51
|
Park SA, Yoo H, Seol JH, Rhee K. HDAC3 and HDAC8 are required for cilia assembly and elongation. Biol Open 2019; 8:bio.043828. [PMID: 31362948 PMCID: PMC6737963 DOI: 10.1242/bio.043828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cilia are extended from mother centrioles in quiescent G0/G1 cells and retracted in dividing cells. Diverse post-translational modifications play roles in the assembly and disassembly of the cilium. Here, we examined class I histone deacetylases (HDACs) as positive regulators of cilia assembly in serum-deprived RPE1 and HK2 cells. We observed that the number of cells with cilia was significantly reduced in HDAC3- and HDAC8-depleted cells. The ciliary length also decreased in HDAC3- and HDAC8-depleted cells compared to that in control cells. A knockdown-rescue experiment showed that wild-type HDAC3 and HDAC8 rescued the cilia assembly and ciliary length in HDAC3- and HDAC8-depleted cells, respectively; however, deacetylase-dead HDAC3 and HDAC8 mutants did not. This suggests that deacetylase activity is critical for both HDAC3 and HDAC8 function in cilia assembly and ciliary length control. This is the first study to report that HDACs are required for the assembly and elongation of the primary cilia. Summary: We identified that HDAC3 and HDAC8 are required for the assembly and elongation of the primary cilia.
Collapse
Affiliation(s)
- Seon-Ah Park
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Hyunjeong Yoo
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Jae Hong Seol
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Kunsoo Rhee
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
52
|
Mirvis M, Siemers KA, Nelson WJ, Stearns TP. Primary cilium loss in mammalian cells occurs predominantly by whole-cilium shedding. PLoS Biol 2019; 17:e3000381. [PMID: 31314751 PMCID: PMC6699714 DOI: 10.1371/journal.pbio.3000381] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 08/19/2019] [Accepted: 07/02/2019] [Indexed: 12/17/2022] Open
Abstract
The primary cilium is a central signaling hub in cell proliferation and differentiation and is built and disassembled every cell cycle in many animal cells. Disassembly is critically important, as misregulation or delay of cilia loss leads to cell cycle defects. The physical means by which cilia are lost are poorly understood but are thought to involve resorption of ciliary components into the cell body. To investigate cilium loss in mammalian cells, we used live-cell imaging to comprehensively characterize individual events. The predominant mode of cilium loss was rapid deciliation, in which the membrane and axoneme of the cilium was shed from the cell. Gradual resorption was also observed, as well as events in which a period of gradual resorption was followed by rapid deciliation. Deciliation resulted in intact shed cilia that could be recovered from culture medium and contained both membrane and axoneme proteins. We modulated levels of katanin and intracellular calcium, two putative regulators of deciliation, and found that excess katanin promotes cilia loss by deciliation, independently of calcium. Together, these results suggest that mammalian ciliary loss involves a tunable decision between deciliation and resorption.
Collapse
Affiliation(s)
- Mary Mirvis
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Kathleen A. Siemers
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - W. James Nelson
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Tim P. Stearns
- Department of Biology, Stanford University, Stanford, California, United States of America
- Department of Genetics, Stanford University, Stanford, California, United States of America
| |
Collapse
|
53
|
Ko JY, Lee EJ, Park JH. Interplay Between Primary Cilia and Autophagy and Its Controversial Roles in Cancer. Biomol Ther (Seoul) 2019; 27:337-341. [PMID: 31042678 PMCID: PMC6609109 DOI: 10.4062/biomolther.2019.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/03/2019] [Accepted: 04/05/2019] [Indexed: 12/27/2022] Open
Abstract
Primary cilia and autophagy are two distinct nutrient-sensing machineries required for maintaining intracellular energy homeostasis, either via signal transduction or recycling of macromolecules from cargo breakdown, respectively. Potential correlations between primary cilia and autophagy have been recently suggested and their relationship may increase our understanding of the pathogenesis of human diseases, including ciliopathies and cancer. In this review, we cover the current issues concerning the bidirectional interaction between primary cilia and autophagy and discuss its role in cancer with cilia defect.
Collapse
Affiliation(s)
- Je Yeong Ko
- Department of Life Systems, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Eun Ji Lee
- Department of Life Systems, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jong Hoon Park
- Department of Life Systems, Sookmyung Women's University, Seoul 04310, Republic of Korea
| |
Collapse
|
54
|
King CR, A A Quadros AR, Chazeau A, Saarloos I, van der Graaf AJ, Verhage M, Toonen RF. Fbxo41 Promotes Disassembly of Neuronal Primary Cilia. Sci Rep 2019; 9:8179. [PMID: 31160656 PMCID: PMC6546786 DOI: 10.1038/s41598-019-44589-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/20/2019] [Indexed: 01/02/2023] Open
Abstract
Neuronal primary cilia are signaling organelles with crucial roles in brain development and disease. Cilia structure is decisive for their signaling capacities but the mechanisms regulating it are poorly understood. We identify Fbxo41 as a novel Skp1/Cullin1/F-box (SCF) E3-ligase complex subunit that targets to neuronal centrioles where its accumulation promotes disassembly of primary cilia, and affects sonic hedgehog signaling, a canonical ciliary pathway. Fbxo41 targeting to centrioles requires its Coiled-coil and F-box domains. Levels of Fbxo41 at the centrioles inversely correlate with neuronal cilia length, and mutations that disrupt Fbxo41 targeting or assembly into SCF-complexes also disturb its function in cilia disassembly and signaling. Fbxo41 dependent cilia disassembly in mitotic and post-mitotic cells requires rearrangements of the actin-cytoskeleton, but requires Aurora A kinase activation only in mitotic cells, highlighting important mechanistical differences controlling cilia size between mitotic and post-mitotic cells. Phorbol esters induce recruitment of overexpressed Fbxo41 to centrioles and cilia disassembly in neurons, but disassembly can also occur in absence of Fbxo41. We propose that Fbxo41 targeting to centrosomes regulates neuronal cilia structure and signaling capacity in addition to Fbxo41-independent pathways controlling cilia size.
Collapse
Affiliation(s)
- Cillian R King
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Ana R A A Quadros
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Anaël Chazeau
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Ingrid Saarloos
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands.,Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Anne Jolien van der Graaf
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands.,Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
55
|
Fabbri L, Bost F, Mazure NM. Primary Cilium in Cancer Hallmarks. Int J Mol Sci 2019; 20:E1336. [PMID: 30884815 PMCID: PMC6471594 DOI: 10.3390/ijms20061336] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/28/2019] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
The primary cilium is a solitary, nonmotile and transitory appendage that is present in virtually all mammalian cells. Our knowledge of its ultrastructure and function is the result of more than fifty years of research that has dramatically changed our perspectives on the primary cilium. The mutual regulation between ciliogenesis and the cell cycle is now well-recognized, as well as the function of the primary cilium as a cellular "antenna" for perceiving external stimuli, such as light, odorants, and fluids. By displaying receptors and signaling molecules, the primary cilium is also a key coordinator of signaling pathways that converts extracellular cues into cellular responses. Given its critical tasks, any defects in primary cilium formation or function lead to a wide spectrum of diseases collectively called "ciliopathies". An emerging role of primary cilium is in the regulation of cancer development. In this review, we seek to describe the current knowledge about the influence of the primary cilium in cancer progression, with a focus on some of the events that cancers need to face to sustain survival and growth in hypoxic microenvironment: the cancer hallmarks.
Collapse
Affiliation(s)
- Lucilla Fabbri
- Université Côte d'Azur (UCA), INSERM U1065, C3M, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice, France.
| | - Frédéric Bost
- Université Côte d'Azur (UCA), INSERM U1065, C3M, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice, France.
| | - Nathalie M Mazure
- Université Côte d'Azur (UCA), INSERM U1065, C3M, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice, France.
| |
Collapse
|
56
|
Kiseleva AA, Korobeynikov VA, Nikonova AS, Zhang P, Makhov P, Deneka AY, Einarson MB, Serebriiskii IG, Liu H, Peterson JR, Golemis EA. Unexpected Activities in Regulating Ciliation Contribute to Off-target Effects of Targeted Drugs. Clin Cancer Res 2019; 25:4179-4193. [PMID: 30867219 DOI: 10.1158/1078-0432.ccr-18-3535] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/14/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE For many tumors, signaling exchanges between cancer cells and other cells in their microenvironment influence overall tumor signaling. Some of these exchanges depend on expression of the primary cilium on nontransformed cell populations, as extracellular ligands including Sonic Hedgehog (SHH), PDGFRα, and others function through receptors spatially localized to cilia. Cell ciliation is regulated by proteins that are themselves therapeutic targets. We investigated whether kinase inhibitors of clinical interest influence ciliation and signaling by proteins with ciliary receptors in cancer and other cilia-relevant disorders, such as polycystic kidney disease (PKD). EXPERIMENTAL DESIGN We screened a library of clinical and preclinical kinase inhibitors, identifying drugs that either prevented or induced ciliary disassembly. Specific bioactive protein targets of the drugs were identified by mRNA depletion. Mechanism of action was defined, and activity of select compounds investigated. RESULTS We identified multiple kinase inhibitors not previously linked to control of ciliation, including sunitinib, erlotinib, and an inhibitor of the innate immune pathway kinase, IRAK4. For all compounds, activity was mediated through regulation of Aurora-A (AURKA) activity. Drugs targeting cilia influenced proximal cellular responses to SHH and PDGFRα. In vivo, sunitinib durably limited ciliation and cilia-related biological activities in renal cells, renal carcinoma cells, and PKD cysts. Extended analysis of IRAK4 defined a subset of innate immune signaling effectors potently affecting ciliation. CONCLUSIONS These results suggest a paradigm by which targeted drugs may have unexpected off-target effects in heterogeneous cell populations in vivo via control of a physical platform for receipt of extracellular ligands.
Collapse
Affiliation(s)
- Anna A Kiseleva
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan, Russian Federation
| | - Vladislav A Korobeynikov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Anna S Nikonova
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Peishan Zhang
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Petr Makhov
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Alexander Y Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan, Russian Federation
| | - Margret B Einarson
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ilya G Serebriiskii
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan, Russian Federation
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Jeffrey R Peterson
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
57
|
Zhang P, Kiseleva AA, Korobeynikov V, Liu H, Einarson MB, Golemis EA. Microscopy-Based Automated Live Cell Screening for Small Molecules That Affect Ciliation. Front Genet 2019; 10:75. [PMID: 30809247 PMCID: PMC6379280 DOI: 10.3389/fgene.2019.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/28/2019] [Indexed: 12/19/2022] Open
Abstract
The primary monocilium, or cilium, is a single antenna-like organelle that protrudes from the surface of most mammalian cell types, and serves as a signaling hub. Mutations of cilia-associated genes result in severe genetic disorders termed ciliopathies. Among these, the most common is autosomal dominant polycystic kidney disease (ADPKD); less common genetic diseases include Bardet–Biedl syndrome, Joubert syndrome, nephronophthisis, and others. Important signaling cascades with receptor systems localized exclusively or in part at cilia include Sonic Hedgehog (SHH), platelet derived growth factor alpha (PDGFRα), WNTs, polycystins, and others. Changes in ciliation during development or in pathological conditions such as cancer impacts signaling by these proteins. Notably, ciliation status of cells is coupled closely to the cell cycle, with cilia protruding in quiescent (G0) or early G1 cells, declining in S/G2, and absent in M phase, and has been proposed to contribute to cell cycle regulation. Because of this complex biology, the elaborate machinery regulating ciliary assembly and disassembly receives input from many cellular proteins relevant to cell cycle control, development, and oncogenic transformation, making study of genetic factors and drugs influencing ciliation of high interest. One of the most effective tools to investigate the dynamics of the cilia under different conditions is the imaging of live cells. However, developing assays to observe the primary cilium in real time can be challenging, and requires a consideration of multiple details related to the cilia biology. With the dual goals of identifying small molecules that may have beneficial activity through action on human diseases, and of identifying ciliary activities of existing agents that are in common use or development, we here describe creation and evaluation of three autofluorescent cell lines derived from the immortalized retinal pigmented epithelium parental cell line hTERT-RPE1. These cell lines stably express the ciliary-targeted fluorescent proteins L13-Arl13bGFP, pEGFP-mSmo, and tdTomato-MCHR1-N-10. We then describe methods for use of these cell lines in high throughput screening of libraries of small molecule compounds to identify positive and negative regulators of ciliary disassembly.
