1
|
Wang ZH, Du WW, Qian FY, Hou HY, Le Deng J, Ren XR, Pan Y, Pan Y, Lee SC, Hu HM, Zhao JZ. Homocapsaicin II induce ferroptosis in colorectal cancer cells via cholesterol-centrosome amplification-multipolarity axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119894. [PMID: 40319933 DOI: 10.1016/j.jep.2025.119894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Homocapsaicin II (Hp II), a natural product abundantly found in Capsicum annuum L., is a structural analog of capsaicin. Traditionally, capsaicin is utilized for managing digestive disorders and analgesic applications. Although emerging evidence suggests anti-cancer properties of capsaicin and its analogs, the specific anti-tumor properties and mechanisms of Hp II remain completely unexplored. Our study demonstrates that Hp II suppresses colorectal cancer progression via ferroptosis induction, both in vitro and in vivo. MATERIALS AND METHODS Anti-cancer activity of Hp II was assessed using colorectal cancer cell lines (in vitro) and subcutaneous tumor xenografts in nude mice (in vivo). Potential protein targets were identified through bioinformatic profiling, with KIF11 validated as a direct binding partner via epoxy-activated sepharose 6B-Hp II pull-down assays and microscale thermophoresis (MST). Ferroptotic mechanisms were dissected using molecular and cellular approaches, including intercellular free Fe2+ and lipid peroxidation quantification. RESULTS Hp II was found to target KIF11 in this study. It bound to KIF11 and stabilized it by reducing its ubiquitination. Increased levels of KIF11 promoted cholesterol production, leading to centrosome amplification (CA). Additionally, KIF11 played a role in centrosome separation, contributing to multipolarity. Ultimately, Cholesterol-induced CA and KIF11-mediated multipolarity synergy led to ferroptosis. This was supported by elevated levels of free Fe2+ and lipid peroxidation, a decreased GSH/GSSG ratio, and changes in the protein levels of ferroptosis markers ACSL5 and STEAP3 (upregulated) as well as FTH1 and GPX4 (downregulated). CONCLUSION In summary, Hp II acts as a dual-pathway inducer targeting both cholesterol-driven CA and KIF11-mediated centrosome separation to trigger ferroptosis. These findings position Hp II-KIF11 as a metabolic-mitotic ferroptosis regulator, connecting cholesterol metabolism, centrosome dynamics, and oxidative cell death.
Collapse
Affiliation(s)
- Zi Han Wang
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Wei Wei Du
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, PR China
| | - Fei Yu Qian
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Hao Yu Hou
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Jia Le Deng
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Xuan Rui Ren
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Yi Pan
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Yan Pan
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Shao Chin Lee
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Hong Mei Hu
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China
| | - Ji Zhong Zhao
- Lab of Cell Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, PR China.
| |
Collapse
|
2
|
Zhang Y, He M, Pan J. Axonemal microtubule dynamics in the assembly and disassembly of cilia. Biochem Soc Trans 2025; 53:BST20240688. [PMID: 39889304 DOI: 10.1042/bst20240688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 02/02/2025]
Abstract
Cilia and eukaryotic flagella (exchangeable terms) function in cell motility and signaling, which are pivotal for development and physiology. Cilia dysfunction can lead to ciliopathies. Cilia are usually assembled in quiescent and/or differentiated cells and undergo disassembly when cells enter cell cycle or in response to environmental stresses. Cilia contain a microtubule-based structure termed axoneme that comprises nine outer doublet microtubules with or without a pair of central microtubules, which is ensheathed by the ciliary membrane. Regulation of the axonemal microtubule dynamics is tightly associated with ciliary assembly and disassembly. In this short review, we discuss recent findings on the regulation of axonemal microtubules by microtubule-binding proteins and microtubule modulating kinesins during ciliary assembly and disassembly.
Collapse
Affiliation(s)
- Yi Zhang
- MOE Key Laboratory of Protein Sciences,State Key Laboratory of Complex, Severe, and Rare Diseases, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Mu He
- School of Biomedical Sciences, The University of Hong Kong, Hongkong, China
| | - Junmin Pan
- MOE Key Laboratory of Protein Sciences,State Key Laboratory of Complex, Severe, and Rare Diseases, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| |
Collapse
|
3
|
Wang Z, Wang W, Liu S, Yang F, Liu X, Hua S, Zhu L, Xu A, Hill DL, Wang D, Jiang K, Lippincott-Schwartz J, Liu X, Yao X. CSPP1 stabilizes microtubules by capping both plus and minus ends. J Mol Cell Biol 2024; 16:mjae007. [PMID: 38389254 PMCID: PMC11285173 DOI: 10.1093/jmcb/mjae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/11/2023] [Accepted: 02/21/2024] [Indexed: 02/24/2024] Open
Abstract
Although the dynamic instability of microtubules (MTs) is fundamental to many cellular functions, quiescent MTs with unattached free distal ends are commonly present and play important roles in various events to power cellular dynamics. However, how these free MT tips are stabilized remains poorly understood. Here, we report that centrosome and spindle pole protein 1 (CSPP1) caps and stabilizes both plus and minus ends of static MTs. Real-time imaging of laser-ablated MTs in live cells showed deposition of CSPP1 at the newly generated MT ends, whose dynamic instability was concomitantly suppressed. Consistently, MT ends in CSPP1-overexpressing cells were hyper-stabilized, while those in CSPP1-depleted cells were much more dynamic. This CSPP1-elicited stabilization of MTs was demonstrated to be achieved by suppressing intrinsic MT catastrophe and restricting polymerization. Importantly, CSPP1-bound MTs were resistant to mitotic centromere-associated kinesin-mediated depolymerization. These findings delineate a previously uncharacterized CSPP1 activity that integrates MT end capping to orchestrate quiescent MTs.
Collapse
Affiliation(s)
- Zhikai Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Wenwen Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Shuaiyu Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Xu Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Shasha Hua
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Lijuan Zhu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Aoqing Xu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Donald L Hill
- Comprehensive Cancer Center, University of Alabama, Birmingham, AL 35233, USA
| | - Dongmei Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Kai Jiang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | | | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| |
Collapse
|
4
|
Deretic J, Odabasi E, Firat-Karalar EN. The multifaceted roles of microtubule-associated proteins in the primary cilium and ciliopathies. J Cell Sci 2023; 136:jcs261148. [PMID: 38095645 DOI: 10.1242/jcs.261148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
The primary cilium is a conserved microtubule-based organelle that is critical for transducing developmental, sensory and homeostatic signaling pathways. It comprises an axoneme with nine parallel doublet microtubules extending from the basal body, surrounded by the ciliary membrane. The axoneme exhibits remarkable stability, serving as the skeleton of the cilium in order to maintain its shape and provide tracks to ciliary trafficking complexes. Although ciliary trafficking and signaling have been exhaustively characterized over the years, less is known about the unique structural and functional complexities of the axoneme. Recent work has yielded new insights into the mechanisms by which the axoneme is built with its proper length and architecture, particularly regarding the activity of microtubule-associated proteins (MAPs). In this Review, we first summarize current knowledge about the architecture, composition and specialized compartments of the primary cilium. Next, we discuss the mechanistic underpinnings of how a functional cilium is assembled, maintained and disassembled through the regulation of its axonemal microtubules. We conclude by examining the diverse localizations and functions of ciliary MAPs for the pathobiology of ciliary diseases.
Collapse
Affiliation(s)
- Jovana Deretic
- Department of Molecular Biology and Genetics, Koç University, Istanbul 34450, Turkey
| | - Ezgi Odabasi
- Department of Molecular Biology and Genetics, Koç University, Istanbul 34450, Turkey
| | - Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Koç University, Istanbul 34450, Turkey
- School of Medicine, Koç University, Istanbul 34450, Turkey
| |
Collapse
|
5
|
Romeiro Motta M, Biswas S, Schaedel L. Beyond uniformity: Exploring the heterogeneous and dynamic nature of the microtubule lattice. Eur J Cell Biol 2023; 102:151370. [PMID: 37922811 DOI: 10.1016/j.ejcb.2023.151370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
A fair amount of research on microtubules since their discovery in 1963 has focused on their dynamic tips. In contrast, the microtubule lattice was long believed to be highly regular and static, and consequently received far less attention. Yet, as it turned out, the microtubule lattice is neither as regular, nor as static as previously believed: structural studies uncovered the remarkable wealth of different conformations the lattice can accommodate. In the last decade, the microtubule lattice was shown to be labile and to spontaneously undergo renovation, a phenomenon that is intimately linked to structural defects and was called "microtubule self-repair". Following this breakthrough discovery, further recent research provided a deeper understanding of the lattice self-repair mechanism, which we review here. Instrumental to these discoveries were in vitro microtubule reconstitution assays, in which microtubules are grown from the minimal components required for their dynamics. In this review, we propose a shift from the term "lattice self-repair" to "lattice dynamics", since this phenomenon is an inherent property of microtubules and can happen without microtubule damage. We focus on how in vitro microtubule reconstitution assays helped us learn (1) which types of structural variations microtubules display, (2) how these structural variations influence lattice dynamics and microtubule damage caused by mechanical stress, (3) how lattice dynamics impact tip dynamics, and (4) how microtubule-associated proteins (MAPs) can play a role in structuring the lattice. Finally, we discuss the unanswered questions about lattice dynamics and how technical advances will help us tackle these questions.
Collapse
Affiliation(s)
- Mariana Romeiro Motta
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany; Laboratoire Reproduction et Développement des Plantes, Université de Lyon, École normale supérieure de Lyon, Lyon 69364, France
| | - Subham Biswas
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany
| | - Laura Schaedel
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany.
| |
Collapse
|
6
|
Westphal D, Meinhardt M, Grützmann K, Schöne L, Steininger J, Neuhaus LT, Wiegel M, Schrimpf D, Aust DE, Schröck E, Baretton GB, Beissert S, Juratli TA, Schackert GG, Gravemeyer J, Becker JC, von Deimling A, Koelsche C, Klink B, Meier F, Schulz A, Muders MH, Seifert M. Identification of Epigenetically Regulated Genes Distinguishing Intracranial from Extracranial Melanoma Metastases. J Invest Dermatol 2023; 143:1233-1245.e17. [PMID: 36716920 DOI: 10.1016/j.jid.2023.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/13/2022] [Accepted: 01/09/2023] [Indexed: 01/29/2023]
Abstract
Despite remarkable advances in treating patients with metastatic melanoma, the management of melanoma brain metastases remains challenging. Recent evidence suggests that epigenetic reprogramming is an important mechanism for the adaptation of melanoma cells to the brain environment. In this study, the methylomes and transcriptomes of a cohort of matched melanoma metastases were evaluated by integrated omics data analysis. The identified 38 candidate genes displayed distinct promoter methylation and corresponding gene expression changes in intracranial compared with extracranial metastases. The 11 most promising genes were validated on protein level in both tumor and surrounding normal tissue using immunohistochemistry. In accordance with the underlying promoter methylation and gene expression changes, a significantly different protein expression was confirmed for STK10, PDXK, WDR24, CSSP1, NMB, RASL11B, phosphorylated PRKCZ, PRKCZ, and phosphorylated GRB10 in the intracranial metastases. The observed changes imply a distinct intracranial phenotype with increased protein kinase B phosphorylation and a higher frequency of proliferating cells. Knockdown of PRKCZ or GRB10 altered the expression of phosphorylated protein kinase B and decreased the viability of a brain-specific melanoma cell line. In summary, epigenetically regulated cancer-relevant alterations were identified that provide insights into the molecular mechanisms that discriminate brain metastases from other organ metastases, which could be exploited by targeting the affected signaling pathways.
