1
|
Purkerson MM, Amend SR, Pienta KJ. Bystanders or active players: the role of extra centrosomes as signaling hubs. Cancer Metastasis Rev 2024; 44:1. [PMID: 39570514 PMCID: PMC11582193 DOI: 10.1007/s10555-024-10224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Centrosomes serve as microtubule-organizing organelles that function in spindle pole organization, cell cycle progression, and cilia formation. A non-canonical role of centrosomes that has gained traction in recent years is the ability to act as signal transduction centers. Centrosome amplification, which includes numerical and structural aberrations of centrosomes, is a candidate hallmark of cancer. The function of centrosomes as signaling centers in cancer cells with centrosome amplification is poorly understood. Establishing a model of how cancer cells utilize centrosomes as signaling platforms will help elucidate the role of extra centrosomes in cancer cell survival and tumorigenesis. Centrosomes act in a diverse array of cellular processes, including cell migration, cell cycle progression, and proteasomal degradation. Given that cancer cells with amplified centrosomes exhibit an increased number and larger area of these signaling platforms, extra centrosomes may be acting to promote tumor development by enhancing signaling kinetics in pathways that are essential for the formation and growth of cancer. In this review, we identify the processes centrosomes are involved in as signal transduction platforms and highlight ways in which cancer cells with centrosome amplification may be taking advantage of these mechanisms.
Collapse
Affiliation(s)
- Madison M Purkerson
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Pharmacology and Molecular Sciences Program, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Sarah R Amend
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kenneth J Pienta
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Pharmacology and Molecular Sciences Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Cheng T, Agwu C, Shim K, Wang B, Jain S, Mahjoub MR. Aberrant centrosome biogenesis disrupts nephron and collecting duct progenitor growth and fate resulting in fibrocystic kidney disease. Development 2023; 150:dev201976. [PMID: 37982452 PMCID: PMC10753588 DOI: 10.1242/dev.201976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Mutations that disrupt centrosome biogenesis or function cause congenital kidney developmental defects and fibrocystic pathologies. Yet how centrosome dysfunction results in the kidney disease phenotypes remains unknown. Here, we examined the consequences of conditional knockout of the ciliopathy gene Cep120, essential for centrosome duplication, in the nephron and collecting duct progenitor niches of the mouse embryonic kidney. Cep120 loss led to reduced abundance of both cap mesenchyme and ureteric bud populations, due to a combination of delayed mitosis, increased apoptosis and premature differentiation of progenitor cells. These defects resulted in dysplastic kidneys at birth, which rapidly formed cysts, displayed increased interstitial fibrosis and decline in kidney function. RNA sequencing of embryonic and postnatal kidneys from Cep120-null mice identified changes in the pathways essential for development, fibrosis and cystogenesis. Our study defines the cellular and developmental defects caused by centrosome dysfunction during kidney morphogenesis and identifies new therapeutic targets for patients with renal centrosomopathies.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Medicine, Division of Nephrology, Washington University in St Louis, St. Louis, MO 63110, USA
| | - Chidera Agwu
- Department of Medicine, Division of Nephrology, Washington University in St Louis, St. Louis, MO 63110, USA
| | - Kyuhwan Shim
- Department of Medicine, Division of Nephrology, Washington University in St Louis, St. Louis, MO 63110, USA
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Sanjay Jain
- Department of Medicine, Division of Nephrology, Washington University in St Louis, St. Louis, MO 63110, USA
| | - Moe R. Mahjoub
- Department of Medicine, Division of Nephrology, Washington University in St Louis, St. Louis, MO 63110, USA
- Department of Cell Biology and Physiology, Washington University in St Louis, St. Louis, MO 63110, USA
| |
Collapse
|
3
|
Wiecek AJ, Cutty SJ, Kornai D, Parreno-Centeno M, Gourmet LE, Tagliazucchi GM, Jacobson DH, Zhang P, Xiong L, Bond GL, Barr AR, Secrier M. Genomic hallmarks and therapeutic implications of G0 cell cycle arrest in cancer. Genome Biol 2023; 24:128. [PMID: 37221612 PMCID: PMC10204193 DOI: 10.1186/s13059-023-02963-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 05/07/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Therapy resistance in cancer is often driven by a subpopulation of cells that are temporarily arrested in a non-proliferative G0 state, which is difficult to capture and whose mutational drivers remain largely unknown. RESULTS We develop methodology to robustly identify this state from transcriptomic signals and characterise its prevalence and genomic constraints in solid primary tumours. We show that G0 arrest preferentially emerges in the context of more stable, less mutated genomes which maintain TP53 integrity and lack the hallmarks of DNA damage repair deficiency, while presenting increased APOBEC mutagenesis. We employ machine learning to uncover novel genomic dependencies of this process and validate the role of the centrosomal gene CEP89 as a modulator of proliferation and G0 arrest capacity. Lastly, we demonstrate that G0 arrest underlies unfavourable responses to various therapies exploiting cell cycle, kinase signalling and epigenetic mechanisms in single-cell data. CONCLUSIONS We propose a G0 arrest transcriptional signature that is linked with therapeutic resistance and can be used to further study and clinically track this state.
Collapse
Affiliation(s)
- Anna J. Wiecek
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Stephen J. Cutty
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Daniel Kornai
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Mario Parreno-Centeno
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Lucie E. Gourmet
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | | | - Daniel H. Jacobson
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
- UCL Cancer Institute, Paul O’Gorman Building, University College London, London, UK
| | - Ping Zhang
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lingyun Xiong
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gareth L. Bond
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Alexis R. Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Cell Cycle Control Team, MRC London Institute of Medical Sciences (LMS), London, UK
| | - Maria Secrier
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| |
Collapse
|
4
|
Cheng T, Agwu C, Shim K, Wang B, Jain S, Mahjoub MR. Aberrant centrosome biogenesis disrupts nephron progenitor cell renewal and fate resulting in fibrocystic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535568. [PMID: 37066373 PMCID: PMC10104032 DOI: 10.1101/2023.04.04.535568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Mutations that disrupt centrosome structure or function cause congenital kidney developmental defects and fibrocystic pathologies. Yet, it remains unclear how mutations in proteins essential for centrosome biogenesis impact embryonic kidney development. Here, we examined the consequences of conditional deletion of a ciliopathy gene, Cep120 , in the two nephron progenitor niches of the embryonic kidney. Cep120 loss led to reduced abundance of both metanephric mesenchyme and ureteric bud progenitor populations. This was due to a combination of delayed mitosis, increased apoptosis, and premature differentiation of progenitor cells. These defects resulted in dysplastic kidneys at birth, which rapidly formed cysts, displayed increased interstitial fibrosis, and decline in filtration function. RNA sequencing of embryonic and postnatal kidneys from Cep120-null mice identified changes in pathways essential for branching morphogenesis, cystogenesis and fibrosis. Our study defines the cellular and developmental defects caused by centrosome dysfunction during kidney development, and identifies new therapeutic targets for renal centrosomopathies. Highlights Defective centrosome biogenesis in nephron progenitors causes:Reduced abundance of metanephric mesenchyme and premature differentiation into tubular structuresAbnormal branching morphogenesis leading to reduced nephron endowment and smaller kidneysChanges in cell-autonomous and paracrine signaling that drive cystogenesis and fibrosisUnique cellular and developmental defects when compared to Pkd1 knockout models.