Collapse
Affiliation(s)
- Peishan Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, China.,Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Anna A Kiseleva
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States.,Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan, Russia
| | - Vladislav Korobeynikov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Margret B Einarson
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| |
Collapse
|
58
|
Nishimura Y, Kasahara K, Shiromizu T, Watanabe M, Inagaki M. Primary Cilia as Signaling Hubs in Health and Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801138. [PMID: 30643718 PMCID: PMC6325590 DOI: 10.1002/advs.201801138] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/20/2018] [Indexed: 05/13/2023]
Abstract
Primary cilia detect extracellular cues and transduce these signals into cells to regulate proliferation, migration, and differentiation. Here, the function of primary cilia as signaling hubs of growth factors and morphogens is in focus. First, the molecular mechanisms regulating the assembly and disassembly of primary cilia are described. Then, the role of primary cilia in mediating growth factor and morphogen signaling to maintain human health and the potential mechanisms by which defects in these pathways contribute to human diseases, such as ciliopathy, obesity, and cancer are described. Furthermore, a novel signaling pathway by which certain growth factors stimulate cell proliferation through suppression of ciliogenesis is also described, suggesting novel therapeutic targets in cancer.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Kousuke Kasahara
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Takashi Shiromizu
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Masatoshi Watanabe
- Department of Oncologic PathologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Masaki Inagaki
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| |
Collapse
|
59
|
Chowdhury P, Powell RT, Stephan C, Uray IP, Talley T, Karki M, Tripathi DN, Park YS, Mancini MA, Davies P, Dere R. Bexarotene - a novel modulator of AURKA and the primary cilium in VHL-deficient cells. J Cell Sci 2018; 131:jcs.219923. [PMID: 30518623 PMCID: PMC6307881 DOI: 10.1242/jcs.219923] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 10/17/2018] [Indexed: 12/22/2022] Open
Abstract
Loss of the gene von Hippel–Lindau (VHL) is associated with loss of primary cilia and is causally linked to elevated levels of Aurora kinase A (AURKA). We developed an image-based high-throughput screening (HTS) assay using a dual-labeling image analysis strategy that identifies both the cilium and the basal body. By using this strategy, we screened small-molecule compounds for the targeted rescue of cilia defects associated with VHL deficiency with high accuracy and reproducibility. Bexarotene was identified and validated as a positive regulator of the primary cilium. Importantly, the inability of an alternative retinoid X receptor (RXR) agonist to rescue ciliogenesis, in contrast to bexarotene, suggested that multiple bexarotene-driven mechanisms were responsible for the rescue. We found that bexarotene decreased AURKA expression in VHL-deficient cells, thereby restoring the ability of these cells to ciliate in the absence of VHL. Finally, bexarotene treatment reduced the propensity of subcutaneous lesions to develop into tumors in a mouse xenograft model of renal cell carcinoma (RCC), with a concomitant decrease in activated AURKA, highlighting the potential of bexarotene treatment as an intervention strategy in the clinic to manage renal cystogenesis associated with VHL deficiency and elevated AURKA expression. Highlighted Article: An image-based screen using a dual labeling strategy identified bexarotene, a rexinoid, as a novel modulator of the primary cilium in VHL-deficient cells.
Collapse
Affiliation(s)
- Pratim Chowdhury
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| | - Reid T Powell
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Clifford Stephan
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Ivan P Uray
- Department of Clinical Oncology, University of Debrecen, Debrecen 4032, Hungary
| | - Tia Talley
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| | - Menuka Karki
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| | - Durga Nand Tripathi
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Sung Park
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Michael A Mancini
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M College of Medicine, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter Davies
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Ruhee Dere
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
60
|
Hoang-Minh LB, Dutra-Clarke M, Breunig JJ, Sarkisian MR. Glioma cell proliferation is enhanced in the presence of tumor-derived cilia vesicles. Cilia 2018; 7:6. [PMID: 30410731 PMCID: PMC6219037 DOI: 10.1186/s13630-018-0060-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/24/2018] [Indexed: 11/10/2022] Open
Abstract
Background The mechanisms by which primary cilia affect glioma pathogenesis are unclear. Depending on the glioma cell line, primary cilia can promote or inhibit tumor development. Here, we used piggyBac-mediated transgenesis to generate patient-derived glioblastoma (GBM) cell lines that stably express Arl13b:GFP in their cilia. This allowed us to visualize and analyze the behavior of cilia and ciliated cells during live GBM cell proliferation. Results Time-lapse imaging of Arl13b:GFP+ cilia revealed their dynamic behaviors, including distal tip excision into the extracellular milieu. Recent studies of non-cancerous cells indicate that this process occurs during the G0 phase, prior to cilia resorption and cell cycle re-entry, and requires ciliary recruitment of F-actin and actin regulators. Similarly, we observed ciliary buds associated with Ki67- cells as well as scattered F-actin+ cilia, suggesting that quiescent GBM cells may also utilize an actin network-based mechanism for ciliary tip excision. Notably, we found that the proliferation of ciliated GBM cells was promoted by exposing them to conditioned media obtained from ciliated cell cultures when compared to conditioned media collected from cilia-defective cell cultures (depleted in either KIF3A or IFT88 using CRISPR/Cas9). These results suggest that GBM cilia may release mitogenic vesicles carrying factors that promote tumor cell proliferation. Although Arl13b is implicated in tumor growth, our data suggest that Arl13b released from GBM cilia does not mediate tumor cell proliferation. Conclusion Collectively, our results indicate that ciliary vesicles may represent a novel mode of intercellular communication within tumors that contributes to GBM pathogenesis. The mitogenic capacity of GBM ciliary vesicles and the molecular mediators of this phenomenon requires further investigation.
Collapse
Affiliation(s)
- Lan B Hoang-Minh
- 1Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA.,2Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA
| | - Marina Dutra-Clarke
- 3Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,4Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,5Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Joshua J Breunig
- 3Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,4Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,5Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Matthew R Sarkisian
- 1Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA.,2Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA
| |
Collapse
|
61
|
Ritter A, Louwen F, Yuan J. Deficient primary cilia in obese adipose-derived mesenchymal stem cells: obesity, a secondary ciliopathy? Obes Rev 2018; 19:1317-1328. [PMID: 30015415 DOI: 10.1111/obr.12716] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 12/14/2022]
Abstract
Obesity alters the composition, structure and function of adipose tissue, characterized by chronic inflammation, insulin resistance and metabolic dysfunction. Adipose-derived mesenchymal stem cells (ASCs) are responsible for cell renewal, spontaneous repair and immunomodulation in adipose tissue. Increasing evidence highlights that ASCs are deficient in obesity, and the underlying mechanisms are not well understood. We have recently shown that obese ASCs have defective primary cilia, which are shortened and unable to properly respond to stimuli. Impaired cilia compromise ASC functions. This work suggests an intertwined connection of obesity, defective cilia and dysfunctional ASCs. We have here discussed the current data regarding defective cilia in various cell types in obesity. Based on these observations, we hypothesize that obesity, a systemic chronic metainflammation, could impair cilia in diverse ciliated cells, like pancreatic islet cells, stem cells and hypothalamic neurons, making these critical cells dysfunctional by shutting down their signal sensors and transducers. In this context, obesity may represent a secondary form of ciliopathy induced by obesity-related inflammation and metabolic dysfunction. Reactivation of ciliated cells might be an alternative strategy to combat obesity and its associated diseases.
Collapse
Affiliation(s)
- A Ritter
- Department of Gynecology and Obstetrics, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - F Louwen
- Department of Gynecology and Obstetrics, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - J Yuan
- Department of Gynecology and Obstetrics, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
62
|
Wang Z, Ma Z, Cao J. Effects of Repeated Aurora-A siRNA Transfection on Cilia Generation and Proliferation of SK-MES-1 or A549 Cells. Cancer Biother Radiopharm 2018; 33:110-117. [PMID: 29641257 DOI: 10.1089/cbr.2017.2297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Suppression of Aurora kinase A (Aurora-A, AURKA) by siRNA of Aurora-A (siAurora-A, siA) has been used in lung tumor treatment. However, the dose and frequency of gene transfection still need to be confirmed further. We imitated multiple administration of solid tumor and attempted to make out the effects of thrice transfection of siAurora-A on cilia generation and apoptosis of SK-MES-1 cells (SK) or A549 cells. METHODS The Aurora-A mRNA levels of cells cultured with serum for 6 d or without serum for 2, 4, or 6 d were examined with real-time quantitative PCR; Cells were transfected single or repeatedly with siAurora-A or siControl (siC), their Aurora-A mRNA levels were determined with PCR; Their cilia were examined with immunohistochemistry. Cell viability was measured with the MTT assay. Protein expression was analyzed with western blot. RESULTS Cell viability showed a downward trend along with the prolongation of starvation time to the second, fourth, and even to the sixth day in both types of cells. But, the expression level of Aurora-A mRNA flipped to rise at the sixth day instead of decreasing at the fourth day. Protein expression trend of total Aurora-A in the two groups was consistent with Aurora-A mRNA expression trend. Compared with siC-3 group (transfected three times with siControl), siAurora-A significantly reduced the Aurora-A mRNA expression in siA-3 group (transfected three times with siAurora-A). Similarly, the cell viability of siA-3 group was lower than that of siC-3 group. The cell viability of siC-3 group was higher than that of serum-free-6d group, but, levels of Aurora-A mRNA expression of siC-3 group had no difference with serum-free-6d group. Finally, among groups transfected once or three times or starved for 6 d, there was no significant difference of ciliated cell proportions in both types of cells respectively. CONCLUSIONS Repeated siAurora-A transfection decreased Aurora-A expression that resulted in effective suppression proliferation of SK-MES-1 or A549 cells, but did not affect cilia generation.