Collapse
Affiliation(s)
- Dana Westphal
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.
| | - Matthias Meinhardt
- Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Konrad Grützmann
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; Institute for Medical Informatics and Biometry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Lisa Schöne
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute for Medical Informatics and Biometry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Julian Steininger
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Lena T Neuhaus
- Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Miriam Wiegel
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Daniel Schrimpf
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela E Aust
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; BioBank Dresden (BBD), Tumor and Normal Tissue Bank (TNTB), National Center for Tumor Diseases (NCT/UCC), University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Evelin Schröck
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Gustavo B Baretton
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; BioBank Dresden (BBD), Tumor and Normal Tissue Bank (TNTB), National Center for Tumor Diseases (NCT/UCC), University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Tareq A Juratli
- Department of Neurosurgery, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Gabriele G Schackert
- Department of Neurosurgery, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jan Gravemeyer
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), Partner Site Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), Partner Site Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Koelsche
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Barbara Klink
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Skin Cancer Center at the University Cancer Center and National Center for Tumor Diseases, Dresden, Germany
| | - Alexander Schulz
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Michael H Muders
- Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Michael Seifert
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute for Medical Informatics and Biometry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
7
|
van den Berg CM, Volkov VA, Schnorrenberg S, Huang Z, Stecker KE, Grigoriev I, Gilani S, Frikstad KAM, Patzke S, Zimmermann T, Dogterom M, Akhmanova A. CSPP1 stabilizes growing microtubule ends and damaged lattices from the luminal side. J Cell Biol 2023; 222:213861. [PMID: 36752787 PMCID: PMC9948759 DOI: 10.1083/jcb.202208062] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/14/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
Microtubules are dynamic cytoskeletal polymers, and their organization and stability are tightly regulated by numerous cellular factors. While regulatory proteins controlling the formation of interphase microtubule arrays and mitotic spindles have been extensively studied, the biochemical mechanisms responsible for generating stable microtubule cores of centrioles and cilia are poorly understood. Here, we used in vitro reconstitution assays to investigate microtubule-stabilizing properties of CSPP1, a centrosome and cilia-associated protein mutated in the neurodevelopmental ciliopathy Joubert syndrome. We found that CSPP1 preferentially binds to polymerizing microtubule ends that grow slowly or undergo growth perturbations and, in this way, resembles microtubule-stabilizing compounds such as taxanes. Fluorescence microscopy and cryo-electron tomography showed that CSPP1 is deposited in the microtubule lumen and inhibits microtubule growth and shortening through two separate domains. CSPP1 also specifically recognizes and stabilizes damaged microtubule lattices. These data help to explain how CSPP1 regulates the elongation and stability of ciliary axonemes and other microtubule-based structures.
Collapse
Affiliation(s)
- Cyntha M. van den Berg
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Vladimir A. Volkov
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands,https://ror.org/02e2c7k09Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands
| | | | - Ziqiang Huang
- EMBL Imaging Centre, EMBL-Heidelberg, Heidelberg, Germany
| | - Kelly E. Stecker
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands,Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Ilya Grigoriev
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Sania Gilani
- https://ror.org/00j9c2840Department of Radiation Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway,Department of Molecular Cell Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kari-Anne M. Frikstad
- https://ror.org/00j9c2840Department of Radiation Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sebastian Patzke
- https://ror.org/00j9c2840Department of Radiation Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | | - Marileen Dogterom
- https://ror.org/02e2c7k09Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands
| | - Anna Akhmanova
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands,Correspondence to Anna Akhmanova:
| |
Collapse
|
8
|
Hua K, Ferland RJ. Fixation methods and immunolabeling for cilia proteins in ciliary and extraciliary locations. Methods Cell Biol 2023; 176:43-57. [PMID: 37164542 DOI: 10.1016/bs.mcb.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Primary cilia are complex organelles, usually singularly located on cell surfaces that are now known to be important for signaling and whose defect is implicated in a category of developmental diseases known as ciliopathies. They are composed of a microtubule axoneme and contain a cilia membrane that is unique and distinct from the plasma membrane. Primary cilia also have their own transport system termed the intraflagellar transport (IFT) system that allows for proteins to be trafficked along the microtubule axoneme in either an anterograde or retrograde manner. Proteins that localize to the primary cilium are referred to as ciliary proteins and have been implicated directly or indirectly in ciliogenesis or ciliary function. It is now recognized that cilia proteins can localize to different compartments of cilia, but can also localize to multiple sites outside of cilia (extraciliary sites). This complexity results in a need for a better understanding of ciliary protein fixation and immunolabeling protocols, as different methods are required to visualize different cilia proteins and reveal novel or unique localizations. Here, we detail a variety of fixation methods and their effects on ciliary protein immunolabeling.
Collapse
Affiliation(s)
- Kiet Hua
- Montefiore Medical Center, Albert Einstein College of Medicine, Department of Neurology, Bronx, NY, United States.
| | - Russell J Ferland
- University of New England, College of Osteopathic Medicine, Department of Biomedical Sciences, Biddeford, ME, United States.
| |
Collapse
|
9
|
Primary Cilia Influence Progenitor Function during Cortical Development. Cells 2022; 11:cells11182895. [PMID: 36139475 PMCID: PMC9496791 DOI: 10.3390/cells11182895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/29/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Corticogenesis is an intricate process controlled temporally and spatially by many intrinsic and extrinsic factors. Alterations during this important process can lead to severe cortical malformations. Apical neuronal progenitors are essential cells able to self-amplify and also generate basal progenitors and/or neurons. Apical radial glia (aRG) are neuronal progenitors with a unique morphology. They have a long basal process acting as a support for neuronal migration to the cortical plate and a short apical process directed towards the ventricle from which protrudes a primary cilium. This antenna-like structure allows aRG to sense cues from the embryonic cerebrospinal fluid (eCSF) helping to maintain cell shape and to influence several key functions of aRG such as proliferation and differentiation. Centrosomes, major microtubule organising centres, are crucial for cilia formation. In this review, we focus on how primary cilia influence aRG function during cortical development and pathologies which may arise due to defects in this structure. Reporting and cataloguing a number of ciliary mutant models, we discuss the importance of primary cilia for aRG function and cortical development.
Collapse
|
10
|
Wang W, Zhang J, Wang Y, Xu Y, Zhang S. Identifies microtubule-binding protein CSPP1 as a novel cancer biomarker associated with ferroptosis and tumor microenvironment. Comput Struct Biotechnol J 2022; 20:3322-3335. [PMID: 35832625 PMCID: PMC9253833 DOI: 10.1016/j.csbj.2022.06.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 12/02/2022] Open
Abstract
Centrosome and spindle pole-associated protein (CSPP1) is a centrosome and microtubule-binding protein that plays a role in cell cycle-dependent cytoskeleton organization and cilia formation. Previous studies have suggested that CSPP1 plays a role in tumorigenesis; however, no pan-cancer analysis has been performed. This study systematically investigates the expression of CSPP1 and its potential clinical outcomes associated with diagnosis, prognosis, and therapy. CSPP1 is widely present in tissues and cells and its aberrant expression serves as a diagnostic biomarker for cancer. CSPP1 dysregulation is driven by multi-dimensional mechanisms involving genetic alterations, DNA methylation, and miRNAs. Phosphorylation of CSPP1 at specific sites may play a role in tumorigenesis. In addition, CSPP1 correlates with clinical features and outcomes in multiple cancers. Take brain low-grade gliomas (LGG) with a poor prognosis as an example, functional enrichment analysis implies that CSPP1 may play a role in ferroptosis and tumor microenvironment (TME), including regulating epithelial-mesenchymal transition, stromal response, and immune response. Further analysis confirms that CSPP1 dysregulates ferroptosis in LGG and other cancers, making it possible for ferroptosis-based drugs to be used in the treatment of these cancers. Importantly, CSPP1-associated tumors are infiltrated in different TMEs, rendering immune checkpoint blockade therapy beneficial for these cancer patients. Our study is the first to demonstrate that CSPP1 is a potential diagnostic and prognostic biomarker associated with ferroptosis and TME, providing a new target for drug therapy and immunotherapy in specific cancers.
Collapse
Key Words
- ACC, adrenocortical carcinoma
- BP, biological pathways
- BRCA, breast invasive carcinoma
- Biomarker
- C-index, concordance index
- CAF, cancer-associated fibroblasts
- CC, cellular component
- CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma
- CHOL, cholangiocarcinoma
- CNA, copy number alteration
- COAD, colon adenocarcinoma
- CPTAC, Clinical Proteomic Tumor Analysis Consortium
- CSPP1
- CSPP1, centrosome and spindle pole-associated protein
- CTL, cytotoxic T lymphocyte
- DEGs, differentially expressed genes
- DLBC, diffuse large B-cell lymphoma
- DSS, disease-specific survival
- EMT, epithelial-mesenchymal transition
- ENCORI, Encyclopedia of RNA Interactomes
- ESCA, esophageal carcinoma
- FAG, ferroptosis-associated gene
- FDG, ferroptosis-driver gene
- FSG, ferroptosis-suppressor gene
- Ferroptosis
- GBM, glioblastoma multiforme
- GO, Gene Ontology
- GSEA, Gene Set Enrichment Analysis
- GSVA, gene set variation analysis
- GTEx, Genotype-Tissue Expression
- HNSC, head and neck squamous cell carcinoma
- ICB, immune checkpoint blockade
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- KICH, kidney chromophobe
- KIRC, renal clear cell carcinoma
- KM, Kaplan-Meier
- LAML, acute myeloid leukemia
- LGG, low-grade gliomas
- LIHC, liver hepatocellular carcinoma
- LUAD, lung adenocarcinoma
- LUSC, lung squamous cell carcinoma
- MF, molecular functions
- MHC, major histocompatibility complex
- MSI, microsatellite instability
- OS, overall survival
- OV, ovarian serous cystadenocarcinoma
- PAAD, pancreatic adenocarcinoma
- PFI, progression-free interval
- PFS, progression-free survival
- PRAD, prostate cancer
- Pan-cancer
- READ, rectum adenocarcinoma
- ROC, receiver operating characteristics
- SKCM, skin cutaneous melanoma
- TCGA, The Cancer Genome Atlas
- TGCT, testicular germ cell tumors, STAD, stomach adenocarcinoma
- THCA, thyroid cancer
- THYM, thymoma
- TIDE, Tumor Immune Dysfunction and Exclusion
- TIMER, Tumor Immune Estimation Resource
- TISIDB, Tumor-Immune System Interactions DataBase
- TMB, tumor mutation burden
- TME, tumor microenvironment
- Tumor microenvironment
- UCEC, endometrial cancer uterine corpus endometrial carcinoma
- UCS, uterine carcinosarcoma
Collapse
Affiliation(s)
- Wenwen Wang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, China
| | - Jingjing Zhang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuqing Wang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yasi Xu
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, China
| | - Shirong Zhang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Behbahanipour M, García-Pardo J, Ventura S. Decoding the role of coiled-coil motifs in human prion-like proteins. Prion 2021; 15:143-154. [PMID: 34428113 PMCID: PMC8386614 DOI: 10.1080/19336896.2021.1961569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 11/28/2022] Open
Abstract
Prions are self-propagating proteins that cause fatal neurodegenerative diseases in humans. However, increasing evidence suggests that eukaryotic cells exploit prion conformational conversion for functional purposes. A recent study delineated a group of twenty prion-like proteins in humans, characterized by the presence of low-complexity glutamine-rich sequences with overlapping coiled-coil (CCs) motifs. This is the case of Mediator complex subunit 15 (MED15), which is overexpressed in a wide range of human cancers. Biophysical studies demonstrated that the prion-like domain (PrLD) of MED15 forms homodimers in solution, sustained by CCs interactions. Furthermore, the same coiled-coil (CC) region plays a crucial role in the PrLD structural transition to a transmissible β-sheet amyloid state. In this review, we discuss the role of CCs motifs and their contribution to amyloid transitions in human prion-like domains (PrLDs), while providing a comprehensive overview of six predicted human prion-like proteins involved in transcription, gene expression, or DNA damage response and associated with human disease, whose PrLDs contain or overlap with CCs sequences. Finally, we try to rationalize how these molecular signatures might relate to both their function and involvement in disease.
Collapse
Affiliation(s)
- Molood Behbahanipour
- Institut De Biotecnologia I De Biomedicina (Ibb) and Departament De Bioquímica I Biologia Molecular, Universitat Autónoma De Barcelona, Barcelona, Spain
| | - Javier García-Pardo
- Institut De Biotecnologia I De Biomedicina (Ibb) and Departament De Bioquímica I Biologia Molecular, Universitat Autónoma De Barcelona, Barcelona, Spain
| | - Salvador Ventura
- Institut De Biotecnologia I De Biomedicina (Ibb) and Departament De Bioquímica I Biologia Molecular, Universitat Autónoma De Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Bauerly E, Akiyama T, Staber C, Yi K, Gibson MC. Impact of cilia-related genes on mitochondrial dynamics during Drosophila spermatogenesis. Dev Biol 2021; 482:17-27. [PMID: 34822845 DOI: 10.1016/j.ydbio.2021.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 11/28/2022]
Abstract
Spermatogenesis is a dynamic process of cellular differentiation that generates the mature spermatozoa required for reproduction. Errors that arise during this process can lead to sterility due to low sperm counts and malformed or immotile sperm. While it is estimated that 1 out of 7 human couples encounter infertility, the underlying cause of male infertility can only be identified in 50% of cases. Here, we describe and examine the genetic requirements for missing minor mitochondria (mmm), sterile affecting ciliogenesis (sac), and testes of unusual size (tous), three previously uncharacterized genes in Drosophila that are predicted to be components of the flagellar axoneme. Using Drosophila, we demonstrate that these genes are essential for male fertility and that loss of mmm, sac, or tous results in complete immotility of the sperm flagellum. Cytological examination uncovered additional roles for sac and tous during cytokinesis and transmission electron microscopy of developing spermatids in mmm, sac, and tous mutant animals revealed defects associated with mitochondria and the accessory microtubules required for the proper elongation of the mitochondria and flagella during ciliogenesis. This study highlights the complex interactions of cilia-related proteins within the cell body and advances our understanding of male infertility by uncovering novel mitochondrial defects during spermatogenesis.