Collapse
|
5
|
Keep Calm and Carry on with Extra Centrosomes. Cancers (Basel) 2022; 14:cancers14020442. [PMID: 35053604 PMCID: PMC8774008 DOI: 10.3390/cancers14020442] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Precise chromosome segregation during mitosis is a vital event orchestrated by formation of bipolar spindle poles. Supernumerary centrosomes, caused by centrosome amplification, deteriorates mitotic processes, resulting in segregation defects leading to chromosomal instability (CIN). Centrosome amplification is frequently observed in various types of cancer and considered as a significant contributor to destabilization of chromosomes. This review provides a comprehensive overview of causes and consequences of centrosome amplification thoroughly describing molecular mechanisms. Abstract Aberrations in the centrosome number and structure can readily be detected at all stages of tumor progression and are considered hallmarks of cancer. Centrosome anomalies are closely linked to chromosome instability and, therefore, are proposed to be one of the driving events of tumor formation and progression. This concept, first posited by Boveri over 100 years ago, has been an area of interest to cancer researchers. We have now begun to understand the processes by which these numerical and structural anomalies may lead to cancer, and vice-versa: how key events that occur during carcinogenesis could lead to amplification of centrosomes. Despite the proliferative advantages that having extra centrosomes may confer, their presence can also lead to loss of essential genetic material as a result of segregational errors and cancer cells must deal with these deadly consequences. Here, we review recent advances in the current literature describing the mechanisms by which cancer cells amplify their centrosomes and the methods they employ to tolerate the presence of these anomalies, focusing particularly on centrosomal clustering.
Collapse
|
6
|
Torban E, Sokol SY. Planar cell polarity pathway in kidney development, function and disease. Nat Rev Nephrol 2021; 17:369-385. [PMID: 33547419 PMCID: PMC8967065 DOI: 10.1038/s41581-021-00395-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Planar cell polarity (PCP) refers to the coordinated orientation of cells in the tissue plane. Originally discovered and studied in Drosophila melanogaster, PCP is now widely recognized in vertebrates, where it is implicated in organogenesis. Specific sets of PCP genes have been identified. The proteins encoded by these genes become asymmetrically distributed to opposite sides of cells within a tissue plane and guide many processes that include changes in cell shape and polarity, collective cell movements or the uniform distribution of cell appendages. A unifying characteristic of these processes is that they often involve rearrangement of actomyosin. Mutations in PCP genes can cause malformations in organs of many animals, including humans. In the past decade, strong evidence has accumulated for a role of the PCP pathway in kidney development including outgrowth and branching morphogenesis of ureteric bud and podocyte development. Defective PCP signalling has been implicated in the pathogenesis of developmental kidney disorders of the congenital anomalies of the kidney and urinary tract spectrum. Understanding the origins, molecular constituents and cellular targets of PCP provides insights into the involvement of PCP molecules in normal kidney development and how dysfunction of PCP components may lead to kidney disease.
Collapse
Affiliation(s)
- Elena Torban
- McGill University and McGill University Health Center Research Institute, 1001 Boulevard Decarie, Block E, Montreal, Quebec, Canada, H4A3J1.,Corresponding authors: Elena Torban (); Sergei Sokol ()
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, USA,Corresponding authors: Elena Torban (); Sergei Sokol ()
| |
Collapse
|
7
|
Bai R, Wu D, Shi Z, Hu W, Li J, Chen Y, Ge W, Yuan Y, Zheng S. Pan-cancer analyses demonstrate that ANKRD6 is associated with a poor prognosis and correlates with M2 macrophage infiltration in colon cancer. Chin J Cancer Res 2021; 33:93-102. [PMID: 33707932 PMCID: PMC7941690 DOI: 10.21147/j.issn.1000-9604.2021.01.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Objective Ankyrin repeat domain-containing protein 6 (ANKRD6) is an ankyrin repeat-containing protein which is structurally related to vertebrate inversin and Drosophila Diego. However, the correlations between ANKRD6 and tumor-infiltrating immune cells in cancers is not clear. Methods ANKRD6 expression was analyzed by Oncomine, Tumor Immune Estimation Resource (TIMER) and Gene Expression Profiling Interactive Analysis (GEPIA). PrognoScan and GEPIA were used to evaluate the influence of ANKRD6 on clinical prognosis. TIMER and CIBERSORT were used to analyze correlations between ANKRD6 expression levels and tumor immune cell infiltrates. Immunohistochemical analysis of the relationship between ANKRD6 expression and overall survival, as well as the relationship between ANKRD6 expression and M2 macrophage infiltration, was performed. Results High level of ANKRD6 expression was associated with poor prognosis of colon cancer. ANKRD6 expression level was positively correlated with infiltrating levels of CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells in colon cancer by using TIMER. Using CIBERSORT, we found that in plasma cells, CD8+ T cells, CD4+ memory resting T cells, follicular helper T cells and activated natural killer cells were significantly lower in the ANKRD6-high group than in the ANKRD6-low group. M0 and M2 macrophages were significantly higher in the ANKRD6-high group than in the ANKRD6-low group. Immunohistochemistry confirmed that M2 macrophage infiltration in the ANKRD6-high group significantly increased. Conclusions The high ANKRD6 expression is associated with poor prognosis of colon cancer. ANKRD6 expression is positively correlated with M2 macrophage infiltration in colon cancer.
Collapse
Affiliation(s)
- Rui Bai
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Dehao Wu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhong Shi
- Department of Medical Oncology, Institute of Cancer and Basic Medicine (ICBM) Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Wangxiong Hu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Juan Li
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Yuanyuan Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Weiting Ge
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Ying Yuan
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Shu Zheng
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
8
|
Vora SM, Fassler JS, Phillips BT. Centrosomes are required for proper β-catenin processing and Wnt response. Mol Biol Cell 2020; 31:1951-1961. [PMID: 32583737 PMCID: PMC7525817 DOI: 10.1091/mbc.e20-02-0139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The Wnt/β-catenin signaling pathway is central to metazoan development and routinely dysregulated in cancer. Wnt/β-catenin signaling initiates transcriptional reprogramming upon stabilization of the transcription factor β-catenin, which is otherwise posttranslationally processed by a destruction complex and degraded by the proteasome. Since various Wnt signaling components are enriched at centrosomes, we examined the functional contribution of centrosomes to Wnt signaling, β-catenin regulation, and posttranslational modifications. In HEK293 cells depleted of centrosomes we find that β-catenin synthesis and degradation rates are unaffected but that the normal accumulation of β-catenin in response to Wnt signaling is attenuated. This is due to accumulation of a novel high-molecular-weight form of phosphorylated β-catenin that is constitutively degraded in the absence of Wnt. Wnt signaling operates by inhibiting the destruction complex and thereby reducing destruction complex–phosphorylated β-catenin, but high-molecular-weight β-catenin is unexpectedly increased by Wnt signaling. Therefore these studies have identified a pool of β-catenin effectively shielded from regulation by Wnt. We present a model whereby centrosomes prevent inappropriate β-catenin modifications that antagonize normal stabilization by Wnt signals.