Collapse
Affiliation(s)
- Zhonghua Wang
- 1 Department of Respiratory Medicine, General Hospital of Command , Shenyang, China .,2 Department of Histology and Embryology, Shenyang Medical , Shenyang, China
| | - Zhuang Ma
- 1 Department of Respiratory Medicine, General Hospital of Command , Shenyang, China
| | - Jianping Cao
- 1 Department of Respiratory Medicine, General Hospital of Command , Shenyang, China
| |
Collapse
|
63
|
Abstract
The primary cilium is an antenna-like organelle assembled on most types of quiescent and differentiated mammalian cells. This immotile structure is essential for interpreting extracellular signals that regulate growth, development and homeostasis. As such, ciliary defects produce a spectrum of human diseases, termed ciliopathies, and deregulation of this important organelle also plays key roles during tumor formation and progression. Recent studies have begun to clarify the key mechanisms that regulate ciliary assembly and disassembly in both normal and tumor cells, highlighting new possibilities for therapeutic intervention. Here, we review these exciting new findings, discussing the molecular factors involved in cilium formation and removal, the intrinsic and extrinsic control of cilium assembly and disassembly, and the relevance of these processes to mammalian cell growth and disease.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Brian D Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
64
|
Abstract
Mitosis is controlled by reversible protein phosphorylation involving specific kinases and phosphatases. A handful of major mitotic protein kinases, such as the cyclin B-CDK1 complex, the Aurora kinases, and Polo-like kinase 1 (PLK1), cooperatively regulate distinct mitotic processes. Research has identified proteins and mechanisms that integrate these kinases into signaling cascades that guide essential mitotic events. These findings have important implications for our understanding of the mechanisms of mitotic regulation and may advance the development of novel antimitotic drugs. We review collected evidence that in vertebrates, the Aurora kinases serve as catalytic subunits of distinct complexes formed with the four scaffold proteins Bora, CEP192, INCENP, and TPX2, which we deem "core" Aurora cofactors. These complexes and the Aurora-PLK1 cascades organized by Bora, CEP192, and INCENP control crucial aspects of mitosis and all pathways of spindle assembly. We compare the mechanisms of Aurora activation in relation to the different spindle assembly pathways and draw a functional analogy between the CEP192 complex and the chromosomal passenger complex that may reflect the coevolution of centrosomes, kinetochores, and the actomyosin cleavage apparatus. We also analyze the roles and mechanisms of Aurora-PLK1 signaling in the cell and centrosome cycles and in the DNA damage response.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, Saint-Petersburg 197758, Russian Federation.
| | | |
Collapse
|
65
|
Abstract
Although tumours initiate from oncogenic changes in a cancer cell, subsequent tumour progression and therapeutic response depend on interactions between the cancer cells and the tumour microenvironment (TME). The primary monocilium, or cilium, provides a spatially localized platform for signalling by Hedgehog, Notch, WNT and some receptor tyrosine kinase pathways and mechanosensation. Changes in ciliation of cancer cells and/or cells of the TME during tumour development enforce asymmetric intercellular signalling in the TME. Growing evidence indicates that some oncogenic signalling pathways as well as some targeted anticancer therapies induce ciliation, while others repress it. The links between the genomic profile of cancer cells, drug treatment and ciliary signalling in the TME likely affect tumour growth and therapeutic response.
Collapse
Affiliation(s)
- Hanqing Liu
- School of Pharmacy, Jiangsu University, Jiangsu, China
| | - Anna A Kiseleva
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
- Kazan Federal University, Kazan, Russia
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
66
|
Loss-of-function of IFT88 determines metabolic phenotypes in thyroid cancer. Oncogene 2018; 37:4455-4474. [DOI: 10.1038/s41388-018-0211-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 01/18/2023]
|
67
|
Wu Q, Gao K, Zheng S, Zhu X, Liang Y, Pan J. Calmodulin regulates a TRP channel (ADF1) and phospholipase C (PLC) to mediate elevation of cytosolic calcium during acidic stress that induces deflagellation in
Chlamydomonas. FASEB J 2018; 32:3689-3699. [DOI: 10.1096/fj.201701396rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Qiong Wu
- Ministry of Education (MOE) Key Laboratory of Protein SciencesTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijingChina
- Laboratory for Marine Biology and BiotechnologyQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Kang Gao
- Hebei Key Laboratory of Molecular and Cellular BiologyCollege of Life ScienceHebei Normal UniversityShijiazhuangChina
| | - Shuzhi Zheng
- Hebei Key Laboratory of Molecular and Cellular BiologyCollege of Life ScienceHebei Normal UniversityShijiazhuangChina
| | - Xin Zhu
- Ministry of Education (MOE) Key Laboratory of Protein SciencesTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijingChina
| | - Yinwen Liang
- Ministry of Education (MOE) Key Laboratory of Protein SciencesTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijingChina
| | - Junmin Pan
- Ministry of Education (MOE) Key Laboratory of Protein SciencesTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijingChina
- Laboratory for Marine Biology and BiotechnologyQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
68
|
Functional interplay between cylindromatosis and histone deacetylase 6 in ciliary homeostasis revealed by phenotypic analysis of double knockout mice. Oncotarget 2018; 7:27527-37. [PMID: 27028867 PMCID: PMC5053669 DOI: 10.18632/oncotarget.8374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/16/2016] [Indexed: 01/04/2023] Open
Abstract
Cilia are present in most vertebrate tissues with a wide variety of functions, and abnormalities of cilia are linked to numerous human disorders. However, the molecular events underlying ciliary homeostasis are poorly understood. In this study, we generated double knockout (DKO) mice for the deubiquitinase cylindromatosis (CYLD) and histone deacetylase 6 (HDAC6), two critical ciliary regulators. The Cyld/Hdac6 DKO mice were phenotypically normal and showed no obvious variances in weight or behavior compared with their wild-type littermates. Strikingly, Cyld loss-induced ciliary defects in the testis, trachea, and kidney were abrogated in the Cyld/Hdac6 DKO mice. In addition, the diminished α-tubulin acetylation and impaired sonic hedgehog signaling caused by loss of Cyld were largely restored by simultaneous deletion of Hdac6. We further found by immunofluorescence microscopy a colocalization of CYLD and HDAC6 at the centrosome/basal body and, interestingly, loss of Cyld promoted the localization of HDAC6 at the centrosome/basal body. These findings provide physiological insight into the ciliary role of the CYLD/HDAC6 axis and suggest a functional interplay between these two proteins in ciliary homeostasis.
Collapse
|
69
|
Nikonova AS, Deneka AY, Kiseleva AA, Korobeynikov V, Gaponova A, Serebriiskii IG, Kopp MC, Hensley HH, Seeger-Nukpezah TN, Somlo S, Proia DA, Golemis EA. Ganetespib limits ciliation and cystogenesis in autosomal-dominant polycystic kidney disease (ADPKD). FASEB J 2018; 32:2735-2746. [PMID: 29401581 DOI: 10.1096/fj.201700909r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is associated with progressive formation of renal cysts, kidney enlargement, hypertension, and typically end-stage renal disease. In ADPKD, inherited mutations disrupt function of the polycystins (encoded by PKD1 and PKD2), thus causing loss of a cyst-repressive signal emanating from the renal cilium. Genetic studies have suggested ciliary maintenance is essential for ADPKD pathogenesis. Heat shock protein 90 (HSP90) clients include multiple proteins linked to ciliary maintenance. We determined that ganetespib, a clinical HSP90 inhibitor, inhibited proteasomal repression of NEK8 and the Aurora-A activator trichoplein, rapidly activating Aurora-A kinase and causing ciliary loss in vitro. Using conditional mouse models for ADPKD, we performed long-term (10 or 50 wk) dosing experiments that demonstrated HSP90 inhibition caused durable in vivo loss of cilia, controlled cystic growth, and ameliorated symptoms induced by loss of Pkd1 or Pkd2. Ganetespib efficacy was not increased by combination with 2-deoxy-d-glucose, a glycolysis inhibitor showing some promise for ADPKD. These studies identify a new biologic activity for HSP90 and support a cilia-based mechanism for cyst repression.-Nikonova, A. S., Deneka, A. Y., Kiseleva, A. A., Korobeynikov, V., Gaponova, A., Serebriiskii, I. G., Kopp, M. C., Hensley, H. H., Seeger-Nukpezah, T. N., Somlo, S., Proia, D. A., Golemis, E. A. Ganetespib limits ciliation and cystogenesis in autosomal-dominant polycystic kidney disease (ADPKD).
Collapse
Affiliation(s)
- Anna S Nikonova
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Alexander Y Deneka
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Kazan Federal University, Kazan, Russia
| | - Anna A Kiseleva
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Kazan Federal University, Kazan, Russia
| | - Vladislav Korobeynikov
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Anna Gaponova
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Laboratory of Genome Engineering, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,Immanuel Kant Baltic Federal University, Konigsberg, Russia
| | - Ilya G Serebriiskii
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Kazan Federal University, Kazan, Russia
| | - Meghan C Kopp
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Cancer Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Harvey H Hensley
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Tamina N Seeger-Nukpezah
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Department I of Internal Medicine and Center for Integrated Oncology, University of Cologne, Cologne, Germany
| | - Stefan Somlo
- Departments of Internal Medicine and Genetics, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - David A Proia
- Synta Pharmaceuticals Corporation, Lexington, Massachusetts, USA
| | - Erica A Golemis
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
70
|
Werner S, Pimenta-Marques A, Bettencourt-Dias M. Maintaining centrosomes and cilia. J Cell Sci 2017; 130:3789-3800. [DOI: 10.1242/jcs.203505] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT
Centrosomes and cilia are present in organisms from all branches of the eukaryotic tree of life. These structures are composed of microtubules and various other proteins, and are required for a plethora of cell processes such as structuring the cytoskeleton, sensing the environment, and motility. Deregulation of centrosome and cilium components leads to a wide range of diseases, some of which are incompatible with life. Centrosomes and cilia are thought to be very stable and can persist over long periods of time. However, these structures can disappear in certain developmental stages and diseases. Moreover, some centrosome and cilia components are quite dynamic. While a large body of knowledge has been produced regarding the biogenesis of these structures, little is known about how they are maintained. In this Review, we propose the existence of specific centrosome and cilia maintenance programs, which are regulated during development and homeostasis, and when deregulated can lead to disease.
Collapse
Affiliation(s)
- Sascha Werner
- Cell Cycle Regulation Lab, Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Ana Pimenta-Marques
- Cell Cycle Regulation Lab, Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Mónica Bettencourt-Dias
- Cell Cycle Regulation Lab, Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| |
Collapse
|
71
|
DeVaul N, Koloustroubis K, Wang R, Sperry AO. A novel interaction between kinase activities in regulation of cilia formation. BMC Cell Biol 2017; 18:33. [PMID: 29141582 PMCID: PMC5688660 DOI: 10.1186/s12860-017-0149-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/01/2017] [Indexed: 01/08/2023] Open
Abstract
Background The primary cilium is an extension of the cell membrane that encloses a microtubule-based axoneme. Primary cilia are essential for transmission of environmental cues that determine cell fate. Disruption of primary cilia function is the molecular basis of numerous developmental disorders. Despite their biological importance, the mechanisms governing their assembly and disassembly are just beginning to be understood. Cilia growth and disassembly are essential events when cells exit and reenter into the cell cycle. The kinases never in mitosis-kinase 2 (Nek2) and Aurora A (AurA) act to depolymerize cilia when cells reenter the cell cycle from G0. Results Coexpression of either kinase with its kinase dead companion [AurA with kinase dead Nek2 (Nek2 KD) or Nek2 with kinase dead AurA (AurA KD)] had different effects on cilia depending on whether cilia are growing or shortening. AurA and Nek2 are individually able to shorten cilia when cilia are growing but both are required when cilia are being absorbed. The depolymerizing activity of each kinase is increased when coexpressed with the kinase dead version of the other kinase but only when cilia are assembling. Additionally, the two kinases act additively when cilia are assembling but not disassembling. Inhibition of AurA increases cilia number while inhibition of Nek2 significantly stimulates cilia length. The complex functional relationship between the two kinases reflects their physical interaction. Further, we identify a role for a PP1 binding protein, PPP1R42, in inhibiting Nek2 and increasing ciliation of ARPE-19 cells. Conclusion We have uncovered a novel functional interaction between Nek2 and AurA that is dependent on the growth state of cilia. This differential interdependence reflects opposing regulation when cilia are growing or shortening. In addition to interaction between the kinases to regulate ciliation, the PP1 binding protein PPP1R42 directly inhibits Nek2 independent of PP1 indicating another level of regulation of this kinase. In summary, we demonstrate a complex interplay between Nek2 and AurA kinases in regulation of ciliation in ARPE-19 cells. Electronic supplementary material The online version of this article (10.1186/s12860-017-0149-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicole DeVaul
- Laboratory of Biochemistry and Genetics, National Institute of Diabetics and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katerina Koloustroubis
- Anatomy and Cell Biology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - Rong Wang
- Anatomy and Cell Biology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - Ann O Sperry
- Anatomy and Cell Biology, East Carolina University, Brody School of Medicine, Greenville, NC, USA.