Collapse
Affiliation(s)
| | - Takuya Akiyama
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Cynthia Staber
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Matthew C Gibson
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA; Department of Anatomy and Cell Biology, The University of Kansas School of Medicine, Kansas City, KS, 66160, USA.
| |
Collapse
|
13
|
Aquino Perez C, Burocziova M, Jenikova G, Macurek L. CK1-mediated phosphorylation of FAM110A promotes its interaction with mitotic spindle and controls chromosomal alignment. EMBO Rep 2021; 22:e51847. [PMID: 34080749 DOI: 10.15252/embr.202051847] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/14/2021] [Accepted: 05/05/2021] [Indexed: 01/10/2023] Open
Abstract
Progression through the cell cycle is driven by cyclin-dependent kinases that control gene expression, orchestration of mitotic spindle, and cell division. To identify new regulators of the cell cycle, we performed transcriptomic analysis of human non-transformed cells expressing a fluorescent ubiquitination-based cell cycle indicator and identified 701 transcripts differentially expressed in G1 and G2 cells. Family with sequence similarity 110 member A (FAM110A) protein is highly expressed in G2 cells and localized at mitotic spindle and spindle poles during mitosis. Depletion of FAM110A impairs chromosomal alignment, delays metaphase-to-anaphase transition, and affects spindle positioning. Using mass spectrometry and immunoprecipitation, we identified casein kinase I (CK1) in complex with FAM110A during mitosis. CK1 phosphorylates the C-terminal domain of FAM110A in vitro, and inhibition of CK1 reduces phosphorylation of mitotic FAM110A. Wild-type FAM110A, but not the FAM110A-S252-S255A mutant deficient in CK1 phosphorylation, rescues the chromosomal alignment, duration of mitosis, and orientation of the mitotic spindle after depletion of endogenous FAM110A. We propose that CK1 regulates chromosomal alignment by phosphorylating FAM110A and promoting its interaction with mitotic spindle.
Collapse
Affiliation(s)
- Cecilia Aquino Perez
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Monika Burocziova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Gabriela Jenikova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
14
|
Xi L, Liu Q, Zhang W, Luo L, Song J, Liu R, Wei S, Wang Y. Circular RNA circCSPP1 knockdown attenuates doxorubicin resistance and suppresses tumor progression of colorectal cancer via miR-944/FZD7 axis. Cancer Cell Int 2021; 21:153. [PMID: 33663510 PMCID: PMC7934234 DOI: 10.1186/s12935-021-01855-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) have been reported to play vital roles in colorectal cancer (CRC). However, only a few circRNAs have been experimentally validated and functionally described. In this research, we aimed to reveal the functional mechanism of circCSPP1 in CRC. METHODS 36 DOX sensitive and 36 resistant CRC cases participated in this study. The expression of circCSPP1, miR-944 and FZD7 were detected by quantitative real time polymerase chain reaction (qRT-PCR) and the protein levels of FZD7, MRP1, P-gp and LRP were detected by western blot. Cell proliferation, migration, invasion, and apoptosis were assessed by 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay, transwell assay, or flow cytometry analysis, respectively. The interaction between miR-944 and circCSPP1 or frizzled-7 (FZD7) was predicted by Starbase 3.0 and verified by the dual luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull down assay. Xenograft tumor assay was performed to examine the effect of circCSPP1 on tumor growth in vivo. RESULTS The expression of circCSPP1 and FZD7 was upregulated while miR-944 expression was downregulated in doxorubicin (DOX)-resistant CRC tissues and cells. CircCSPP1 knockdown significantly downregulated enhanced doxorubicin sensitivity, suppressed proliferation, migration, invasion, and induced apoptosis in DOX-resistant CRC cells. Interestingly, we found that circCSPP1 directly downregulated miR-944 expression and miR-944 decreased FZD7 level through targeting to 3' untranslated region (UTR) of FZD7. Furthermore, circCSPP1 mediated DOX-resistant CRC cell progression and doxorubicin sensitivity by regulating miR-944/FZD7 axis. Besides, circCSPP1 downregulation dramatically repressed CRC tumor growth in vivo. CONCLUSION Our data indicated that circCSPP1 knockdown inhibited DOX-resistant CRC cell growth and enhanced doxorubicin sensitivity by miR-944/FZD7 axis, providing a potential target for CRC therapy.
Collapse
Affiliation(s)
- Lanlan Xi
- Department of Surgery of Traditional Chinese Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Quanlin Liu
- Department of Colorectal Surgery, Zhengzhou Anorectal Hospital, No. 51, Longhai East Road, Zhengzhou, 450004, China.
| | - Wei Zhang
- Department of Colorectal Surgery, Zhengzhou Anorectal Hospital, No. 51, Longhai East Road, Zhengzhou, 450004, China
| | - Linshan Luo
- Department of Colorectal Surgery, Zhengzhou Anorectal Hospital, No. 51, Longhai East Road, Zhengzhou, 450004, China
| | - Jingfeng Song
- Department of Colorectal Surgery, Zhengzhou Anorectal Hospital, No. 51, Longhai East Road, Zhengzhou, 450004, China
| | - Ruitao Liu
- Department of Large Intestine, Zhengzhou Anorectal Hospital, Zhengzhou, China
| | - Shue Wei
- Department of Large Intestine, Zhengzhou Anorectal Hospital, Zhengzhou, China
| | - Yong Wang
- Department of Colorectal Surgery, Zhengzhou Anorectal Hospital, No. 51, Longhai East Road, Zhengzhou, 450004, China
| |
Collapse
|
15
|
A CEP104-CSPP1 Complex Is Required for Formation of Primary Cilia Competent in Hedgehog Signaling. Cell Rep 2020; 28:1907-1922.e6. [PMID: 31412255 PMCID: PMC6702141 DOI: 10.1016/j.celrep.2019.07.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 05/21/2019] [Accepted: 07/10/2019] [Indexed: 12/25/2022] Open
Abstract
CEP104 is an evolutionarily conserved centrosomal and ciliary tip protein. CEP104 loss-of-function mutations are reported in patients with Joubert syndrome, but their function in the etiology of ciliopathies is poorly understood. Here, we show that cep104 silencing in zebrafish causes cilia-related manifestations: shortened cilia in Kupffer’s vesicle, heart laterality, and cranial nerve development defects. We show that another Joubert syndrome-associated cilia tip protein, CSPP1, interacts with CEP104 at microtubules for the regulation of axoneme length. We demonstrate in human telomerase reverse transcriptase-immortalized retinal pigmented epithelium (hTERT-RPE1) cells that ciliary translocation of Smoothened in response to Hedgehog pathway stimulation is both CEP104 and CSPP1 dependent. However, CEP104 is not required for the ciliary recruitment of CSPP1, indicating that an intra-ciliary CEP104-CSPP1 complex controls axoneme length and Hedgehog signaling competence. Our in vivo and in vitro analyses of CEP104 define its interaction with CSPP1 as a requirement for the formation of Hedgehog signaling-competent cilia, defects that underlie Joubert syndrome. cep104-depleted zebrafish display shortened KV cilia and defective brain development CEP104 interacts with CSPP1 at the tip of the primary cilium to regulate cilia length CEP104 or CSPP1 loss in human cells leads to defective Hedgehog signaling Impaired signaling is linked to reduction of ciliary SMO but not ARL13B or INPP5E
Collapse
|
16
|
Yang X, Mei J, Wang H, Gu D, Ding J, Liu C. The emerging roles of circular RNAs in ovarian cancer. Cancer Cell Int 2020; 20:265. [PMID: 32587475 PMCID: PMC7313187 DOI: 10.1186/s12935-020-01367-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Circular RNA (circRNA) is a novel class of regulatory noncoding RNA (ncRNA) molecules with a unique covalently closed loop structure. Next-generation sequencing shows that thousands of circRNAs are widely and stably expressed in multiple eukaryotes. As novel regulatory ncRNAs, circRNAs possess several specific molecular functions, including regulating gene transcription and translation, acting as miRNA sponges, and interacting with functional proteins. Ovarian cancer (OvCa) is one of the most aggressive malignant diseases affecting the lives of thousands of women worldwide, and the majority of OvCa cases are diagnosed at advanced stages. Accumulating evidence has revealed the significant roles of circRNAs in the occurrence and progression of OvCa, indicating the function of circRNAs as promising biomarkers and their therapeutic relevance in this disease. This review aims to summarize the mechanisms by which circRNAs mediate OvCa progression as well as their diagnostic and prognostic values in OvCa.
Collapse
Affiliation(s)
- Xuejing Yang
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023 China
| | - Jie Mei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023 China
| | - Huiyu Wang
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023 China
| | - Dingyi Gu
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023 China
| | - Junli Ding
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023 China
| | - Chaoying Liu
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023 China
| |
Collapse
|
17
|
Cebeci AN, Zou M, BinEssa HA, Alzahrani AS, Al-Rijjal RA, Al-Enezi AF, Al-Mohanna FA, Cavalier E, Meyer BF, Shi Y. Mutation of SGK3, a Novel Regulator of Renal Phosphate Transport, Causes Autosomal Dominant Hypophosphatemic Rickets. J Clin Endocrinol Metab 2020; 105:5672651. [PMID: 31821448 DOI: 10.1210/clinem/dgz260] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/09/2019] [Indexed: 11/19/2022]
Abstract
CONTEXT Hypophosphatemic rickets (HR) is a group of rare hereditary renal phosphate wasting disorders caused by mutations in PHEX, FGF23, DMP1, ENPP1, CLCN5, SLC9A3R1, SLC34A1, or SLC34A3. OBJECTIVE A large kindred with 5 HR patients was recruited with dominant inheritance. The study was undertaken to investigate underlying genetic defects in HR patients. DESIGN Patients and their family members were initially analyzed for PHEX and FGF23 mutations using polymerase chain reaction sequencing and copy number analysis. Exome sequencing was subsequently performed to identify novel candidate genes. RESULTS PHEX and FGF23 mutations were not detected in the patients. No copy number variation was observed in the genome using CytoScan HD array analysis. Mutations in DMP1, ENPP1, CLCN5, SLC9A3R1, SLC34A1, or SLC34A3 were also not found by exome sequencing. A novel c.979-96 T>A mutation in the SGK3 gene was found to be strictly segregated in a heterozygous pattern in patients and was not present in normal family members. The mutation is located 1 bp downstream of a highly conserved adenosine branch point, resulted in exon 13 skipping and in-frame deletion of 29 amino acids, which is part of the protein kinase domain and contains a Thr-320 phosphorylation site that is required for its activation. Protein tertiary structure modelling showed significant structural change in the protein kinase domain following the deletion. CONCLUSIONS The c.979-96 T>A splice mutation in the SGK3 gene causes exon 13 skipping and deletion of 29 amino acids in the protein kinase domain. The SGK3 mutation may cause autosomal dominant HR.