Collapse
Affiliation(s)
- Setu M Vora
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324
| | - Jan S Fassler
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324
| | - Bryan T Phillips
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324
| |
Collapse
|
9
|
Yang K, Tylkowski MA, Hüber D, Contreras CT, Hoyer-Fender S. ODF2/Cenexin maintains centrosome cohesion by restricting β-catenin accumulation. J Cell Sci 2018; 131:jcs.220954. [DOI: 10.1242/jcs.220954] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/10/2018] [Indexed: 12/22/2022] Open
Abstract
The centrosome, as the main microtubule organizing center, safeguards chromosome segregation by constituting the bipolar spindle. Centrosome aberrations are causally related to chromosome segregation disorders, both characterizing cancer cells. Thus, restriction to only one centrosome per cell, and cell cycle dependent duplication is mandatory. Duplicated centrosomes remain physically connected to function as a single entity, until onset of mitosis when centrosome disjunction is licensed by disassembly of linker proteins and accumulation of β-catenin. The crucial role β-catenin plays in centrosome disjunction inevitably demands for restricting its premature accumulation. ODF2/Cenexin is an essential centrosomal component but its relevance for the interphase centrosome has not been elucidated. We show here, that ODF2/Cenexin plays a central role in centrosome cohesion. Depletion of ODF2/Cenexin induces premature centrosome splitting and formation of tripolar spindles that are likely caused by the observed accumulation of centrosomal β-catenin. Our data collectively indicate that ODF2/Cenexin restricts β-catenin accumulation at the centrosome thus preventing premature centrosome disjunction.
Collapse
Affiliation(s)
- Kefei Yang
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology – Developmental Biology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| | - Marco Andreas Tylkowski
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology – Developmental Biology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| | - Daniela Hüber
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology – Developmental Biology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| | - Constanza Tapia Contreras
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology – Developmental Biology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| | - Sigrid Hoyer-Fender
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology – Developmental Biology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| |
Collapse
|
10
|
Harrison LE, Bleiler M, Giardina C. A look into centrosome abnormalities in colon cancer cells, how they arise and how they might be targeted therapeutically. Biochem Pharmacol 2017; 147:1-8. [PMID: 29128368 DOI: 10.1016/j.bcp.2017.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/07/2017] [Indexed: 02/06/2023]
Abstract
Cancer cells have long been noted for alterations in centrosome structure, number, and function. Colorectal cancers are interesting in this regard since two frequently mutated genes, APC and CTNNB1 (β-catenin), encode proteins that directly interact with the centrosome and affect its ability to direct microtubule growth and establish cell polarity. Colorectal cancers also frequently display centrosome over-duplication and clustering. Efforts have been directed toward understanding how supernumerary centrosomes cluster and whether disrupting this clustering may be a way to induce aberrant/lethal mitoses of cancer cells. Given the important role of the centrosome in establishing spindle polarity and regulating some apoptotic signaling pathways, other approaches to centrosome targeting may be fruitful as well. Basic information on the nature and extent of centrosome defects in colorectal cancer, including why they over-duplicate and whether this over-duplication compensates for their functional defects, could provide a framework for the development of novel approaches for the therapeutic targeting of colorectal cancer.
Collapse
Affiliation(s)
- Lauren E Harrison
- Department of Molecular and Cell Biology, 91 North Eagleville Road, U3125, University of Connecticut, Storrs, CT 06269, United States
| | - Marina Bleiler
- Department of Molecular and Cell Biology, 91 North Eagleville Road, U3125, University of Connecticut, Storrs, CT 06269, United States
| | - Charles Giardina
- Department of Molecular and Cell Biology, 91 North Eagleville Road, U3125, University of Connecticut, Storrs, CT 06269, United States.
| |
Collapse
|
11
|
Bryja V, Červenka I, Čajánek L. The connections of Wnt pathway components with cell cycle and centrosome: side effects or a hidden logic? Crit Rev Biochem Mol Biol 2017; 52:614-637. [PMID: 28741966 DOI: 10.1080/10409238.2017.1350135] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Wnt signaling cascade has developed together with multicellularity to orchestrate the development and homeostasis of complex structures. Wnt pathway components - such as β-catenin, Dishevelled (DVL), Lrp6, and Axin-- are often dedicated proteins that emerged in evolution together with the Wnt signaling cascade and are believed to function primarily in the Wnt cascade. It is interesting to see that in recent literature many of these proteins are connected with cellular functions that are more ancient and not limited to multicellular organisms - such as cell cycle regulation, centrosome biology, or cell division. In this review, we summarize the recent literature describing this crosstalk. Specifically, we attempt to find the answers to the following questions: Is the response to Wnt ligands regulated by the cell cycle? Is the centrosome and/or cilium required to activate the Wnt pathway? How do Wnt pathway components regulate the centrosomal cycle and cilia formation and function? We critically review the evidence that describes how these connections are regulated and how they help to integrate cell-to-cell communication with the cell and the centrosomal cycle in order to achieve a fine-tuned, physiological response.
Collapse
Affiliation(s)
- Vítězslav Bryja
- a Department of Experimental Biology, Faculty of Science , Masaryk University , Brno , Czech Republic
| | - Igor Červenka
- b Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology , Karolinska Institutet , Stockholm , Sweden
| | - Lukáš Čajánek
- c Department of Histology and Embryology, Faculty of Medicine , Masaryk University , Brno , Czech Republic
| |
Collapse
|
12
|
Chu CW, Ossipova O, Ioannou A, Sokol SY. Prickle3 synergizes with Wtip to regulate basal body organization and cilia growth. Sci Rep 2016; 6:24104. [PMID: 27062996 PMCID: PMC4827067 DOI: 10.1038/srep24104] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/21/2016] [Indexed: 12/21/2022] Open
Abstract
PCP proteins maintain planar polarity in many epithelial tissues and have been implicated in cilia development in vertebrate embryos. In this study we examine Prickle3 (Pk3), a vertebrate homologue of Drosophila Prickle, in Xenopus gastrocoel roof plate (GRP). GRP is a tissue equivalent to the mouse node, in which cilia-generated flow promotes left-right patterning. We show that Pk3 is enriched at the basal body of GRP cells but is recruited by Vangl2 to anterior cell borders. Interference with Pk3 function disrupted the anterior polarization of endogenous Vangl2 and the posterior localization of cilia in GRP cells, demonstrating its role in PCP. Strikingly, in cells with reduced Pk3 activity, cilia growth was inhibited and γ-tubulin and Nedd1 no longer associated with the basal body, suggesting that Pk3 has a novel function in basal body organization. Mechanistically, this function of Pk3 may involve Wilms tumor protein 1-interacting protein (Wtip), which physically associates with and cooperates with Pk3 to regulate ciliogenesis. We propose that, in addition to cell polarity, PCP components control basal body organization and function.