| |
Collapse
|
72
|
Walz G. Role of primary cilia in non-dividing and post-mitotic cells. Cell Tissue Res 2017; 369:11-25. [PMID: 28361305 PMCID: PMC5487853 DOI: 10.1007/s00441-017-2599-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/20/2017] [Accepted: 02/27/2017] [Indexed: 12/12/2022]
Abstract
The essential role of primary (non-motile) cilia during the development of multi-cellular tissues and organs is well established and is underlined by severe disease manifestations caused by mutations in cilia-associated molecules that are collectively termed ciliopathies. However, the role of primary cilia in non-dividing and terminally differentiated, post-mitotic cells is less well understood. Although the prevention of cells from re-entering the cell cycle may represent a major chore, primary cilia have recently been linked to DNA damage responses, autophagy and mitochondria. Given this connectivity, primary cilia in non-dividing cells are well positioned to form a signaling hub outside of the nucleus. Such a center could integrate information to initiate responses and to maintain cellular homeostasis if cell survival is jeopardized. These more discrete functions may remain undetected until differentiated cells are confronted with emergencies.
Collapse
Affiliation(s)
- Gerd Walz
- Renal Division, Department of Medicine, University Freiburg Medical Center, Hugstetter Strasse 55, 79106, Freiburg, Germany.
| |
Collapse
|
73
|
Abstract
The primary cilium is an antenna-like, immotile organelle present on most types of mammalian cells, which interprets extracellular signals that regulate growth and development. Although once considered a vestigial organelle, the primary cilium is now the focus of considerable interest. We now know that ciliary defects lead to a panoply of human diseases, termed ciliopathies, and the loss of this organelle may be an early signature event during oncogenic transformation. Ciliopathies include numerous seemingly unrelated developmental syndromes, with involvement of the retina, kidney, liver, pancreas, skeletal system and brain. Recent studies have begun to clarify the key mechanisms that link cilium assembly and disassembly to the cell cycle, and suggest new possibilities for therapeutic intervention.
Collapse
Affiliation(s)
- Irma Sánchez
- Department of Pathology, NYU School of Medicine, Smilow Research Building, 522 First Avenue, New York, New York 10016, USA
| | - Brian David Dynlacht
- Department of Pathology, NYU School of Medicine, Smilow Research Building, 522 First Avenue, New York, New York 10016, USA
| |
Collapse
|
74
|
Wang L, Gu L, Meng D, Wu Q, Deng H, Pan J. Comparative Proteomics Reveals Timely Transport into Cilia of Regulators or Effectors as a Mechanism Underlying Ciliary Disassembly. J Proteome Res 2017; 16:2410-2418. [DOI: 10.1021/acs.jproteome.6b01048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Limei Wang
- MOE
Key Laboratory of Protein Sciences, Tsinghua-Peking Center for Life
Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lixiao Gu
- MOE
Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Dan Meng
- Tianjin
Key Laboratory of Food and Biotechnology, School of Biotechnology
and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Qiong Wu
- MOE
Key Laboratory of Protein Sciences, Tsinghua-Peking Center for Life
Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haiteng Deng
- MOE
Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Junmin Pan
- MOE
Key Laboratory of Protein Sciences, Tsinghua-Peking Center for Life
Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Laboratory
for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong Province 266237, China
| |
Collapse
|
75
|
Christensen ST, Morthorst SK, Mogensen JB, Pedersen LB. Primary Cilia and Coordination of Receptor Tyrosine Kinase (RTK) and Transforming Growth Factor β (TGF-β) Signaling. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028167. [PMID: 27638178 DOI: 10.1101/cshperspect.a028167] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Since the beginning of the millennium, research in primary cilia has revolutionized our way of understanding how cells integrate and organize diverse signaling pathways during vertebrate development and in tissue homeostasis. Primary cilia are unique sensory organelles that detect changes in their extracellular environment and integrate and transmit signaling information to the cell to regulate various cellular, developmental, and physiological processes. Many different signaling pathways have now been shown to rely on primary cilia to function properly, and mutations that lead to ciliary dysfunction are at the root of a pleiotropic group of diseases and syndromic disorders called ciliopathies. In this review, we present an overview of primary cilia-mediated regulation of receptor tyrosine kinase (RTK) and transforming growth factor β (TGF-β) signaling. Further, we discuss how defects in the coordination of these pathways may be linked to ciliopathies.
Collapse
Affiliation(s)
- Søren T Christensen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| | - Stine K Morthorst
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| | - Johanne B Mogensen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| | - Lotte B Pedersen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| |
Collapse
|
76
|
Urrego D, Sánchez A, Tomczak AP, Pardo LA. The electric fence to cell-cycle progression: Do local changes in membrane potential facilitate disassembly of the primary cilium? Bioessays 2017; 39. [DOI: 10.1002/bies.201600190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Diana Urrego
- Max-Planck-Institut für experimentelle Medizin; AG Oncophysiology; Göttingen Germany
| | - Araceli Sánchez
- Max-Planck-Institut für experimentelle Medizin; AG Oncophysiology; Göttingen Germany
| | - Adam P. Tomczak
- Max-Planck-Institut für experimentelle Medizin; AG Oncophysiology; Göttingen Germany
| | - Luis A. Pardo
- Max-Planck-Institut für experimentelle Medizin; AG Oncophysiology; Göttingen Germany
| |
Collapse
|
77
|
Mizuno K, Sloboda RD. Protein arginine methyltransferases interact with intraflagellar transport particles and change location during flagellar growth and resorption. Mol Biol Cell 2017; 28:1208-1222. [PMID: 28298486 PMCID: PMC5415017 DOI: 10.1091/mbc.e16-11-0774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/03/2017] [Accepted: 03/08/2017] [Indexed: 01/09/2023] Open
Abstract
Protein arginine methyl transferases are located at specific sites in the flagellum. They change location during changes in flagellar dynamics (i.e., resorption and regeneration) via interaction with intraflagellar transport trains. Changes in protein by posttranslational modifications comprise an important mechanism for the control of many cellular processes. Several flagellar proteins are methylated on arginine residues during flagellar resorption; however, the function is not understood. To learn more about the role of protein methylation during flagellar dynamics, we focused on protein arginine methyltransferases (PRMTs) 1, 3, 5, and 10. These PRMTs localize to the tip of flagella and in a punctate pattern along the length, very similar, but not identical, to that of intraflagellar transport (IFT) components. In addition, we found that PRMT 1 and 3 are also highly enriched at the base of the flagella, and the basal localization of these PRMTs changes during flagellar regeneration and resorption. Proteins with methyl arginine residues are also enriched at the tip and base of flagella, and their localization also changes during flagellar assembly and disassembly. PRMTs are lost from the flagella of fla10-1 cells, which carry a temperature-sensitive mutation in the anterograde motor for IFT. The data define the distribution of specific PRMTs and their target proteins in flagella and demonstrate that PRMTs are cargo for translocation within flagella by the process of IFT.
Collapse
Affiliation(s)
- Katsutoshi Mizuno
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755
| | - Roger D Sloboda
- Marine Biological Laboratory, Woods Hole, MA 02543 .,Marine Biological Laboratory, Woods Hole, MA 02543
| |
Collapse
|
78
|
Korobeynikov V, Deneka AY, Golemis EA. Mechanisms for nonmitotic activation of Aurora-A at cilia. Biochem Soc Trans 2017; 45:37-49. [PMID: 28202658 PMCID: PMC5860652 DOI: 10.1042/bst20160142] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/19/2016] [Accepted: 10/24/2016] [Indexed: 12/12/2022]
Abstract
Overexpression of the Aurora kinase A (AURKA) is oncogenic in many tumors. Many studies of AURKA have focused on activities of this kinase in mitosis, and elucidated the mechanisms by which AURKA activity is induced at the G2/M boundary through interactions with proteins such as TPX2 and NEDD9. These studies have informed the development of small molecule inhibitors of AURKA, of which a number are currently under preclinical and clinical assessment. While the first activities defined for AURKA were its control of centrosomal maturation and organization of the mitotic spindle, an increasing number of studies over the past decade have recognized a separate biological function of AURKA, in controlling disassembly of the primary cilium, a small organelle protruding from the cell surface that serves as a signaling platform. Importantly, these activities require activation of AURKA in early G1, and the mechanisms of activation are much less well defined than those in mitosis. A better understanding of the control of AURKA activity and the role of AURKA at cilia are both important in optimizing the efficacy and interpreting potential downstream consequences of AURKA inhibitors in the clinic. We here provide a current overview of proteins and mechanisms that have been defined as activating AURKA in G1, based on the study of ciliary disassembly.
Collapse
Affiliation(s)
- Vladislav Korobeynikov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, U.S.A
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, U.S.A
| | - Alexander Y Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, U.S.A
- Kazan Federal University, Kazan 420000, Russian Federation
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, U.S.A.
| |
Collapse
|
79
|
Wang GF, Dong Q, Bai Y, Yuan J, Xu Q, Cao C, Liu X. Oxidative stress induces mitotic arrest by inhibiting Aurora A-involved mitotic spindle formation. Free Radic Biol Med 2017; 103:177-187. [PMID: 28017898 DOI: 10.1016/j.freeradbiomed.2016.12.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/05/2016] [Accepted: 12/21/2016] [Indexed: 01/11/2023]
Abstract
Oxidative stress contributes to the oxidative modification of cellular components, including lipids, proteins and DNA, and results in DNA damage, cell cycle arrest, cellular dysfunction and apoptosis. However, the mechanism underlying oxidative stress-induced mitotic abnormalities is not fully understood. In this study, we demonstrated that exogenous and endogenous reactive oxygen species (ROS) promoted mitotic arrest. Delayed formation and abnormal function of the mitotic spindle, which directly impeded mitosis and promoted abnormal chromosome separation, was responsible for ROS-induced mitotic arrest. As a key regulator of mitotic spindle assembly, Aurora A kinase was hyperphosphorylated in early mitosis under oxidative stress, which may disturb the function of Aurora A in mitotic spindle formation. Our findings identified a mechanism by which ROS regulate mitotic progression and indicated a potential molecular target for the treatment of oxidative stress-related diseases.