Collapse
Affiliation(s)
- Ayşe Nurcan Cebeci
- Department of Pediatric Endocrinology, Istanbul Bilim University, Istanbul, Turkey
| | - Minjing Zou
- Department of Genetics, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Huda A BinEssa
- Department of Genetics, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Ali S Alzahrani
- Department of Medicine King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Roua A Al-Rijjal
- Department of Genetics, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Anwar F Al-Enezi
- Department of Genetics, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU de Liège, Liège, Belgium
| | - Brian F Meyer
- Department of Genetics, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Yufei Shi
- Department of Genetics, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
18
|
Sun Q, Yu R, Wang C, Yao J, Zhang L. Circular RNA circ-CSPP1 regulates CCNE2 to facilitate hepatocellular carcinoma cell growth via sponging miR-577. Cancer Cell Int 2020; 20:202. [PMID: 32514247 PMCID: PMC7260814 DOI: 10.1186/s12935-020-01287-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Circ-centro-some/spindle pole-associated protein (CSPP1) has been confirmed to be characterized in diverse human malignancies and its ectopic expression may regulate tumor progression and development. However, in hepatocellular carcinoma (HCC), its biological role, clinical significance and molecular mechanism are still unclear. METHODS Circ-CSPP1 expression and its prognostic values in HCC tissues were detected by qRT-PCR or in situ hybridization (ISH), and enriched by using Rnase R. The functional experiments (Circ-CSPP1 was overexpressed or knocked down) were performed in HCC cells. The HCC cell growth was analyzed by CCK-8 assay, transwell, wound healing and colony formation assays. The interation between circ-CSPP1 and miR-577/miR-577 and cyclin E2 (CCNE2) were determined by dual luciferase assay or RNA binding protein immunoprecipitation (RIP) assay. The RNA fluorescence in situ hybridization (FISH) assay was used to detect the subcellular distribution. Finally, an in vivo nude mouse tumor model was constructed. RESULTS In HCC patients and cells, circ-CSPP1 was aberrantly expressed, and its upregulation predicted poor prognosis, and closely correlated with tumor size and TNM stage. Circ-CSPP1 resisted RnaseR digestion, indicating it is a circular RNA structure. Moreover, overexpression of circ-CSPP1 promoted HCC cell viability, colony formation, migration, and invasion in vitro. Knockdown of circ-CSPP1 showed contrary results. Circ-CSPP1 acts as a miR-577 sponge and positively regulated the target of miR-577, CCNE2. Besides, miR-577 inhibitor rescued the suppressive effects of circ-CSPP1 knockdown on HCC cell growth, whereas was completely reversed by silencing of CCNE2. Finally, the in vivo experiments confirmed that circ-CSPP1 knockdown regulated xenograft tumor volume and downregulated CCNE2, p-Rb, E2F1 and c-myc expression. CONCLUSION These findings revealed that circ-CSPP1 contributed to HCC progression by positively regulating CCNE2 via miR-577, thus established its potential as new a prognostic and therapeutic marker for HCC patients.
Collapse
Affiliation(s)
- Qian Sun
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Dong Lu, Erqi District, Zhengzhou, Henan 450052 China
| | - Rui Yu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Dong Lu, Erqi District, Zhengzhou, Henan 450052 China
| | - Chunfeng Wang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Dong Lu, Erqi District, Zhengzhou, Henan 450052 China
| | - Jianning Yao
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Dong Lu, Erqi District, Zhengzhou, Henan 450052 China
| | - Lianfeng Zhang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Dong Lu, Erqi District, Zhengzhou, Henan 450052 China
| |
Collapse
|
19
|
Peng Y, Zhao W, Qu F, Jing J, Hu Y, Liu Y, Ding Z. Proteomic alterations underlie an association with teratozoospermia in obese mice sperm. Reprod Biol Endocrinol 2019; 17:82. [PMID: 31651332 PMCID: PMC6813985 DOI: 10.1186/s12958-019-0530-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/02/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Obesity is a worldwide crisis impairing human health. In this condition, declines in sperm quality stem from reductions in sperm concentration, motility and increase in sperm deformity. The mechanism underlying these alterations remains largely unknown. This study, determined if obesity-associated proteomic expression patterns in mice sperm parallel those in spermatozoa obtained from obese humans. METHODS An obese mouse model was established via feeding a high-fat diet (HFD). Histological analysis identified testicular morphology and a computer assisted semen analyzer (CASA) evaluated sperm parameters. Proteome analysis was performed using a label-free quantitative LC-MS/MS system. Western blot, immunohistochemical and immunofluorescent analyses characterized protein expression levels and localization in testis, sperm and clinical samples. RESULTS Bodyweight gains on the HFD induced hepatic steatosis. Declines in sperm motility accompanied sperm deformity development. Differential proteomic analysis identified reduced cytoskeletal proteins, centrosome and spindle pole associated protein 1 (CSPP1) and Centrin 1 (CETN1), in sperm from obese mice. In normal weight mice, both CSPP1 and CETN1 were localized in the spermatocytes and spermatids. Their expression was appreciable in the post-acrosomal region parallel to the microtubule tracks of the manchette structure in spermatids, which affects spermatid head shaping and morphological maintenance. Moreover, CSPP1 was localized in the head-tail coupling apparatus of the mature sperm, while CETN1 expression was delimited to the post-acrosomal region within the sperm head. Importantly, sperm CSPP1 and CETN1 abundance in both the overweight and obese males decreased in comparison with that in normal weight men. CONCLUSION These findings show that regionally distinct expression and localization of CETN1 and CSPP1 is strongly related to spermiogenesis and sperm morphology maintaining. Obesity is associated with declines in the CETN1 and CSPP1 abundance and compromise of both sperm morphology in mice and relevant clinical samples. This parallelism between altered protein expression in mice and humans suggests that these effects may contribute to poor sperm quality including increased deformity.
Collapse
Affiliation(s)
- Yuanhong Peng
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenzhen Zhao
- Department of Histology and Embryology, School of Basic Medical Science, Dali University, Dali, 671000, Yunnan, China
- Institute of Reproductive Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Fei Qu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia Jing
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanqin Hu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yue Liu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
20
|
Shabaninejad Z, Vafadar A, Movahedpour A, Ghasemi Y, Namdar A, Fathizadeh H, Pourhanifeh MH, Savardashtaki A, Mirzaei H. Circular RNAs in cancer: new insights into functions and implications in ovarian cancer. J Ovarian Res 2019; 12:84. [PMID: 31481095 PMCID: PMC6724287 DOI: 10.1186/s13048-019-0558-5] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of long non-coding RNAs (lncRNAs) which have a circular and closed loop structure. They are ubiquitous, stable, conserved and diverse RNA molecules with a range of activities such as translation and splicing regulation, which are able to interacting with RNA-binding proteins and specially miRNA sponge. The expression patterns of the circRNAs exhibited tissue specificity and also, step and stage specificity. Accumulating evidences approved the critical role of circular RNAs in many cancers such as ovarian cancer. Given that these molecules exert their effects through multiple cellular and molecular mechanisms (i.e., angiogenesis, apoptosis, growth, and metastasis) which are involved in cancer pathogenesis, circular RNAs, in particular, act by controlling cell proliferation in ovarian cancer, so that, it has been shown that the deregulation of these molecules is associated with initiation and progression of ovarian cancer. Therefore, they are attractive molecules which have introduced them as cancer biomarkers. Moreover, they could be used as new therapeutic candidates for developing novel treatment strategies. Here, for first time, we have provided a comprehensive review on the recent knowledge of circular RNAs and their pathological roles in the ovarian cancer.
Collapse
Affiliation(s)
- Zahra Shabaninejad
- Department of Nanobiotechnology, School of Basic Sciences, Tarbiat Modares University, Tehran, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afshin Namdar
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Hadis Fathizadeh
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
21
|
Gheiratmand L, Coyaud E, Gupta GD, Laurent EMN, Hasegan M, Prosser SL, Gonçalves J, Raught B, Pelletier L. Spatial and proteomic profiling reveals centrosome-independent features of centriolar satellites. EMBO J 2019; 38:e101109. [PMID: 31304627 PMCID: PMC6627244 DOI: 10.15252/embj.2018101109] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/19/2022] Open
Abstract
Centriolar satellites are small electron-dense granules that cluster in the vicinity of centrosomes. Satellites have been implicated in multiple critical cellular functions including centriole duplication, centrosome maturation, and ciliogenesis, but their precise composition and assembly properties have remained poorly explored. Here, we perform in vivo proximity-dependent biotin identification (BioID) on 22 human satellite proteins, to identify 2,113 high-confidence interactions among 660 unique polypeptides. Mining this network, we validate six additional satellite components. Analysis of the satellite interactome, combined with subdiffraction imaging, reveals the existence of multiple unique microscopically resolvable satellite populations that display distinct protein interaction profiles. We further show that loss of satellites in PCM1-depleted cells results in a dramatic change in the satellite interaction landscape. Finally, we demonstrate that satellite composition is largely unaffected by centriole depletion or disruption of microtubules, indicating that satellite assembly is centrosome-independent. Together, our work offers the first systematic spatial and proteomic profiling of human centriolar satellites and paves the way for future studies aimed at better understanding the biogenesis and function(s) of these enigmatic structures.
Collapse
Affiliation(s)
- Ladan Gheiratmand
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - Etienne Coyaud
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoONCanada
| | - Gagan D Gupta
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
- Present address:
Department of Chemistry and BiologyRyerson UniversityTorontoONCanada
| | | | - Monica Hasegan
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - Suzanna L Prosser
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - João Gonçalves
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - Brian Raught
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoONCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
| | - Laurence Pelletier
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| |
Collapse
|
22
|
Busselez J, Chichón FJ, Rodríguez MJ, Alpízar A, Gharbi SI, Franch M, Melero R, Paradela A, Carrascosa JL, Carazo JM. Cryo-Electron Tomography and Proteomics studies of centrosomes from differentiated quiescent thymocytes. Sci Rep 2019; 9:7187. [PMID: 31076588 PMCID: PMC6510768 DOI: 10.1038/s41598-019-43338-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/08/2019] [Indexed: 02/02/2023] Open
Abstract
We have used cryo Electron Tomography, proteomics and immunolabeling to study centrosomes isolated from the young lamb thymus, an efficient source of quiescent differentiated cells. We compared the proteome of thymocyte centrosomes to data published for KE37 cells, focusing on proteins associated with centriole disengagement and centrosome separation. The data obtained enhances our understanding of the protein system joining the centrioles, a system comprised of a branched network of fibers linked to an apparently amorphous density that was partially characterized here. A number of proteins were localized to the amorphous density by immunolabeling (C-NAP1, cohesin SMC1, condensin SMC4 and NCAPD2), yet not DNA. In conjuction, these data not only extend our understanding of centrosomes but they will help refine the model that focus on the protein system associated with the centriolar junction.
Collapse
Affiliation(s)
- Johan Busselez
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain. .,Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 Rue Laurent Fries, 67400, Illkirch-Graffenstaden, France.
| | - Francisco Javier Chichón
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - Maria Josefa Rodríguez
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - Adan Alpízar
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - Séverine Isabelle Gharbi
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - Mònica Franch
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - Roberto Melero
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - Alberto Paradela
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - José L Carrascosa
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain
| | - José-Maria Carazo
- Centro Nacional de Biotecnologia (CNB-CSIC), Darwin 3, Campus de Cantoblanco 28049, Madrid, Spain.
| |
Collapse
|
23
|
Li QH, Liu Y, Chen S, Zong ZH, Du YP, Sheng XJ, Zhao Y. circ-CSPP1 promotes proliferation, invasion and migration of ovarian cancer cells by acting as a miR-1236-3p sponge. Biomed Pharmacother 2019; 114:108832. [PMID: 30965236 DOI: 10.1016/j.biopha.2019.108832] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 01/09/2023] Open
Abstract
Circular RNAs are known to participate in tumorigenesis through a variety of pathways, and as such, have potential to serve as molecular markers in tumor diagnosis and treatment. Here, using quantitative reverse transcription (qRT)-PCR, we showed that circ-CSPP1 is highly expressed in ovarian cancer (OC) tissues. Particularly, we detected circ-CSPP1 expression in three OC cell lines; of which, OVCAR3 and A2780 demonstrated higher levels of circ-CSPP1 expression, and CAOV3 showed lower circ-CSPP1 expression level. Subsequent silencing of circ-CSPP1 in OVCAR3 and A2780 cell lines revealed decreased cell growth, migration and invasion, while overexpression of circ-CSPP1 caused opposite results We also found that miR-1236-3p is a target of circ-CSPP1. Circ-CSPP1 silencing increased the expression of miR-1236-3p, and circ-CSPP1 overexpression decreased miR-1236-3p expression. MiR-1236-3p reportedly plays a tumor-suppressor role in OC by targeting zinc finger E-box binding homeobox 1 (ZEB1). In agreement with this, we showed that silencing circ-CSPP1 significantly decreased ZEB1 expression at both RNA and protein levels, and epithelial-mesenchymal transition (EMT) related markers (E-cadherin and N-cadherin) varied with ZEB1 expression. Circ-CSPP1 silencing also caused decreased expression of matrix metalloproteinase-2 (MMP-2) and vascular endothelial growth factor A (VEGFA), both of which are related to tumorigenesis. Overexpression of circ-CSPP1 had opposite effects. In addition, we indicated that the tumor-promoting effect was inhibited after we transfected miR-1236-3p into circ-CSPP1 overexpressing OC cells. Altogether, our findings suggest that by acting as a miR-1236-3p sponge, circ-CSPP1 impairs the inhibitory effect of miR-1236-3p on ZEB1, which subsequently promotes EMT and OC development.