Collapse
Affiliation(s)
- Chih-Wen Chu
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andriani Ioannou
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
13
|
Ossipova O, Chu CW, Fillatre J, Brott BK, Itoh K, Sokol SY. The involvement of PCP proteins in radial cell intercalations during Xenopus embryonic development. Dev Biol 2015; 408:316-27. [PMID: 26079437 PMCID: PMC4810801 DOI: 10.1016/j.ydbio.2015.06.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 06/10/2015] [Accepted: 06/11/2015] [Indexed: 11/19/2022]
Abstract
The planar cell polarity (PCP) pathway orients cells in diverse epithelial tissues in Drosophila and vertebrate embryos and has been implicated in many human congenital defects and diseases, such as ciliopathies, polycystic kidney disease and malignant cancers. During vertebrate gastrulation and neurulation, PCP signaling is required for convergent extension movements, which are primarily driven by mediolateral cell intercalations, whereas the role for PCP signaling in radial cell intercalations has been unclear. In this study, we examine the function of the core PCP proteins Vangl2, Prickle3 (Pk3) and Disheveled in the ectodermal cells, which undergo radial intercalations during Xenopus gastrulation and neurulation. In the epidermis, multiciliated cell (MCC) progenitors originate in the inner layer, but subsequently migrate to the embryo surface during neurulation. We find that the Vangl2/Pk protein complexes are enriched at the apical domain of intercalating MCCs and are essential for the MCC intercalatory behavior. Addressing the underlying mechanism, we identified KIF13B, as a motor protein that binds Disheveled. KIF13B is required for MCC intercalation and acts synergistically with Vangl2 and Disheveled, indicating that it may mediate microtubule-dependent trafficking of PCP proteins necessary for cell shape regulation. In the neural plate, the Vangl2/Pk complexes were also concentrated near the outermost surface of deep layer cells, suggesting a general role for PCP in radial intercalation. Consistent with this hypothesis, the ectodermal tissues deficient in Vangl2 or Disheveled functions contained more cell layers than normal tissues. We propose that PCP signaling is essential for both mediolateral and radial cell intercalations during vertebrate morphogenesis. These expanded roles underscore the significance of vertebrate PCP proteins as factors contributing to a number of diseases, including neural tube defects, tumor metastases, and various genetic syndromes characterized by abnormal migratory cell behaviors.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chih-Wen Chu
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan Fillatre
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Barbara K Brott
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keiji Itoh
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
14
|
Ossipova O, Kim K, Sokol SY. Planar polarization of Vangl2 in the vertebrate neural plate is controlled by Wnt and Myosin II signaling. Biol Open 2015; 4:722-30. [PMID: 25910938 PMCID: PMC4467192 DOI: 10.1242/bio.201511676] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The vertebrate neural tube forms as a result of complex morphogenetic movements, which require the functions of several core planar cell polarity (PCP) proteins, including Vangl2 and Prickle. Despite the importance of these proteins for neurulation, their subcellular localization and the mode of action have remained largely unknown. Here we describe the anteroposterior planar cell polarity (AP-PCP) of the cells in the Xenopus neural plate. At the neural midline, the Vangl2 protein is enriched at anterior cell edges and that this localization is directed by Prickle, a Vangl2-interacting protein. Our further analysis is consistent with the model, in which Vangl2 AP-PCP is established in the neural plate as a consequence of Wnt-dependent phosphorylation. Additionally, we uncover feedback regulation of Vangl2 polarity by Myosin II, reiterating a role for mechanical forces in PCP. These observations indicate that both Wnt signaling and Myosin II activity regulate cell polarity and cell behaviors during vertebrate neurulation.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kyeongmi Kim
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
15
|
Godinho SA, Pellman D. Causes and consequences of centrosome abnormalities in cancer. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0467. [PMID: 25047621 DOI: 10.1098/rstb.2013.0467] [Citation(s) in RCA: 261] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Centrosome amplification is a hallmark of cancer. However, despite significant progress in recent years, we are still far from understanding how centrosome amplification affects tumorigenesis. Boveri's hypothesis formulated more than 100 years ago was that aneuploidy induced by centrosome amplification promoted tumorigenesis. Although the hypothesis remains appealing 100 years later, it is also clear that the role of centrosome amplification in cancer is more complex than initially thought. Here, we review how centrosome abnormalities are generated in cancer and the mechanisms cells employ to adapt to centrosome amplification, in particular centrosome clustering. We discuss the different mechanisms by which centrosome amplification could contribute to tumour progression and the new advances in the development of therapies that target cells with extra centrosomes.
Collapse
Affiliation(s)
- S A Godinho
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - D Pellman
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute, and Pediatric Hematology/Oncology, Children's Hospital, Boston, MA 02115, USA Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Pediatric Hematology/Oncology, Children's Hospital, Boston, MA 02115, USA Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Ossipova O, Chuykin I, Chu CW, Sokol SY. Vangl2 cooperates with Rab11 and Myosin V to regulate apical constriction during vertebrate gastrulation. Development 2014; 142:99-107. [PMID: 25480917 DOI: 10.1242/dev.111161] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Core planar cell polarity (PCP) proteins are well known to regulate polarity in Drosophila and vertebrate epithelia; however, their functions in vertebrate morphogenesis remain poorly understood. In this study, we describe a role for PCP signaling in the process of apical constriction during Xenopus gastrulation. The core PCP protein Vangl2 is detected at the apical surfaces of cells at the blastopore lip, and it functions during blastopore formation and closure. Further experiments show that Vangl2, as well as Daam1 and Rho-associated kinase (Rock), regulate apical constriction of bottle cells at the blastopore and ectopic constriction of ectoderm cells triggered by the actin-binding protein Shroom3. At the blastopore lip, Vangl2 is required for the apical accumulation of the recycling endosome marker Rab11. We also show that Rab11 and the associated motor protein Myosin V play essential roles in both endogenous and ectopic apical constriction, and might be involved in Vangl2 trafficking to the cell surface. Overexpression of Rab11 RNA was sufficient to partly restore normal blastopore formation in Vangl2-deficient embryos. These observations suggest that Vangl2 affects Rab11 to regulate apical constriction during blastopore formation.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ilya Chuykin
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chih-Wen Chu
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
17
|
Sebbagh M, Borg JP. Insight into planar cell polarity. Exp Cell Res 2014; 328:284-95. [PMID: 25236701 DOI: 10.1016/j.yexcr.2014.09.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 08/30/2014] [Accepted: 09/01/2014] [Indexed: 11/16/2022]
Abstract
Planar cell polarity or PCP refers to a uniform cellular organization within the plan, typically orthogonal to the apico-basal polarity axis. As such, PCP provides directional cues that control and coordinate the integration of cells in tissues to build a living organism. Although dysfunctions of this fundamental cellular process have been convincingly linked to the etiology of various pathologies such as cancer and developmental defects, the molecular mechanisms governing its establishment and maintenance remain poorly understood. Here, we review some aspects of invertebrate and vertebrate PCPs, highlighting similarities and differences, and discuss the prevalence of the non-canonical Wnt signaling as a central PCP pathway, as well as recent findings on the importance of cell contractility and cilia as promising avenues of investigation.