Collapse
Affiliation(s)
- Guang-Fei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China
| | - Qincai Dong
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Yuanyuan Bai
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Jing Yuan
- Beijing Institute of Disease Control and Prevention, Beijing 100071, China
| | - Quanbin Xu
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Cheng Cao
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China.
| | - Xuan Liu
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China.
| |
Collapse
|
80
|
Blas-Rus N, Bustos-Morán E, Martín-Cófreces NB, Sánchez-Madrid F. Aurora-A shines on T cell activation through the regulation of Lck. Bioessays 2016; 39. [PMID: 27910998 DOI: 10.1002/bies.201600156] [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] [Indexed: 01/01/2023]
Abstract
Different protein kinases control signaling emanating from the T cell receptor (TCR) during antigen-specific T cell activation. Mitotic kinases, e.g. Aurora-A, have been widely studied in the context of mitosis due to their role during microtubule (MT) nucleation, becoming critical regulators of cell cycle progression. We have recently described a specific role for Aurora-A kinase in antigenic T cell activation. Blockade of Aurora-A in T cells severely disrupts the dynamics of MTs and CD3ζ-bearing signaling vesicles during T cell activation. Furthermore, Aurora-A deletion impairs the activation of signaling molecules downstream of the TCR. Targeting Aurora-A disturbs the activation of Lck, which is one of the first signals that drive T cell activation in an antigen-dependent manner. This work describes possible models of regulation of Lck by Aurora-A during T cell activation. We also discuss possible roles for Aurora-A in other systems similar to the IS, and its putative functions in cell polarization.
Collapse
Affiliation(s)
- Noelia Blas-Rus
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain
| | - Eugenio Bustos-Morán
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Noa B Martín-Cófreces
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain.,Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
81
|
Burgess SG, Grazia Concilio M, Bayliss R, Fielding AJ. Detection of Ligand-induced Conformational Changes in the Activation Loop of Aurora-A Kinase by PELDOR Spectroscopy. ChemistryOpen 2016; 5:531-534. [PMID: 28032021 PMCID: PMC5167317 DOI: 10.1002/open.201600101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Indexed: 11/10/2022] Open
Abstract
The structure of protein kinases has been extensively studied by protein crystallography. Conformational movement of the kinase activation loop is thought to be crucial for regulation of activity; however, in many cases the position of the activation loop in solution is unknown. Protein kinases are an important class of therapeutic target and kinase inhibitors are classified by their effect on the activation loop. Here, we report the use of pulsed electron double resonance (PELDOR) and site-directed spin labeling to monitor conformational changes through the insertion of MTSL [S-(1-oxyl-2,2,5,5-tetramethyl-2,5-dihydro-1 H-pyrrol-3-yl)methyl methanesulfonothioate] on the dynamic activation loop and a stable site on the outer surface of the enzyme. The action of different ligands such as microtubule-associated protein (TPX2) and inhibitors could be discriminated as well as their ability to lock the activation loop in a fixed conformation. This study provides evidence for structural adaptations that could be used for drug design and a methodological approach that has potential to characterize inhibitors in development.
Collapse
Affiliation(s)
- Selena G. Burgess
- Astbury Centre for Structural and Molecular BiologyFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUnited Kingdom
| | - Maria Grazia Concilio
- The Photon Science Institute and School of ChemistryUniversity of ManchesterManchesterM13 9PLUnited Kingdom
| | - Richard Bayliss
- Astbury Centre for Structural and Molecular BiologyFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUnited Kingdom
| | - Alistair J. Fielding
- The Photon Science Institute and School of ChemistryUniversity of ManchesterManchesterM13 9PLUnited Kingdom
| |
Collapse
|
82
|
Goto H, Inaba H, Inagaki M. Mechanisms of ciliogenesis suppression in dividing cells. Cell Mol Life Sci 2016; 74:881-890. [PMID: 27669693 PMCID: PMC5306231 DOI: 10.1007/s00018-016-2369-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/05/2016] [Accepted: 09/14/2016] [Indexed: 12/26/2022]
Abstract
The primary cilium is a non-motile and microtubule-enriched protrusion ensheathed by plasma membrane. Primary cilia function as mechano/chemosensors and signaling hubs and their disorders predispose to a wide spectrum of human diseases. Most types of cells assemble their primary cilia in response to cellular quiescence, whereas they start to retract the primary cilia upon cell-cycle reentry. The retardation of ciliary resorption process has been shown to delay cell-cycle progression to the S or M phase after cell-cycle reentry. Apart from this conventional concept of ciliary disassembly linked to cell-cycle reentry, recent studies have led to a novel concept, suggesting that cells can suppress primary cilia assembly during cell proliferation. Accumulating evidence has also demonstrated the importance of Aurora-A (a protein originally identified as one of mitotic kinases) not only in ciliary resorption after cell-cycle reentry but also in the suppression of ciliogenesis in proliferating cells, whereas Aurora-A activators are clearly distinct in both phenomena. Here, we summarize the current knowledge of how cycling cells suppress ciliogenesis and compare it with mechanisms underlying ciliary resorption after cell-cycle reentry. We also discuss a reciprocal relationship between primary cilia and cell proliferation.
Collapse
Affiliation(s)
- Hidemasa Goto
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan. .,Department of Cellular Oncology, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan.
| | - Hironori Inaba
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Masaki Inagaki
- Department of Physiology, Mie University School of Medicine, Tsu, Mie, Japan.
| |
Collapse
|
83
|
Inaba H, Goto H, Kasahara K, Kumamoto K, Yonemura S, Inoko A, Yamano S, Wanibuchi H, He D, Goshima N, Kiyono T, Hirotsune S, Inagaki M. Ndel1 suppresses ciliogenesis in proliferating cells by regulating the trichoplein-Aurora A pathway. J Cell Biol 2016; 212:409-23. [PMID: 26880200 PMCID: PMC4754717 DOI: 10.1083/jcb.201507046] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Ndel1, a protein located at the subdistal appendage of mother centriole, functions as an upstream regulator of the trichoplein–Aurora A pathway that suppresses ciliogenesis in proliferating cells. Primary cilia protrude from the surface of quiescent cells and disassemble at cell cycle reentry. We previously showed that ciliary reassembly is suppressed by trichoplein-mediated Aurora A activation pathway in growing cells. Here, we report that Ndel1, a well-known modulator of dynein activity, localizes at the subdistal appendage of the mother centriole, which nucleates a primary cilium. In the presence of serum, Ndel1 depletion reduces trichoplein at the mother centriole and induces unscheduled primary cilia formation, which is reverted by forced trichoplein expression or coknockdown of KCTD17 (an E3 ligase component protein for trichoplein). Serum starvation induced transient Ndel1 degradation, subsequent to the disappearance of trichoplein at the mother centriole. Forced expression of Ndel1 suppressed trichoplein degradation and axonemal microtubule extension during ciliogenesis, similar to trichoplein induction or KCTD17 knockdown. Most importantly, the proportion of ciliated and quiescent cells was increased in the kidney tubular epithelia of newborn Ndel1-hypomorphic mice. Thus, Ndel1 acts as a novel upstream regulator of the trichoplein–Aurora A pathway to inhibit primary cilia assembly.
Collapse
Affiliation(s)
- Hironori Inaba
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Hidemasa Goto
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan Department of Cellular Oncology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Kousuke Kasahara
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan Department of Oncology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Kanako Kumamoto
- Department of Genetic Disease Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Shigenobu Yonemura
- Center for Life Science Technologies (Ultrastructural Research Team), Institute of Physical and Chemical Research, Kobe 650-0047, Japan
| | - Akihito Inoko
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Shotaro Yamano
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Dongwei He
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Naoki Goshima
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Tohru Kiyono
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Shinji Hirotsune
- Department of Genetic Disease Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Masaki Inagaki
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan Department of Cellular Oncology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| |
Collapse
|
84
|
Yan M, Wang C, He B, Yang M, Tong M, Long Z, Liu B, Peng F, Xu L, Zhang Y, Liang D, Lei H, Subrata S, Kelley KW, Lam EWF, Jin B, Liu Q. Aurora-A Kinase: A Potent Oncogene and Target for Cancer Therapy. Med Res Rev 2016; 36:1036-1079. [PMID: 27406026 DOI: 10.1002/med.21399] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/18/2016] [Accepted: 06/08/2016] [Indexed: 02/06/2023]
Abstract
The Aurora kinase family is comprised of three serine/threonine kinases, Aurora-A, Aurora-B, and Aurora-C. Among these, Aurora-A and Aurora-B play central roles in mitosis, whereas Aurora-C executes unique roles in meiosis. Overexpression or gene amplification of Aurora kinases has been reported in a broad range of human malignancies, pointing to their role as potent oncogenes in tumorigenesis. Aurora kinases therefore represent promising targets for anticancer therapeutics. A number of Aurora kinase inhibitors (AKIs) have been generated; some of which are currently undergoing clinical evaluation. Recent studies have unveiled novel unexpected functions of Aurora kinases during cancer development and the mechanisms underlying the anticancer actions of AKIs. In this review, we discuss the most recent advances in Aurora-A kinase research and targeted cancer therapy, focusing on the oncogenic roles and signaling pathways of Aurora-A kinases in promoting tumorigenesis, the recent preclinical and clinical AKI data, and potential alternative routes for Aurora-A kinase inhibition.
Collapse
Affiliation(s)
- Min Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunli Wang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Mengying Yang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Mengying Tong
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Zijie Long
- Institute of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bing Liu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Fei Peng
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Lingzhi Xu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Yan Zhang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Dapeng Liang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Haixin Lei
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Sen Subrata
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith W Kelley
- Laboratory of Immunophysiology, Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Bilian Jin
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.
| | - Quentin Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China. .,Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China. .,Institute of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
85
|
Barretta ML, Spano D, D'Ambrosio C, Cervigni RI, Scaloni A, Corda D, Colanzi A. Aurora-A recruitment and centrosomal maturation are regulated by a Golgi-activated pool of Src during G2. Nat Commun 2016; 7:11727. [PMID: 27242098 PMCID: PMC4895030 DOI: 10.1038/ncomms11727] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 04/25/2016] [Indexed: 02/02/2023] Open
Abstract
The Golgi apparatus is composed of stacks of cisternae laterally connected by tubules to form a ribbon-like structure. At the onset of mitosis, the Golgi ribbon is broken down into discrete stacks, which then undergo further fragmentation. This ribbon cleavage is required for G2/M transition, which thus indicates that a ‘Golgi mitotic checkpoint' couples Golgi inheritance with cell cycle transition. We previously showed that the Golgi-checkpoint regulates the centrosomal recruitment of the mitotic kinase Aurora-A; however, how the Golgi unlinking regulates this recruitment was unknown. Here we show that, in G2, Aurora-A recruitment is promoted by activated Src at the Golgi. Our data provide evidence that Src and Aurora-A interact upon Golgi ribbon fragmentation; Src phosphorylates Aurora-A at tyrosine 148 and this specific phosphorylation is required for Aurora-A localization at the centrosomes. This process, pivotal for centrosome maturation, is a fundamental prerequisite for proper spindle formation and chromosome segregation. The Golgi mitotic checkpoint couples Golgi inheritance with cell cycle transition, and regulates centrosomal recruitment of the mitotic kinase Aurora-A. Here the authors show that upon Golgi ribbon fragmentation in G2, Src phosphorylates Aurora-A at the Golgi, driving its localization to the centrosomes.