Collapse
Affiliation(s)
- Qian-Hui Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institute in Guangdong Province, Guangzhou, 510150, China
| | - Yao Liu
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Shuo Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institute in Guangdong Province, Guangzhou, 510150, China
| | - Zhi-Hong Zong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, China Medical University, Shenyang, 110001, China
| | - Yu-Ping Du
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiu-Jie Sheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institute in Guangdong Province, Guangzhou, 510150, China.
| | - Yang Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institute in Guangdong Province, Guangzhou, 510150, China.
| |
Collapse
|
24
|
Hua K, Ferland RJ. Primary Cilia Reconsidered in the Context of Ciliopathies: Extraciliary and Ciliary Functions of Cilia Proteins Converge on a Polarity theme? Bioessays 2018; 40:e1700132. [PMID: 29882973 PMCID: PMC6239423 DOI: 10.1002/bies.201700132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Once dismissed as vestigial organelles, primary cilia have garnered the interest of scientists, given their importance in development/signaling, and for their implication in a new disease category known as ciliopathies. However, many, if not all, "cilia" proteins also have locations/functions outside of the primary cilium. These extraciliary functions can complicate the interpretation of a particular ciliopathy phenotype: it may be a result of defects at the cilium and/or at extraciliary locations, and it could be broadly related to a unifying cellular process for these proteins, such as polarity. Assembly of a cilium has many similarities to the development of other polarized structures. This evolutionarily preserved process for the assembly of polarized cell structures offers a perspective on how the cilium may have evolved. We hypothesize that cilia proteins are critical for cell polarity, and that core polarity proteins may have been specialized to form various cellular protrusions, including primary cilia.
Collapse
Affiliation(s)
- Kiet Hua
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
- Department of Neurology, Albany Medical College, Albany, New York, USA, 12208
| |
Collapse
|
25
|
Shearer RF, Frikstad KAM, McKenna J, McCloy RA, Deng N, Burgess A, Stokke T, Patzke S, Saunders DN. The E3 ubiquitin ligase UBR5 regulates centriolar satellite stability and primary cilia. Mol Biol Cell 2018; 29:1542-1554. [PMID: 29742019 PMCID: PMC6080653 DOI: 10.1091/mbc.e17-04-0248] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Primary cilia are crucial for signal transduction in a variety of pathways, including hedgehog and Wnt. Disruption of primary cilia formation (ciliogenesis) is linked to numerous developmental disorders (known as ciliopathies) and diseases, including cancer. The ubiquitin-proteasome system (UPS) component UBR5 was previously identified as a putative positive regulator of ciliogenesis in a functional genomics screen. UBR5 is an E3 ubiquitin ligase that is frequently deregulated in tumors, but its biological role in cancer is largely uncharacterized, partly due to a lack of understanding of interacting proteins and pathways. We validated the effect of UBR5 depletion on primary cilia formation using a robust model of ciliogenesis, and identified CSPP1, a centrosomal and ciliary protein required for cilia formation, as a UBR5-interacting protein. We show that UBR5 ubiquitylates CSPP1, and that UBR5 is required for cytoplasmic organization of CSPP1-comprising centriolar satellites in centrosomal periphery, suggesting that UBR5-mediated ubiquitylation of CSPP1 or associated centriolar satellite constituents is one underlying requirement for cilia expression. Hence, we have established a key role for UBR5 in ciliogenesis that may have important implications in understanding cancer pathophysiology.
Collapse
Affiliation(s)
- Robert F Shearer
- Garvan Institute of Medical Research, Kinghorn Cancer Centre, Darlinghurst 2010, Australia.,Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales, Sydney 2052, Australia
| | - Kari-Anne Myrum Frikstad
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0310 Oslo, Norway
| | - Jessie McKenna
- Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | - Rachael A McCloy
- Garvan Institute of Medical Research, Kinghorn Cancer Centre, Darlinghurst 2010, Australia
| | - Niantao Deng
- Garvan Institute of Medical Research, Kinghorn Cancer Centre, Darlinghurst 2010, Australia
| | - Andrew Burgess
- Garvan Institute of Medical Research, Kinghorn Cancer Centre, Darlinghurst 2010, Australia.,Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales, Sydney 2052, Australia
| | - Trond Stokke
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0310 Oslo, Norway
| | - Sebastian Patzke
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0310 Oslo, Norway
| | - Darren N Saunders
- Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
26
|
Hua K, Ferland RJ. Primary cilia proteins: ciliary and extraciliary sites and functions. Cell Mol Life Sci 2018; 75:1521-1540. [PMID: 29305615 PMCID: PMC5899021 DOI: 10.1007/s00018-017-2740-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023]
Abstract
Primary cilia are immotile organelles known for their roles in development and cell signaling. Defects in primary cilia result in a range of disorders named ciliopathies. Because this organelle can be found singularly on almost all cell types, its importance extends to most organ systems. As such, elucidating the importance of the primary cilium has attracted researchers from all biological disciplines. As the primary cilia field expands, caution is warranted in attributing biological defects solely to the function of this organelle, since many of these "ciliary" proteins are found at other sites in cells and likely have non-ciliary functions. Indeed, many, if not all, cilia proteins have locations and functions outside the primary cilium. Extraciliary functions are known to include cell cycle regulation, cytoskeletal regulation, and trafficking. Cilia proteins have been observed in the nucleus, at the Golgi apparatus, and even in immune synapses of T cells (interestingly, a non-ciliated cell). Given the abundance of extraciliary sites and functions, it can be difficult to definitively attribute an observed phenotype solely to defective cilia rather than to some defective extraciliary function or a combination of both. Thus, extraciliary sites and functions of cilia proteins need to be considered, as well as experimentally determined. Through such consideration, we will understand the true role of the primary cilium in disease as compared to other cellular processes' influences in mediating disease (or through a combination of both). Here, we review a compilation of known extraciliary sites and functions of "cilia" proteins as a means to demonstrate the potential non-ciliary roles for these proteins.
Collapse
Affiliation(s)
- Kiet Hua
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
- Department of Neurology, Albany Medical College, Albany, NY, 12208, USA.
| |
Collapse
|
27
|
Hua K, Ferland RJ. Fixation methods can differentially affect ciliary protein immunolabeling. Cilia 2017; 6:5. [PMID: 28352462 PMCID: PMC5366141 DOI: 10.1186/s13630-017-0045-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 01/28/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Primary cilia are immotile, microtubule-based organelles present on most cells. Defects in primary cilia presence/function result in a category of developmental diseases referred to as ciliopathies. As the cilia field progresses, there is a need to consider both the ciliary and extraciliary roles of cilia proteins. However, traditional fixation methods are not always suitable for examining the full range of localizations of cilia proteins. Here, we tested a variety of fixation methods with commonly used cilia markers to determine the most appropriate fixation method for different cilia proteins. METHODS Mouse inner medullary collecting duct and human retinal pigmented epithelial cells were grown to confluence, serum starved, and fixed with one of the following fixation agents: paraformaldehyde-sucrose, paraformaldehyde-PBS, methanol, cytoskeletal buffer followed by methanol, or three variations of cytoskeletal buffer-paraformaldehyde fixation. Each cell type and fixation method combination was probed with the following ciliary markers: acetylated α-tubulin, detyrosinated tubulin, polyglutamylated tubulin, β-tubulin, adenylyl cyclase 3 (AC3), ADP-ribosylation factor-like protein 13b (Arl13b), centrosome and spindle pole associated protein 1 (CSPP1), or intraflagellar transport protein 20 (IFT20). Intraflagellar transport protein 88 (IFT88) and GM130 (Golgi marker) were also used. We assessed actin (via phalloidin) and microtubule integrity, centrioles, cilia, and two extraciliary sites (mitotic figures and Golgi). RESULTS For the cilia markers examined, paraformaldehyde fixation preserved cilia immunolabeling of cilia-membrane proteins (AC3 and Arl13b), but failed to reveal cilia immunostaining of axonemal proteins (CSPP1 and IFT20). Methanol revealed cilia labeling for some axonemal proteins, but not others, and this depended on cell type. Generally, any method that first included a wash in cytoskeletal buffer, before fixing, revealed more distinct cilia immunolabeling for axonemal proteins (CSPP1, IFT20, and IFT88), but resulted in the loss of cilia labeling for cilia-membrane proteins (AC3 and Arl13b). All three different post-translational modifications of tubulin antibodies positively immunolabeled cilia in all fixation methods tested. Ultimately, we found that fixing cells in a solution of paraformaldehyde prepared in cytoskeletal buffer allowed for the preservation of cilia immunolabeling for most cilia proteins tested and allowed visualization of two extraciliary sites (mitotic figures and Golgi). CONCLUSION Some general patterns were observed to guide in the choice of a fixation agent. Cilia-membrane proteins generally benefit from quick fixation with no prior permeabilization, whereas axonemal proteins tend to benefit from permeabilization and use of cytoskeletal buffer.
Collapse
Affiliation(s)
- Kiet Hua
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208 USA
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208 USA.,Department of Neurology, Albany Medical College, Albany, NY 12208 USA
| |
Collapse
|
28
|
Zhu L, Wang Z, Wang W, Wang C, Hua S, Su Z, Brako L, Garcia-Barrio M, Ye M, Wei X, Zou H, Ding X, Liu L, Liu X, Yao X. Mitotic Protein CSPP1 Interacts with CENP-H Protein to Coordinate Accurate Chromosome Oscillation in Mitosis. J Biol Chem 2015; 290:27053-27066. [PMID: 26378239 DOI: 10.1074/jbc.m115.658534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Indexed: 12/23/2022] Open
Abstract
Mitotic chromosome segregation is orchestrated by the dynamic interaction of spindle microtubules with the kinetochores. During chromosome alignment, kinetochore-bound microtubules undergo dynamic cycles between growth and shrinkage, leading to an oscillatory movement of chromosomes along the spindle axis. Although kinetochore protein CENP-H serves as a molecular control of kinetochore-microtubule dynamics, the mechanistic link between CENP-H and kinetochore microtubules (kMT) has remained less characterized. Here, we show that CSPP1 is a kinetochore protein essential for accurate chromosome movements in mitosis. CSPP1 binds to CENP-H in vitro and in vivo. Suppression of CSPP1 perturbs proper mitotic progression and compromises the satisfaction of spindle assembly checkpoint. In addition, chromosome oscillation is greatly attenuated in CSPP1-depleted cells, similar to what was observed in the CENP-H-depleted cells. Importantly, CSPP1 depletion enhances velocity of kinetochore movement, and overexpression of CSPP1 decreases the speed, suggesting that CSPP1 promotes kMT stability during cell division. Specific perturbation of CENP-H/CSPP1 interaction using a membrane-permeable competing peptide resulted in a transient mitotic arrest and chromosome segregation defect. Based on these findings, we propose that CSPP1 cooperates with CENP-H on kinetochores to serve as a novel regulator of kMT dynamics for accurate chromosome segregation.