Collapse
Affiliation(s)
- Michael Sebbagh
- CRCM, "Equipe labellisée Ligue Contre le Cancer", Inserm, U1068, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; CNRS, UMR7258, Marseille F-13009, France; Aix-Marseille University, F-13284 Marseille, France.
| | - Jean-Paul Borg
- CRCM, "Equipe labellisée Ligue Contre le Cancer", Inserm, U1068, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; CNRS, UMR7258, Marseille F-13009, France; Aix-Marseille University, F-13284 Marseille, France.
| |
Collapse
|
18
|
Jones C, Qian D, Kim SM, Li S, Ren D, Knapp L, Sprinzak D, Avraham KB, Matsuzaki F, Chi F, Chen P. Ankrd6 is a mammalian functional homolog of Drosophila planar cell polarity gene diego and regulates coordinated cellular orientation in the mouse inner ear. Dev Biol 2014; 395:62-72. [PMID: 25218921 DOI: 10.1016/j.ydbio.2014.08.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 01/23/2023]
Abstract
The coordinated polarization of neighboring cells within the plane of the tissue, known as planar cell polarity (PCP), is a recurring theme in biology. It is required for numerous developmental processes for the form and function of many tissues and organs across species. The genetic pathway regulating PCP was first discovered in Drosophila, and an analogous but distinct pathway is emerging in vertebrates. It consists of membrane protein complexes known as core PCP proteins that are conserved across species. Here we report that the over-expression of the murine Ankrd6 (mAnkrd6) gene that shares homology with Drosophila core PCP gene diego causes a typical PCP phenotype in Drosophila, and mAnkrd6 can rescue the loss of function of diego in Drosophila. In mice, mAnkrd6 protein is asymmetrically localized in cells of the inner ear sensory organs, characteristic of components of conserved core PCP complexes. The loss of mAnkrd6 causes PCP defects in the inner ear sensory organs. Moreover, canonical Wnt signaling is significantly increased in mouse embryonic fibroblasts from mAnkrd6 knockout mice in comparison to wild type controls. Together, these results indicated that mAnkrd6 is a functional homolog of the Drosophila diego gene for mammalian PCP regulation and act to suppress canonical Wnt signaling.
Collapse
Affiliation(s)
- Chonnettia Jones
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA.
| | - Dong Qian
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Sun Myoung Kim
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Shuangding Li
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Dongdong Ren
- Department of Otolaryngology, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai, China
| | - Lindsey Knapp
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - David Sprinzak
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv, Israel
| | - Karen B Avraham
- Department of Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, Israel
| | - Fumio Matsuzaki
- Laboratory of Cell Asymmetry, Center for Developmental Biology, Riken, Kobe, Japan
| | - Fanglu Chi
- Department of Otolaryngology, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai, China
| | - Ping Chen
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
19
|
Yu X, Wang M, Dong Q, Jin F. Diversin is overexpressed in breast cancer and accelerates cell proliferation and invasion. PLoS One 2014; 9:e98591. [PMID: 24858714 PMCID: PMC4032268 DOI: 10.1371/journal.pone.0098591] [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: 11/26/2013] [Accepted: 05/05/2014] [Indexed: 12/22/2022] Open
Abstract
Diversin was recently reported to play roles in Wnt and JNK pathways. However, the expression pattern and biological roles of diversin in human breast cancer have not been reported. In the present study, we found that diversin was overexpressed in breast cancer specimens by immunohistochemistry and western blot. Significant association was observed between diversin overexpression and TNM stage (p = 0.0036), nodal metastasis (p = 0.0033), negative estrogen receptor expression (p = 0.0012) and triple-negative status (p = 0.0017). Furthermore, colony formation assay and matrigel invasion assay showed that knockdown of diversin expression in MDA-MB-231 cell line with high endogenous expression decreased cell proliferation and cell invasion. Transfection of diversin plasmid in MCF-7 cell line increased cell proliferation and invasion. Further analysis showed that diversin depletion downregulated JNK phosphorylation while its overexpression upregulated JNK phosphorylation. In conclusion, our study demonstrated that diversin was overexpressed in human breast cancers. Diversin could contribute to breast cancer cell proliferation and invasion.
Collapse
Affiliation(s)
- Xinmiao Yu
- Department of Surgical Oncology and Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Minghao Wang
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qianze Dong
- Department of pathology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Ossipova O, Kim K, Lake BB, Itoh K, Ioannou A, Sokol SY. Role of Rab11 in planar cell polarity and apical constriction during vertebrate neural tube closure. Nat Commun 2014; 5:3734. [PMID: 24818582 PMCID: PMC4097039 DOI: 10.1038/ncomms4734] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/27/2014] [Indexed: 12/31/2022] Open
Abstract
Epithelial folding is a critical process underlying many morphogenetic events including vertebrate neural tube closure, however, its spatial regulation is largely unknown. Here we show that during neural tube formation Rab11-positive recycling endosomes acquire bilaterally symmetric distribution in the Xenopus neural plate, being enriched at medial apical cell junctions. This mediolateral polarization was under the control of planar cell polarity (PCP) signalling, was necessary for neural plate folding and was accompanied by the polarization of the exocyst component Sec15. Our further experiments demonstrate that similar PCP-dependent polarization of Rab11 is essential for ectopic apical constriction driven by the actin-binding protein Shroom and during embryonic wound repair. We propose that anisotropic membrane trafficking has key roles in diverse morphogenetic behaviours of individual cells and propagates in a tissue by a common mechanism that involves PCP.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Kyeongmi Kim
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Blue B Lake
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Keiji Itoh
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Andriani Ioannou
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
21
|
Liu YP, Tsai IC, Morleo M, Oh EC, Leitch CC, Massa F, Lee BH, Parker DS, Finley D, Zaghloul NA, Franco B, Katsanis N. Ciliopathy proteins regulate paracrine signaling by modulating proteasomal degradation of mediators. J Clin Invest 2014; 124:2059-70. [PMID: 24691443 PMCID: PMC4001542 DOI: 10.1172/jci71898] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 02/06/2014] [Indexed: 02/06/2023] Open
Abstract
Cilia are critical mediators of paracrine signaling; however, it is unknown whether proteins that contribute to ciliopathies converge on multiple paracrine pathways through a common mechanism. Here, we show that loss of cilopathy-associated proteins Bardet-Biedl syndrome 4 (BBS4) or oral-facial-digital syndrome 1 (OFD1) results in the accumulation of signaling mediators normally targeted for proteasomal degradation. In WT cells, several BBS proteins and OFD1 interacted with proteasomal subunits, and loss of either BBS4 or OFD1 led to depletion of multiple subunits from the centrosomal proteasome. Furthermore, overexpression of proteasomal regulatory components or treatment with proteasomal activators sulforaphane (SFN) and mevalonolactone (MVA) ameliorated signaling defects in cells lacking BBS1, BBS4, and OFD1, in morphant zebrafish embryos, and in induced neurons from Ofd1-deficient mice. Finally, we tested the hypothesis that other proteasome-dependent pathways not known to be associated with ciliopathies are defective in the absence of ciliopathy proteins. We found that loss of BBS1, BBS4, or OFD1 led to decreased NF-κB activity and concomitant IκBβ accumulation and that these defects were ameliorated with SFN treatment. Taken together, our data indicate that basal body proteasomal regulation governs paracrine signaling pathways and suggest that augmenting proteasomal function might benefit ciliopathy patients.