Collapse
Affiliation(s)
- Maria Luisa Barretta
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Daniela Spano
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Chiara D'Ambrosio
- Proteomics and Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, ISPAAM, National Research Council (CNR), Via Argine 1085, 80147 Naples, Italy
| | - Romina Ines Cervigni
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Andrea Scaloni
- Proteomics and Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, ISPAAM, National Research Council (CNR), Via Argine 1085, 80147 Naples, Italy
| | - Daniela Corda
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Antonino Colanzi
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| |
Collapse
|
86
|
Liang Y, Meng D, Zhu B, Pan J. Mechanism of ciliary disassembly. Cell Mol Life Sci 2016; 73:1787-802. [PMID: 26869233 PMCID: PMC11108551 DOI: 10.1007/s00018-016-2148-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/19/2022]
Abstract
As motile organelles and sensors, cilia play pivotal roles in cell physiology, development and organ homeostasis. Ciliary defects are associated with a class of cilia-related diseases or developmental disorders, termed ciliopathies. Even though the presence of cilia is required for diverse functions, cilia can be removed through ciliary shortening or resorption that necessitates disassembly of the cilium, which occurs normally during cell cycle progression, cell differentiation and in response to cellular stress. The functional significance of ciliary resorption is highlighted in controlling the G1-S transition during cell cycle progression. Internal or external cues that trigger ciliary resorption initiate signaling cascades that regulate several downstream events including depolymerization of axonemal microtubules, dynamic changes in actin and the ciliary membrane, regulation of intraflagellar transport and posttranslational modifications of ciliary proteins. To ensure ciliary resorption, both the active disassembly of the cilium and the simultaneous inhibition of ciliary assembly must be coordinately regulated.
Collapse
Affiliation(s)
- Yinwen Liang
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dan Meng
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bing Zhu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junmin Pan
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China.
| |
Collapse
|
87
|
Blas-Rus N, Bustos-Morán E, Pérez de Castro I, de Cárcer G, Borroto A, Camafeita E, Jorge I, Vázquez J, Alarcón B, Malumbres M, Martín-Cófreces NB, Sánchez-Madrid F. Aurora A drives early signalling and vesicle dynamics during T-cell activation. Nat Commun 2016; 7:11389. [PMID: 27091106 PMCID: PMC4838898 DOI: 10.1038/ncomms11389] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 03/21/2016] [Indexed: 01/09/2023] Open
Abstract
Aurora A is a serine/threonine kinase that contributes to the progression of mitosis by inducing microtubule nucleation. Here we have identified an unexpected role for Aurora A kinase in antigen-driven T-cell activation. We find that Aurora A is phosphorylated at the immunological synapse (IS) during TCR-driven cell contact. Inhibition of Aurora A with pharmacological agents or genetic deletion in human or mouse T cells severely disrupts the dynamics of microtubules and CD3ζ-bearing vesicles at the IS. The absence of Aurora A activity also impairs the activation of early signalling molecules downstream of the TCR and the expression of IL-2, CD25 and CD69. Aurora A inhibition causes delocalized clustering of Lck at the IS and decreases phosphorylation levels of tyrosine kinase Lck, thus indicating Aurora A is required for maintaining Lck active. These findings implicate Aurora A in the propagation of the TCR activation signal. Aurora A is a protein kinase that contributes to the progression of mitosis by stimulating microtubule nucleation. Here the authors show that Aurora A also functions during T cell activation by maintaining TCR signaling through Lck activation.
Collapse
Affiliation(s)
- Noelia Blas-Rus
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, C/ Diego de León 62, Madrid 28006, Spain
| | - Eugenio Bustos-Morán
- Cell-cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Ignacio Pérez de Castro
- Cell Division and Cancer Group, Centro Nacional de Investigaciones Oncológicas (CNIO), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Guillermo de Cárcer
- Cell Division and Cancer Group, Centro Nacional de Investigaciones Oncológicas (CNIO), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Aldo Borroto
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/ Nicolás cabrera 1, Madrid 28049, Spain
| | - Emilio Camafeita
- Laboratory of Cardiovascular Proteomics, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Inmaculada Jorge
- Laboratory of Cardiovascular Proteomics, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/ Nicolás cabrera 1, Madrid 28049, Spain
| | - Marcos Malumbres
- Cell Division and Cancer Group, Centro Nacional de Investigaciones Oncológicas (CNIO), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Noa B Martín-Cófreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, C/ Diego de León 62, Madrid 28006, Spain.,Cell-cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, C/ Diego de León 62, Madrid 28006, Spain.,Cell-cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| |
Collapse
|
88
|
Khan NA, Willemarck N, Talebi A, Marchand A, Binda MM, Dehairs J, Rueda-Rincon N, Daniels VW, Bagadi M, Raj DBTG, Vanderhoydonc F, Munck S, Chaltin P, Swinnen JV. Identification of drugs that restore primary cilium expression in cancer cells. Oncotarget 2016; 7:9975-92. [PMID: 26862738 PMCID: PMC4891097 DOI: 10.18632/oncotarget.7198] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 12/08/2015] [Indexed: 12/19/2022] Open
Abstract
The development of cancer is often accompanied by a loss of the primary cilium, a microtubule-based cellular protrusion that functions as a cellular antenna and that puts a break on cell proliferation. Hence, restoration of the primary cilium in cancer cells may represent a novel promising approach to attenuate tumor growth. Using a high content analysis-based approach we screened a library of clinically evaluated compounds and marketed drugs for their ability to restore primary cilium expression in pancreatic ductal cancer cells. A diverse set of 118 compounds stimulating cilium expression was identified. These included glucocorticoids, fibrates and other nuclear receptor modulators, neurotransmitter regulators, ion channel modulators, tyrosine kinase inhibitors, DNA gyrase/topoisomerase inhibitors, antibacterial compounds, protein inhibitors, microtubule modulators, and COX inhibitors. Certain compounds also dramatically affected the length of the cilium. For a selection of compounds (Clofibrate, Gefitinib, Sirolimus, Imexon and Dexamethasone) their ability to restore ciliogenesis was confirmed in a panel of human cancer cell line models representing different cancer types (pancreas, lung, kidney, breast). Most compounds attenuated cell proliferation, at least in part through induction of the primary cilium, as demonstrated by cilium removal using chloral hydrate. These findings reveal that several commonly used drugs restore ciliogenesis in cancer cells, and warrant further investigation of their antineoplastic properties.
Collapse
Affiliation(s)
- Niamat Ali Khan
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Nicolas Willemarck
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Ali Talebi
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | | | - Maria Mercedes Binda
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Jonas Dehairs
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Natalia Rueda-Rincon
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Veerle W. Daniels
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Muralidhararao Bagadi
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Deepak Balaji Thimiri Govinda Raj
- European Molecular Biology Laboratory (EMBL), Grenoble Outstation and Unit of Virus Host-Cell Interactions (UVHCI), UJF-EMBL-CNRS, CS 90181, France
| | - Frank Vanderhoydonc
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| | - Sebastian Munck
- VIB Bio Imaging Core and Center for the Biology of Disease, 3000 Leuven, Belgium
- KU Leuven - University of Leuven, Center for Human Genetics, 3000 Leuven, Belgium
| | - Patrick Chaltin
- Cistim Leuven vzw, Bioincubator 2, 3001 Leuven, Belgium
- Centre for Drug Design and Discovery (CD3) KU Leuven R & D, Bioincubator 2, 3001 Leuven, Belgium
| | - Johannes V. Swinnen
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, 3000 Leuven, Belgium
| |
Collapse
|
89
|
Lindon C, Grant R, Min M. Ubiquitin-Mediated Degradation of Aurora Kinases. Front Oncol 2016; 5:307. [PMID: 26835416 PMCID: PMC4716142 DOI: 10.3389/fonc.2015.00307] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/25/2015] [Indexed: 11/18/2022] Open
Abstract
The Aurora kinases are essential regulators of mitosis in eukaryotes. In somatic cell divisions of higher eukaryotes, the paralogs Aurora kinase A (AurA) and Aurora kinase B (AurB) play non-overlapping roles that depend on their distinct spatiotemporal activities. These mitotic roles of Aurora kinases depend on their interactions with different partners that direct them to different mitotic destinations and different substrates: AurB is a component of the chromosome passenger complex that orchestrates the tasks of chromosome segregation and cytokinesis, while AurA has many known binding partners and mitotic roles, including a well-characterized interaction with TPX2 that mediates its role in mitotic spindle assembly. Beyond the spatial control conferred by different binding partners, Aurora kinases are subject to temporal control of their activation and inactivation. Ubiquitin-mediated proteolysis is a critical route to irreversible inactivation of these kinases, which must occur for ordered transition from mitosis back to interphase. Both AurA and AurB undergo targeted proteolysis after anaphase onset as substrates of the anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase, even while they continue to regulate steps during mitotic exit. Temporal control of Aurora kinase destruction ensures that AurB remains active at the midbody during cytokinesis long after AurA activity has been largely eliminated from the cell. Differential destruction of Aurora kinases is achieved despite the fact that they are targeted at the same time and by the same ubiquitin ligase, making these substrates an interesting case study for investigating molecular determinants of ubiquitin-mediated proteolysis in higher eukaryotes. The prevalence of Aurora overexpression in cancers and their potential as therapeutic targets add importance to the task of understanding the molecular determinants of Aurora kinase stability. Here, we review what is known about ubiquitin-mediated targeting of these critical mitotic regulators and discuss the different factors that contribute to proteolytic control of Aurora kinase activity in the cell.
Collapse
Affiliation(s)
- Catherine Lindon
- Department of Pharmacology, University of Cambridge , Cambridge , UK
| | - Rhys Grant
- Department of Pharmacology, University of Cambridge , Cambridge , UK
| | - Mingwei Min
- Department of Cell Biology, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
90
|
Izawa I, Goto H, Kasahara K, Inagaki M. Current topics of functional links between primary cilia and cell cycle. Cilia 2015; 4:12. [PMID: 26719793 PMCID: PMC4696186 DOI: 10.1186/s13630-015-0021-1] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/10/2015] [Indexed: 12/31/2022] Open
Abstract
Primary cilia, microtubule-based sensory structures, orchestrate various critical signals during development and tissue homeostasis. In view of the rising interest into the reciprocal link between ciliogenesis and cell cycle, we discuss here several recent advances to understand the molecular link between the individual step of ciliogenesis and cell cycle control. At the onset of ciliogenesis (the transition from centrosome to basal body), distal appendage proteins have been established as components indispensable for the docking of vesicles at the mother centriole. In the initial step of axonemal extension, CP110, Ofd1, and trichoplein, key negative regulators of ciliogenesis, are found to be removed by a kinase-dependent mechanism, autophagy, and ubiquitin–proteasome system, respectively. Of note, their disposal functions as a restriction point to decide that the axonemal nucleation and extension begin. In the elongation step, Nde1, a negative regulator of ciliary length, is revealed to be ubiquitylated and degraded by CDK5-SCFFbw7 in a cell cycle-dependent manner. With regard to ciliary length control, it has been uncovered in flagellar shortening of Chlamydomonas that cilia itself transmit a ciliary length signal to cytoplasm. At the ciliary resorption step upon cell cycle re-entry, cilia are found to be disassembled not only by Aurora A-HDAC6 pathway but also by Nek2-Kif24 and Plk1-Kif2A pathways through their microtubule-depolymerizing activity. On the other hand, it is becoming evident that the presence of primary cilia itself functions as a structural checkpoint for cell cycle re-entry. These data suggest that ciliogenesis and cell cycle intimately link each other, and further elucidation of these mechanisms will contribute to understanding the pathology of cilia-related disease including cancer and discovering targets of therapeutic interventions.