Collapse
Affiliation(s)
- Lijuan Zhu
- Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China
| | - Zhikai Wang
- Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China; the Morehouse School of Medicine and Atlanta Cardiovascular Research Institute, Atlanta, Georgia 30310
| | - Wenwen Wang
- Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China; the Morehouse School of Medicine and Atlanta Cardiovascular Research Institute, Atlanta, Georgia 30310,; the Airforce General Hospital, Beijing 100036, China
| | - Chunli Wang
- the National Chromatographic Research and Analysis Center, Chinese Academy of Sciences, Dalian 116023, China
| | - Shasha Hua
- Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China; the Airforce General Hospital, Beijing 100036, China
| | - Zeqi Su
- the Beijing University of Chinese Medicine, Beijing 100029, China
| | - Larry Brako
- the Morehouse School of Medicine and Atlanta Cardiovascular Research Institute, Atlanta, Georgia 30310
| | - Minerva Garcia-Barrio
- the Morehouse School of Medicine and Atlanta Cardiovascular Research Institute, Atlanta, Georgia 30310
| | - Mingliang Ye
- the National Chromatographic Research and Analysis Center, Chinese Academy of Sciences, Dalian 116023, China
| | - Xuan Wei
- the Airforce General Hospital, Beijing 100036, China
| | - Hanfa Zou
- the National Chromatographic Research and Analysis Center, Chinese Academy of Sciences, Dalian 116023, China
| | - Xia Ding
- the Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lifang Liu
- the Airforce General Hospital, Beijing 100036, China.
| | - Xing Liu
- Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China; the Morehouse School of Medicine and Atlanta Cardiovascular Research Institute, Atlanta, Georgia 30310,.
| | - Xuebiao Yao
- Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
29
|
Sternemalm J, Geimer S, Frikstad KAM, Schink KO, Stokke T, Patzke S. CSPP-L Associates with the Desmosome of Polarized Epithelial Cells and Is Required for Normal Spheroid Formation. PLoS One 2015; 10:e0134789. [PMID: 26241740 PMCID: PMC4524657 DOI: 10.1371/journal.pone.0134789] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/14/2015] [Indexed: 11/20/2022] Open
Abstract
Deleterious mutations of the Centrosome/Spindle Pole associated Protein 1 gene, CSPP1, are causative for Joubert-syndrome and Joubert-related developmental disorders. These disorders are defined by a characteristic mal-development of the brain, but frequently involve renal and hepatic cyst formation. CSPP-L, the large protein isoform of CSPP1 localizes to microtubule ends of the mitotic mid-spindle and the ciliary axoneme, and is required for ciliogenesis. We here report the microtubule independent but Desmoplakin dependent localization of CSPP-L to Desmosomes in apical-basal polarized epithelial cells. Importantly, siRNA conferred depletion of CSPP-L or Desmoplakin promoted multi-lumen spheroid formation in 3D-cultures of non-ciliated human colon carcinoma Caco-2 cells. Multi-lumen spheroids of CSPP1 siRNA transfectants showed disrupted apical cell junction localization of the cytoskeleton organizing RhoGEF ECT2. Our results hence identify a novel, non-ciliary role for CSPP-L in epithelial morphogenesis.
Collapse
Affiliation(s)
- Johan Sternemalm
- Department of Radiation Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals-Norwegian Radium Hospital, Oslo, Norway
| | - Stefan Geimer
- Cell Biology/Electron Microscopy, University of Bayreuth, Bayreuth, Germany
| | - Kari-Anne M Frikstad
- Department of Radiation Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals-Norwegian Radium Hospital, Oslo, Norway
| | - Kay O Schink
- Department of Molecular Cell Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals-Norwegian Radium Hospital, Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trond Stokke
- Department of Radiation Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals-Norwegian Radium Hospital, Oslo, Norway
| | - Sebastian Patzke
- Department of Radiation Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals-Norwegian Radium Hospital, Oslo, Norway
| |
Collapse
|
30
|
Sternemalm J, Russnes HG, Zhao X, Risberg B, Nord S, Caldas C, Børresen-Dale AL, Stokke T, Patzke S. Nuclear CSPP1 expression defined subtypes of basal-like breast cancer. Br J Cancer 2014; 111:326-38. [PMID: 24901235 PMCID: PMC4102947 DOI: 10.1038/bjc.2014.297] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/24/2014] [Accepted: 05/09/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The multi-exon CSPP1 gene, encoding for centrosome and microtubule-associated proteins involved in ciliogenesis and cell division, is a candidate oncogene in luminal breast cancer but expression of CSPP1 proteins remained unexplored. METHODS CSPP1 gene and protein expression was examined in normal mammary tissue, human breast cancer cell lines, and primary breast cancer biopsies from two patient cohorts. Cell type and epitope-dependent subcellular-specific CSPP1 staining pattern in normal mammary gland epithelium and cancer biopsies were correlated to molecular and clinical parameters. RESULTS A novel, nuclear localised CSPP1 isoform was exclusively detected in luminal epithelial cells, whereas cytoplasmic CSPP-L was generally expressed in normal mammary epithelium. Luminal cell-related nuclear CSPP1 expression was preserved in type-matched cell lines and carcinomas, and correlated to gene copy number and mRNA expression. In contrast, basal-like carcinomas displayed generally lower CSPP1 mRNA expression. Yet, a subgroup of basal-like breast carcinomas depicted nuclear CSPP1 expression, displayed luminal traits, and differed from nuclear CSPP1 devoid counterparts in expression of eight genes. Eight-gene signature defined groups of basal-like tumours from an independent cohort showed significant differences in survival. CONCLUSIONS Differential expression of a nuclear CSPP1 isoform identified biologically and clinically distinct subgroups of basal-like breast carcinoma.
Collapse
Affiliation(s)
- J Sternemalm
- Department of Radiation Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway
| | - H G Russnes
- 1] Departments of Genetics, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway [2] Department of Pathology, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway [3] K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, N-0310 Oslo, Norway
| | - X Zhao
- Center for Cancer Systems Biology, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - B Risberg
- 1] Department of Pathology, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway [2] Institute for Medical Informatics, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway
| | - S Nord
- 1] Departments of Genetics, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway [2] K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, N-0310 Oslo, Norway
| | - C Caldas
- 1] Breast Cancer Functional Genomics, Cancer Research UK Cambridge Research Institute, Cambridge CB2 0RE, UK [2] Department of Oncology, University of Cambridge, Li Ka-Shing Centre, Robinson Way, Cambridge CB2 0RE, UK [3] Cambridge Breast Unit, Addenbrooke's Hospital and Cambridge National Institute for Health Research Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - A L Børresen-Dale
- 1] Departments of Genetics, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway [2] K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, N-0310 Oslo, Norway
| | - T Stokke
- Department of Radiation Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway
| | - S Patzke
- Department of Radiation Biology, Division of Cancer Medicine, Surgery and Transplantation, Institute for Cancer Research, Oslo University Hospitals - Norwegian Radium Hospital, N-0310 Oslo, Norway
| |
Collapse
|
31
|
Shaheen R, Shamseldin HE, Loucks CM, Seidahmed MZ, Ansari S, Ibrahim Khalil M, Al-Yacoub N, Davis EE, Mola NA, Szymanska K, Herridge W, Chudley AE, Chodirker BN, Schwartzentruber J, Majewski J, Katsanis N, Poizat C, Johnson CA, Parboosingh J, Boycott KM, Innes AM, Alkuraya FS. Mutations in CSPP1, encoding a core centrosomal protein, cause a range of ciliopathy phenotypes in humans. Am J Hum Genet 2014; 94:73-9. [PMID: 24360803 PMCID: PMC3882732 DOI: 10.1016/j.ajhg.2013.11.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/13/2013] [Indexed: 11/22/2022] Open
Abstract
Ciliopathies are characterized by a pattern of multisystem involvement that is consistent with the developmental role of the primary cilium. Within this biological module, mutations in genes that encode components of the cilium and its anchoring structure, the basal body, are the major contributors to both disease causality and modification. However, despite rapid advances in this field, the majority of the genes that drive ciliopathies and the mechanisms that govern the pronounced phenotypic variability of this group of disorders remain poorly understood. Here, we show that mutations in CSPP1, which encodes a core centrosomal protein, are disease causing on the basis of the independent identification of two homozygous truncating mutations in three consanguineous families (one Arab and two Hutterite) affected by variable ciliopathy phenotypes ranging from Joubert syndrome to the more severe Meckel-Gruber syndrome with perinatal lethality and occipital encephalocele. Consistent with the recently described role of CSPP1 in ciliogenesis, we show that mutant fibroblasts from one affected individual have severely impaired ciliogenesis with concomitant defects in sonic hedgehog (SHH) signaling. Our results expand the list of centrosomal proteins implicated in human ciliopathies.
Collapse
Affiliation(s)
- Ranad Shaheen
- Department of Genetics, King Faisal Specialist Hospital, Riyadh 11211, Saudi Arabia
| | - Hanan E Shamseldin
- Department of Genetics, King Faisal Specialist Hospital, Riyadh 11211, Saudi Arabia
| | - Catrina M Loucks
- Department of Medical Genetics, University of Calgary, Alberta Children's Hospital, Calgary, AB T3B 6A8, Canada
| | | | - Shinu Ansari
- Department of Genetics, King Faisal Specialist Hospital, Riyadh 11211, Saudi Arabia
| | - Mohamed Ibrahim Khalil
- Department of Obstetrics and Gynecology, Security Forces Hospital, Riyadh 11481, Saudi Arabia
| | - Nadya Al-Yacoub
- Cardiovascular Research Program, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Erica E Davis
- Center for Human Disease Modeling, Duke University, Durham, NC 22710, USA
| | - Natalie A Mola
- Center for Human Disease Modeling, Duke University, Durham, NC 22710, USA
| | - Katarzyna Szymanska
- Leeds Institute of Molecular Medicine, St. James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Warren Herridge
- Leeds Institute of Molecular Medicine, St. James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Albert E Chudley
- Department of Paediatrics and Child Health and Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3A 1R9, Canada
| | - Bernard N Chodirker
- Department of Paediatrics and Child Health and Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3A 1R9, Canada
| | | | - Jacek Majewski
- McGill University and Genome Quebec Innovation Center, Montreal, QC H3A 0G4, Canada
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University, Durham, NC 22710, USA
| | - Coralie Poizat
- Cardiovascular Research Program, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Colin A Johnson
- Leeds Institute of Molecular Medicine, St. James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Jillian Parboosingh
- Department of Medical Genetics, University of Calgary, Alberta Children's Hospital, Calgary, AB T3B 6A8, Canada; Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - A Micheil Innes
- Department of Medical Genetics, University of Calgary, Alberta Children's Hospital, Calgary, AB T3B 6A8, Canada; Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary, Calgary, AB T3B 6A8, Canada.
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital, Riyadh 11211, Saudi Arabia; Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.
| |
Collapse
|
32
|
Akizu N, Silhavy JL, Rosti RO, Scott E, Fenstermaker AG, Schroth J, Zaki MS, Sanchez H, Gupta N, Kabra M, Kara M, Ben-Omran T, Rosti B, Guemez-Gamboa A, Spencer E, Pan R, Cai N, Abdellateef M, Gabriel S, Halbritter J, Hildebrandt F, van Bokhoven H, Gunel M, Gleeson JG. Mutations in CSPP1 lead to classical Joubert syndrome. Am J Hum Genet 2014; 94:80-6. [PMID: 24360807 DOI: 10.1016/j.ajhg.2013.11.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 11/19/2013] [Indexed: 12/24/2022] Open
Abstract
Joubert syndrome and related disorders (JSRDs) are genetically heterogeneous and characterized by a distinctive mid-hindbrain malformation. Causative mutations lead to primary cilia dysfunction, which often results in variable involvement of other organs such as the liver, retina, and kidney. We identified predicted null mutations in CSPP1 in six individuals affected by classical JSRDs. CSPP1 encodes a protein localized to centrosomes and spindle poles, as well as to the primary cilium. Despite the known interaction between CSPP1 and nephronophthisis-associated proteins, none of the affected individuals in our cohort presented with kidney disease, and further, screening of a large cohort of individuals with nephronophthisis demonstrated no mutations. CSPP1 is broadly expressed in neural tissue, and its encoded protein localizes to the primary cilium in an in vitro model of human neurogenesis. Here, we show abrogated protein levels and ciliogenesis in affected fibroblasts. Our data thus suggest that CSPP1 is involved in neural-specific functions of primary cilia.