Collapse
Affiliation(s)
- Yangfan P. Liu
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - I-Chun Tsai
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Manuela Morleo
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Edwin C. Oh
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Carmen C. Leitch
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Filomena Massa
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Byung-Hoon Lee
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - David S. Parker
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Daniel Finley
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Norann A. Zaghloul
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Brunella Franco
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.
Telethon Institute of Genetics and Medicine, Naples, Italy.
Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA.
Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
Department of Medical Translational Sciences, Federico II University, Naples, Italy
| |
Collapse
|
22
|
Acentriolar mitosis activates a p53-dependent apoptosis pathway in the mouse embryo. Proc Natl Acad Sci U S A 2014; 111:E1491-500. [PMID: 24706806 DOI: 10.1073/pnas.1400568111] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Centrosomes are the microtubule-organizing centers of animal cells that organize interphase microtubules and mitotic spindles. Centrioles are the microtubule-based structures that organize centrosomes, and a defined set of proteins, including spindle assembly defective-4 (SAS4) (CPAP/CENPJ), is required for centriole biogenesis. The biological functions of centrioles and centrosomes vary among animals, and the functions of mammalian centrosomes have not been genetically defined. Here we use a null mutation in mouse Sas4 to define the cellular and developmental functions of mammalian centrioles in vivo. Sas4-null embryos lack centrosomes but survive until midgestation. As expected, Sas4(-/-) mutants lack primary cilia and therefore cannot respond to Hedgehog signals, but other developmental signaling pathways are normal in the mutants. Unlike mutants that lack cilia, Sas4(-/-) embryos show widespread apoptosis associated with global elevated expression of p53. Cell death is rescued in Sas4(-/-) p53(-/-) double-mutant embryos, demonstrating that mammalian centrioles prevent activation of a p53-dependent apoptotic pathway. Expression of p53 is not activated by abnormalities in bipolar spindle organization, chromosome segregation, cell-cycle profile, or DNA damage response, which are normal in Sas4(-/-) mutants. Instead, live imaging shows that the duration of prometaphase is prolonged in the mutants while two acentriolar spindle poles are assembled. Independent experiments show that prolonging spindle assembly is sufficient to trigger p53-dependent apoptosis. We conclude that a short delay in the prometaphase caused by the absence of centrioles activates a previously undescribed p53-dependent cell death pathway in the rapidly dividing cells of the mouse embryo.
Collapse
|
23
|
Luan L, Zhao Y, Xu Z, Jiang G, Zhang X, Fan C, Liu D, Zhao H, Xu K, Wang M, Yu X, Wang E. Diversin increases the proliferation and invasion ability of non-small-cell lung cancer cells via JNK pathway. Cancer Lett 2014; 344:232-8. [DOI: 10.1016/j.canlet.2013.10.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 10/25/2013] [Accepted: 10/28/2013] [Indexed: 10/26/2022]
|
24
|
Singh J, Mlodzik M. Planar cell polarity signaling: coordination of cellular orientation across tissues. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 1:479-99. [PMID: 23066429 DOI: 10.1002/wdev.32] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Establishment of Planar Cell Polarity (PCP) in epithelia, in the plane of an epithelium, is an important feature of the development and homeostasis of most organs. Studies in different model organisms have contributed a wealth of information regarding the mechanisms that govern PCP regulation. Genetic studies in Drosophila have identified two signaling systems, the Fz/PCP and Fat/Dachsous system, which are both required for PCP establishment in many different tissues in a largely non-redundant manner. Recent advances in vertebrate PCP studies have added novel factors of PCP regulation and also new cellular features requiring PCP-signaling input, including the positioning and orientation of the primary cilium of many epithelial cells. This review focuses mostly on several recent advances made in the Drosophila and vertebrate PCP field and integrates these within the existing PCP-signaling framework.
Collapse
Affiliation(s)
- Jaskirat Singh
- Department of Developmental & Regenerative Biology, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
25
|
Abstract
The canonical Wnt/β-catenin pathway is an ancient and evolutionarily conserved signaling pathway that is required for the proper development of all metazoans, from the basal demosponge Amphimedon queenslandica to humans. Misregulation of Wnt signaling is implicated in many human diseases, making this pathway an intense area of research in industry as well as academia. In this review, we explore our current understanding of the molecular steps involved in the transduction of a Wnt signal. We will focus on how the critical Wnt pathway component, β-catenin, is in a "futile cycle" of constant synthesis and degradation and how this cycle is disrupted upon pathway activation. We describe the role of the Wnt pathway in major human cancers and in the control of stem cell self-renewal in the developing organism and in adults. Finally, we describe well-accepted criteria that have been proposed as evidence for the involvement of a molecule in regulating the canonical Wnt pathway.
Collapse
Affiliation(s)
- Kenyi Saito-Diaz
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232-8240, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The Wnt pathway is a major embryonic signaling pathway that controls cell proliferation, cell fate, and body-axis determination in vertebrate embryos. Soon after egg fertilization, Wnt pathway components play a role in microtubule-dependent dorsoventral axis specification. Later in embryogenesis, another conserved function of the pathway is to specify the anteroposterior axis. The dual role of Wnt signaling in Xenopus and zebrafish embryos is regulated at different developmental stages by distinct sets of Wnt target genes. This review highlights recent progress in the discrimination of different signaling branches and the identification of specific pathway targets during vertebrate axial development.