Collapse
Affiliation(s)
- Ichiro Izawa
- Division of Biochemistry, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681 Japan
| | - Hidemasa Goto
- Division of Biochemistry, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681 Japan ; Department of Cellular Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550 Japan
| | - Kousuke Kasahara
- Division of Biochemistry, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681 Japan ; Department of Oncology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi 467-8603 Japan
| | - Masaki Inagaki
- Division of Biochemistry, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681 Japan ; Department of Cellular Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550 Japan
| |
Collapse
|
91
|
Asteriti IA, De Mattia F, Guarguaglini G. Cross-Talk between AURKA and Plk1 in Mitotic Entry and Spindle Assembly. Front Oncol 2015; 5:283. [PMID: 26779436 PMCID: PMC4688340 DOI: 10.3389/fonc.2015.00283] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
The Aurora kinase A (AURKA) is involved in different aspects of mitotic control, from mitotic entry to cytokinesis. Consistent with its pleiotropic roles, several AURKA interactors are able to modulate its activity, the best characterized being the microtubule-binding protein TPX2, the centrosomal protein Cep192, and Bora. Bora has been described as an essential cofactor of AURKA for phosphorylation-mediated activation of the mitotic kinase polo-like kinase 1 (Plk1) at the G2/M transition. A complex AURKA/Plk1 signaling axis is emerging, with multiple involved actors; recent data suggest that this control network is not restricted to mitotic entry only, but operates throughout mitosis. Here, we integrate available data from the literature to depict the complex interplay between AURKA and Plk1 in G2 and mitosis and how it contributes to their mitotic functions. We will particularly focus on how the activity of specifically localized AURKA/Plk1 pools is modulated in time and space by their reciprocal regulation to ensure the timely and coordinated unfolding of downstream mitotic events.
Collapse
Affiliation(s)
- Italia Anna Asteriti
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| | - Fabiola De Mattia
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
92
|
Yu F, Ran J, Zhou J. Ciliopathies: Does HDAC6 Represent a New Therapeutic Target? Trends Pharmacol Sci 2015; 37:114-119. [PMID: 26651415 DOI: 10.1016/j.tips.2015.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/28/2015] [Accepted: 11/02/2015] [Indexed: 02/02/2023]
Abstract
Cilia are cellular appendages with critical roles in sensing and transducing environmental signals and guiding fluid flow. Consistent with these diverse activities, defects in ciliary structure or function have been implicated in a variety of human diseases, collectively known as 'ciliopathies'. Histone deacetylase 6 (HDAC6) is a unique cytoplasmic enzyme that regulates many biological processes through its deacetylase and ubiquitin-binding activities. There is accumulating evidence that HDAC6 is a major driver of ciliary disassembly. Small-molecule compounds that inhibit HDAC6 have been demonstrated to restore ciliary structure and function in several different ciliopathies. Here, we discuss recent findings that highlight the important role for HDAC6 in mediating ciliary disassembly and the potential for HDAC6-selective inhibitors as therapeutics for specific ciliopathies.
Collapse
Affiliation(s)
- Fan Yu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jie Ran
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
93
|
Hu Z, Liang Y, He W, Pan J. Cilia disassembly with two distinct phases of regulation. Cell Rep 2015; 10:1803-10. [PMID: 25801021 DOI: 10.1016/j.celrep.2015.02.044] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/30/2014] [Accepted: 02/17/2015] [Indexed: 01/23/2023] Open
Abstract
Cilia and flagella are dynamic organelles that undergo assembly and disassembly during each cell cycle. They are structurally polarized, and the mechanisms by which these organelles are disassembled are incompletely understood. Here, we show that flagellar resorption occurs in two distinct phases of length-dependent regulation. A CDK-like kinase, encoded by flagellar shortening 1 (FLS1), is required for the normal rate of disassembly of only the distal part of the flagellum. Mechanistically, loss of function of FLS1 prevents the initial phosphorylation of CALK, an aurora-like kinase that regulates flagellar shortening, and induces the earlier onset of the inhibitory phosphorylation of CrKinesin13, a microtubule depolymerase, which is involved in flagellar shortening. In addition, CALK and CrKinesin13 phosphorylation can also be induced by the process of flagellar shortening itself, demonstrating an example of cilia-generated signaling not requiring the binding of a ligand or the stimulation of an ion channel.
Collapse
|
94
|
Nikonova AS, Deneka AY, Eckman L, Kopp MC, Hensley HH, Egleston BL, Golemis EA. Opposing Effects of Inhibitors of Aurora-A and EGFR in Autosomal-Dominant Polycystic Kidney Disease. Front Oncol 2015; 5:228. [PMID: 26528438 PMCID: PMC4607875 DOI: 10.3389/fonc.2015.00228] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/01/2015] [Indexed: 01/24/2023] Open
Abstract
Aurora-A kinase (AURKA) overexpression in numerous tumors induces aneuploidy, in part because of cytokinetic defects. Alisertib and other small-molecule inhibitors targeting AURKA are effective in some patients as monotherapies or combination therapies. Epidermal growth factor receptor (EGFR) pro-proliferative signaling activity is commonly elevated in cancer, and the EGFR inhibitor erlotinib is commonly used as a standard of care agent for cancer. An erlotinib/alisertib combination therapy is currently under assessment in clinical trials, following pre-clinical studies that indicated synergy of these drugs in cancer. We were interested in further exploring the activity of this drug combination. Beyond well-established functions for AURKA in mitotic progression, additional non-mitotic AURKA functions include control of ciliary stability and calcium signaling. Interestingly, alisertib exacerbates the disease phenotype in mouse models for autosomal-dominant polycystic kidney disease (ADPKD), a common inherited syndrome induced by aberrant signaling from PKD1 and PKD2, cilia-localized proteins that have calcium channel activity. EGFR is also more active in ADPKD, making erlotinib also of potential interest in this disease setting. In this study, we have explored the interaction of alisertib and erlotinib in an ADPKD model. These experiments indicated erlotinib-restrained cystogenesis, opposing alisertib action. Erlotinib also interacted with alisertib to regulate proliferative signaling proteins, albeit in a complicated manner. Results suggest a nuanced role of AURKA signaling in different pathogenic conditions and inform the clinical use of AURKA inhibitors in cancer patients with comorbidities.
Collapse
Affiliation(s)
- Anna S Nikonova
- Program in Molecular Therapeutics, Fox Chase Cancer Center , Philadelphia, PA , USA
| | - Alexander Y Deneka
- Program in Molecular Therapeutics, Fox Chase Cancer Center , Philadelphia, PA , USA ; Cancer Biology, Drexel University College of Medicine , Philadelphia, PA , USA
| | - Louisa Eckman
- Program in Molecular Therapeutics, Fox Chase Cancer Center , Philadelphia, PA , USA
| | - Meghan C Kopp
- Cancer Biology, Drexel University College of Medicine , Philadelphia, PA , USA
| | - Harvey H Hensley
- Program in Molecular Therapeutics, Fox Chase Cancer Center , Philadelphia, PA , USA
| | - Brian L Egleston
- Program in Molecular Therapeutics, Fox Chase Cancer Center , Philadelphia, PA , USA
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center , Philadelphia, PA , USA
| |
Collapse
|
95
|
Phua SC, Lin YC, Inoue T. An intelligent nano-antenna: Primary cilium harnesses TRP channels to decode polymodal stimuli. Cell Calcium 2015; 58:415-22. [PMID: 25828566 PMCID: PMC4564334 DOI: 10.1016/j.ceca.2015.03.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/30/2022]
Abstract
The primary cilium is a solitary hair-like organelle on the cell surface that serves as an antenna sensing ever-changing environmental conditions. In this review, we will first recapitulate the molecular basis of the polymodal sensory function of the primary cilia, specifically focusing on transient receptor potential (TRP) channels that accumulate inside the organelle and conduct calcium ions (Ca(2+)). Each subfamily member, namely TRPP2 TRPP3, TRPC1 and TRPV4, is gated by multiple environmental factors, including chemical (receptor ligands, intracellular second messengers such as Ca(2+)), mechanical (fluid shear stress, hypo-osmotic swelling), or physical (temperature, voltage) stimuli. Both activity and heterodimer compositions of the TRP channels may be dynamically regulated for precise tuning to the varying dynamic ranges of the individual input stimuli. We will thus discuss the potential regulation of TRP channels by local second messengers. Despite its reported importance in embryonic patterning and tissue morphogenesis, the precise functional significance of the downstream Ca(2+) signals of the TRP channels remains unknown. We will close our review by featuring recent technological advances in visualizing and analyzing signal transduction inside the primary cilia, together with current perspectives illuminating the functional significance of intraciliary Ca(2+) signals.
Collapse
Affiliation(s)
- Siew Cheng Phua
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA.
| | - Yu-Chun Lin
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Takanari Inoue
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Precursory Research for Embryonic Science and Technology (PRESTO) Investigator, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan.
| |
Collapse
|
96
|
Nielsen BS, Malinda RR, Schmid FM, Pedersen SF, Christensen ST, Pedersen LB. PDGFRβ and oncogenic mutant PDGFRα D842V promote disassembly of primary cilia through a PLCγ- and AURKA-dependent mechanism. J Cell Sci 2015; 128:3543-9. [PMID: 26290382 DOI: 10.1242/jcs.173559] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/17/2015] [Indexed: 01/04/2023] Open
Abstract
Primary cilia are microtubule-based sensory organelles projecting from most quiescent mammalian cells, which disassemble in cells cultured in serum-deprived conditions upon re-addition of serum or growth factors. Platelet-derived growth factors (PDGF) are implicated in deciliation, but the specific receptor isoforms and mechanisms involved are unclear. We report that PDGFRβ promotes deciliation in cultured cells and provide evidence implicating PLCγ and intracellular Ca(2+) release in this process. Activation of wild-type PDGFRα alone did not elicit deciliation. However, expression of constitutively active PDGFRα D842V mutant receptor, which potently activates PLCγ (also known as PLCG1), caused significant deciliation, and this phenotype was rescued by inhibiting PDGFRα D842V kinase activity or AURKA. We propose that PDGFRβ and PDGFRα D842V promote deciliation through PLCγ-mediated Ca(2+) release from intracellular stores, causing activation of calmodulin and AURKA-triggered deciliation.
Collapse
Affiliation(s)
- Brian S Nielsen
- Department of Biology, Section of Cell and Developmental Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, Copenhagen OE DK-2100, Denmark
| | - Raj R Malinda
- Department of Biology, Section of Cell and Developmental Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, Copenhagen OE DK-2100, Denmark
| | - Fabian M Schmid
- Department of Biology, Section of Cell and Developmental Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, Copenhagen OE DK-2100, Denmark
| | - Stine F Pedersen
- Department of Biology, Section of Cell and Developmental Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, Copenhagen OE DK-2100, Denmark
| | - Søren T Christensen
- Department of Biology, Section of Cell and Developmental Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, Copenhagen OE DK-2100, Denmark
| | - Lotte B Pedersen
- Department of Biology, Section of Cell and Developmental Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, Copenhagen OE DK-2100, Denmark
| |
Collapse
|
97
|
Ran J, Yang Y, Li D, Liu M, Zhou J. Deacetylation of α-tubulin and cortactin is required for HDAC6 to trigger ciliary disassembly. Sci Rep 2015; 5:12917. [PMID: 26246421 PMCID: PMC4526867 DOI: 10.1038/srep12917] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/14/2015] [Indexed: 12/21/2022] Open
Abstract
Cilia play important roles in sensing extracellular signals and directing fluid flow. Ciliary dysfunction is associated with a variety of diseases known as ciliopathies. Histone deacetylase 6 (HDAC6) has recently emerged as a major driver of ciliary disassembly, but little is known about the downstream players. Here we provide the first evidence that HDAC6-mediated deacetylation of α-tubulin and cortactin is critical for its induction of ciliary disassembly. HDAC6 is localized in the cytoplasm and enriched at the centrosome and basal body. Overexpression of HDAC6 decreases the levels of acetylated α-tubulin and cortactin without affecting the expression or localization of known ciliary regulators. We also find that overexpression of α-tubulin or cortactin or their acetylation-deficient mutants enhances the ability of HDAC6 to induce ciliary disassembly. In addition, acetylation-mimicking mutants of α-tubulin and cortactin counteract HDAC6-induced ciliary disassembly. Furthermore, HDAC6 stimulates actin polymerization, and inhibition of actin polymerization abolishes the activity of HDAC6 to trigger ciliary disassembly. These findings provide mechanistic insight into the ciliary role of HDAC6 and underscore the importance of reversible acetylation in regulating ciliary homeostasis.
