency in certain developmental contexts. Overall, our findings support a model whereby β-catenin-binding sites on APC do not provide a critical mechanistic function per se, but rather dock β-catenin in the destruction complex to increase the efficiency of β-catenin destruction. Furthermore, in Drosophila embryos expressing some APC2 mutant transgenes we observe a separation of β-catenin destruction and Wg/Wnt signaling outputs and suggest that cytoplasmic retention of β-catenin likely accounts for this difference.
Collapse
|
22
|
Poulton JS, Mu FW, Roberts DM, Peifer M. APC2 and Axin promote mitotic fidelity by facilitating centrosome separation and cytoskeletal regulation. Development 2013; 140:4226-36. [PMID: 24026117 DOI: 10.1242/dev.094425] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To ensure the accurate transmission of genetic material, chromosome segregation must occur with extremely high fidelity. Segregation errors lead to chromosomal instability (CIN), with deleterious consequences. Mutations in the tumor suppressor adenomatous polyposis coli (APC) initiate most colon cancers and have also been suggested to promote disease progression through increased CIN, but the mechanistic role of APC in preventing CIN remains controversial. Using fly embryos as a model, we investigated the role of APC proteins in CIN. Our findings suggest that APC2 loss leads to increased rates of chromosome segregation error. This occurs through a cascade of events beginning with incomplete centrosome separation leading to failure to inhibit formation of ectopic cleavage furrows, which result in mitotic defects and DNA damage. We test several hypotheses related to the mechanism of action of APC2, revealing that APC2 functions at the embryonic cortex with several protein partners, including Axin, to promote mitotic fidelity. Our in vivo data demonstrate that APC2 protects genome stability by modulating mitotic fidelity through regulation of the cytoskeleton.
Collapse
Affiliation(s)
- John S Poulton
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
23
|
Behrens J. Everything You Would Like to Know About Wnt SignalingReview and commentary on
Wnt Signaling
, edited by Roel Nusse, Xi He, and Renée van Amerongen. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2013, 454 pp. ISBN: 978-1-936113-23-1. Sci Signal 2013. [DOI: 10.1126/scisignal.2004167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jürgen Behrens
- Friedrich-Alexander University Erlangen-Nuremberg, Nikolaus-Fiebiger-Center, Glückstrasse 6, D-91054 Erlangen, Germany
| |
Collapse
|
24
|
Gonzalez C. Drosophila melanogaster: a model and a tool to investigate malignancy and identify new therapeutics. Nat Rev Cancer 2013; 13:172-83. [PMID: 23388617 DOI: 10.1038/nrc3461] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
For decades, lower-model organisms such as Drosophila melanogaster have often provided the first glimpse into the mechanism of action of human cancer-related proteins, thus making a substantial contribution to elucidating the molecular basis of the disease. More recently, D. melanogaster strains that are engineered to recapitulate key aspects of specific types of human cancer have been paving the way for the future role of this 'workhorse' of biomedical research, helping to further investigate the process of malignancy, and serving as platforms for therapeutic drug discovery.
Collapse
Affiliation(s)
- Cayetano Gonzalez
- IRB-Barcelona, c/Baldiri Reixac 10-12, Barcelona, Spain. gonzalez@ irbbarcelona.org
| |
Collapse
|
25
|
Abstract
The Wnt/β-catenin pathway is highly regulated to insure the correct temporal and spatial activation of its target genes. In the absence of a Wnt stimulus, the transcriptional coactivator β-catenin is degraded by a multiprotein "destruction complex" that includes the tumor suppressors Axin and adenomatous polyposis coli (APC), the Ser/Thr kinases GSK-3 and CK1, protein phosphatase 2A (PP2A), and the E3-ubiquitin ligase β-TrCP. The complex generates a β-TrCP recognition site by phosphorylation of a conserved Ser/Thr-rich sequence near the β-catenin amino terminus, a process that requires scaffolding of the kinases and β-catenin by Axin. Ubiquitinated β-catenin is degraded by the proteasome. The molecular mechanisms that underlie several aspects of destruction complex function are poorly understood, particularly the role of APC. Here we review the molecular mechanisms of destruction complex function and discuss several potential roles of APC in β-catenin destruction.
Collapse
Affiliation(s)
- Jennifer L Stamos
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|