Collapse
Affiliation(s)
- Naiara Akizu
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Jennifer L Silhavy
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Rasim Ozgur Rosti
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Eric Scott
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Ali G Fenstermaker
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Jana Schroth
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo 12311, Egypt
| | - Henry Sanchez
- Pediatrics Department, Palo Alto Medical Foundation, Fremont, CA 94538, USA
| | - Neerja Gupta
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Madhulika Kabra
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Majdi Kara
- Department of Pediatrics, Tripoli Children's Hospital, PO Box 2214, Tripoli, Libya
| | - Tawfeg Ben-Omran
- Clinical and Metabolic Genetics Division, Department of Pediatrics, Hamad Medical Corporation, Doha 3050, Qatar
| | - Basak Rosti
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Alicia Guemez-Gamboa
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Emily Spencer
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Roger Pan
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Na Cai
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Mostafa Abdellateef
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute
| | - Stacey Gabriel
- The Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
| | - Jan Halbritter
- Howard Hughes Medical Institute; Division of Nephrology, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Friedhelm Hildebrandt
- Howard Hughes Medical Institute; Division of Nephrology, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hans van Bokhoven
- Department of Human Genetics, Radboud University Medical Centre, 6500 HB Nijmegen, the Netherlands
| | - Murat Gunel
- Yale Program on Neurogenetics, Departments of Neurosurgery, Neurobiology, and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joseph G Gleeson
- Neurogenetics Laboratory, Institute for Genomic Medicine and Departments of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Howard Hughes Medical Institute.
| |
Collapse
|
33
|
Li F, Jang H, Puttabyatappa M, Jo M, Curry TE. Ovarian FAM110C (family with sequence similarity 110C): induction during the periovulatory period and regulation of granulosa cell cycle kinetics in rats. Biol Reprod 2012; 86:185. [PMID: 22460667 DOI: 10.1095/biolreprod.112.099259] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
FAM110C belongs to a family of proteins that regulates cell proliferation. In the present study, the spatiotemporal expression pattern of FAM110C and its potential role were examined during the periovulatory period. Immature female rats were injected with equine chorionic gonadotropin (eCG) followed by human chorionic gonadotropin (hCG) and ovaries or granulosa cells were collected at various times after hCG administration (n = 3/time point). Expression levels of Fam110c mRNA and protein were highly induced both in intact ovaries and granulosa cells at 8 to 12 h after hCG treatment. In situ hybridization analysis demonstrated Fam110c mRNA expression was induced in theca and granulosa cells at 4 h after hCG, primarily localized to granulosa cells at 8 h and 12 h, and decreased at 24 h after hCG. There was negligible Fam110c mRNA detected in newly forming corpora lutea. In rat granulosa cell cultures, hCG induced expression of Fam110c mRNA was inhibited by RU486, whereas NS398 and AG1478 had no effect, suggesting that Fam110c expression is regulated in part by the progesterone receptor pathway. Promoter activity analysis revealed that an Sp1 site was important for the induction of Fam110c expression by hCG. Overexpression of FAM110C promoted granulosa cells to arrest at the G(1) phase of the cell cycle but did not change progesterone levels. In summary, hCG induces Fam110c mRNA expression in granulosa cells by activation of an Sp1-binding site and the actions of progesterone. Our findings suggest that FAM110C may control granulosa cell differentiation into luteal cells by arresting cell cycle progression.
Collapse
Affiliation(s)
- Feixue Li
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, People's Republic of China
| | | | | | | | | |
Collapse
|
34
|
Zeng F, Tian Y, Shi S, Wu Q, Liu S, Zheng H, Yue L, Li Y. Identification of mouse MARVELD1 as a microtubule associated protein that inhibits cell cycle progression and migration. Mol Cells 2011; 31:267-74. [PMID: 21347699 PMCID: PMC3932696 DOI: 10.1007/s10059-011-0037-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 12/15/2010] [Accepted: 12/24/2010] [Indexed: 12/20/2022] Open
Abstract
MARVEL domain-containing 1 (MARVELD1) is a newly identified nuclear protein; however its function has not been clear until now. Here, we report that mouse MARVELD1 (mMARVELD1), which is highly conserved between mice and humans, exhibits cell cycle-dependent cellular localization. In NIH3T3 cells, MARVELD1 was observed in the nucleus and at the perinuclear region during interphase, but was localized at the mitotic spindle and midbody at metaphase, and a significant fraction of mMARVELD1 translocated to the plasma membrane during anaphase. In addition, treatment of cells with colchicine, a microtubule-depolymerizing agent, resulted in translocation of mMARVELD1 to the plasma membrane, and association of mMARVELD1 and α-tubulin was confirmed by co-immunoprecipitation. Finally, overexpression of mMARVELD1 resulted in a remarkable inhibition of cell proliferation, G1-phase arrest, and reduced cell migration. These findings indicate that mMARVELD1 is a microtubule-associated protein that plays an important role in cell cycle progression and migration.
Collapse
Affiliation(s)
- Fanli Zeng
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Yanyan Tian
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Shuliang Shi
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Qiong Wu
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Shanshan Liu
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Hongxia Zheng
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Lei Yue
- The Academy of Fundamental and Interdisciplinary Science, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Yu Li
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
- The Academy of Fundamental and Interdisciplinary Science, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| |
Collapse
|
35
|
Patzke S, Redick S, Warsame A, Murga-Zamalloa CA, Khanna H, Doxsey S, Stokke T. CSPP is a ciliary protein interacting with Nephrocystin 8 and required for cilia formation. Mol Biol Cell 2010; 21:2555-67. [PMID: 20519441 PMCID: PMC2912343 DOI: 10.1091/mbc.e09-06-0503] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
We described previously the cell cycle- and microtubule-related functions of two splice isoforms of the centrosome spindle pole-associated protein (CSPP and CSPP-L). Here, we show that endogenous CSPP isoforms not only localize to centrosomes and the midbody in cycling cells but also extend to the cilia axoneme in postmitotic resting cells. They are required for ciliogenesis in hTERT-RPE1 cells in vitro and are expressed in ciliated renal, retinal, and respiratory cells in vivo. We report that CSPP isoforms require their common C-terminal domain to interact with Nephrocystin 8 (NPHP8/RPGRIP1L) and to form a ternary complex with NPHP8 and NPHP4. We find CSPP-L to be required for the efficient localization of NPHP8 but not NPHP4 to the basal body. The ciliogenesis defect in hTERT-RPE1 cells is, however, not mediated through loss of NPHP8. Similar to the effects of ectopical expression of CSPP-L, cilia length increased in NPHP8-depleted cells. Our results thus suggest that CSPP proteins may be involved in further cytoskeletal organization of the basal body and its primary cilium. To conclude, we have identified a novel, nonmitotic function of CSPP proteins placing them into a ciliary protein network crucial for normal renal and retinal tissue architecture and physiology.
Collapse
Affiliation(s)
- Sebastian Patzke
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, N-0310 Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
36
|
Difilippantonio MJ, Ghadimi BM, Howard T, Camps J, Nguyen QT, Ferris DK, Sackett DL, Ried T. Nucleation capacity and presence of centrioles define a distinct category of centrosome abnormalities that induces multipolar mitoses in cancer cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2009; 50:672-696. [PMID: 19768832 PMCID: PMC4322947 DOI: 10.1002/em.20532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Analysis of centrosome number and structure has become one means of assessing the potential for aberrant chromosome segregation and aneuploidy in tumor cells. Centrosome amplification directly causes multipolar catastrophic mitoses in mouse embryonic fibroblasts (MEFs) deficient for the tumor suppressor genes Brca1 or Trp53. We observed supernumerary centrosomes in cell lines established from aneuploid, but not from diploid, colorectal carcinomas; however, multipolar mitoses were never observed. This discrepancy prompted us to thoroughly characterize the centrosome abnormalities in these and other cancer cell lines with respect to both structure and function. The most striking result was that supernumerary centrosomes in aneuploid colorectal cancer cell lines were unable to nucleate microtubules, despite the presence of gamma-tubulin, pericentrin, PLK1, and AURKA. Analysis by scanning electron microscopy revealed that these supernumerary structures are devoid of centrioles, a result significantly different from observations in aneuploid pancreatic cancer cell lines and in Trp53 or Brca1 deficient MEFs. Thus, multipolar mitoses are dependent upon the ability of extra gamma-tubulin containing structures to nucleate microtubules, and this correlated with the presence of centrioles. The assessment of centrosome function with respect to chromosome segregation must therefore take into consideration the presence of centrioles and the capacity to nucleate microtubules. The patterns and mechanisms of chromosomal aberrations in hematologic malignancies and solid tumors are fundamentally different. The former is characterized by specific chromosome translocations, whose consequence is the activation of oncogenes. Most carcinomas, however, reveal variations in the nuclear DNA content. The observed genomic imbalances and gross variations in chromosome number can result from unequal chromosome segregation during mitotic cell division. It is therefore fundamental to elucidate mechanisms involved in distribution of the genome to daughter cells. Prior to cell division, the centrosome organizes microtubules and the mitotic spindle. Deciphering the consequences of alterations in centrosome number, structure, and function is an important step towards understanding how a diploid genome is maintained. Although extra centrosomes have now been observed in carcinomas and were correlated with aneuploidy, a careful functional investigation of these structures and their role in generating chromosome imbalances may lead to the identification of distinct mechanistic pathways of genomic instability. Understanding these pathways will also be important in determining whether they are potential molecular targets of therapeutic intervention.
Collapse
Affiliation(s)
- Michael J Difilippantonio
- Genetics Branch, Center for Cancer Research, National Cancer Institute/NIH, 50 South Drive, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Asiedu M, Wu D, Matsumura F, Wei Q. Centrosome/spindle pole-associated protein regulates cytokinesis via promoting the recruitment of MyoGEF to the central spindle. Mol Biol Cell 2009; 20:1428-40. [PMID: 19129481 DOI: 10.1091/mbc.e08-01-0001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cooperative communications between the central spindle and the contractile ring are critical for the spatial and temporal regulation of cytokinesis. Here we report that MyoGEF, a guanine nucleotide exchange factor that localizes to the central spindle and cleavage furrow, interacts with centrosome/spindle pole-associated protein (CSPP), which is concentrated at the spindle pole and central spindle during mitosis and cytokinesis. Both in vitro and in vivo pulldown assays show that MyoGEF interacts with CSPP. The C-terminus of MyoGEF and N-terminus of CSPP are required for their interaction. Immunofluorescence analysis indicates that MyoGEF and CSPP colocalize at the central spindle. Depletion of CSPP or MyoGEF by RNA-interference (RNAi) not only causes defects in mitosis and cytokinesis, such as metaphase arrest and furrow regression, but also mislocalization of nonmuscle myosin II with a phosphorylated myosin regulatory light chain (p-MRLC). Importantly, CSPP depletion by RNAi interferes with MyoGEF localization at the central spindle. Finally, MyoGEF interacts with ECT2, and RNAi-mediated depletion of MyoGEF leads to mislocalization of ECT2 and RhoA during cytokinesis. Therefore, we propose that CSPP interacts with and recruits MyoGEF to the central spindle, where MyoGEF contributes to the spatiotemporal regulation of cytokinesis.
Collapse
Affiliation(s)
- Michael Asiedu
- Department of Biochemistry, Kansas State University, Manhattan, KS 66506, USA
| | | | | | | |
Collapse
|
38
|
Hauge H, Patzke S, Aasheim HC. Characterization of the FAM110 gene family. Genomics 2007; 90:14-27. [PMID: 17499476 DOI: 10.1016/j.ygeno.2007.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 03/05/2007] [Accepted: 03/08/2007] [Indexed: 01/14/2023]
Abstract
We have previously characterized the centrosome/spindle pole-associated protein (CSPP) involved in cell cycle progression. The open reading frame C20orf55 was identified in a yeast two-hybrid screen in a search for CSPP-interacting proteins. A homology search revealed that C20orf55 belongs to a gene family consisting of three members that have not yet been described. The HUGO Nomenclature Committee has assigned these genes the names FAM110A-FAM110C. Studies of transfectants showed that the FAM110 proteins localized to centrosomes and accumulated at the microtubule organization center in interphase and at spindle poles in mitosis. In addition, overexpression of FAM110C induced microtubule aberrancies. Our data also indicated a cell cycle-regulated expression of FAM110A. Moreover, ectopic expression of FAM110B and FAM110C proteins impaired cell cycle progression in G1 phase. To summarize, we have characterized a novel family of genes encoding proteins with distinct conserved motifs, of which all members localize to centrosomes and spindle poles.
Collapse
Affiliation(s)
- Helena Hauge
- Faculty of Medicine, University of Oslo, and Department of Immunology, Institute for Cancer Research, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway
| | | | | |
Collapse
|
39
|
Abstract
RNA interference is an evolutionarily conserved gene silencing process triggered by double-stranded RNAs. Common to all cell types, is the production of 21-24 nucleotide small interfering RNA (siRNAs), which guide the RNA-induced silencing complex (RISC) to identify and cleave target mRNA sequences. Presently, this biological breakthrough method has revolutionised gene function studies and holds great promise as validating drug targets and treating human diseases. However, despite the success that has been achieved by this technology, studies carried in human blood cells have revealed that siRNAs could generate bystander effects, including the activation of innate immunity and inhibition of unintended target genes. Interestingly, 2' uridine-modified siRNAs did not trigger TLR signalling, but they totally suppressed immune activation by immunostimulatory siRNAs when both molecules where delivered to the same endosomes. This review describes the recent advances in understanding the innate immune response to both single and double-stranded siRNAs. Also, it highlights the spectrum of molecular strategies allowing the design of therapeutic siRNAs with minimal side effects.