Collapse
Affiliation(s)
- Jie Ran
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yunfan Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Min Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
98
|
Roosing S, Hofree M, Kim S, Scott E, Copeland B, Romani M, Silhavy JL, Rosti RO, Schroth J, Mazza T, Miccinilli E, Zaki MS, Swoboda KJ, Milisa-Drautz J, Dobyns WB, Mikati MA, İncecik F, Azam M, Borgatti R, Romaniello R, Boustany RM, Clericuzio CL, D'Arrigo S, Strømme P, Boltshauser E, Stanzial F, Mirabelli-Badenier M, Moroni I, Bertini E, Emma F, Steinlin M, Hildebrandt F, Johnson CA, Freilinger M, Vaux KK, Gabriel SB, Aza-Blanc P, Heynen-Genel S, Ideker T, Dynlacht BD, Lee JE, Valente EM, Kim J, Gleeson JG. Functional genome-wide siRNA screen identifies KIAA0586 as mutated in Joubert syndrome. eLife 2015; 4:e06602. [PMID: 26026149 PMCID: PMC4477441 DOI: 10.7554/elife.06602] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/28/2015] [Indexed: 12/14/2022] Open
Abstract
Defective primary ciliogenesis or cilium stability forms the basis of human ciliopathies, including Joubert syndrome (JS), with defective cerebellar vermis development. We performed a high-content genome-wide small interfering RNA (siRNA) screen to identify genes regulating ciliogenesis as candidates for JS. We analyzed results with a supervised-learning approach, using SYSCILIA gold standard, Cildb3.0, a centriole siRNA screen and the GTex project, identifying 591 likely candidates. Intersection of this data with whole exome results from 145 individuals with unexplained JS identified six families with predominantly compound heterozygous mutations in KIAA0586. A c.428del base deletion in 0.1% of the general population was found in trans with a second mutation in an additional set of 9 of 163 unexplained JS patients. KIAA0586 is an orthologue of chick Talpid3, required for ciliogenesis and Sonic hedgehog signaling. Our results uncover a relatively high frequency cause for JS and contribute a list of candidates for future gene discoveries in ciliopathies. DOI:http://dx.doi.org/10.7554/eLife.06602.001 Joubert syndrome is a rare disorder that affects the brain and causes physical, mental, and sometimes visual impairments. In individuals with this condition, two parts of the brain called the cerebellar vermis and the brainstem do not develop properly. This is thought to be due to defects in the development and maintenance of tiny hair-like structures called cilia, which are found on the surface of cells. Currently, mutations in 25 different genes are known to be able to cause Joubert syndrome. However, these mutations only account for around 50% of the cases that have been studied, and the ‘unexplained’ cases suggest that mutations in other genes may also cause the disease. Here, Roosing et al. used a technique called a ‘genome-wide siRNA screen’ to identify other genes regulating the formation of cilia that might also be connected with Joubert syndrome. This approach identified almost 600 candidate genes. The data from the screen were combined with gene sequence data from 145 individuals with unexplained Joubert syndrome. Roosing et al. found that individuals with Joubert syndrome from 15 different families had mutations in a gene called KIAA0586. In chickens and mice, this gene—known as Talpid3—is required for the formation of cilia. Roosing et al.'s findings reveal a new gene that is involved in Joubert syndrome and also provides a list of candidate genes for future studies of other conditions caused by defects in the formation of cilia. The next challenges are to find out what causes the remaining unexplained cases of the disease and to understand what roles the genes identified in this study play in cilia. DOI:http://dx.doi.org/10.7554/eLife.06602.002
Collapse
Affiliation(s)
- Susanne Roosing
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Matan Hofree
- Department of Computer Science and Engineering, University of California, San Diego, San Diego, United States
| | - Sehyun Kim
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, United States
| | - Eric Scott
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Brett Copeland
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Marta Romani
- IRCCS Casa Sollievo della Sofferenza, Mendel Institute, San Giovanni Rotondo, Italy
| | - Jennifer L Silhavy
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Rasim O Rosti
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Jana Schroth
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Tommaso Mazza
- IRCCS Casa Sollievo della Sofferenza, Mendel Institute, San Giovanni Rotondo, Italy
| | - Elide Miccinilli
- IRCCS Casa Sollievo della Sofferenza, Mendel Institute, San Giovanni Rotondo, Italy
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo, Egypt
| | - Kathryn J Swoboda
- Departments of Neurology and Pediatrics, University of Utah School of Medicine, Salt Lake City, United States
| | - Joanne Milisa-Drautz
- Department of Pediatric Genetics, University of New Mexico, Albuquerque, United States
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Hospital, Seattle, United States
| | - Mohamed A Mikati
- Division of Pediatric Neurology, Department of Pediatrics, Duke Institute for Brain Sciences, Duke University Medical Center, Durham, United States
| | - Faruk İncecik
- Department of Pediatric Neurology, Cukurova University Medical Faculty, Balcali, Turkey
| | - Matloob Azam
- Department of Pediatrics and Child Neurology, Wah Medical College, Wah Cantt, Pakistan
| | - Renato Borgatti
- Neuropsychiatry and Neurorehabilitation Unit, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Romina Romaniello
- Neuropsychiatry and Neurorehabilitation Unit, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Rose-Mary Boustany
- Departments of Pediatrics, Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Carol L Clericuzio
- Division of Genetics/Dysmorphology, Department Pediatrics, University of New Mexico, Albuquerque, United States
| | - Stefano D'Arrigo
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Petter Strømme
- Women and Children's Division, Oslo University Hospital, Oslo, Norway
| | - Eugen Boltshauser
- Department of Pediatric Neurology, University Children's Hospital, Zurich, Switzerland
| | - Franco Stanzial
- Department of Pediatrics, Genetic Counselling Service, Regional Hospital of Bolzano, Bolzano, Italy
| | - Marisol Mirabelli-Badenier
- Child Neuropsychiatry Unit, Department of Neurosciences and Rehabilitation, Istituto G. Gaslini, Genoa, Italy
| | - Isabella Moroni
- Unit of Child Neurology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Francesco Emma
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Colin A Johnson
- Section of Ophthalmology and Neurosciences, Wellcome Trust Brenner Building, Leeds Institute of Molecular Medicine, University of Leeds, St. James's University Hospital, Leeds, United Kingdom
| | - Michael Freilinger
- Neuropediatric group, Department of Paediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Keith K Vaux
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Stacey B Gabriel
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, United States
| | - Pedro Aza-Blanc
- High Content Screening Systems, Sanford-Burnham Institute, La Jolla, United States
| | - Susanne Heynen-Genel
- High Content Screening Systems, Sanford-Burnham Institute, La Jolla, United States
| | - Trey Ideker
- Department of Computer Science and Engineering, University of California, San Diego, San Diego, United States
| | - Brian D Dynlacht
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, United States
| | - Ji Eun Lee
- Samsung Genome Institute, Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Enza Maria Valente
- IRCCS Casa Sollievo della Sofferenza, Mendel Institute, San Giovanni Rotondo, Italy
| | - Joon Kim
- Korea Advanced Institute of Science and Technology, School of Medical Science and Engineering, Daejeon, Republic of Korea
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, New York Genome Center, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| |
Collapse
|
99
|
Shagisultanova E, Gaponova AV, Gabbasov R, Nicolas E, Golemis EA. Preclinical and clinical studies of the NEDD9 scaffold protein in cancer and other diseases. Gene 2015; 567:1-11. [PMID: 25967390 DOI: 10.1016/j.gene.2015.04.086] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/20/2015] [Accepted: 04/21/2015] [Indexed: 12/17/2022]
Abstract
Cancer progression requires a significant reprogramming of cellular signaling to support the essential tumor-specific processes that include hyperproliferation, invasion (for solid tumors) and survival of metastatic colonies. NEDD9 (also known as CasL and HEF1) encodes a multi-domain scaffolding protein that assembles signaling complexes regulating multiple cellular processes relevant to cancer. These include responsiveness to signals emanating from the T and B cell receptors, integrins, chemokine receptors, and receptor tyrosine kinases, as well as cytoplasmic oncogenes such as BCR-ABL and FAK- and SRC-family kinases. Downstream, NEDD9 regulation of partners including CRKL, WAVE, PI3K/AKT, ERK, E-cadherin, Aurora-A (AURKA), HDAC6, and others allow NEDD9 to influence functions as pleiotropic as migration, invasion, survival, ciliary resorption, and mitosis. In this review, we summarize a growing body of preclinical and clinical data that indicate that while NEDD9 is itself non-oncogenic, changes in expression of NEDD9 (most commonly elevation of expression) are common features of tumors, and directly impact tumor aggressiveness, metastasis, and response to at least some targeted agents inhibiting NEDD9-interacting proteins. These data strongly support the relevance of further development of NEDD9 as a biomarker for therapeutic resistance. Finally, we briefly discuss emerging evidence supporting involvement of NEDD9 in additional pathological conditions, including stroke and polycystic kidney disease.
Collapse
Affiliation(s)
- Elena Shagisultanova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Anna V Gaponova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Rashid Gabbasov
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Genetics, Kazan Federal University (Volga Region), Kazan, Tatarstan, Russia
| | - Emmanuelle Nicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
100
|
Gottardo M, Callaini G, Riparbelli MG. Aurora A inhibition by MNL8054 promotes centriole elongation during Drosophila male meiosis. Cell Cycle 2015; 14:2844-52. [PMID: 25785740 DOI: 10.1080/15384101.2015.1026488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Aurora A kinase plays an important role in several aspects of cell division, including centrosome maturation and separation, a crucial step for the correct organization of the bipolar spindle. Although it has long been showed that this kinase accumulates at the centrosome throughout mitosis its precise contribution to centriole biogenesis and structure has until now not been reported. It is not surprising that so little is known, due to the small size of somatic centrioles, where only dramatic structural changes may be identified by careful electron microscopy analysis. Conversely, centrioles of Drosophila primary spermatocytes increase tenfold in length during the first prophase, thus making any change easily detectable. Therefore, we examined the consequence of the pharmacological inhibition of Aurora A by MLN8054 on centriole biogenesis during early Drosophila gametogenesis. Here, we show that depletion of this kinase results in longer centrioles, mainly during transition from prophase to prometaphase of the first meiosis. We also found abnormal ciliogenesis characterized by irregularly growing axonemal doublets. Our results represent the first documentation of a potential requirement of Aurora A in centriole integrity and elongation.
Collapse
Affiliation(s)
- Marco Gottardo
- a Department of Life Sciences ; University of Siena ; Siena , Italy
| | | | | |
Collapse
|