Collapse
Affiliation(s)
- Hiroki Hikasa
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | | |
Collapse
|
27
|
Mahuzier A, Gaudé HM, Grampa V, Anselme I, Silbermann F, Leroux-Berger M, Delacour D, Ezan J, Montcouquiol M, Saunier S, Schneider-Maunoury S, Vesque C. Dishevelled stabilization by the ciliopathy protein Rpgrip1l is essential for planar cell polarity. ACTA ACUST UNITED AC 2012; 198:927-40. [PMID: 22927466 PMCID: PMC3432770 DOI: 10.1083/jcb.201111009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Rpgrip1l is required for planar localization of the basal body and acts within a ciliopathy protein complex by stabilizing dishevelled. Cilia are at the core of planar polarity cellular events in many systems. However, the molecular mechanisms by which they influence the polarization process are unclear. Here, we identify the function of the ciliopathy protein Rpgrip1l in planar polarity. In the mouse cochlea and in the zebrafish floor plate, Rpgrip1l was required for positioning the basal body along the planar polarity axis. Rpgrip1l was also essential for stabilizing dishevelled at the cilium base in the zebrafish floor plate and in mammalian renal cells. In rescue experiments, we showed that in the zebrafish floor plate the function of Rpgrip1l in planar polarity was mediated by dishevelled stabilization. In cultured cells, Rpgrip1l participated in a complex with inversin and nephrocystin-4, two ciliopathy proteins known to target dishevelled to the proteasome, and, in this complex, Rpgrip1l prevented dishevelled degradation. We thus uncover a ciliopathy protein complex that finely tunes dishevelled levels, thereby modulating planar cell polarity processes.
Collapse
Affiliation(s)
- Alexia Mahuzier
- Centre National de la Recherche Scientifique UMR 7622, Institut National de la Santé et de la Recherche Médicale U969, 75005 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
A Wnt/beta-catenin pathway antagonist Chibby binds Cenexin at the distal end of mother centrioles and functions in primary cilia formation. PLoS One 2012; 7:e41077. [PMID: 22911743 PMCID: PMC3401179 DOI: 10.1371/journal.pone.0041077] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 06/18/2012] [Indexed: 11/25/2022] Open
Abstract
The mother centriole of the centrosome is distinguished from immature daughter centrioles by the presence of accessory structures (distal and subdistal appendages), which play an important role in the organization of the primary cilium in quiescent cells. Primary cilia serve as sensory organelles, thus have been implicated in mediating intracellular signal transduction pathways. Here we report that Chibby (Cby), a highly conserved antagonist of the Wnt/β-catenin pathway, is a centriolar component specifically located at the distal end of the mother centriole and essential for assembly of the primary cilium. Cby appeared as a discrete dot in the middle of a ring-like structure revealed by staining with a distal appendage component of Cep164. Cby interacted with one of the appendage components, Cenexin (Cnx), which thereby abrogated the inhibitory effect of Cby on β-catenin-mediated transcriptional activation in a dose-dependent manner. Cby and Cnx did not precisely align, as Cby was detected at a more distal position than Cnx. Cnx emerged earlier than Cby during the cell cycle and was required for recruitment of Cby to the mother centriole. However, Cby was dispensable for Cnx localization to the centriole. During massive centriogenesis in in vitro cultured mouse tracheal epithelial cells, Cby and Cnx were expressed in a similar pattern, which was coincident with the expression of Foxj1. Our results suggest that Cby plays an important role in organization of both primary and motile cilia in collaboration with Cnx.
Collapse
|
29
|
Gordon LR, Gribble KD, Syrett CM, Granato M. Initiation of synapse formation by Wnt-induced MuSK endocytosis. Development 2012; 139:1023-33. [PMID: 22318632 DOI: 10.1242/dev.071555] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In zebrafish, the MuSK receptor initiates neuromuscular synapse formation by restricting presynaptic growth cones and postsynaptic acetylcholine receptors (AChRs) to the center of skeletal muscle cells. Increasing evidence suggests a role for Wnts in this process, yet how muscle cells respond to Wnt signals is unclear. Here, we show that in vivo, wnt11r and wnt4a initiate MuSK translocation from muscle membranes to recycling endosomes and that this transition is crucial for AChR accumulation at future synaptic sites. Moreover, we demonstrate that components of the planar cell polarity pathway colocalize to recycling endosomes and that this localization is MuSK dependent. Knockdown of several core components disrupts MuSK translocation to endosomes, AChR localization and axonal guidance. We propose that Wnt-induced trafficking of the MuSK receptor to endosomes initiates a signaling cascade to align pre- with postsynaptic elements. Collectively, these findings suggest a general mechanism by which Wnt signals shape synaptic connectivity through localized receptor endocytosis.
Collapse
Affiliation(s)
- Laura R Gordon
- Department of Cell and Developmental Biology, University of Pennsylvania. Philadelphia, PA 19104-6058, USA
| | | | | | | |
Collapse
|
30
|
Planar cell polarity signaling pathway in congenital heart diseases. J Biomed Biotechnol 2011; 2011:589414. [PMID: 22131815 PMCID: PMC3205795 DOI: 10.1155/2011/589414] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 08/31/2011] [Indexed: 12/14/2022] Open
Abstract
Congenital heart disease (CHD) is a common cardiac disorder in humans. Despite many advances in the understanding of CHD and the identification of many associated genes, the fundamental etiology for the majority of cases remains unclear. The planar cell polarity (PCP) signaling pathway, responsible for tissue polarity in Drosophila and gastrulation movements and cardiogenesis in vertebrates, has been shown to play multiple roles during cardiac differentiation and development. The disrupted function of PCP signaling is connected to some CHDs. Here, we summarize our current understanding of how PCP factors affect the pathogenesis of CHD.
Collapse
|
31
|
Roles of planar cell polarity pathways in the development of neural [correction of neutral] tube defects. J Biomed Sci 2011; 18:66. [PMID: 21864354 PMCID: PMC3175158 DOI: 10.1186/1423-0127-18-66] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 08/24/2011] [Indexed: 02/08/2023] Open
Abstract
Neural tube defects (NTDs) are the second most common birth defect in humans. Despite many advances in the understanding of NTDs and the identification of many genes related to NTDs, the fundamental etiology for the majority of cases of NTDs remains unclear. Planar cell polarity (PCP) signaling pathway, which is important for polarized cell movement (such as cell migration) and organ morphogenesis through the activation of cytoskeletal pathways, has been shown to play multiple roles during neural tube closure. The disrupted function of PCP pathway is connected with some NTDs. Here, we summarize our current understanding of how PCP factors affect the pathogenesis of NTDs.