Collapse
Affiliation(s)
- Mouldy Sioud
- Institute for Cancer Research, Department of Immunology, Molecular Medicine Group, Montebello, N-0310 Oslo, Norway.
| |
Collapse
|
40
|
Huynh T, Wälchli S, Sioud M. Transcriptional targeting of small interfering RNAs into cancer cells. Biochem Biophys Res Commun 2006; 350:854-9. [PMID: 17034763 DOI: 10.1016/j.bbrc.2006.09.127] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 09/20/2006] [Indexed: 12/27/2022]
Abstract
Small interfering RNAs (siRNAs) are widely used for analyzing gene function and have the potential to be developed into human therapeutics. However, persistent siRNA expression in normal cells may cause toxic side effects. Therefore, the therapeutic applications of RNAi in cancer require either the specific delivery of synthetic siRNAs into cancer cells or the control of siRNA expression. Accordingly, we have developed a cancer-specific vector that expresses siRNAs from the human survivin promoter. A plasmid vector expressing siRNAs under this promoter enabled efficient gene silencing of gene expression in different cancer cell lines. The levels of inhibition were comparable to that obtained with the constitutively active U6 promoter. By contrast to U6 promoter, no significant gene silencing was obtained with the Survivin promoter in normal mammary epithelial cells. Collectively, these data indicate that the survivin promoter is suitable for directing siRNA expression in cancer cells, but not normal cells.
Collapse
Affiliation(s)
- Trang Huynh
- Department of Immunology, Molecular Medicine Group, University Department, University of Oslo, The Norwegian Radium Hospital, Montebello, N-310 Oslo, Norway
| | | | | |
Collapse
|
41
|
Patzke S, Stokke T, Aasheim HC. CSPP and CSPP-L associate with centrosomes and microtubules and differently affect microtubule organization. J Cell Physiol 2006; 209:199-210. [PMID: 16826565 DOI: 10.1002/jcp.20725] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We recently described the identification of a centrosome/spindle pole associated protein, CSPP, involved in cell cycle progression. Here we report a CSPP isoform denoted CSPP-L, with a 294 amino acids longer N-terminus and a 51 amino acids insertion located in the coiled-coil mid-domain. Expression analysis indicates an inverse cell cycle dependent regulation. CSPP mRNA expression is highest in G1 whereas CSPP-L expression is highest in G2/M. Ectopic expression of CSPP-L impairs cell cycle progression weaker in G1 than CSPP. Furthermore, normal mitotic phenotypes were observed in CSPP-L but not in CSPP transfectants. CSPP-L relocates from spindle microtubules and poles in metaphase to the mid-spindle in anaphase and concentrates at the mid-body in telophase/cytokinesis. CSPP-L high-expressing mitotic cells were predominantly characterized by lagging chromosomes or monopolar spindles, in contrast to the predominant multipolar spindles observed with CSPP expression. The different effects of CSPP and CSPP-L on microtubule organization in mitosis depend on the coiled-coil mid-domain insertion. The common C-terminal domain is required to repress that activity until mitosis. Notably, this C-terminal domain alone can associate with centrosomes in a microtubule independent manner. Taken together, CSPP and CSPP-L interact with centrosomes and microtubules and can differently affect microtubule organization.
Collapse
Affiliation(s)
- Sebastian Patzke
- Department of Immunology, The Norwegian Radium Hospital, Montebello, Oslo, Norway
| | | | | |
Collapse
|
42
|
de Nijs L, Lakaye B, Coumans B, Léon C, Ikeda T, Delgado-Escueta AV, Grisar T, Chanas G. EFHC1, a protein mutated in juvenile myoclonic epilepsy, associates with the mitotic spindle through its N-terminus. Exp Cell Res 2006; 312:2872-9. [PMID: 16824517 DOI: 10.1016/j.yexcr.2006.05.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Revised: 04/25/2006] [Accepted: 05/22/2006] [Indexed: 01/23/2023]
Abstract
A novel gene, EFHC1, mutated in juvenile myoclonic epilepsy (JME) encodes a protein with three DM10 domains of unknown function and one putative EF-hand motif. To study the properties of EFHC1, we expressed EGFP-tagged protein in various cell lines. In interphase cells, the fusion protein was present in the cytoplasm and in the nucleus with specific accumulation at the centrosome. During mitosis EGFP-EFHC1 colocalized with the mitotic spindle, especially at spindle poles and with the midbody during cytokinesis. Using a specific antibody, we demonstrated the same distribution of the endogenous protein. Deletion analyses revealed that the N-terminal region of EFHC1 is crucial for the association with the mitotic spindle and the midbody. Our results suggest that EFHC1 could play an important role during cell division.
Collapse
Affiliation(s)
- Laurence de Nijs
- Center for Cellular and Molecular Neurobiology, University of Liège, Avenue de l'Hôpital 1, B-36, 4000 Liège, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Chen Y, Kong Q. Cell brain: insight into hepatocarcinogenesis. Med Hypotheses 2006; 67:44-52. [PMID: 16600524 DOI: 10.1016/j.mehy.2005.09.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Revised: 09/25/2005] [Accepted: 09/27/2005] [Indexed: 11/15/2022]
Abstract
Although great effort has been made, the understanding of the mechanisms of hepatocarcinogenesis is still limited. Among all the related hypotheses, the cell brain theory, which emphasized the integrate roles of the complex consisting of centrosome, the embedded centrioles and connecting microtubules (MTs) and interpreted cancer as a cell brain illness rather than a genetic disease, emerges to be more logic and recognizable. According to cell brain theory, all the cellular procedures are coordinated as a whole by the "brain" of a cell determining a cell's fate. Structural and functional abnormalities in the cell brain may result in unequal or multipolar segregation of the chromosomes, thereby causing cell cycle disorder, centrosome amplification, and genomic instability. Although there lacking of direct evidence associating cell brain defects and hepatocarcinogenesis, latest understanding of the roles of the cells brain in cell control does teach us that any defects in the cell brain may contribute to hepatocarcinogenesis. Briefly, more than 100 key proteins involved in DNA synthesis, DNA repair, cell cycle, and apoptosis have been localized to the cell brain. Specifically, more and more novel proteins associated with centrosome such as centrin, centriolin and cenexin are located in the centriole, a core component of cenrtrosome. Aberrant phosphorylation of these proteins and/or mutation of the coding genes may inevitably cause supernumerary centrioles and/or excess pericentriolar material. Modifications of any MT proteins such as tyrosinated tubulin (Tyr-tubulin), detyrosinated tubulin (Glu-tubulin) and Delta2-tubulin may change the structure and function of MTs, thereby interfering with G1 phase progression, altering the dynamics of some key proteins, and mis-regulating signal transduction and transcription. Although little work has been done, we intend to believe, based on the latest understanding of the novel roles of the cell brain in cell control, that defects in any part of the cell brain either in the structure or in the function may result in changes of the genes, eventually leading to the development of liver cancer, which is discussed in this paper and is expected to be helpful in shedding light on the often paradoxical observations seen in the development of cancer, including HCC. It also teaches us that when treating cancerous problems therapeutically or prophylactically, great attention should be given to the centrosome/cell brain, instead of gene alone. More specifically, the centrosome-centered cell brain may come to be novel targets in the treatment of cancer including HCC.
Collapse
Affiliation(s)
- Ying Chen
- Cell Brain Research Center, South Biology Building, Room 128, Shandong University School of Life Science, Jinan, Shandong Province 250100, PR China
| | | |
Collapse
|
44
|
Zhao Y, Zhang N, Kong Q. Does the cell-brain theory work in explaining carcinogenesis? Med Hypotheses 2006; 65:708-15. [PMID: 15975733 DOI: 10.1016/j.mehy.2005.04.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Revised: 03/25/2005] [Accepted: 04/12/2005] [Indexed: 10/25/2022]
Abstract
As a major microtubule-organizing center, the centrosome, together with the embedded centrioles and connecting filaments (or microtubules), has lately been proposed to be the "brain" of a cell. Although there are a lot of works to be done to test this hypothesis, emerging data have suggested that this centrosome-centered "cell brain" is playing increasingly important roles in cell control. Genes seem not to tell the whole story, despite the commonly held view that genetic alteration is the cause of most medical problems including cancer development. Although the mechanisms through which gene expression and protein synthesis are regulated remain to be studied, current advances in our understanding of the roles of the centrosome in the regulation of DNA synthesis, DNA repair, cell cycle, apoptosis and in the maintenance of genetic stability are challenging our tradition thoughts. Genetic alterations may be repaired by the centrosome-centered "cell brain"-mediated self-defense, but the cell brain defects intend to cause genetic alterations, which, in turn, may result in cancer development. Further understanding of the roles of the centrosome/cell brain in these and other new aspects are becoming very helpful in comprehending why and how medical problems including tumors develop. Meanwhile, it suggests that great attention should be given to the centrosome/cell brain, instead of gene alone when treating medical problems, which is discussed in this paper on the basis of cell brain theory and may prove helpful in shedding light on the often paradoxical observations seen in cell control, particularly in cancer development.
Collapse
Affiliation(s)
- Yunfeng Zhao
- Cell Brain Research Center, School of Life Science, Shandong University, Room 128, Biology Building, Jinan, Shandong Province 250100, China
| | | | | |
Collapse
|
45
|
Abstract
The discovery that nucleic acids mediated the inhibition of gene expression in a sequence-specific manner has provided the scientific community with a potentially important tool to analyse gene function and validate drug targets. Selective inhibition of gene expression by ribozymes and small interfering RNAs (siRNAs) is being explored for potential therapeutics against viral infections, inflammatory disorders, haematological diseases and cancer. In order to be used as pharmaceutical drugs, chemical modifications are necessary to increase their stability in vivo. However, such modifications should not affect either the ribozyme cleavage activity or the incorporation of the siRNAs into the RNA interference (RNAi) targeting complex and subsequent mRNA cleavage. To attain stability, ribozymes and siRNAs must also overcome several other problems, including accessibility to target messenger RNAs (mRNAs), efficient delivery to target cells and unwanted non-specific effects.
Collapse
Affiliation(s)
- M Sioud
- Institute for Cancer Research, Department of Immunology, Molecular Medicine Group, The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway.
| |
Collapse
|
46
|
Zhang Z, Yang Y, Gong A, Wang C, Liang Y, Chen Y. Localization of NGF and TrkA at mitotic apparatus in human glioma cell line U251. Biochem Biophys Res Commun 2005; 337:68-74. [PMID: 16181609 DOI: 10.1016/j.bbrc.2005.08.265] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Accepted: 08/30/2005] [Indexed: 10/25/2022]
Abstract
It has previously been implicated that nerve growth factor (NGF) with its high-affinity receptor tyrosine kinase A (TrkA) could play an important role in the growth modulation of human tumor cells, such as glioblastoma multiform cell lines and human breast cancer cell lines. However, the direct mitogenic effects of NGF and TrkA in these tumor cells still remain to be elucidated. Herein we show, by immunofluorescence staining, that NGF was colocalized with gamma-tubulin at the centrosomes or the spindle poles throughout the cell cycle and phosphorylated TrkA was colocalized with alpha-tubulin at mitotic spindle in the glioma cell line U251. The results suggest that NGF concentrated to centrosome can recruit its receptor TrkA there and cause phosphorylation of the latter. The phosphorylated TrkA with the tyrosine kinase activity may phosphorylate the tubulin and promote the mitotic spindle assembly. By these mechanisms, NGF can modulate the mitosis of human glioma cells.
Collapse
Affiliation(s)
- Zhijian Zhang
- School of Medicine, Jiangsu University, Jiangsu Zhenjiang 212001, China
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
The well recognized activities of the mammalian centrosome--microtubule nucleation, duplication, and organization of the primary cilium--are under the control of the cell cycle. However, the centrosome is more than just a follower of the cell cycle; it can also be essential for the cell to transit G1 and enter S phase. How the centrosome influences G1 progression is a mystery.
Collapse
Affiliation(s)
- Greenfield Sluder
- Department of Cell Biology, University of Massachusetts Medical School, Worcester 01605, USA.
| |
Collapse
|