Collapse
|
32
|
Yasunaga T, Itoh K, Sokol SY. Regulation of basal body and ciliary functions by Diversin. Mech Dev 2011; 128:376-86. [PMID: 21843637 DOI: 10.1016/j.mod.2011.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 07/23/2011] [Accepted: 07/25/2011] [Indexed: 11/19/2022]
Abstract
The centrosome is essential for the formation of the cilia and has been implicated in cell polarization and signaling during early embryonic development. A number of Wnt pathway components were found to localize at the centrosome, but how this localization relates to their signaling functions is unclear. In this study, we assessed a role for Diversin, a putative Wnt pathway mediator, in developmental processes that involve cilia. We find that Diversin is specifically localized to the basal body compartment near the base of the cilium in Xenopus multi-ciliated skin cells. Overexpression of Diversin RNA disrupted basal body polarization in these cells, suggesting that tightly regulated control of Diversin levels is crucial for this process. In cells depleted of endogenous Diversin, basal body structure appeared abnormal and this was accompanied by disrupted polarity, shortened or absent cilia and defective ciliary flow. These results are consistent with the involvement of Diversin in processes that are related to the acquisition of cell polarity and require ciliary functions.
Collapse
Affiliation(s)
- Takayuki Yasunaga
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
33
|
Itoh K, Sokol SY. Polarized translocation of fluorescent proteins in Xenopus ectoderm in response to Wnt signaling. J Vis Exp 2011:2700. [PMID: 21654626 PMCID: PMC3197113 DOI: 10.3791/2700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Cell polarity is a fundamental property of eukaryotic cells that is dynamically regulated by both intrinsic and extrinsic factors during embryonic development. One of the signaling pathways involved in this regulation is the Wnt pathway, which is used many times during embryogenesis and critical for human disease. Multiple molecular components of this pathway coordinately regulate signaling in a spatially-restricted manner, but the underlying mechanisms are not fully understood. Xenopus embryonic epithelial cells is an excellent system to study subcellular localization of various signaling proteins. Fluorescent fusion proteins are expressed in Xenopus embryos by RNA microinjection, ectodermal explants are prepared and protein localization is evaluated by epifluorescence. In this experimental protocol we describe how subcellular localization of Diversin, a cytoplasmic protein that has been implicated in signaling and cell polarity determination is visualized in Xenopus ectodermal cells to study Wnt signal transduction. Coexpression of a Wnt ligand or a Frizzled receptor alters the distribution of Diversin fused with red fluorescent protein, RFP, and recruits it to the cell membrane in a polarized fashion. This ex vivo protocol should be a useful addition to in vitro studies of cultured mammalian cells, in which spatial control of signaling differs from that of the intact tissue and is much more difficult to analyze.
Collapse
Affiliation(s)
- Keiji Itoh
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine
| | | |
Collapse
|
34
|
Dishevelled is essential for neural connectivity and planar cell polarity in planarians. Proc Natl Acad Sci U S A 2011; 108:2813-8. [PMID: 21282632 DOI: 10.1073/pnas.1012090108] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Wingless/Integrated (Wnt) signaling pathway controls multiple events during development and homeostasis. It comprises multiple branches, mainly classified according to their dependence on β-catenin activation. The Wnt/β-catenin branch is essential for the establishment of the embryonic anteroposterior (AP) body axis throughout the phylogenetic tree. It is also required for AP axis establishment during planarian regeneration. Wnt/β-catenin-independent signaling encompasses several different pathways, of which the most extensively studied is the planar cell polarity (PCP) pathway, which is responsible for planar polarization of cell structures within an epithelial sheet. Dishevelled (Dvl) is the hub of Wnt signaling because it regulates and channels the Wnt signal into every branch. Here, we analyze the role of Schmidtea mediterranea Dvl homologs (Smed-dvl-1 and Smed-dvl-2) using gene silencing. We demonstrate that in addition to a role in AP axis specification, planarian Dvls are involved in at least two different β-catenin-independent processes. First, they are essential for neural connectivity through Smed-wnt5 signaling. Second, Smed-dvl-2, together with the S. mediterranea homologs of Van-Gogh (Vang) and Diversin (Div), is required for apical positioning of the basal bodies of epithelial cells. These data represent evidence not only of the function of the PCP network in lophotrocozoans but of the involvement of the PCP core elements Vang and Div in apical positioning of the cilia.
Collapse
|
35
|
Rack1 is required for Vangl2 membrane localization and planar cell polarity signaling while attenuating canonical Wnt activity. Proc Natl Acad Sci U S A 2011; 108:2264-9. [PMID: 21262816 DOI: 10.1073/pnas.1013170108] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The vertebrate planar cell polarity (PCP) pathway shares molecular components with the β-catenin-mediated canonical Wnt pathway but acts through membrane complexes containing Vang or Frizzled to orient neighboring cells coordinately. The molecular interactions underlying the action of Vang in PCP signaling and specification, however, are yet to be delineated. Here, we report the identification of Rack1 as an interacting protein of a vertebrate Vang protein, Vangl2. We demonstrate that Rack1 is required in zebrafish for PCP-regulated processes, including oriented cell division, cellular polarization, and convergent extension during gastrulation. We further show that the knockdown of Rack1 affects membrane localization of Vangl2 and that the Vangl2-interacting domain of Rack1 has a dominant-negative effect on Vangl2 localization and gastrulation. Moreover, Rack1 antagonizes canonical Wnt signaling. Together, our data suggest that Rack1 regulates the localization of an essential PCP protein and acts as a molecular switch to promote PCP signaling.
Collapse
|
36
|
Luyten A, Su X, Gondela S, Chen Y, Rompani S, Takakura A, Zhou J. Aberrant regulation of planar cell polarity in polycystic kidney disease. J Am Soc Nephrol 2010; 21:1521-32. [PMID: 20705705 DOI: 10.1681/asn.2010010127] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mutations in PKD1, which encodes polycystin-1 (PC1), contribute to >85% of cases of autosomal dominant polycystic kidney disease (ADPKD). The planar cell polarity (PCP) pathway is necessary for the oriented cell division and convergent extension that establishes and maintains the structure of kidney tubules, but the role of this pathway in the pathophysiology of ADPKD is incompletely understood. Here, we show that inactivation of Pkd1 in postnatal developing mouse kidneys leads to a defect in oriented cell division in precystic kidney tubules. We also observed this defect in precystic Pkd1-inactivated mature kidneys subjected to ischemia-reperfusion injury as a "third hit." Cystic kidneys exhibited striking upregulation and activation of Frizzled 3 (Fz3), a regulator of PCP, and its downstream effector, CDC42. Precystic kidneys demonstrated upregulation of CDC42, but the localization of the polarity proteins Par3 and Par6 was similar to control. Fz3 was expressed on the cilia of cystic kidneys but barely detected on the cilia of normal kidneys. In vitro, PC1 and Fz3 antagonized each other to control CDC42 expression and the rate of cell migration in HEK293T cells. Taken together, our data suggest that PC1 controls oriented cell division and that aberrant PCP signaling contributes to cystogenesis.
Collapse
Affiliation(s)
- Annouck Luyten
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|